51
|
Zhang H, Fan J, Qu J, Han Q, Zhou H, Song X. Predictive markers for anti-inflammatory treatment response in thyroid eye disease. Front Endocrinol (Lausanne) 2023; 14:1292519. [PMID: 38111706 PMCID: PMC10726127 DOI: 10.3389/fendo.2023.1292519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/15/2023] [Indexed: 12/20/2023] Open
Abstract
Anti-inflammatory treatment is the primary and vital therapeutic approach for active, moderate-to-severe thyroid eye disease (TED). Accurate pretreatment prediction of treatment response is of paramount importance for the prognosis of patients. However, relying solely on the clinical activity score asa determinant of activity has led to unsatisfactory treatment outcomes. In recent years, significant advancements have been made in identifying predictive markers for anti-inflammatory treatment response in TED, clinical markers, body fluid biomarkers and imaging biomarkers. Several clinical studies have developed prediction models based on these markers. However, there is still a lack of comprehensive elucidation or comparison between the different markers. Therefore, this review aims to provide a detailed analysis of the definition, characteristics, and application of predictive markers for anti-inflammatory treatment response in TED. Through detailed literature search, 26 articles applying anti-inflammatory treatment effect prediction with a total of 1948 TED patients were used for analysis and discussion. By gaining a better understanding of the current research on predictive markers, we can accelerate and guide the exploration of treatment prediction strategies, leading us towards an era of precise therapy for TED.
Collapse
Affiliation(s)
- Haiyang Zhang
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jingyuan Fan
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jialu Qu
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Qinghe Han
- Department of Radiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Huifang Zhou
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xuefei Song
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
52
|
Gu J, Xie S, Li X, Wu Z, Xue L, Wang S, Wei W. Identification of plasma proteomic signatures associated with the progression of cardia gastric cancer and precancerous lesions. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:286-294. [PMID: 39036665 PMCID: PMC11256680 DOI: 10.1016/j.jncc.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 07/23/2024] Open
Abstract
Objective Considering that there are no effective biomarkers for the screening of cardia gastric cancer (CGC), we developed a noninvasive diagnostic approach, employing data-independent acquisition (DIA) proteomics to identify candidate protein markers. Methods Plasma samples were obtained from 40 subjects, 10 each for CGC, cardia high-grade dysplasia (CHGD), cardia low-grade dysplasia (CLGD), and healthy controls. Proteomic profiles were obtained through liquid chromatography-mass spectrometry (LC-MS/MS-based DIA proteomics. Candidate plasma proteins were identified by weighted gene co-expression network analysis (WGCNA) combined with machine learning and further validated by the Human Protein Atlas (HPA) database. The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of the biomarker panel. Results There was a clear distinction in proteomic features among CGC, CHGD, CLGD, and the healthy controls. According to the WGCNA, we found 42 positively associated and 164 inversely associated proteins related to CGC progression and demonstrated several canonical cancer-associated pathways. Combined with the results from random forests, LASSO regression, and immunohistochemical results from the HPA database, we identified three candidate proteins (GSTP1, CSRP1, and LY6G6F) that could together distinguish CLGD (AUC = 0.91), CHGD (AUC = 0.99) and CGC (AUC = 0.98) from healthy controls with excellent accuracy. Conclusions The panel of protein biomarkers showed promising diagnostic potential for CGC and precancerous lesions. Further validation and a larger-scale study are warranted to assess its potential clinical applications, suggesting a potential avenue for CGC prevention in the future.
Collapse
Affiliation(s)
- Jianhua Gu
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Shuanghua Xie
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xinqing Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeming Wu
- iPhenome Biotechnology (Dalian), Inc., Dalian, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaoming Wang
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqiang Wei
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
53
|
Wong MCS, Leung EY, Yau STY, Chan SC, Xie S, Xu W, Huang J. Prediction algorithm for gastric cancer in a general population: A validation study. Cancer Med 2023; 12:20544-20553. [PMID: 37855240 PMCID: PMC10660462 DOI: 10.1002/cam4.6629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/04/2023] [Accepted: 09/30/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Worldwide, gastric cancer is a leading cause of cancer incidence and mortality. This study aims to devise and validate a scoring system based on readily available clinical data to predict the risk of gastric cancer in a large Chinese population. METHODS We included a total of 6,209,697 subjects aged between 18 and 70 years who have received upper digestive endoscopy in Hong Kong from 1997 to 2018. A binary logistic regression model was constructed to examine the predictors of gastric cancer in a derivation cohort (n = 4,347,224), followed by model evaluation in a validation cohort (n = 1,862,473). The algorithm's discriminatory ability was evaluated as the area under the curve (AUC) of the mathematically constructed receiver operating characteristic (ROC) curve. RESULTS Age, male gender, history of Helicobacter pylori infection, use of proton pump inhibitors, non-use of aspirin, non-steroidal anti-inflammatory drugs (NSAIDs), and statins were significantly associated with gastric cancer. A scoring of ≤8 was designated as "average risk (AR)". Scores at 9 or above were assigned as "high risk (HR)". The prevalence of gastric cancer was 1.81% and 0.096%, respectively, for the HR and LR groups. The AUC for the risk score in the validation cohort was 0.834, implying an excellent fit of the model. CONCLUSIONS This study has validated a simple, accurate, and easy-to-use scoring algorithm which has a high discriminatory capability to predict gastric cancer. The score could be adopted to risk stratify subjects suspected as having gastric cancer, thus allowing prioritized upper digestive tract investigation.
Collapse
Affiliation(s)
- Martin C. S. Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineChinese University of Hong KongHong KongSARChina
- Centre for Health Education and Health Promotion, Faculty of MedicineChinese University of Hong KongHong KongSARChina
- School of Public HealthThe Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- School of Public HealthThe Peking UniversityBeijingChina
- School of Public HealthFudan UniversityShanghaiChina
| | - Eman Yee‐man Leung
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineChinese University of Hong KongHong KongSARChina
| | - Sarah T. Y. Yau
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineChinese University of Hong KongHong KongSARChina
| | - Sze Chai Chan
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineChinese University of Hong KongHong KongSARChina
| | - Shaohua Xie
- Department of Molecular medicine and SurgeryKarolinska InstitutetSweden
| | - Wanghong Xu
- School of Public HealthFudan UniversityShanghaiChina
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineChinese University of Hong KongHong KongSARChina
- Centre for Health Education and Health Promotion, Faculty of MedicineChinese University of Hong KongHong KongSARChina
| |
Collapse
|
54
|
Wang S, Qian M, Wu M, Feng S, Zhang K. The prediction model of operative link on gastric intestinal metaplasia stage III-IV: A multicenter study. Heliyon 2023; 9:e21905. [PMID: 38027917 PMCID: PMC10665748 DOI: 10.1016/j.heliyon.2023.e21905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Intestinal metaplasia plays a crucial role in the risk stratification of gastric cancer development. The objective of the study was to develop a prediction model for Operative Link on Gastric Intestinal Metaplasia (OLGIM) Stage III-IV. Methods We analyzed 7945 high-risk gastric cancer individuals from 115 hospitals who underwent questionnaires and gastroscope. The participants were assigned to either the development or validation cohort randomly. Demographics and clinical characteristics were obtained. The outcome measurement was OLGIM III-IV. Univariate logistic regression was used for feature selection and multivariate logistic analysis was performed to develop the nomogram. Area under the curves, calibration plots, decision curve and clinical impact analysis were used to assess the performance of the nomogram. Results 4600 individuals and 3345 individuals were included in the development and validation cohort, of which 124 and 86 individuals were diagnosed with OLGIM III-IV, respectively. Parameters in the training validation cohort matched well and there was no significant difference between two cohorts. A nomogram model for predicting OLGIM Stage III-IV consisted of 4 significantly associated variables, including age, gender, PG I and G-17 (AUC 0.723 and 0.700 for the 2 cohorts). The nomogram demonstrated excellent performance in the calibration curve. Decision curve and clinical impact analysis suggested clinical benefit of the prediction model. Conclusions This reliable individualized nomogram might contribute to more accurate management for patients with OLGIM III-IV. Therefore, we suggest that this study be used as an incentive to promote the application.
Collapse
Affiliation(s)
- Song Wang
- Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meng Qian
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Min Wu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuo Feng
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
55
|
Wang M, Liu H, Fan K. Signal Amplification Strategy Design in Nanozyme-Based Biosensors for Highly Sensitive Detection of Trace Biomarkers. SMALL METHODS 2023; 7:e2301049. [PMID: 37817364 DOI: 10.1002/smtd.202301049] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/12/2023] [Indexed: 10/12/2023]
Abstract
Nanozymes show great promise in enhancing disease biomarker sensing by leveraging their physicochemical properties and enzymatic activities. These qualities facilitate signal amplification and matrix effects reduction, thus boosting biomarker sensing performance. In this review, recent studies from the last five years, concentrating on disease biomarker detection improvement through nanozyme-based biosensing are examined. This enhancement primarily involves the modulations of the size, morphology, doping, modification, electromagnetic mechanisms, electron conduction efficiency, and surface plasmon resonance effects of nanozymes for increased sensitivity. In addition, a comprehensive description of the synthesis and tuning strategies employed for nanozymes has been provided. This includes a detailed elucidation of their catalytic mechanisms in alignment with the fundamental principles of enhanced sensing technology, accompanied by the presentation of quantitatively analyzed results. Moreover, the diverse applications of nanozymes in strip sensing, colorimetric sensing, electrochemical sensing, and surface-enhanced Raman scattering have been outlined. Additionally, the limitations, challenges, and corresponding recommendations concerning the application of nanozymes in biosensing have been summarized. Furthermore, insights have been offered into the future development and outlook of nanozymes for biosensing. This review aims to serve not only as a reference for enhancing the sensitivity of nanozyme-based biosensors but also as a catalyst for exploring nanozyme properties and their broader applications in biosensing.
Collapse
Affiliation(s)
- Mengting Wang
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Hongxing Liu
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
56
|
Yu QY, Zhang HC, Jin H. Prediction of canceration of gastric ulcer with serum pepsinogen Ⅰ/Ⅱ ratio and gastrin-17. Shijie Huaren Xiaohua Zazhi 2023; 31:846-851. [DOI: 10.11569/wcjd.v31.i20.846] [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: 08/31/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Screening sensitivity markers for canceration of gastric ulcer is of great significance for the prevention and treatment of gastric cancer. Previous studies have suggested that pepsinogen and gastrin-17 (G-17) may have appreciated value in the prediction of early gastric cancer, but there is still no definitive consensus on this.
AIM To assess the clinical value of serum pepsinogen I (PGⅠ)/pepsinogen Ⅱ (PGⅡ) ratio combined with G-17 in predicting canceration of gastric ulcer to provide sensitive biochemical markers for early diagnosis of gastric cancer.
METHODS A retrospective analysis was conducted on 215 patients with gastric ulcer at our hospital from July 2020 to April 2023. According to the histopathological diagnosis by gastroscopy, the patients were divided into either a simple ulcer group (184 cases) or a gastric cancer group (31 cases). Serum PGⅠ, PGⅡ, G-17, and tumor markers [including carbohydrate antigen (CA)724, CA199, carcinoembryonic antigen (CEA), and ferritin] were detected on admission. The delta over baseline (DOB) value of Helicobacter pylori (H. pylori) infection was measured by the 13C breath test.
RESULTS There were no differences in gender, age, DOB value, or disease course between the two groups (P > 0.05). Compared with the simple ulcer group, serum levels of PGⅡ, G-17, CA724, CA199, and CEA in the gastric cancer group were increased, while PGⅠ, ferritin, and PGⅠ/PGⅡ ratio were decreased (P < 0.05). Spearman test showed that PGⅠ/PGⅡ ratio was negatively correlated with G-17, CA724, CA199, and CEA, and positively correlated with ferritin (P < 0.05). G-17 was positively correlated with CA724, CA199, and CEA, and negatively correlated with ferritin (P < 0.05). Receiver operating curve (ROC) analysis showed that the area under the ROC curve (AUC) of PGⅠ/PGⅡ ratio and G-17 for diagnosing canceration of gastric ulcer was 0.804 and 0.742, respectively. The AUC of PGⅠ/PGⅡ ratio combined with G-17 was 0.899, significantly higher than that of either indicator alone (P < 0.05).
CONCLUSION The decrease of serum PGⅠ/PGⅡ ratio and increase of G-17 are closely related to the canceration of gastric ulcer. The combination of PGⅠ/PGⅡ ratio and G-17 has good predictive performance for canceration of gastric ulcer. PGⅠ/PGⅡ ratio and G-17 can serve as sensitive biomarkers for the early diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Qiao-Yan Yu
- Department of Gastroenterology, Jinhua Guangfu Cancer Hospital, Jinhua 321000, Zhejiang Province, China
| | - Hong-Cheng Zhang
- Department of Gastroenterology, Jinhua Guangfu Cancer Hospital, Jinhua 321000, Zhejiang Province, China
| | - Hao Jin
- Department of Gastroenterology, Jinhua Guangfu Cancer Hospital, Jinhua 321000, Zhejiang Province, China
| |
Collapse
|
57
|
Xu W, Yang B, Lin S, Zhan F, Chen H, Qiu X, Liu C. Sex-specific disparities of serum pepsinogen I in relation to body mass index. Clin Chem Lab Med 2023; 61:2010-2016. [PMID: 37171227 DOI: 10.1515/cclm-2023-0236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
OBJECTIVES The clinical significance of serum pepsinogen (PG) for screening gastric cancer has been a controversial topic. Serum PG I levels have been demonstrated to be correlated with age, sex, and the Helicobacter pylori (HP) infection. However, the underlying factors that influence serum PG I variations remain to be fully elucidated. We aimed to evaluate the impacts of sex and body mass index (BMI) on PG I in Chinese population. METHODS The cross-sectional study recruited 4,299 apparently healthy participants in Fujian Province. Serum PG levels were automatically measured using ELISA method. Serum H. pylori-IgG antibody was detected by the colloidal gold immunoassay. Clinical characteristics were obtained by questionnaire. RESULTS Totally, 2,263 participants who had tests of serum PG and anti-HP IgG antibody were enrolled. Increased BMI and serum uric acid were observed in males with low PG I value (<70 μg/L). Multiple logistic regression showed the presence of overweight was the independent risk factor for male participants with low PG I level (odds ratio [OR] 1.519; p=0.017). However, the association was not found in females. CONCLUSIONS Sex-specific association of serum low PG I with overweight was observed in the southeast coastal areas of China. Thus, effects of sexual dimorphism should not be ignored during the clinical utilization of serum PG I.
Collapse
Affiliation(s)
- Wenshen Xu
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Bin Yang
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Fujian Key Laboratory of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, P.R. China
- Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, P.R. China
- Fujian Clinical Research Center for Clinical Immunology Laboratory Test, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Sheng Lin
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Fujian Key Laboratory of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, P.R. China
- Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, P.R. China
- Fujian Clinical Research Center for Clinical Immunology Laboratory Test, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Fuguo Zhan
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, P.R. China
| | - Huijuan Chen
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, P.R. China
| | - Xiaoming Qiu
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, P.R. China
| | - Can Liu
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
- Fujian Key Laboratory of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, P.R. China
- Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, P.R. China
- Fujian Clinical Research Center for Clinical Immunology Laboratory Test, the First Affiliated Hospital, Fujian Medical University, Fuzhou, P.R. China
| |
Collapse
|
58
|
Adams A, Gandhi A, In H. Gastric cancer: A unique opportunity to shift the paradigm of cancer disparities in the United States. Curr Probl Surg 2023; 60:101382. [PMID: 37993211 DOI: 10.1016/j.cpsurg.2023.101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/04/2023] [Indexed: 11/24/2023]
Affiliation(s)
- Alexandra Adams
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
| | - Atish Gandhi
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Haejin In
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Health Behavior, Society and Policy, Rutgers School of Public Health, New Brunswick, New Jersey
| |
Collapse
|
59
|
Nguyen NLT, Dang NDT, Vu QVAN, Dang AK, Ta TVAN. A Model for Gastric Cancer Risk Prediction Based on MUC1 Polymorphisms and Health-risk Behaviors in a Vietnamese Population. In Vivo 2023; 37:2347-2356. [PMID: 37652501 PMCID: PMC10500499 DOI: 10.21873/invivo.13339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND/AIM Although the expression of mucin 1(MUC1) and prostate stem cell antigen (PSCA) genes is correlated with gastric cancer development and progression, the utility of these two genes as biomarkers of gastric cancer prognosis still needs to be confirmed in clinical practice. This study aimed to develop a model predictive of gastric cancer that integrates several significant single nucleotide polymorphisms (SNPs) of MUC1 and PSCA genes, and some health-risk behavior factors in a Vietnamese population. PATIENTS AND METHODS A total of 302 patients with primary gastric carcinoma and 304 healthy persons were included in a case-control study. The generalized linear model was used with the profile of age, sex, history of smoking and using alcohol, personal and family medical history of stomach diseases, and the SNPs of MUC1 and PSCA. The prognostic value of the model was assessed by the area under a receiver operating characteristic curve (AUC) and Akaike Information Criterion (AIC) values. RESULTS In male participants, the final model, consisting of age, sex, history of smoking and using alcohol, personal and family medical history of stomach diseases and SNP MUC1 rs4072037, provided acceptable discrimination, with an AUC of 0.6374 and the lowest AIC value (539.53). In female participants, the predictive model including age, sex, history of smoking and using alcohol, personal and family medical history of stomach diseases, SNPs MUC1 rs4072037 and rs2070803 had an AUC of 0.6937 and AIC of 266.80. The calibration plots of the male model approximately fitted the ideal calibration line. CONCLUSION The predictive model based on age, sex, medical history, and genetic and health-risk behavior factors has a high potential in determining gastric cancer. Further studies that elucidate other genetic variants should be carried out to define high-risk gastric cancer groups and propose appropriate personalized prevention.
Collapse
Affiliation(s)
| | - Ngoc Dung Thi Dang
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam;
| | - Quy VAN Vu
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Anh Kim Dang
- School of Preventive Medicine and Public Health, Hanoi Medical University, Hanoi, Vietnam
| | - Thanh-VAN Ta
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam;
| |
Collapse
|
60
|
Fu XY, Mao XL, Wu HW, Lin JY, Ma ZQ, Liu ZC, Cai Y, Yan LL, Sun Y, Ye LP, Li SW. Development and validation of LightGBM algorithm for optimizing of Helicobacter pylori antibody during the minimum living guarantee crowd based gastric cancer screening program in Taizhou, China. Prev Med 2023; 174:107605. [PMID: 37419420 DOI: 10.1016/j.ypmed.2023.107605] [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: 01/22/2023] [Revised: 06/22/2023] [Accepted: 07/02/2023] [Indexed: 07/09/2023]
Abstract
Gastric cancer continues to be a significant health concern in China, with a high incidence rate. To mitigate its impact, early detection and treatment is key. However, conducting large-scale endoscopic gastric cancer screening is not feasible in China. Instead, a more appropriate approach would be to initially screen high-risk groups and follow up with endoscopic testing as needed. We conducted a study on 25,622 asymptomatic participants aged 45-70 years from a free gastric cancer screening program in the Taizhou city government's Minimum Living Guarantee Crowd (MLGC) initiative. Participants completed questionnaires, blood tests, and underwent gastrin-17 (G-17), pepsinogen I and II (PGI and PGII), and H. pylori IgG antibody (IgG) assessments. Using the light gradient boosting machine (lightGBM) algorithm, we developed a predictive model for gastric cancer risk. In the full model, F1 score was 2.66%, precision was 1.36%, and recall was 58.14%. In the high-risk model, F1 score was 2.51%, precision was 1.27%, and recall was 94.55%. Excluding IgG, the F1 score was 2.73%, precision was 1.40%, and recall was 68.62%. We conclude that H. pylori IgG appears to be able to be excluded from the prediction model without significantly affecting its performance, which is important from a health economic point of view. It suggests that screening indicators can be optimized, and expenditures reduced. These findings can have important implications for policymakers, as we can focus resources on other important aspects of gastric cancer prevention and control.
Collapse
Affiliation(s)
- Xin-Yu Fu
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xin-Li Mao
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Hao-Wen Wu
- Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Jia-Ying Lin
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Zong-Qing Ma
- Information center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Zhi-Cheng Liu
- Information center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yue Cai
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Ling-Ling Yan
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yi Sun
- Department of Neurology, Faculty of Medical, University of Toyama, Toyama, Toyama Ken, Japan.
| | - Li-Ping Ye
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China.
| | - Shao-Wei Li
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China.
| |
Collapse
|
61
|
Xu Q, Cui F, Li X, Wang N, Gao Y, Yin S, Hu F. Dangshen Huangjiu prevents gastric mucosal injury and inhibits Akt/NF-κB pathway. Food Funct 2023; 14:7897-7911. [PMID: 37491882 DOI: 10.1039/d3fo00489a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
One of the top ten tonic herbs, Dangshen is frequently found in Chinese functional foods. With the inclusion of Dangshen in the list of food and medicine substances in 2020, the Dangshen Huangjiu (DHJ) emerged. In the Bencao, it is written that Huangjiu can "open up the curved veins and thicken the stomach and intestines". Furthermore, increasing investigations have verified the protective effect of Dangshen on the gastric mucosa. Therefore, we propose the hypothesis that the stomach mucosa might be protected by the DHJ. To demonstrate that the effect of solids in Dangshen Huangjiu (DHJG) on damaged human gastric mucosal epithelial cells (GES-1) was reversed, the study used ethanol to induce injury to GES-1 and then used protein immunoblotting (western blotting) to determine the expression levels of p-Akt, p-NF-κB-p65, and NF-κB-p65 proteins in the cells. 0.04 mol L-1 MNNG (5 mL kg-1 body weight) mixed with eating disorders(2 d satiety, l d starvation, 3 d cycle) was used to further establish a chronic non-atrophic gastritis (CNAG) model in Wistar rats, at the same time, the experimental rats were given DHJ and DHJG gavage. Cellular assays confirmed that DHJG (25-100 μg mL-1) dose-dependently increased the viability of ethanol-injured GES-1 and lowered p-Akt and p-NF-κB-p65/NF-κB-p65 protein expression. Animal experiments revealed that 10 mL kg-1 and 20 mL kg-1 DHJ had no significant effect on the basic activity and gastric tissues and related biochemical indices of healthy rats; DHJ (10 mL kg-1, 20 mL kg-1) and DHJG (2.8 g kg-1, 11.4 g kg-1) resulted in some improvement in weight loss and significant improvement in gastric mucosal pathology in CNAG rats with damage. Particularly, DHJ and DHJG significantly decreased the expression of p-Akt, p-NF-κB-p65/NF-κB-p65 and Bcl-2/Bax proteins and Akt, IKKβ, IκBα and NF-κB mRNA in the gastric tissues of CNAG rats. These results showed that DHJG ameliorates ethanol-induced GES-1 cell injury; both DHJ and DHJG alleviate CNAG, and the mechanisms by which they do so may be related to DHJ and DHJG increasing the antioxidant capacity (elevating SOD, decreasing MDA), attenuating inflammatory responses (decreasing IL-1β, IL-6, and TNF-α), reversing apoptosis (reducing the Bcl-2/Bax ratio) and down-regulating gastric tissue p-Akt and p-NF-κB-p65/NF-κB-p65 protein expression as well as Akt, IKKβ, IκBα and NF-κB mRNA expression. This study indicates that the interventional effects of DHJ and DHJG in CNAG may act through the Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Qiaohong Xu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- State Key Laboratory of Applied Organic Chemistry, Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Fang Cui
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- Codonopsis Radix Research Institute, Lanzhou University, Lanzhou, 730000, China
| | - Xiaodong Li
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Nan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- State Key Laboratory of Applied Organic Chemistry, Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yingrui Gao
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- State Key Laboratory of Applied Organic Chemistry, Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Shiping Yin
- Gansu Wushanchi Huangjiu Co. Ltd, Linxia, 731804, China
| | - Fangdi Hu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- State Key Laboratory of Applied Organic Chemistry, Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, 730000, China
- Codonopsis Radix Research Institute, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
62
|
Guo X, Peng Y, Song Q, Wei J, Wang X, Ru Y, Xu S, Cheng X, Li X, Wu D, Chen L, Wei B, Lv X, Ji G. A Liquid Biopsy Signature for the Early Detection of Gastric Cancer in Patients. Gastroenterology 2023; 165:402-413.e13. [PMID: 36894035 DOI: 10.1053/j.gastro.2023.02.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND & AIMS Diagnosing gastric cancer (GC) while the disease remains eligible for surgical resection is challenging. In view of this clinical challenge, novel and robust biomarkers for early detection thus improving prognosis of GC are necessary. The present study is to develop a blood-based long noncoding RNA (LR) signature for the early-detection of GC. METHODS The present 3-step study incorporated data from 2141 patients, including 888 with GC, 158 with chronic atrophic gastritis, 193 with intestinal metaplasia, 501 healthy donors, and 401 with other gastrointestinal cancers. The LR profile of stage I GC tissue samples were analyzed using transcriptomic profiling in discovery phase. The extracellular vesicle (EV)-derived LR signature was identified with a training cohort (n = 554) and validated with 2 external cohorts (n = 429 and n = 504) and a supplemental cohort (n = 69). RESULTS In discovery phase, one LR (GClnc1) was found to be up-regulated in both tissue and circulating EV samples with an area under the curve (AUC) of 0.9369 (95% confidence interval [CI], 0.9073-0.9664) for early-stage GC (stage I/II). The diagnostic performance of this biomarker was further confirmed in 2 external validation cohorts (Xi'an cohort, AUC: 0.8839; 95% CI: 0.8336-0.9342; Beijing cohort, AUC: 0.9018; 95% CI: 0.8597-0.9439). Moreover, EV-derived GClnc1 robustly distinguished early-stage GC from precancerous lesions (chronic atrophic gastritis and intestinal metaplasia) and GC with negative traditional gastrointestinal biomarkers (CEA, CA72-4, and CA19-9). The low levels of this biomarker in postsurgery and other gastrointestinal tumor plasma samples indicated its GC specificity. CONCLUSIONS EV-derived GClnc1 serves as a circulating biomarker for the early detection of GC, thus providing opportunities for curative surgery and improved survival outcomes.
Collapse
Affiliation(s)
- Xin Guo
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Department of Endoscopic Surgery, Air Force 986(th) Hospital, Fourth Military Medical University, Xi'an, China; Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yunhua Peng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Qiying Song
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jiangpeng Wei
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinxin Wang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yi Ru
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Shenhui Xu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Cheng
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaohua Li
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Di Wu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lubin Chen
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Department of Endoscopic Surgery, Air Force 986(th) Hospital, Fourth Military Medical University, Xi'an, China
| | - Bo Wei
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Xiaohui Lv
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Gang Ji
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
63
|
Zhang T, Song SS, Liu M, Park S. Association of Fried Food Intake with Gastric Cancer Risk: A Systemic Review and Meta-Analysis of Case-Control Studies. Nutrients 2023; 15:2982. [PMID: 37447308 PMCID: PMC10347084 DOI: 10.3390/nu15132982] [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: 05/31/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Gastric cancer is one of the most prevalent cancers in Asia, and has a significant global incidence. However, the impact of fried food consumption on gastric cancer risk remains uncertain, mainly due to the limited number of participants in previous studies. To address this knowledge gap, we aimed to examine the association between fried food intake and gastric cancer incidence through a comprehensive meta-analysis. We conducted a thorough search across multiple databases, including PubMed, EMBASE, Google Scholar, Cochrane Library, China National Knowledge Infrastructure (CNKI), Korean Information Service System (KISS), and Research Information Service System (RISS), to collect studies. The newly analyzed results of the Korean Genome and Epidemiology Study (KoGES) findings were added. We assessed integrated odds ratios (ORs) and their corresponding 95% confidence intervals (CIs) from the selected studies using Cochrane RevMan 5.0 for the meta-analysis. The quality of the studies included in the meta-analysis was assessed using the Study Quality Assessment Tool of the National Heart, Lung, and Blood Institute (NHLBI). We included 18 studies in the analysis, which compared the impact of fried food intake in gastric cancer patients (n = 5739) and healthy adults (control, n = 70,933). There was a significant positive association between gastric cancer risk and fried food intake (OR = 1.52, 95% CI = 1.23-1.87, I2 = 76%, p = 0.0001). The relationship was found to be significant in both non-East Asians (OR = 1.48, 95% CI = 1.18-1.85, I2 = 31%, p = 0.0006) and East Asians (OR = 1.54, 95% CI = 1.14-2.08, I2 = 83%, p = 0.005). In conclusion, this meta-analysis supports the notion that fried food intake is associated with an increased risk of gastric cancer in both non-Asians and Asians. Promoting a reduction in fried food consumption as a measure against gastric cancer risk is recommended.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Bioconvergence, Hoseo University, Asan 31499, Republic of Korea;
| | - Sang Shin Song
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Republic of Korea;
| | - Meiling Liu
- Department of Chemical Engineering, Shanxi Institute of Science and Technology, Jincheng 048011, China;
| | - Sunmin Park
- Department of Bioconvergence, Hoseo University, Asan 31499, Republic of Korea;
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Republic of Korea;
| |
Collapse
|
64
|
Aziz S, König S, Umer M, Akhter TS, Iqbal S, Ibrar M, Ur-Rehman T, Ahmad T, Hanafiah A, Zahra R, Rasheed F. Risk factor profiles for gastric cancer prediction with respect to Helicobacter pylori: A study of a tertiary care hospital in Pakistan. Artif Intell Gastroenterol 2023; 4:10-27. [DOI: 10.35712/aig.v4.i1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/01/2023] [Accepted: 04/20/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fourth leading cause of cancer-related deaths worldwide. Diagnosis relies on histopathology and the number of endoscopies is increasing. Helicobacter pylori (H. pylori) infection is a major risk factor.
AIM To develop an in-silico GC prediction model to reduce the number of diagnostic surgical procedures. The meta-data of patients with gastroduodenal symptoms, risk factors associated with GC, and H. pylori infection status from Holy Family Hospital Rawalpindi, Pakistan, were used with machine learning.
METHODS A cohort of 341 patients was divided into three groups [normal gastric mucosa (NGM), gastroduodenal diseases (GDD), and GC]. Information associated with socioeconomic and demographic conditions and GC risk factors was collected using a questionnaire. H. pylori infection status was determined based on urea breath test. The association of these factors and histopathological grades was assessed statistically. K-Nearest Neighbors and Random Forest (RF) machine learning models were tested.
RESULTS This study reported an overall frequency of 64.2% (219/341) of H. pylori infection among enrolled subjects. It was higher in GC (74.2%, 23/31) as compared to NGM and GDD and higher in males (54.3%, 119/219) as compared to females. More abdominal pain (72.4%, 247/341) was observed than other clinical symptoms including vomiting, bloating, acid reflux and heartburn. The majority of the GC patients experienced symptoms of vomiting (91%, 20/22) with abdominal pain (100%, 22/22). The multinomial logistic regression model was statistically significant and correctly classified 80% of the GDD/GC cases. Age, income level, vomiting, bloating and medication had significant association with GDD and GC. A dynamic RF GC-predictive model was developed, which achieved > 80% test accuracy.
CONCLUSION GC risk factors were incorporated into a computer model to predict the likelihood of developing GC with high sensitivity and specificity. The model is dynamic and will be further improved and validated by including new data in future research studies. Its use may reduce unnecessary endoscopic procedures. It is freely available.
Collapse
Affiliation(s)
- Shahid Aziz
- Patients Diagnostic Lab, Isotope Application Division, Pakistan Institute of Nuclear Science and Technology, Islamabad 44000, Pakistan
- Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Interdisciplinary Center for Clinical Research, Core Unit Proteomics, University of Münster, Münster 48149, Germany
| | - Simone König
- Interdisciplinary Center for Clinical Research, Core Unit Proteomics, University of Münster, Münster 48149, Germany
| | - Muhammad Umer
- Management Information System Division, Pakistan Institute of Nuclear Science and Technology, Islamabad 44000, Pakistan
| | - Tayyab Saeed Akhter
- Centre for Liver and Digestive Diseases, Holy Family Hospital, Rawalpindi 46300, Pakistan
| | - Shafqat Iqbal
- Centre for Liver and Digestive Diseases, Holy Family Hospital, Rawalpindi 46300, Pakistan
| | - Maryum Ibrar
- Pakistan Scientific and Technological Information Centre, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Tofeeq Ur-Rehman
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Tanvir Ahmad
- Patients Diagnostic Lab, Isotope Application Division, Pakistan Institute of Nuclear Science and Technology, Islamabad 44000, Pakistan
| | - Alfizah Hanafiah
- Faculty of Medicine, Department of Medical Microbiology and Immunology, Universiti Kebangsan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Rabaab Zahra
- Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Faisal Rasheed
- Patients Diagnostic Lab, Isotope Application Division, Pakistan Institute of Nuclear Science and Technology, Islamabad 44000, Pakistan
| |
Collapse
|
65
|
Gao Y, Xin L, Lin H, Yao B, Zhang T, Zhou AJ, Huang S, Wang JH, Feng YD, Yao SH, Guo Y, Dang T, Meng XM, Yang ZZ, Jia WQ, Pang HF, Tian XJ, Deng B, Wang JP, Fan WC, Wang J, Shi LH, Yang GY, Sun C, Wang W, Zang JC, Li SY, Shi RH, Li ZS, Wang LW. Machine learning-based automated sponge cytology for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction: a nationwide, multicohort, prospective study. Lancet Gastroenterol Hepatol 2023; 8:432-445. [PMID: 36931287 DOI: 10.1016/s2468-1253(23)00004-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction have a dismal prognosis, and early detection is key to reduce mortality. However, early detection depends on upper gastrointestinal endoscopy, which is not feasible to implement at a population level. We aimed to develop and validate a fully automated machine learning-based prediction tool integrating a minimally invasive sponge cytology test and epidemiological risk factors for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction before endoscopy. METHODS For this multicohort prospective study, we enrolled participants aged 40-75 years undergoing upper gastrointestinal endoscopy screening at 39 tertiary or secondary hospitals in China for model training and testing, and included community-based screening participants for further validation. All participants underwent questionnaire surveys, sponge cytology testing, and endoscopy in a sequential manner. We trained machine learning models to predict a composite outcome of high-grade lesions, defined as histology-confirmed high-grade intraepithelial neoplasia and carcinoma of the oesophagus and oesophagogastric junction. The predictive features included 105 cytological and 15 epidemiological features. Model performance was primarily measured with the area under the receiver operating characteristic curve (AUROC) and average precision. The performance measures for cytologists with AI assistance was also assessed. FINDINGS Between Jan 1, 2021, and June 30, 2022, 17 498 eligible participants were involved in model training and validation. In the testing set, the AUROC of the final model was 0·960 (95% CI 0·937 to 0·977) and the average precision was 0·482 (0·470 to 0·494). The model achieved similar performance to consensus of cytologists with AI assistance (AUROC 0·955 [95% CI 0·933 to 0·975]; p=0·749; difference 0·005, 95% CI, -0·011 to 0·020). If the model-defined moderate-risk and high-risk groups were referred for endoscopy, the sensitivity was 94·5% (95% CI 88·8 to 97·5), specificity was 91·9% (91·2 to 92·5), and the predictive positive value was 18·4% (15·6 to 21·6), and 90·3% of endoscopies could be avoided. Further validation in community-based screening showed that the AUROC of the model was 0·964 (95% CI 0·920 to 0·990), and 92·8% of endoscopies could be avoided after risk stratification. INTERPRETATION We developed a prediction tool with favourable performance for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction. This approach could prevent the need for endoscopy screening in many low-risk individuals and ensure resource optimisation by prioritising high-risk individuals. FUNDING Science and Technology Commission of Shanghai Municipality.
Collapse
Affiliation(s)
- Ye Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Lei Xin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Han Lin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Bin Yao
- School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Tao Zhang
- Department of Gastroenterology, Nanchong Central Hospital, Nanchong, Sichuan Province, China
| | - Ai-Jun Zhou
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Jian-Hua Wang
- Department of Gastroenterology, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Ya-Dong Feng
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Sheng-Hua Yao
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, Jiangsu Province, China
| | - Yan Guo
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, Jiangsu Province, China
| | - Tong Dang
- Department of Digestive Diseases, Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Xian-Mei Meng
- Department of Digestive Diseases, Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Zeng-Zhou Yang
- Digestive Endoscopy Unit, Linzhou People's Hospital, Anyang, Henan Province, China
| | - Wan-Qi Jia
- Gastrointestinal Endoscopy Center, Nanyang Second People's Hospital, Nanyang, Henan Province, China
| | - Hui-Fang Pang
- Department of Gastroenterology, Digestive Endoscopy Unit, Tongliao City Hospital, Tongliao, Inner Mongolia, China
| | - Xiao-Juan Tian
- Department of Gastroenterology, Xixia County People's Hospital, Nanyang, Henan Province, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Jun-Ping Wang
- Department of Gastroenterology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi Province, China
| | - Wen-Chuan Fan
- Department of Gastroenterology, Digestive Endoscopy Center, The People's Hospital of Yanting City, Mianyang, Sichuan Province, China
| | - Jun Wang
- Department of Gastroenterology, Jinhu County People's Hospital, Huaian, Jiangsu Province, China
| | - Li-Hong Shi
- Department of Gastroenterology, the Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Guan-Yu Yang
- School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Chang Sun
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Wei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Jun-Cai Zang
- Harbor Scientific Instrument, Xiangtan, Hunan, China
| | - Song-Yang Li
- Harbor Scientific Instrument, Xiangtan, Hunan, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Luo-Wei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China.
| |
Collapse
|
66
|
Zhu X, Lv J, Zhu M, Yan C, Deng B, Yu C, Guo Y, Ni J, She Q, Wang T, Wang J, Jiang Y, Chen J, Hang D, Song C, Gao X, Wu J, Dai J, Ma H, Yang L, Chen Y, Song M, Wei Q, Chen Z, Hu Z, Shen H, Ding Y, Li L, Jin G. Development, validation, and evaluation of a risk assessment tool for personalized screening of gastric cancer in Chinese populations. BMC Med 2023; 21:159. [PMID: 37106459 PMCID: PMC10142220 DOI: 10.1186/s12916-023-02864-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Effective risk prediction models are lacking for personalized endoscopic screening of gastric cancer (GC). We aimed to develop, validate, and evaluate a questionnaire-based GC risk assessment tool for risk prediction and stratification in the Chinese population. METHODS In this three-stage multicenter study, we first selected eligible variables by Cox regression models and constructed a GC risk score (GCRS) based on regression coefficients in 416,343 subjects (aged 40-75 years) from the China Kadoorie Biobank (CKB, development cohort). In the same age range, we validated the GCRS effectiveness in 13,982 subjects from another independent Changzhou cohort (validation cohort) as well as in 5348 subjects from an endoscopy screening program in Yangzhou. Finally, we categorized participants into low (bottom 20%), intermediate (20-80%), and high risk (top 20%) groups by the GCRS distribution in the development cohort. RESULTS The GCRS using 11 questionnaire-based variables demonstrated a Harrell's C-index of 0.754 (95% CI, 0.745-0.762) and 0.736 (95% CI, 0.710-0.761) in the two cohorts, respectively. In the validation cohort, the 10-year risk was 0.34%, 1.05%, and 4.32% for individuals with a low (≤ 13.6), intermediate (13.7~30.6), and high (≥ 30.7) GCRS, respectively. In the endoscopic screening program, the detection rate of GC varied from 0.00% in low-GCRS individuals, 0.27% with intermediate GCRS, to 2.59% with high GCRS. A proportion of 81.6% of all GC cases was identified from the high-GCRS group, which represented 28.9% of all the screened participants. CONCLUSIONS The GCRS can be an effective risk assessment tool for tailored endoscopic screening of GC in China. Risk Evaluation for Stomach Cancer by Yourself (RESCUE), an online tool was developed to aid the use of GCRS.
Collapse
Affiliation(s)
- Xia Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Caiwang Yan
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Bin Deng
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 88 Daxue South Rd, Yangzhou, 225000, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Yu Guo
- Fuwai Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Ni
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
| | - Qiang She
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 88 Daxue South Rd, Yangzhou, 225000, China
| | - Tianpei Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Public Health Institute of Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jiayu Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Yue Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Jiaping Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Dong Hang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Ci Song
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Xuefeng Gao
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 88 Daxue South Rd, Yangzhou, 225000, China
| | - Jian Wu
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 88 Daxue South Rd, Yangzhou, 225000, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard University T.H. Chan School of Public Health, Boston, MA, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
- Public Health Institute of Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China.
| | - Yanbing Ding
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 88 Daxue South Rd, Yangzhou, 225000, China.
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China.
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, 211166, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China.
- Public Health Institute of Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
67
|
Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat Rev Clin Oncol 2023; 20:338-349. [PMID: 36959359 DOI: 10.1038/s41571-023-00747-0] [Citation(s) in RCA: 271] [Impact Index Per Article: 135.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/25/2023]
Abstract
Gastric cancer remains a major cause of cancer-related mortality worldwide. The temporal trends for this malignancy, however, are dynamic, and reports from the past decade indicate important declines in some regions and demographic groups, as well as a few notable exceptions in which gastric cancer rates are either stable or increasing. Two main anatomical subtypes of gastric cancer exist, non-cardia and cardia, with different temporal trends and risk factors (such as obesity and reflux for cardia gastric cancer and Helicobacter pylori infection for non-cardia gastric cancer). Shifts in the distribution of anatomical locations have been detected in several high-incidence regions. H. pylori is an important aetiological factor for gastric cancer; importantly, the anticipated long-term findings from studies examining the effect of H. pylori eradication on the risk of (re)developing gastric cancer have emerged in the past few years. In this Review, we highlight the latest trends in incidence and mortality using an evidence-based approach. We make the best possible inferences, including clinical and public health inference, on the basis of the quality of the evidence available, and highlight burning questions as well as gaps in knowledge and public health practice that need to be addressed to reduce gastric cancer burden worldwide.
Collapse
Affiliation(s)
- Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Theresa Nguyen Wenker
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
68
|
Sheng C, Sun L, Lyu Z, Li L, Zhang Y, Zhang Y, Zhang Y, Dai H, Huang Y, Song F, Yuan Y, Chen K. Development of a modified ABC method among Helicobacter pylori infected but serum pepsinogen test-negative individuals. Helicobacter 2023; 28:e12966. [PMID: 36941759 DOI: 10.1111/hel.12966] [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: 10/07/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Although the ABC method for gastric cancer (GC) screening has been widely adopted in Japan, it may not be suitable for other countries due to population heterogeneity and different tumor histology. We aim to develop a modified ABC method to improve GC screening performance, especially among Helicobacter pylori (Hp) infected but serum pepsinogen (sPG) test-negative individuals. METHODS A total of 4745 participants were recruited from Tianjin, China, and were classified into four groups by combined assay for Hp infection and sPG concentrations: Group A (Hp [-], PG [-]), Group B (Hp [+], PG [-]), Group C (Hp [+], PG [+]), and Group D (Hp [-], PG [+]). We used receiver-operating characteristic (ROC) curves analysis and minimum p value method to determine the optimal cutoff point for PG II in Group B. We performed logistic regressions to examine the risk of GC across different subgroups. In addition to the derivation set, the performance of the modified ABC method was also evaluated in an external set involving 16,292 participants from Liaoning, China. RESULTS In the modified ABC method, we further classified Group B as low-risk (Group B1) and high-risk subgroups (Group B2) using optimal sPG II cutoff point (20.0 ng/mL) by ROC curves analysis and minimum p value method. Compared with Group B1, Group B2 had a significantly higher risk of GC (adjusted OR = 2.54, 95% CI = 1.94-3.33). The modified ABC method showed good discrimination for GC (AUC = 0.61, 95% CI = 0.59-0.63) and improved risk reclassification (NRI = 0.11, p < .01). Similar results were observed in the validation dataset. CONCLUSIONS The modified ABC method can effectively identify high-risk population for GC among Hp-infected but sPG test-negative participants in China.
Collapse
Affiliation(s)
- Chao Sheng
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Zhangyan Lyu
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Limin Li
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yuhao Zhang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yu Zhang
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yacong Zhang
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Hongji Dai
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yubei Huang
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| |
Collapse
|
69
|
Wang XY, Wang LL, Xu L, Liang SZ, Yu MC, Zhang QY, Dong QJ. Evaluation of polygenic risk score for risk prediction of gastric cancer. World J Gastrointest Oncol 2023; 15:276-285. [PMID: 36908320 PMCID: PMC9994049 DOI: 10.4251/wjgo.v15.i2.276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/11/2023] [Accepted: 02/02/2023] [Indexed: 02/14/2023] Open
Abstract
Genetic variations are associated with individual susceptibility to gastric cancer. Recently, polygenic risk score (PRS) models have been established based on genetic variants to predict the risk of gastric cancer. To assess the accuracy of current PRS models in the risk prediction, a systematic review was conducted. A total of eight eligible studies consisted of 544842 participants were included for evaluation of the performance of PRS models. The overall accuracy was moderate with Area under the curve values ranging from 0.5600 to 0.7823. Incorporation of epidemiological factors or Helicobacter pylori (H. pylori) status increased the accuracy for risk prediction, while selection of single nucleotide polymorphism (SNP) and number of SNPs appeared to have little impact on the model performance. To further improve the accuracy of PRS models for risk prediction of gastric cancer, we summarized the association between gastric cancer risk and H. pylori genomic variations, cancer associated bacteria members in the gastric microbiome, discussed the potentials for performance improvement of PRS models with these microbial factors. Future studies on comprehensive PRS models established with human SNPs, epidemiological factors and microbial factors are indicated.
Collapse
Affiliation(s)
- Xiao-Yu Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Li-Li Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Lin Xu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Shu-Zhen Liang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Meng-Chao Yu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Qiu-Yue Zhang
- Department of Clinical Laboratory, the Eighth Medical Center of the General Hospital of the People’s Liberation Army, Beijing 100000, China
| | - Quan-Jiang Dong
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| |
Collapse
|
70
|
A DSC Test for the Early Detection of Neoplastic Gastric Lesions in a Medium-Risk Gastric Cancer Area. Int J Mol Sci 2023; 24:ijms24043290. [PMID: 36834698 PMCID: PMC9966253 DOI: 10.3390/ijms24043290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
In this study, we aimed to assess the accuracy of the proposed novel, noninvasive serum DSC test in predicting the risk of gastric cancer before the use of upper endoscopy. To validate the DSC test, we enrolled two series of individuals living in Veneto and Friuli-Venezia Giulia, Italy (n = 53 and n = 113, respectively), who were referred for an endoscopy. The classification used for the DSC test to predict gastric cancer risk combines the coefficient of the patient's age and sex and serum pepsinogen I and II, gastrin 17, and anti-Helicobacter pylori immunoglobulin G concentrations in two equations: Y1 and Y2. The coefficient of variables and the Y1 and Y2 cutoff points (>0.385 and >0.294, respectively) were extrapolated using regression analysis and an ROC curve analysis of two retrospective datasets (300 cases for the Y1 equation and 200 cases for the Y2 equation). The first dataset included individuals with autoimmune atrophic gastritis and first-degree relatives with gastric cancer; the second dataset included blood donors. Demographic data were collected; serum pepsinogen, gastrin G17, and anti-Helicobacter pylori IgG concentrations were assayed using an automatic Maglumi system. Gastroscopies were performed by gastroenterologists using an Olympus video endoscope with detailed photographic documentation during examinations. Biopsies were taken at five standardized mucosa sites and were assessed by a pathologist for diagnosis. The accuracy of the DSC test in predicting neoplastic gastric lesions was estimated to be 74.657% (65%CI; 67.333% to 81.079%). The DSC test was found to be a useful, noninvasive, and simple approach to predicting gastric cancer risk in a population with a medium risk of developing gastric cancer.
Collapse
|
71
|
Development of amplified luminescent proximity homogeneous assay for quantitation of gastrin-17. Anal Biochem 2023; 662:115016. [PMID: 36502889 DOI: 10.1016/j.ab.2022.115016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
A highly sensitive and convenient amplified luminescent proximity homogeneous assay (AlphaLISA) method with high throughput and automation potential was developed for quantitation of serum Gastrin-17 (G-17) levels, which can facilitate the early diagnosis of atrophic gastritis in people at high risk of gastric cancer using a non-invasive approach. In this study, donor and acceptor beads with modified carboxyl groups on the surface were directly coupled to anti-G-17 antibodies through activation was proposed for application in the development of the new AlphaLISA, which can effectively simplify the steps and shorten the reaction time to achieve faster detection. Therefore, the G-17-AlphaLISA only needs to react for 15 min to obtain good analysis results. The proposed method has a wider detection range than commercial enzyme-linked immunosorbent assay (ELISA) kits (0.12-112.8 pmol/L > 0.5-40 pmol/L). In addition, results of G-17-AlphaLISA and ELISA had good correlation and agreement (ρ = 0.936). Importantly, the developed method may be more suitable for the large-scale screening of people at high risk for gastric cancer than traditional ELISA and provides a novel solution for other biomarkers that require accurate, highly sensitive, and high throughput detection.
Collapse
|
72
|
He S, Sun D, Li H, Cao M, Yu X, Lei L, Peng J, Li J, Li N, Chen W. Real-World Practice of Gastric Cancer Prevention and Screening Calls for Practical Prediction Models. Clin Transl Gastroenterol 2023; 14:e00546. [PMID: 36413795 PMCID: PMC9944379 DOI: 10.14309/ctg.0000000000000546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Some gastric cancer prediction models have been published. Still, the value of these models for application in real-world practice remains unclear. We aim to summarize and appraise modeling studies for gastric cancer risk prediction and identify potential barriers to real-world use. METHODS This systematic review included studies that developed or validated gastric cancer prediction models in the general population. RESULTS A total of 4,223 studies were screened. We included 18 development studies for diagnostic models, 10 for prognostic models, and 1 external validation study. Diagnostic models commonly included biomarkers, such as Helicobacter pylori infection indicator, pepsinogen, hormone, and microRNA. Age, sex, smoking, body mass index, and family history of gastric cancer were frequently used in prognostic models. Most of the models were not validated. Only 25% of models evaluated the calibration. All studies had a high risk of bias, but over half had acceptable applicability. Besides, most studies failed to clearly report the application scenarios of prediction models. DISCUSSION Most gastric cancer prediction models showed common shortcomings in methods, validation, and reports. Model developers should further minimize the risk of bias, improve models' applicability, and report targeting application scenarios to promote real-world use.
Collapse
Affiliation(s)
- Siyi He
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| | - Dianqin Sun
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| | - He Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| | - Maomao Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| | - Xinyang Yu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| | - Lin Lei
- Department of Cancer Prevention and Control, Shenzhen Center for Chronic Disease Control, Shenzhen, Guangdong Province, China
| | - Ji Peng
- Department of Cancer Prevention and Control, Shenzhen Center for Chronic Disease Control, Shenzhen, Guangdong Province, China
| | - Jiang Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| | - Ni Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
| |
Collapse
|
73
|
Ma C, Zhang P, Du S, Li Y, Li S. Construction of Tongue Image-Based Machine Learning Model for Screening Patients with Gastric Precancerous Lesions. J Pers Med 2023; 13:jpm13020271. [PMID: 36836505 PMCID: PMC9968136 DOI: 10.3390/jpm13020271] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Screening patients with precancerous lesions of gastric cancer (PLGC) is important for gastric cancer prevention. The accuracy and convenience of PLGC screening could be improved with the use of machine learning methodologies to uncover and integrate valuable characteristics of noninvasive medical images related to PLGC. In this study, we therefore focused on tongue images and for the first time constructed a tongue image-based PLGC screening deep learning model (AITongue). The AITongue model uncovered potential associations between tongue image characteristics and PLGC, and integrated canonical risk factors, including age, sex, and Hp infection. Five-fold cross validation analysis on an independent cohort of 1995 patients revealed the AITongue model could screen PLGC individuals with an AUC of 0.75, 10.3% higher than that of the model with only including canonical risk factors. Of note, we investigated the value of the AITongue model in predicting PLGC risk by establishing a prospective PLGC follow-up cohort, reaching an AUC of 0.71. In addition, we developed a smartphone-based app screening system to enhance the application convenience of the AITongue model in the natural population from high-risk areas of gastric cancer in China. Collectively, our study has demonstrated the value of tongue image characteristics in PLGC screening and risk prediction.
Collapse
Affiliation(s)
- Changzheng Ma
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing 100084, China
| | - Peng Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing 100084, China
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Chaoyang District, Beijing 100029, China
| | - Yan Li
- Department of Traditional Chinese Medicine, Yijishan Hospital of Wannan Medical College, Wuhu 241000, China
| | - Shao Li
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing 100084, China
- Correspondence:
| |
Collapse
|
74
|
Kotelevets SM, Chekh SA, Chukov SZ. Cancer risk stratification system and classification of gastritis: Perspectives. World J Meta-Anal 2023; 11:18-28. [DOI: 10.13105/wjma.v11.i1.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/17/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
Kyoto global consensus reports that the current ICD-10 classification for gastritis is obsolete. The Kyoto classification of gastritis states that severe mucosal atrophy has a high risk of gastric cancer, while mild to moderate atrophy has a low risk. The updated Kimura-Takemoto classification of atrophic gastritis considers five histological types of multifocal corpus atrophic gastritis according to stages C2 to O3. This method of morphological diagnosis of atrophic gastritis increases sensitivity by 2.4 times for severe atrophy compared to the updated Sydney system. This advantage should be considered when stratifying the high risk of gastric cancer. The updated Kimura-Takemoto classification of atrophic gastritis should be used as a reference standard (gold standard) in studies of morpho-functional relationships to identify serological markers of atrophic gastritis with evidence-based effectiveness. The use of artificial intelligence in the serological screening of atrophic gastritis makes it possible to screen a large number of the population. During serological screening of atrophic gastritis and risk stratification of gastric cancer, it is advisable to use the Kyoto classification of gastritis with updated Kimura-Takemoto classification of atrophic gastritis.
Collapse
Affiliation(s)
- Sergey M Kotelevets
- Department of Therapy, North Caucasus State Academy, Cherkessk 369000, Karachay-Cherkess Republic, Russia
| | - Sergey A Chekh
- Department of Mathematics, North Caucasus State Academy, Cherkessk 369000, Karachay-Cherkess Republic, Russia
| | - Sergey Z Chukov
- Department of Pathological Anatomy, Stavropol State Medical University, Stavropol 355017, Stavropol region, Russia
| |
Collapse
|
75
|
Afrash MR, Shafiee M, Kazemi-Arpanahi H. Establishing machine learning models to predict the early risk of gastric cancer based on lifestyle factors. BMC Gastroenterol 2023; 23:6. [PMID: 36627564 PMCID: PMC9832798 DOI: 10.1186/s12876-022-02626-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Gastric cancer is one of the leading causes of death worldwide. Screening for gastric cancer greatly relies on endoscopy and pathology biopsy, which are invasive and pose financial burdens. Thus, the prevention of the disease by modifying lifestyle-related behaviors and dietary habits or even the prevention of risk factor formation is of great importance. This study aimed to construct an inexpensive, non-invasive, fast, and high-precision diagnostic model using six machine learning (ML) algorithms to classify patients at high or low risk of developing gastric cancer by analyzing individual lifestyle factors. METHODS This retrospective study used the data of 2029 individuals from the gastric cancer database of Ayatollah Taleghani Hospital in Abadan City, Iran. The data were randomly separated into training and test sets (ratio 0.7:0.3). Six ML methods, including multilayer perceptron (MLP), support vector machine (SVM) (linear kernel), SVM (RBF kernel), k-nearest neighbors (KNN) (K = 1, 3, 7, 9), random forest (RF), and eXtreme Gradient Boosting (XGBoost), were trained to construct prognostic models before and after performing the relief feature selection method. Finally, to evaluate the models' performance, the metrics derived from the confusion matrix were calculated via a test split and cross-validation. RESULTS This study found 11 important influence factors for the risk of gastric cancer, such as Helicobacter pylori infection, high salt intake, and chronic atrophic gastritis, among other factors. Comparisons indicated that the XGBoost had the best performance for the risk prediction of gastric cancer. CONCLUSIONS The results suggest that based on simple baseline patient data, the ML techniques have the potential to start the prescreening of gastric cancer and identify high-risk individuals who should proceed with invasive examinations. Our model could also considerably lessen the number of cases that need endoscopic surveillance. Future studies are required to validate the efficacy of the models in a larger and multicenter population.
Collapse
Affiliation(s)
- Mohammad Reza Afrash
- grid.411705.60000 0001 0166 0922Department of Artificial Intelligence, Smart University of Medical Sciences, Tehran, Iran
| | - Mohsen Shafiee
- Department of Nursing, Abadan University of Medical Sciences, Abadan, Iran
| | - Hadi Kazemi-Arpanahi
- Department of Health Information Technology, Abadan University of Medical Sciences, Abadan, Iran
| |
Collapse
|
76
|
Wang X, Zhang Q, Han F, Wu Y. The application of new gastric cancer screening score system for gastric cancer screening and risk assessment of gastric precancerous lesions in China. Scand J Gastroenterol 2023; 58:34-37. [PMID: 35868004 DOI: 10.1080/00365521.2022.2099761] [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] [Indexed: 02/04/2023]
Abstract
Objective: To evaluate the value of new gastric cancer screening score system for risk assessment of gastric precancerous lesions.Methods: A total of 520 patients were enrolled after the examination of endoscopy at Endoscopy Center, Department of Gastroenterology, from June 2018 to December 2021. The patients were divided into three groups according to age, gender, serum helicobacter pylori antibody test, pepsinogen I (PGI), pepsinogen II (PGII), pepsinogen I/II ratio (PGR) and gastrin-17 test results before endoscopy: Group A defined as low-risk group (0-11 points), Group B defined as middle-risk group (12-16 points), Group C defined as high-risk group (17-23 points). The detection rates of gastric cancer and atrophic gastritis in three groups were analyzed. According to the range and degree of atrophy/intestinal metaplasia, patients were divided into five groups on the basis of OLGA/OLGIM staging system. The levels of PG I, PG II and PGR were compared between different groups, and the correlation between new gastric cancer screening score system and OLGA/OLGIM staging system were evaluated. Statistical analysis was accomplished by ANOVA, chi-square test and Gamma coefficient analysis.Results: A total of 520 patients were enrolled. 268 patients were classified into group A,222 patients into group B and 30 patients into group C, respectively. According to the pathological results, 281 cases were non-atrophic gastritis, 230 cases atrophic gastritis and 9 cases gastric cancer. For OLGA staging system, 281 patients were divided into stage-0 group, 121 patients into stage-I group, 72 patients into stage-II group, 33 patients into stage-III group and 13 patients into stage-IV groups. The PGI and PGR level correlated inversely with the rising OLGA stages (F = 3.028, p = .016, F = 6.036, p < .001). For OLGIM staging system, 252 patients were divided into stage-0 group, 137 patients into stage-I group, 80 patients into stage-II group, 36 patients into stage-III group and 15 patients into stage-IV group. The PGR level correlated inversely with the rising OLGIM stages (F = 3.466, p=.007). The detection rates of gastric cancer and atrophic gastritis in Group C were much higher than other groups. (X2 = 14.727, p < .001; X2 = 51.280, p < .001). Gamma coefficient analysis showed significant correlations between OLGA/OLGIM and the new gastric cancer screening score system (p < .001).Conclusions: The new gastric cancer screening score system is closely linked with histological OLGA/OLGIM staging system in the risk assessment of gastric precancerous lesions. The role of new gastric cancer screening score system in future gastric precancerous lesions screening and high risk population identifying was promising.
Collapse
Affiliation(s)
- Xiaoteng Wang
- Department of Gastroenterology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| | - Qiping Zhang
- Department of Infection Control and Public Health, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| | - Feng Han
- Department of Gastroenterology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| | - Yiming Wu
- Department of Gastroenterology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| |
Collapse
|
77
|
Liao F, Zhu Z, Zhu S, Wan J, Fan C, Zhang X, Shu X, Lu N. Investigation of the prevalence and risk factors of Helicobacter pylori infection and the value of different gastric cancer screening methods in a low-risk region of gastric cancer in China. Ann Med 2023; 55:2243988. [PMID: 37669097 PMCID: PMC10481759 DOI: 10.1080/07853890.2023.2243988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND The aim of this current study was to identify the prevalence and risk factors of H. pylori infection in the low-risk area of gastric cancer in China, and evaluate the value of different gastric cancer screening methods. METHODS An epidemiological study was conducted in Yudu County, Jiangxi, China, and participants were followed up for 6 years. All participants completed a questionnaire, laboratory tests and endoscopy. Patients were divided into H. pylori positive and negative groups, and risk factors for H. pylori infection were identified using multivariate logistic regression analysis. RESULTS A total of 1962 residents were included, the prevalence of H. pylori infection was 33.8%. Multivariate analysis showed that annual income ≤20,000 yuan (OR: 1.44, 95% CI: 1.18-1.77, p < 0.001), loss of appetite (OR: 1.71, 95% CI: 1.29-2.26, p < 0.001), PG II >37.23 ng/mL (OR: 2.11, 95% CI: 1.50-2.97, p < 0.001), G-17 > 1.5 and ≤5.7 pmol/L (OR: 2.52, 95% CI: 1.93-3.30, p < 0.001), and G-17 > 5.7 pmol/L (OR: 1.96, 95% CI: 1.48-2.60, p < 0.001) were risk factors of H. pylori infection, while alcohol consumption (OR: 0.70, 95% CI: 0.54-0.91, p = 0.006) was a protective factor. According to the new gastric cancer screening method, the prevalence of low-grade intraepithelial neoplasia in the low-risk group, medium-risk group and high-risk group was 4.4%, 7.7% and 12.5% respectively (p < 0.001). CONCLUSIONS In a low-risk area of gastric cancer in China, the infection rate of H. pylori is relatively low. Low income, loss of appetite, high PG II, and high G-17 were risk factors for H. pylori infection, while alcohol consumption was a protective factor. Moreover, the new gastric cancer screening method better predicted low-grade intraepithelial neoplasia than the ABC method and the new ABC method.
Collapse
Affiliation(s)
- Foqiang Liao
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Zhenhua Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Shusheng Zhu
- Department of Gastroenterology, Yudu County People’s Hospital, Ganzhou, Jiangxi, P.R.China
| | - Jianhua Wan
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Chenglai Fan
- Department of Gastroenterology, Yudu County People’s Hospital, Ganzhou, Jiangxi, P.R.China
| | - Xu Zhang
- Department of Gastroenterology, Yudu County People’s Hospital, Ganzhou, Jiangxi, P.R.China
| | - Xu Shu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Nonghua Lu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
78
|
Fei L, Hou G, Lu Z, Yang X, Ji Z. High expression of pituitary tumor gene family is a predictor for poor prognosis of gastric cancer. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2101548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Affiliation(s)
- Lihong Fei
- Department of Gastroenterology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| | - Guoxin Hou
- Department of Oncology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| | - Zhimin Lu
- Department of outpatient, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| | - Xinmei Yang
- Department of Oncology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| | - Zizhong Ji
- Department of Gastroenterology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| |
Collapse
|
79
|
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
|
80
|
Xu H, Li W. Early detection of gastric cancer in China: progress and opportunities. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0655. [PMID: 36514907 PMCID: PMC9755958 DOI: 10.20892/j.issn.2095-3941.2022.0655] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 12/14/2022] Open
Affiliation(s)
- Hengmin Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Wenqing Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
81
|
Wang Z, Han W, Xue F, Zhao Y, Wu P, Chen Y, Yang C, Gu W, Jiang J. Nationwide gastric cancer prevention in China, 2021-2035: a decision analysis on effect, affordability and cost-effectiveness optimisation. Gut 2022; 71:2391-2400. [PMID: 35902213 DOI: 10.1136/gutjnl-2021-325948] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 07/08/2022] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To project future trajectories of the gastric cancer (GC) burden in China under different scenarios of GC prevention and identify strategies to improve affordability and cost-effectiveness. DESIGN Using a cohort of Chinese men and women born during 1951-1980, we assumed that different prevention strategies were conducted, including eradication of Helicobacter pylori (Hp) and endoscopy screening (one-time, annual, biennial, triennial or stratified according to personal risk). We performed a literature search to identify up-to-date data and populate a Markov model to project the number of new GC cases and deaths during 2021-2035, as well as resource requirements and quality-adjusted life-years (QALYs). We examined the impacts of general (among the whole population) and targeted (high-risk population) prevention. RESULTS During 2021-2035, 10.0 million new GC cases and 5.6 million GC deaths would occur, with 7.6%-35.5% and 6.9%-44.5%, respectively, being avoidable through various prevention strategies. Relative to the status quo, Hp eradication was a cost-saving strategy. General annual screening dominated other screening strategies, but cost more than CNY 70 000 per QALY gained (willingness-to-pay) compared with Hp eradication. Among endoscopy strategies, targeted screening resulted in 44%-49% lower cost per QALY gained over the status quo than general screening. Among high-risk population, tailoring the screening frequency according to personal risk could reduce endoscopy-related resources by 22% compared with biennial screening and by 55% compared with annual screening, CONCLUSION: Our findings provide important input for future decision-making and investment, highlighting the need and feasibility for China to include GC prevention in its national health plans.
Collapse
Affiliation(s)
- Zixing Wang
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Wei Han
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Fang Xue
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Yujie Zhao
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Peng Wu
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Yali Chen
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Cuihong Yang
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Wentao Gu
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| | - Jingmei Jiang
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
82
|
Hu Y, Zhu Y, Lu NH. The management of Helicobacter pylori infection and prevention and control of gastric cancer in China. Front Cell Infect Microbiol 2022; 12:1049279. [PMID: 36530421 PMCID: PMC9751207 DOI: 10.3389/fcimb.2022.1049279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/14/2022] [Indexed: 12/04/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection, a type-1 carcinogen, was closely associated with gastric cancer (GC). Successfully eradicating H. pylori infection could reduce the incidence of GC. China was a country with high incidence of GC and high prevalence of H. pylori infection. Nearly half of worldwide GC new cases and deaths attributed to H. pylori infection occurred in China. H. pylori prevalence varied over time with the improvement of socioeconomic status and sanitary conditions. The knowledge of antibiotic resistance rate in time was important to guide the clinical choice of antibiotics use in the regimens. With the publication of five Chinese consensus reports on the management of H. pylori infection and the effort of public preach of H. pylori-related knowledge, the standardization of H. pylori diagnosis and treatment by clinicians was improved. Bismuth-containing quadruple therapy was widely applied in clinical practice of H. pylori eradication because of high efficacy and safety. High-dose Proton Pump Inhibitor-amoxicillin dual therapy or vonoprazan-amoxicillin dual therapy showed comparable efficacy and lower side effects than bismuth-containing quadruple therapy, which were the alternative choice. The diagnosis rate of early GC was low and distinguishing Chinese GC risk population for the further endoscopy screening was important. Efforts have been done to establish prediction models to stratify GC risk in the Chinese GC risk population. We reviewed the current situation of the management of H. pylori infection and prevention and control of GC in China here.
Collapse
Affiliation(s)
- Yi Hu
- Department Of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China,JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department Of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China,JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China,*Correspondence: Nong-Hua Lu, ; Yin Zhu,
| | - Nong-Hua Lu
- Department Of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China,JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China,*Correspondence: Nong-Hua Lu, ; Yin Zhu,
| |
Collapse
|
83
|
Chen X, Pan T, Guo G, Chen G, Cai Y, Tang Y, Wang Y, Wang Y, Deng Z, Li L, Zhou Y. LncRNA NORAD mediates KMT2D expression by targeting miR-204-5p and affects the growth of gastric cancer. J Gastrointest Oncol 2022; 13:2832-2844. [PMID: 36636064 PMCID: PMC9830370 DOI: 10.21037/jgo-22-1014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Long non-coding ribonucleic acids (lncRNAs) are a class of non-coding RNAs implicated in the development of many malignancies, including gastric cancer (GC). In this study, we investigated the functions and molecular mechanisms of non-coding RNA activated by deoxyribonucleic acid damage (NORAD) in GC. METHODS NORAD expression at the messenger RNA levels was determined by quantitative reverse transcriptase (RT)-polymerase chain reaction assays. Cell proliferation, migration, and invasion were detected by Cell Counting Kit-8 assays, in-vivo tumor formation assays, and Transwell assays. Cell-cycle distribution was detected by a flow cytometry analysis. NORAD location was detected by nucleocytoplasmic fractionation assays. The interaction between NORAD and the microRNA-204-5p (miR-204-5p)/Lysine Methyltransferase 2D (KMT2D) axis was verified by dual-luciferase reporter gene assays and RNA binding protein immunoprecipitation (RIP) assays. Western blot was used to study the phosphatase and tensin homolog (PTEN)/phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT) signaling pathway. RESULTS NORAD was upregulated in the GC tissues and cell lines. The silencing of NORAD repressed cell proliferation and the Growth 2 (G2)/Mitosis (M) cell-cycle transition in GC. NORAD also regulated KMT2D expression by targeting miR-204-5p and mediated PTEN/PI3K/AKT signaling in GC. CONCLUSIONS We found that NORAD acts as an oncogene in GC. Our findings might provide a novel therapeutic target for GC.
Collapse
Affiliation(s)
- Xinqi Chen
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Ting Pan
- Department of Blood Transfusion, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Guohu Guo
- Department of General Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guohao Chen
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yongchang Cai
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yuxin Tang
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yijun Wang
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yucheng Wang
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Zhenwei Deng
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Libo Li
- Department of General Surgery, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yan Zhou
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
84
|
Wu M, Feng S, Qian M, Wang S, Zhang K. Helicobacter pylori Infection Combined with OLGA and OLGIM Staging Systems for Risk Assessment of Gastric Cancer: A Retrospective Study in Eastern China. Risk Manag Healthc Policy 2022; 15:2243-2255. [PMID: 36475275 PMCID: PMC9719712 DOI: 10.2147/rmhp.s391386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/17/2022] [Indexed: 09/25/2024] Open
Abstract
PURPOSE Helicobacter pylori (H. pylori) infection is a high-risk factor for gastric cancer (GC). The main aim of this study was to evaluate the effect of H. pylori on gastritis staging systems and the value of H. pylori combined with gastritis staging systems in predicting GC risk. PATIENTS AND METHODS This study enrolled 609 patients with gastric atrophy (GA) and 527 patients with gastric intestinal metaplasia (GIM), who were graded by the OLGA and OLGIM staging systems, respectively. Each individual underwent serum pepsinogen (PG) test, H. pylori detection and questionnaire investigation. We did a real-world retrospective follow-up survey for them in April 2022. RESULTS Compared with H. pylori-negative patients, H. pylori-positive patients had higher serum PGs/gastrin-17 (G-17) levels and lower PGR levels, regardless of OLGA/OLGIM stages I-II or III-IV. Furthermore, eight patients with atrophic gastritis who progressed to GC were previously in OLGA stages III-IV and OLGIM stages II-IV. The average duration of this process was 2.19±1.03 years. Logistic regression analysis indicated that PGI and H. pylori infection were independent risk factors of individuals with OLGA stages III-IV. Age and PGR were independent risk factors of patients with OLGIM stages III-IV. PGI and PGR had good clinical diagnostic values for OLGA stages III-IV and OLGIM stages III-IV, respectively. CONCLUSION Patients with OLGA/OLGIM stages III-IV should undergo endoscopic surveillance regardless of H. pylori infection. H. pylori-positive patients with OLGIM stage II also have a high risk of GC. H. pylori combined with PGI and PGR is helpful to evaluate the severity of chronic gastritis.
Collapse
Affiliation(s)
- Min Wu
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Anhui, People’s Republic of China
| | - Shuo Feng
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Anhui, People’s Republic of China
| | - Meng Qian
- Graduate School of Bengbu Medical College, Anhui, People’s Republic of China
| | - Song Wang
- Department of Gastroenterology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, People’s Republic of China
| | - Kaiguang Zhang
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Anhui, People’s Republic of China
| |
Collapse
|
85
|
Hua Z, Shen R, Lu B, Li M, Zhou P, Wu J, Dong W, Zhou Q, Zhang J. Weifuchun alters tongue flora and decreases serum trefoil factor I levels in gastric intestinal metaplasia: A CONSORT-compliant article. Medicine (Baltimore) 2022; 101:e31407. [PMID: 36397419 PMCID: PMC9666156 DOI: 10.1097/md.0000000000031407] [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: 02/02/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To explore the molecular mechanisms of Weifuchun in the treatment of gastric intestinal metaplasia (GIM), we designed a preclinical pilot study to examine potential markers of disease progression based on alterations in the tongue flora. METHODS Total 27 patients with GIM were treated with Weifuchun for 4 weeks and 26 volunteers as controls. Tongue coating bacteria were profiled using 16S rDNA high-throughput sequencing. Serum pepsinogen I and II levels were detected using the latex immunoturbidimetric assay. The levels of serum trefoil factor I was detected by ELISA. Microplate-based quantification was used to detect serum total bile acid (TBA). RESULTS After treatment, the relative abundance of 4 dominant tongue coating genera (Granulicatella, Gemella, Lachnoanaerobaculum, and Neisseria) increased significantly wheras Alloprevotella, [Eubacterium] nodatum group, Prevotell, and Ruminococcaceae UCG-014 decreased (P < .05). The results showed that Alloprevotella and 3 rare tongue coating genera (Lautropia, Treponema 2, and Aliihoeflea) might be potential markers or target flora for the treatment of GIM. Kyoto encyclopedia of genes and genomes (KEGG) function prediction analysis showed that Weifuchun may regulate bile secretion and folate biosynthesis in patients with GIM. The level of serum trefoil factor I decreased significantly in response to Weifuchun treatment, which was consistent with the decrease in folate biosynthesis predicted by KEGG. CONCLUSION Weifuchun may restore the balance of tongue flora by decreasing the levels of serum trefoil factor I, thereby providing a new way to measuring the underlying effectiveness and potential mechanisms of action of this traditional Chinese medicinal compound in the treatment of GIM.
Collapse
Affiliation(s)
- Zhaolai Hua
- Institute of Tumor Prevention and Control, People’s Hospital of Yangzhong City, Yangzhong, China
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
| | - Rui Shen
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medical, Nanjing, China
| | - Bin Lu
- Department of Oncology, People’s Hospital of Yangzhong City, Yangzhong, China
| | - Meifeng Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medical, Nanjing, China
| | - Ping Zhou
- Institute of Tumor Prevention and Control, People’s Hospital of Yangzhong City, Yangzhong, China
| | - Juan Wu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medical, Nanjing, China
| | - Wei Dong
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medical, Nanjing, China
| | - Qihai Zhou
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
| | - Junfeng Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medical, Nanjing, China
| |
Collapse
|
86
|
Diagnostic performances of pepsinogens and gastrin-17 for atrophic gastritis and gastric cancer in Mongolian subjects. PLoS One 2022; 17:e0274938. [PMID: 36251649 PMCID: PMC9576095 DOI: 10.1371/journal.pone.0274938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/08/2022] [Indexed: 11/06/2022] Open
Abstract
In Mongolia, gastric cancer morbidity and mortality are high, and more than 80 percent of cases are diagnosed at an advanced stage. This study aimed to evaluate pepsinogens (PGIs) and gastrin-17 (G-17) levels and to determine the diagnostic performances for gastric cancer and chronic atrophic gastritis among Mongolian individuals. We enrolled a total of 120 subjects, including gastric cancer (40), atrophic gastritis (40), and healthy control (40), matched by age (±2) and sex. Pepsinogen I (PGI), Pepsinogen II (PGII), G-17, and H. pylori IgG levels were measured using GastroPanel ELISA kit (Biohit, Helsinki, Finland). Also, PGI to PGII ratio (PGR) was calculated. For atrophic gastritis, when the optimal cut-off value of PGI was ≤75.07 ng/ml, the sensitivity and specificity were 75% and 50%, respectively; when the optimal cut-off value of PGR was ≤6.25, sensitivity and specificity were 85% and 44.7%, respectively. For gastric cancer, when the optimal cut-off value of PGI was ≤35.25 ng/ml, the sensitivity and specificity were 47.2% and 86.8%, respectively; when the optimal cut-off value of PGR was ≤5.27, sensitivity and specificity were 75% and 60.5%, respectively. Combinations of biomarkers with risk factors could improve diagnostic accuracy (AUC for atrophic gastritis 74.8, 95% CI 64.0–85.7, p<0.001; AUC for gastric cancer 75.5, 95% CI 64.2–86.8, p<0.001). PGI, PGR biomarkers combined with the risk of age, family history of gastric cancer, and previous gastric disease could not be an alternative test for upper endoscopy but might be a supportive method which is identifying individuals at medium- and high risk of gastric cancer and precancerous lesions who may need upper endoscopy.
Collapse
|
87
|
Baranovsky AY, Tsvetkova TL. The stomach cancer prognosis map is the basis for the formation of a register of patients with precancerous diseases. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2022:39-45. [DOI: 10.31146/1682-8658-ecg-205-9-39-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
A single-stage retrospective observational comparative study was conducted to find the most significant risk factors for stomach cancer. The analysis of 36 risk factors for stomach cancer in 143 patients aged 32 to 83 years, indigenous residents of cities, regions and republics of the Northwestern Federal District of Russia who underwent complex, including surgical treatment of this disease. The control group consisted of 128 people who underwent in-depth medical examination in the amount necessary for the program of this study. The ranking of the studied risk factors for gastric cancer according to their degree of prognostic significance is presented as follows: the presence of precancerous diseases accompanied by progressive atrophy of the gastric mucosa, intestinal metaplasia and/or dysplasia in combination with prolonged gastric helicobacteriosis; a decrease in the blood content of pepsinogen I and stimulated gastrin-17, as well as a decrease in the ratio of PG I/PG II; prolonged presence of anemia, leukopenia, neutropenia, lymphopenia, thrombocytopenia, especially in men over 50 years of age with bad habits (smoking, alcohol abuse); presence in patients with diabetes mellitus and obesity, especially when combined with them and under the condition of an increased index of glycosed hemoglobin; gross and multiple disorders of the rhythm of nutrition and the content of the diet. Using the obtained data, a mathematical assessment of the prognostic significance of each of the studied signs can be carried out, an algorithm for predicting stomach cancer and making individualized medical decisions is developed, without which it is impossible to create an effective and convenient register of patients with precancerous diseases at all stages of medical care for organizing and conducting personalized and effective cancer prevention measures.
Collapse
Affiliation(s)
- A. Yu. Baranovsky
- Federal State Budgetary Educational Institution of Higher Education “Saint Petersburg State University”
| | | |
Collapse
|
88
|
Baranovskiy AY, Tcvetkova TL. Risk factors of gastric cancer as a basis for the development of a prognostic questionnaire for the register of patients with precancerous gastroduodenal diseases. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2022:29-38. [DOI: 10.31146/1682-8658-ecg-205-9-29-38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The article is a critical analysis of the world scientific literature devoted to the search for risk factors for stomach cancer for the timely prognosis of this disease and the implementation of cancer prevention measures. The paper presents data from numerous studies to determine the role of environmental factors, including unfavorable ecology, as well as gender, age, smoking, alcohol abuse. The authors’ opinions are presented on the essential role of the alimentary factor in the genesis of neoplasms in the stomach, including the predominance of animal fats in food, the abuse of overcooked, pickled foods rich in nitrosoamines, foods saturated with spices, the use of too hot food, the use of foods infected with mycotoxins in nutrition. The role of environmental factors in the prognosis of gastric cancer is noted: the state of secretory activity of the stomach, the dynamics of inflammatory and atrophic processes in the mucous membrane. A special role for the prognosis of stomach cancer is assigned by many authors to the pyloric helicobacter, as well as the quantitative indicator of glycated blood hemoglobin and its dynamics. The significance of genetic changes in the genesis of gastric cancer and their role as prognostic factors of the disease is ambiguous. The article draws attention to the multidirectional results of many authors in understanding a large number of factors they have studied that could be used as prognostic witnesses of stomach cancer. The expediency of searching for the most significant regional factors for the prognosis of gastric cancer is substantiated, on the basis of which it is very important to create registers of patients with precancerous diseases of the stomach for the organization and implementation of personalized and effective measures of cancer prevention.
Collapse
|
89
|
Shen L, Zhang D, Huang Y, Liu L, Zheng Y, Zhang L, Lu D, Cai J, Zhu R, Sun H, Shi M, Ni Y, Zhang J. Initial screening of gastric cancer using oral contrast-enhanced trans-abdominal ultrasonography in rural asymptomatic individuals. Cancer Epidemiol 2022; 80:102236. [PMID: 35939867 DOI: 10.1016/j.canep.2022.102236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/27/2022] [Accepted: 07/30/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND & AIMS Initial screening for high-risk population of gastric cancer (GC) is needed in rural areas of large-population countries. This study aims to explore the feasibility of applying noninvasive ultrasonography as an initial screening strategy to improve the early diagnosis and prevention of GC. METHODS Oral contrast-enhanced trans-abdominal ultrasonography (OCTU) was initially applied to screen around 15,000 residents from 24 different rural villages of Changxing Island in Shanghai, China, facilitating the identification of high-risk population for further endoscopy examination. RESULTS 176 subjects (1.18 %) were initially identified with gastric diseases using OCTU while 14,787 ones (98.93 %) were normal with negative results. 145 out of 176 individuals (82.39 %) took further endoscopy examination, and 16 were diagnosed with GC with biopsy examination, with 9 of them at the early stage. We followed up with the Center for Disease Control and Prevention, and identified another 6 GC cases occurred within one year among OCTU-negative population, serving as an adjustment factor for sensitivity analysis. As a result, with a total of 22 GC cases included in this cohort, the positive predictive rate, the negative predictive value, sensitivity, and specificity were 9.09%, 99.96 %, 75.5 %, and 98.93 %, respectively. CONCLUSIONS OCTU is feasible, non-invasive, low-cost, and widely acceptable in rural area, thus we proposed that OCTU is practicable to serve as a supplementary screening method to improve the early detection of GC in rural area of China and other developing countries with large population.
Collapse
Affiliation(s)
- Li Shen
- Department of Ultrasonography, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, No. 25 Nanmen Road, Chongming, Shanghai 202150, China
| | - Danni Zhang
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yaoping Huang
- Community Health Service Center, No. 59 Fengnan Road, Changxing town, Chongming district, Shanghai 201913, China
| | - Lan Liu
- Department of Ultrasonography, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, No. 25 Nanmen Road, Chongming, Shanghai 202150, China
| | - Yin Zheng
- Department of Cancer prevention, Fudan University Shanghai Cancer Center, No. 270 Dongan Road, Shanghai 200032, China
| | - Li Zhang
- Department of Ultrasonography, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, No. 25 Nanmen Road, Chongming, Shanghai 202150, China
| | - Dianyuan Lu
- Department of Ultrasonography, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, No. 25 Nanmen Road, Chongming, Shanghai 202150, China
| | - Jianrong Cai
- Department of Ultrasonography, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, No. 25 Nanmen Road, Chongming, Shanghai 202150, China
| | - Rongrong Zhu
- Department of Ultrasonography, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, No. 25 Nanmen Road, Chongming, Shanghai 202150, China
| | - Huixiang Sun
- Community Health Service Center, No. 59 Fengnan Road, Changxing town, Chongming district, Shanghai 201913, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai 200025, China
| | - Yan Ni
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai 200025, China.
| |
Collapse
|
90
|
Wu D, Shi A, Wang H, Yu X. Panel of serum biomarkers (GastroPanel) in diagnosis of atrophic gastritis and Helicobacter pylori infection: a protocol of systematic review and meta-analysis. BMJ Open 2022; 12:e062849. [PMID: 36171026 PMCID: PMC9528581 DOI: 10.1136/bmjopen-2022-062849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION The aetiology of gastric cancer is still unclear but Helicobacter pylori (HP) infection and chronic atrophic gastritis (AG) are recognised as two major risk factors for gastric cancer. GastroPanel (GP) test is the first non-invasive diagnostic tool to detect AG and HP infection.The aim of the study is to conduct a systematic review and meta-analysis to review published literature about the GP test for diagnosing AG and HP infection, with the objective of estimating the diagnostic performance indices of GP for AG and HP infection. METHODS AND ANALYSIS This protocol of systematic review and meta-analysis is reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols statement guidelines. PubMed, Embase, Web of Science and Cochrane Library databases will be systematically searched from inception to March 2022 for eligible studies. No language limitations were imposed. The studies will be downloaded into the EndNote V.X9 software and duplicates will be removed. Two review authors independently screened the full text against the inclusion criteria, extracted the data from each included study by using a piloted data extraction form and conducted risk of bias assessment, resolving disagreement by discussion. Results will be synthesised narratively in summary tables, using a random-effect bivariate model, and we fit a hierarchical summary receiver operating characteristic curve. ETHICS AND DISSEMINATION This systematic review will include data extracted form published studies, therefore, does not require ethics approval. The results of this study will be submitted to a peer-reviewed journal. PROSPERO REGISTRATION NUMBER CRD42021282616.
Collapse
Affiliation(s)
- Dan Wu
- Clinical Laboratory, People's Hospital of Xinjin District Chengdu (Emergency Command Sub-center of Xinjin District, Chengdu), Chengdu, Sichuan, China
| | - Anya Shi
- The Second Clinical Medical Hospital, Lanzhou University, Lanzhou, Gansu, China
| | - Haiping Wang
- Gansu Provincial Key Laboratory of Biotherapy and Regenerative Medicine, The first hospital of Lanzhou university, Lanzhou, Gansu, China
- The First Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Xiuzhong Yu
- Clinical Laboratory, People's Hospital of Xinjin District Chengdu (Emergency Command Sub-center of Xinjin District, Chengdu), Chengdu, Sichuan, China
| |
Collapse
|
91
|
Lai Y, Wei W, Du Y, Gao J, Li Z. Biomaterials for Helicobacter pylori therapy: therapeutic potential and future perspectives. Gut Microbes 2022; 14:2120747. [PMID: 36070564 PMCID: PMC9467593 DOI: 10.1080/19490976.2022.2120747] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Helicobacter pylori (H. pylori) is the main cause of gastric adenocarcinoma. However, the traditional antibiotic treatment of H. pylori is limited due to increased antibiotic resistance and low efficacy; low drug delivery efficiency and difficulties in eradicating H. pylori that is present intracellularly or in biofilms cause further setbacks. Biomaterials that can protect drugs against stomach acid, target lesions, control drug release, destroy biofilms, and exhibit unique antibacterial mechanisms and excellent biocompatibility have emerged as attractive tools for H. pylori eradication, particularly for drug-resistant strains. Herein, we review the virulence mechanisms, current drug treatments, and antibiotic resistance of H. pylori strains. Furthermore, recent advances in the development of biomaterials, including nanoparticles (such as lipid-based nanoparticles, polymeric nanoparticles, and inorganic nanoparticles), microspheres, and hydrogels, for effective and precise therapy of H. pylori and different types of therapeutic mechanisms, as well as future perspectives, have also been summarized.
Collapse
Affiliation(s)
- Yongkang Lai
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China,Department of Gastroenterology, Ganzhou People’s Hospital Affiliated to Nanchang University, Ganzhou, China
| | - Wei Wei
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yiqi Du
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China,Jie Gao Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zhaoshen Li
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China,CONTACT Zhaoshen Li Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
92
|
Endoscopic methods for the detection and treatment of gastric cancer. Curr Opin Gastroenterol 2022; 38:436-442. [PMID: 35881962 DOI: 10.1097/mog.0000000000000867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
PURPOSE OF REVIEW The intent of this review is to describe new advances in endoscopic approaches to surveillance and management of gastric cancer. RECENT FINDINGS There are new endoscopic techniques and approaches that have improved the detection of gastric cancer, including narrow band imaging, confocal laser endocytomicroscopy and magnetically controlled capsule endoscopy. This article highlights the role of endoscopic submucosal dissection in the treatment of focal and diffuse gastric dysplasia and early gastric cancer with a discussion of indications, complications and outcomes. We review several recent guidelines addressing the surveillance strategies for individuals at high-risk for developing gastric cancer, such as those with atrophic gastritis and intestinal metaplasia, how gastric dysplasia and early gastric cancer can be endoscopically managed, and recommended surveillance after endoscopic intervention. SUMMARY Endoscopic approaches are evolving rapidly that will improve detection of dysplasia and early gastric cancer in high-risk individuals. Surveillance guidelines from various international societies reflect differences in local experience and prevalence of gastric cancer. Endoscopic submucosal dissection is now widely accepted as a first-line approach to early gastric cancers that can be resected en-bloc .
Collapse
|
93
|
Zhou JP, Liu CH, Liu BW, Wang YJ, Benghezal M, Marshall BJ, Tang H, Li H. Association of serum pepsinogens and gastrin-17 with Helicobacter pylori infection assessed by urea breath test. Front Cell Infect Microbiol 2022; 12:980399. [PMID: 36051244 PMCID: PMC9425458 DOI: 10.3389/fcimb.2022.980399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background Association of gastric atrophy or cancer with levels of serum pepsinogens, gastrin-17 and anti-Helicobacter pylori IgG antibody have been extensively studied. However, the association of serum pepsinogen and gastrin-17 with H. pylori infection has not been studied in a large population. Aim To investigate the impact of H. pylori infection on serum levels of pepsinogens and gastrin-17. Methods A total of 354, 972 subjects who underwent health check-ups were included. Serum levels of pepsinogens and gastrin-17 were measured using the enzyme-linked immunosorbent assay. H. pylori infection was detected using 14C-urea breath test (UBT). Multivariable logistic regression analysis was used to investigate the association of serum pepsinogen and gastrin-17 with H. pylori infection. Results H. pylori prevalence was 33.18% in this study. The mean levels of pepsinogens and gastrin-17 were higher, while the mean pepsinogen-I/II ratio were lower among H. pylori-positive than -negative subjects. In H. pylori-positive subjects, pepsinogen and gastrin-17 levels correlated positively, whereas the pepsinogen-I/II ratio correlated negatively with UBT values (e.g., the mean serum level of pepsinogen-I in subjects with UBT values in the range of 100-499dpm, 500-1499dpm, and ≥1500dpm was 94.77 ± 38.99, 102.77 ± 43.59, and 111.53 ± 47.47 ng/mL, respectively). Compared with H. pylori-negative subjects, the adjusted odds ratio (aOR) of having pepsinogen-I ≤ 70 ng/mL in the three H. pylori-positive but with different UBT value groups was 0.31 (p<0.001), 0.16 (p<0.001), and 0.08 (p<0.001), respectively; while the aOR of having G-17>5.70 pmol/L was 4.56 (p<0.001), 7.43 (p<0.001), and 7.12 (p<0.001). This suggested that H. pylori-positive subjects with higher UBT values were less likely to have pepsinogen-I ≤70 ng/mL (a serum marker for gastric atrophy), but more likely to have gastrin-17 >5.70 pmol/L (a marker for peptic ulcer). Conclusions H. pylori-positive subjects with higher UBT values are unlikely to have gastric atrophy, but may have greater risk of severe gastritis or peptic ulcers. Our study suggests that H. pylori-positive patients with high UBT values may benefit the most from H. pylori eradication.
Collapse
Affiliation(s)
- Jun-peng Zhou
- West China Marshall Research Center for Infectious Diseases, Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-hai Liu
- West China Marshall Research Center for Infectious Diseases, Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Bo-wen Liu
- Dental Department, 363 Hospital, Chengdu, China
| | - You-juan Wang
- Health Management Center, West China Hospital of Sichuan University, Chengdu, China
| | - Mohammed Benghezal
- West China Marshall Research Center for Infectious Diseases, Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Barry James Marshall
- West China Marshall Research Center for Infectious Diseases, Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Helicobacter pylori Research Laboratory, School of Biomedical Sciences, Marshall Centre for Infectious Disease Research and Training, University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Engineering, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Hong Tang
- West China Marshall Research Center for Infectious Diseases, Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Hong Li, ; Hong Tang,
| | - Hong Li
- West China Marshall Research Center for Infectious Diseases, Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Hong Li, ; Hong Tang,
| |
Collapse
|
94
|
Liu D, Zhang R, Chen S, Sun B, Zhang K. Analysis of gastric microbiome reveals three distinctive microbial communities associated with the occurrence of gastric cancer. BMC Microbiol 2022; 22:184. [PMID: 35870901 PMCID: PMC9308235 DOI: 10.1186/s12866-022-02594-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/13/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Gastric microbial dysbiosis were reported to be associated with gastric cancer (GC). This study aimed to explore the variation, diversity, and composition patterns of gastric bacteria in stages of gastric carcinogenesis based on the published datasets. METHODS We conducted a gastric microbial analysis using 10 public datasets based on 16S rRNA sequencing, including 1270 gastric biopsies of 109 health control, 183 superficial gastritis (SG), 135 atrophic gastritis (AG), 124 intestinal metaplasia (IM), 94 intraepithelial neoplasia (IN), 344 GC, and 281 adjacent normal tissues. And QIIME2-pipeline, DESeq2, NetMoss2, vegan, igraph, and RandomForest were used for the data processing and analysis. RESULTS We identified three gastric microbial communities among all the gastric tissues. The first community (designate as GT-H) was featured by the high abundance of Helicobacter. The other two microbial communities, namely GT-F, and GT-P, were featured by the enrichment of phylum Firmicutes and Proteobacteria, respectively. The distribution of GC-associated bacteria, such as Fusobacterium, Peptostreptococcus, Streptococcus, and Veillonella were enriched in tumor tissues, and mainly distributed in GT-F type microbial communities. Compared with SG, AG, and IM, the bacterial diversity in GC was significantly reduced. And the strength of microbial interaction networks was initially increased in IM but gradually decreased from IN to GC. In addition, Randomforest models constructed in in GT-H and GT-F microbial communities showed excellent performance in distinguishing GC from SG and precancerous stages, with varied donated bacteria. CONCLUSIONS This study identified three types of gastric microbiome with different patterns of composition which helps to clarify the potential key bacteria in the development of gastric carcinogenesis.
Collapse
Affiliation(s)
- Dehua Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Rutong Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Si Chen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Baolin Sun
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Kaiguang Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
95
|
Qin S, Wang X, Li S, Tan C, Zeng X, Luo X, Yi L, Peng L, Wu M, Peng Y, Wang L, Wan X. Clinical Benefit and Cost Effectiveness of Risk-Stratified Gastric Cancer Screening Strategies in China: A Modeling Study. PHARMACOECONOMICS 2022; 40:725-737. [PMID: 35701687 DOI: 10.1007/s40273-022-01160-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE A new gastric cancer screening scoring system (NGCS) strategy was recommended for the early gastric cancer (GC) screening process in China. The current study aimed to assess the clinical benefits and the cost effectiveness of the NGCS strategy in GC high-risk areas of China from a societal perspective. METHODS A Markov microsimulation model was developed to evaluate 30 alternative screening strategies with varying initiation age, including the NGCS strategy, the modified NGCS strategy, and the endoscopic screening strategy with various screening intervals. The primary outcomes included GC mortality, number of endoscopies, quality-adjusted life-years (QALYs), costs, and incremental cost-effectiveness ratios (ICERs). Cost estimates were reported in 2021 USD (US$) and both costs and benefits were discounted at 5% annually. Deterministic and probabilistic sensitivity analyses were performed to evaluate model uncertainty. RESULTS Screening with the NGCS strategy from age 40 years (40-NGCS) reduced the GC incidence by 86.4%, which provided the greatest benefit across strategies. Compared with all strategies, at a willingness-to pay threshold of US$17,922 per QALY, the 40-NGCS strategy was a leading cost-effective strategy, with an ICER of US$15,668 per QALY. Results were robust in univariate and probabilistic sensitivity analyses. The probability of the 40-NGCS strategy being cost effective was 0.863. CONCLUSIONS The 40-NGCS strategy was an effective and cost-effective strategy to reduce GC incidence and mortality in China. The findings provide important evidence for decision makers to formulate and optimize targeted approaches for GC prevention and control policies in China.
Collapse
Affiliation(s)
- Shuxia Qin
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xuehong Wang
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Sini Li
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China
- Faculty of Medicine, Dentistry and Health, School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - Chongqing Tan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiaohui Zeng
- PET-CT Center, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xia Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lidan Yi
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Liubao Peng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Meiyu Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ye Peng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Liting Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiaomin Wan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
96
|
Zhou MJ, Huang RJ. Catching Up with the World: Pepsinogen Screening for Gastric Cancer in the United States. Cancer Epidemiol Biomarkers Prev 2022; 31:1257-1258. [PMID: 35775231 DOI: 10.1158/1055-9965.epi-22-0372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 01/15/2023] Open
Abstract
Gastric cancer remains a deadly cancer with poor outcomes in the United States. There is a need for screening strategies for gastric cancer in the U.S. population. With progressive Helicobacter pylori-mediated inflammation of the gastric mucosa, pepsinogen I levels decrease and the pepsinogen I/II ratio decreases. Pepsinogen test positivity (PG+) has been evaluated as a promising screening test among Asian and European populations; however, its utility in multiethnic U.S. populations is poorly described. In this case-control study nested within the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial, In and colleagues evaluate the discrimination of PG+ in serum collected from individuals prior to the development of gastric cancer. The authors find that PG+ individuals were at nearly 10-fold increased risk for developing gastric cancer, and this effect remained robust after adjusting for Helicobacter pylori status, family history, education, smoking, and obesity. In subgroup analysis, the predictive ability of the test was particularly robust for noncardia gastric cancers, and nonpredictive of cardia gastric cancers. Serum pepsinogen testing holds promise as a noninvasive screening strategy to triage individuals at heightened risk for gastric cancer, and may help to improve early diagnosis in the United States. See related article by In et al., p. 1426.
Collapse
Affiliation(s)
- Margaret J Zhou
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Redwood City, California
| | - Robert J Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Redwood City, California
| |
Collapse
|
97
|
Clinical applicability of a new scoring system for population-based screening and risk factors of gastric cancer in the Wannan region. BMC Gastroenterol 2022; 22:306. [PMID: 35739473 PMCID: PMC9219187 DOI: 10.1186/s12876-022-02384-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background We aimed to evaluate the clinical applicability of a new scoring system that comprises the variables age, sex, pepsinogen ratio (PGR), gastrin-17 (G-17), and Helicobacter pylori (Hp) infection for gastric cancer (GC) screening in the Wannan region, China. We also explored the risk factors of GC in the Wannan region. Methods We prospectively enrolled asymptomatic participants from January 1, 2019 to June 30, 2021 at the First Affiliated Hospital of Wannan Medical College. We used a receiver operating characteristic (ROC) curve to estimate the screening value of combined measurements of pepsinogen I, PGII, PGR, G-17, and Hp. Univariate analysis and multivariate analysis were used to explore the independent risk factors of GC. Results A total of 25,194 asymptomatic patients were eventually screened. The area under the ROC curve (AUC) of combined measurements was 0.817 (95% confidence interval [CI] 0.721–0.913), the sensitivity was 81.5%, and the specificity was 77.8%. The detection rate of this new scoring system for GC screening in low-, medium-, and high-risk groups was 0%, 1.63%, and 9%, respectively (P < 0.001). Multivariate analysis showed that age (odds ratio [OR], 5.934; 95% CI 3.695–9.529; P < 0.001), sex (OR 5.721; 95% CI 2.579–12.695; P < 0.001), Hp infection (OR 1.992; 95% CI 1.255–3.163; P = 0.003), a history of smoking (OR 2.028; 95% CI 1.213–3.392; P = 0.007), consuming a high-salt diet (OR 2.877; 95% CI 1.807–4.580; P < 0.001), frequently eating pickled foods (OR 1.873; 95% CI 1.125–3.120; P = 0.016), and frequently eating fried foods (OR 2.459; 95% CI 1.384–4.369; P = 0.002) were independent risk factors for GC and precancerous lesions. However, frequent consumption of green vegetables (OR 0.388; 95% CI 0.242–0.620; P < 0.001) was an independent protective factor against GC and precancerous lesions. Conclusion The new scoring system for GC screening was feasible in the Wannan region, especially in high-risk populations. Frequent consumption of green vegetables was an independent protective factor against GC and precancerous lesions. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02384-w.
Collapse
|
98
|
Pei B, Wen Z, Yang Q, Wang J, Cao Q, Dai L, Li X. Risk Factors Analysis and Prediction Model Establishment of Intestinal Metaplasia or Dysplasia in Patients With Chronic Atrophic Gastritis: A Multi-Center Retrospective Study. Front Med (Lausanne) 2022; 9:912331. [PMID: 35665336 PMCID: PMC9157492 DOI: 10.3389/fmed.2022.912331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To investigate the risk factors and construct a prediction model of chronic atrophic gastritis (CAG) patients with intestinal metaplasia or dysplasia. Method The clinical data of 450 patients with CAG who were diagnosed and treated in the Department of Gastroenterology of the Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine from June 2016 to February 2022 were collected. Single and multiple factors logistic regression analysis were used to explore the risk factors of intestinal metaplasia or dysplasia in patients of training cohort. Then, we constructed a model to predict the onset of intestinal metaplasia or dysplasia based on the data of training cohort, following which we tested the model in an external validation cohort of 193 patients from a local university teaching hospital. The ROC curve, calibration curve, and decision curve analysis were used to evaluate the accuracy of the prediction model. Result Helicobacter pylori (H. pylori, HP) infection, pepsinogen I, gastrin-17, and the number of lesions were found to be independent rick factors of the model. The liner prediction model showed excellent predictive value in both training cohort and validation cohort. Conclusion HP infection, pepsinogen I, gastrin-17, and the number of lesions are independent risk factors for intestinal metaplasia or dysplasia in patients with CAG. The prediction model constructed based on these factors has a high accuracy and excellent calibration, which can provide a great basis for condition assessment and individualized treatment of the patients.
Collapse
Affiliation(s)
- Bei Pei
- The Graduated School, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Ziang Wen
- The Graduated School, Anhui Medical University, Hefei, China
| | - Qi Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Jieyu Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Qinglin Cao
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Longfei Dai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuejun Li
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
99
|
Sun D, Lei L, Xia C, Li H, Cao M, He S, Zhang Z, Guo G, Song G, Peng J, Chen W. Sociodemographic disparities in gastric cancer and the gastric precancerous cascade: A population-based study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 23:100437. [PMID: 35355616 PMCID: PMC8958536 DOI: 10.1016/j.lanwpc.2022.100437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
BACKGROUND Gastric carcinogenesis is a multistep process initiating with chronic gastritis and progressing through atrophy, intestinal metaplasia, and dysplasia to carcinoma. This study aims to comprehensively investigate sociodemographic disparities in each stage of gastric carcinogenesis and estimate to what extent the inequalities could be ascribed to risk factors of gastric cancer (GC). METHODS We used the baseline data from a community-based study in China's high-risk areas, totalling 27094 participants. Gastric mucosa status was ascertained by endoscopy and biopsies. An overall socioeconomic status (SES) variable was generated by latent class analysis. We calculated relative risks (RRs) and 95% confidence intervals (CIs) using modified Poisson regression to assess associations of sociodemographic factors with each cascade stage. We estimated the percentage of the excess risk for neoplastic lesions among vulnerable populations that can be explained by established risk factors. RESULTS Age and sex showed associations with all gastric lesions, whose RRs increased with lesion progressing. Compared with individuals without schooling, the RRs of neoplastic lesions for people with primary, secondary, and post-secondary education were 0·86 (95% CI 0·76-0·97), 1·00 (95% CI 0·88-1·13), and 0·70 (95% CI 0·47-1·03), respectively. Participants with medium SES had a lower risk of neoplastic lesions than people in the low SES group (RR 0·83, 95% CI 0·74-0·93). GC risk factors could explain 33·6% of the excess risk of neoplastic lesions among men and a small proportion of the disparities among SES groups. INTERPRETATION Age and sex were essential sociodemographic factors for GC and precursor diseases. Individuals with low educational levels or SES were more likely to have neoplastic lesions. About one-third of the sex difference and a slight fraction of the socioeconomic inequalities could be attributed to included risk factors. FUNDING Sanming Project of Medicine in Shenzhen, National Natural Science Foundation, and Special Project of Bejing-Tianjin-Hebei Basic Research Cooperation.
Collapse
Affiliation(s)
- Dianqin Sun
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Pan-jia-yuan South Lane, Chaoyang District, Beijing 100021, China
| | - Lin Lei
- Department of Cancer Prevention and Control, Shenzhen Center for Chronic Disease Control, Shenzhen 518020, China
| | - Changfa Xia
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Pan-jia-yuan South Lane, Chaoyang District, Beijing 100021, China
| | - He Li
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Pan-jia-yuan South Lane, Chaoyang District, Beijing 100021, China
| | - Maomao Cao
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Pan-jia-yuan South Lane, Chaoyang District, Beijing 100021, China
| | - Siyi He
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Pan-jia-yuan South Lane, Chaoyang District, Beijing 100021, China
| | - Zhiyi Zhang
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital, Wuwei, China
| | - Guizhou Guo
- Linzhou Cancer Hospital, Anyang 456500, China
| | - Guohui Song
- Cixian Cancer Institute, Handan 056500, China
| | - Ji Peng
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital, Wuwei, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Pan-jia-yuan South Lane, Chaoyang District, Beijing 100021, China
- Corresponding author.
| |
Collapse
|
100
|
Gong Y, Kang J, Wu R, Ge F, Zheng YS, Zeng Q. Gastroscopic results for the asymptomatic, average-risk population in Northern China: a cross-sectional study of 60,519 adults. Scand J Gastroenterol 2022; 57:748-756. [PMID: 35132926 DOI: 10.1080/00365521.2022.2035810] [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: 10/24/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Studies on average-risk individuals undergoing gastroscopy screening in China are scarce. OBJECTIVE To determine and compare the prevalence of lesions found by gastroscopy and the association between sex, age, Helicobacter pylori infection and gastric premalignant lesions. METHODS Gastroscopy results were analysed for 60,519 individuals enrolled from January 2013 to December 2019. RESULTS The median age was 49.84 years (SD, 9.47 years) for women and 48.90 years (SD, 8.82 years) for men, and the ratio of females to males was 35.10% (n = 21,240) to 64.90% (n = 39,279). The most common lesions detected by endoscopy were chronic gastritis, reflux oesophagitis, duodenitis and gastric polyps, detected in 24.48%, 10.28%, 3.96% and 3.61%, respectively. Oesophageal cancer and gastric cancer were detected in 0.33% and 0.47% of patients, respectively. The prevalence of chronic gastritis increased with age and was higher in males than in females (26.47% [n = 10396] versus 20.80% [n = 4417], p < .001). The prevalence of gastric ulcers was highest in the elderly group, and the H. pylori infection rate of gastric ulcer patients was 47.28%. The prevalence of gastric polyps was higher in females than in males (5.47% [n = 1161] versus 2.61% [n = 1024], p < .001), and the H. pylori infection rate in inflammatory polyp patients was higher than that in fundic gland polyp patients (28.32% [n = 442] versus 7.29% [n = 29], p < .001). CONCLUSION The prevalence of upper gastrointestinal endoscopic lesions is high in the asymptomatic population undergoing physical examination and is associated with sex, age, and H. pylori infection.
Collapse
Affiliation(s)
- Yan Gong
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Juan Kang
- Department of Emergency, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Rilige Wu
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, China
| | - Fulin Ge
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Yan-Song Zheng
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Qiang Zeng
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|