1
|
Wang SN, Wang YK, Jiang B, Bu JX, Li YY. Computer analysis of abnormal proliferation and transformation cells in gastric mucosa and its clinical significance. Biotechnol Genet Eng Rev 2024; 40:1998-2011. [PMID: 37067362 DOI: 10.1080/02648725.2023.2197382] [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: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 04/18/2023]
Abstract
To investigate the image computer analysis of abnormally proliferating transformed cells of gastric mucosa and its clinical significance. The pathological pictures of gastric adenomatous polyp cells, abnormally proliferating altered cells, and tubular adenocarcinoma cells in the stomach mucosa were assessed by image computer method on a total of 96 gastroscopic biopsy and ESD resection specimens. The data of cytoplasmic area, nuclear area, nuclear-cytoplasmic ratio, nuclear factor and N-heterotypic index of gastric adenomatous polyps, abnormal proliferative transformation and gastric intramucosal tubular adenocarcinoma were collected, and the mean, standard deviation and variance were calculated respectively. Standard Error, Maximum, Minimum Parameters and Statistical Structure. There were substantial discrepancies between gastric mucosal gastric adenomatous polyp cells and gastric mucosal abnormally proliferating transformed cells, according to the five data in the abnormal cells in the stomach mucosal proliferation area (p < 0.01); There was no significant difference between cells (p > 0.05). Computer analysis of cell images can provide quantitative values for the pathological diagnosis of gastric adenomatous polyp cells, abnormally proliferating transformed cells and tubular adenocarcinoma cells in the gastric mucosa, especially the degree of atypical proliferation. The monitoring of abnormally proliferated and transformed cells in gastric mucosa is of great significance for clinicians to accurately treat and track cell transformation, and to control the occurrence and development of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Su-Nan Wang
- Department of Electronic & Communication Engineering, Shenzhen Polytechnic, shenzhen, China
| | - Yang-Kun Wang
- Department of Pathology, Shenzhen Longgang District Fourth People's Hospital, Shenzhen, China
| | - Bo Jiang
- 990 Hospital of PLA Joint Logistics Support Force, Zhumadian, China
| | - Jian-Xue Bu
- 989 Hospital of PLA Joint Logistics Support Force, Luoyang, China
| | - Ying-Ying Li
- Department of Electronic & Communication Engineering, Shenzhen Polytechnic, shenzhen, China
| |
Collapse
|
2
|
Mamun TI, Younus S, Rahman MH. Gastric cancer-Epidemiology, modifiable and non-modifiable risk factors, challenges and opportunities: An updated review. Cancer Treat Res Commun 2024; 41:100845. [PMID: 39357127 DOI: 10.1016/j.ctarc.2024.100845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Gastric cancer represents a significant global health challenge due to its high mortality and incidence rates, particularly in Eastern Asia, Eastern Europe, and South America. This comprehensive review synthesizes the latest epidemiological data and explores both modifiable and non-modifiable risk factors associated with gastric cancer, aiming to delineate the multifactorial etiology of this disease. Modifiable risk factors include Helicobacter pylori infection, obesity, dietary habits, smoking and alcohol consumption, whereas nonmodifiable factors comprise genetic predispositions, age, family history and male gender. The interplay of these factors significantly impacts the risk and progression of gastric cancer, suggesting potential preventive strategies. The challenges in treating gastric cancer are considerable, largely because of the late-stage diagnosis and the heterogeneity of the disease, which complicate effective treatment regimens. Current treatment strategies involve a combination of surgery, chemotherapy, radiotherapy, and targeted therapies. The FLOT regimen (5-FU, Leucovorin, Oxaliplatin and Docetaxel) is now a standard for resectable cases in Europe and the US, showing superior survival and response rates over ECF and ECX regimens. For HER2-positive gastric cancer, trastuzumab combined with chemotherapy improves overall survival, as demonstrated by the ToGA trial. Additionally, immune checkpoint inhibitors like pembrolizumab and nivolumab offer promising results. However, the five-year survival rate remains low, underscoring the urgency for improved therapeutic approaches. Recent advancements in molecular biology and cancer genomics have begun to pave the way for personalized medicine in gastric cancer care, focusing on molecular targeted therapies and immunotherapy. This review also highlights the critical need for better screening methods that could facilitate early detection and treatment, potentially improving the prognosis. By integrating epidemiological insights with new therapeutic strategies, this article aims to thoroughly understand of gastric cancer's dynamics and outline a framework for future research and clinical management, advocating for a multidisciplinary approach to tackle this formidable disease.
Collapse
Affiliation(s)
- Tajul Islam Mamun
- Department of Epidemiology and Public Health, Sylhet Agricultural University, Sylhet 3100, Bangladesh.
| | - Sabrina Younus
- Department of Pharmacy, University of Chittagong, Chattogram 4331, Bangladesh
| | - Md Hashibur Rahman
- Department of Physiology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| |
Collapse
|
3
|
Puhr HC, Berchtold L, Zingerle L, Felfernig M, Weissenbacher L, Jomrich G, Asari R, Schoppmann SF, Prager GW, Bergen ES, Berghoff AS, Preusser M, Ilhan-Mutlu A. Association of family history with patient characteristics and prognosis in a large European gastroesophageal cancer cohort. Wien Klin Wochenschr 2024:10.1007/s00508-024-02432-3. [PMID: 39235615 DOI: 10.1007/s00508-024-02432-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/10/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The role of the family history in the development and prognosis of gastroesophageal cancer is a controversially discussed topic as appropriate data from western cohorts are lacking. This study aims to explore its associations with disease and outcome parameters in a large European cohort. METHODS We retrospectively analyzed self-reported family history in patients with gastroesophageal cancer treated between 1 January 1990 and 31 December 2021 at the Medical University of Vienna. Association analyses with patient characteristics, tumor characteristics, symptoms and overall survival (OS) were performed. RESULTS In our cohort of 1762 gastroesophageal cancer patients, 592 (34%) reported a positive family history of cancer (159, 9%, gastroesophageal cancer). No associations were found with histopathological parameters or initial symptoms; however, a positive family history correlated with female gender (cancer in general: p = 0.011; gastroesophageal cancer: p = 0.015). Family history of cancer in general was associated with earlier cancer stages (p = 0.04), higher BMI (p = 0.005), and alcohol consumption (p = 0.010), while a positive history for gastroesophageal cancer was associated with higher age at diagnosis (p = 0.002) and stomach cancer (p = 0.002). There was no statistically significant association of positive family history with OS (p = 0.1, p = 0.45), also not in subgroups for histology (adeno and squamous cell), number of family members and degree of relative. CONCLUSION Our results emphasize that a positive family history is neither statistically significantly associated with prognosis nor with specific histopathological features in patients with gastroesophageal cancer. Yet, associations with distinct patient characteristics and positive family history indicate that specific subgroups might profit from endoscopic surveillance. Prospective studies are warranted to investigate these findings further.
Collapse
Affiliation(s)
- Hannah C Puhr
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Luzia Berchtold
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Institute for Medical Statistics, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Linda Zingerle
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Melanie Felfernig
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Lisa Weissenbacher
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Gerd Jomrich
- Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Reza Asari
- Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Sebastian F Schoppmann
- Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Gerald W Prager
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Elisabeth S Bergen
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Anna S Berghoff
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Aysegül Ilhan-Mutlu
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
4
|
Oh H, Cho S, Lee JA, Ryu S, Chang Y. Risk prediction model for gastric cancer within 5 years in healthy Korean adults. Gastric Cancer 2024; 27:675-683. [PMID: 38561527 DOI: 10.1007/s10120-024-01488-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Although endoscopy is commonly used for gastric cancer screening in South Korea, predictive models that integrate endoscopy results are scarce. We aimed to develop a 5-year gastric cancer risk prediction model using endoscopy results as a predictor. METHODS We developed a predictive model using the cohort data of the Kangbuk Samsung Health Study from 2011 to 2019. Among the 260,407 participants aged ≥20 years who did not have any previous history of cancer, 435 cases of gastric cancer were observed. A Cox proportional hazard regression model was used to evaluate the predictors and calculate the 5-year risk of gastric cancer. Harrell's C-statistics and Nam-D'Agostino χ2 test were used to measure the quality of discrimination and calibration ability, respectively. RESULTS We included age, sex, smoking status, alcohol consumption, family history of cancer, and previous results for endoscopy in the risk prediction model. This model showed sufficient discrimination ability [development cohort: C-Statistics: 0.800, 95% confidence interval (CI) 0.770-0.829; validation cohort: C-Statistics: 0.799, 95% CI 0.743-0.856]. It also performed well with effective calibration (development cohort: χ2 = 13.65, P = 0.135; validation cohort: χ2 = 15.57, P = 0.056). CONCLUSION Our prediction model, including young adults, showed good discrimination and calibration. Furthermore, this model considered a fixed time interval of 5 years to predict the risk of developing gastric cancer, considering endoscopic results. Thus, it could be clinically useful, especially for adults with endoscopic results.
Collapse
Affiliation(s)
- Hyungseok Oh
- Workplace Health Institute, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sunwoo Cho
- Workplace Health Institute, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Jung Ah Lee
- Department of Family Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea.
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Huang D, Song M, Abe SK, Rahman MS, Islam MR, Saito E, De la Torre K, Sawada N, Tamakoshi A, Shu XO, Cai H, Hozawa A, Kanemura S, Kim J, Chen Y, Ito H, Sugawara Y, Park SK, Shin MH, Hirabayashi M, Kimura T, Gao YT, Wen W, Oze I, Shin A, Ahn YO, Ahsan H, Boffetta P, Chia KS, Matsuo K, Qiao YL, Rothman N, Zheng W, Inoue M, Kang D. Family history and gastric cancer incidence and mortality in Asia: a pooled analysis of more than half a million participants. Gastric Cancer 2024; 27:701-713. [PMID: 38649672 PMCID: PMC11193690 DOI: 10.1007/s10120-024-01499-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND The family history of gastric cancer holds important implications for cancer surveillance and prevention, yet existing evidence predominantly comes from case-control studies. We aimed to investigate the association between family history of gastric cancer and gastric cancer risk overall and by various subtypes in Asians in a prospective study. METHODS We included 12 prospective cohorts with 550,508 participants in the Asia Cohort Consortium. Cox proportional hazard regression was used to estimate study-specific adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between family history of gastric cancer and gastric cancer incidence and mortality, then pooled using random-effects meta-analyses. Stratified analyses were performed for the anatomical subsites and histological subtypes. RESULTS During the mean follow-up of 15.6 years, 2258 incident gastric cancers and 5194 gastric cancer deaths occurred. The risk of incident gastric cancer was higher in individuals with a family history of gastric cancer (HR 1.44, 95% CI 1.32-1.58), similarly in males (1.44, 1.31-1.59) and females (1.45, 1.23-1.70). Family history of gastric cancer was associated with both cardia (HR 1.26, 95% CI 1.00-1.60) and non-cardia subsites (1.49, 1.35-1.65), and with intestinal- (1.48, 1.30-1.70) and diffuse-type (1.59, 1.35-1.87) gastric cancer incidence. Positive associations were also found for gastric cancer mortality (HR 1.30, 95% CI 1.19-1.41). CONCLUSIONS In this largest prospective study to date on family history and gastric cancer, a familial background of gastric cancer increased the risk of gastric cancer in the Asian population. Targeted education, screening, and intervention in these high-risk groups may reduce the burden of gastric cancer.
Collapse
Affiliation(s)
- Dan Huang
- Department of Preventive Medicine, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea
| | - Minkyo Song
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Baltimore, MD, USA
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institute of Health, Bethesda, MD, USA
| | - Sarah Krull Abe
- Division of Prevention, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | - Md Shafiur Rahman
- Division of Prevention, National Cancer Center Institute for Cancer Control, Tokyo, Japan
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Tokyo, Japan
| | - Md Rashedul Islam
- Division of Prevention, National Cancer Center Institute for Cancer Control, Tokyo, Japan
- Hitotsubashi Institute for Advanced Study, Hitotsubashi University, Tokyo, Japan
| | - Eiko Saito
- Institute for Global Health Policy Research, National Center for Global Health and Medicine, Tokyo, Japan
| | - Katherine De la Torre
- Department of Preventive Medicine, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Norie Sawada
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | - Akiko Tamakoshi
- Department of Public Health, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hui Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Atsushi Hozawa
- Tohoku University Graduate School of Medicine, Sendai, Miyagi Prefecture, Japan
| | - Seiki Kanemura
- Tohoku University Graduate School of Medicine, Sendai, Miyagi Prefecture, Japan
| | - Jeongseon Kim
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Yu Chen
- Departments of Population Health and Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Hidemi Ito
- Division of Cancer Information and Control, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Descriptive Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yumi Sugawara
- Tohoku University Graduate School of Medicine, Sendai, Miyagi Prefecture, Japan
| | - Sue K Park
- Department of Preventive Medicine, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Myung-Hee Shin
- Department of Social and Preventive Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mayo Hirabayashi
- Division of Prevention, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | - Takashi Kimura
- Department of Public Health, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
- Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Aesun Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Yoon-Ok Ahn
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Habibul Ahsan
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Paolo Boffetta
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Kee Seng Chia
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - You-Lin Qiao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Manami Inoue
- Division of Prevention, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | - Daehee Kang
- Department of Preventive Medicine, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea.
| |
Collapse
|
6
|
Abdi E, Latifi-Navid S, Zahri S, Mostafaiy B, Yazdanbod A, Pourfarzi F. Risk of Gastric Cancer is Highly Dependent on Type of First-Degree Family Member Affected by Cancer: Lessons from a High-Risk Population in Iran. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:2004-2013. [PMID: 38033849 PMCID: PMC10682579 DOI: 10.18502/ijph.v52i9.13582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/16/2022] [Indexed: 12/02/2023]
Abstract
Background Family history of gastric cancer (GC) in first-degree relatives may increase the risk of GC. This study aimed to assess how family history of GC in first-degree relatives really affects the risk of GC in an extremely high-risk population. Methods A large population-based case-control study was carried out on 1222 incident GC cases and 1235 controls in Ardabil Province-a high-risk area in North-West Iran-to assess the associations of GC family history in first-degree relatives with the risk of GC (2003-2017). Results GC family history did not significantly associate with the risk of GC overall (ORadj=1.09, 95% CI: 0.80-1.47, P=0.589). It found no significant association of GC family history in a parent, and in a father, mother, and sister separately, with the risk of GC. However, GC risk was significantly associated with a history of GC in a sibling (ORadj=1.61, 95% CI: 1.11-2.35, P=0.013), especially brother (ORadj=2.24, 95% CI: 1.41-3.64, P=0.0008). The risk was greatly increased in subjects with two or more affected brothers (ORadj =5.56, 95% CI: 2.33-14.20, P=0.0002). Conclusion We did not find a familial tendency to cardia GC and non-cardia GC as well as histopathologic features. Determining the type of first-degree relationships with GC may, therefore, be more important than assessing family history alone for predicting the risk of GC in this high-risk area.
Collapse
Affiliation(s)
- Esmat Abdi
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Saeid Latifi-Navid
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Saber Zahri
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Behdad Mostafaiy
- Department of Statistics, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Abbas Yazdanbod
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farhad Pourfarzi
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
7
|
Kendrick P, Kelly YO, Baumann MM, Compton K, Blacker BF, Daoud F, Li Z, Mouhanna F, Nassereldine H, Schmidt C, Sylte DO, Force LM, Hay SI, Rodriquez EJ, Mensah GA, Nápoles AM, Pérez-Stable EJ, Murray CJ, Mokdad AH, Dwyer-Lindgren L. The burden of stomach cancer mortality by county, race, and ethnicity in the USA, 2000-2019: a systematic analysis of health disparities. LANCET REGIONAL HEALTH. AMERICAS 2023; 24:100547. [PMID: 37600165 PMCID: PMC10435837 DOI: 10.1016/j.lana.2023.100547] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 08/22/2023]
Abstract
Background There are persistent disparities in stomach cancer mortality among racial-ethnic groups in the USA, but the extent to which these patterns vary geographically is not well understood. This analysis estimated age-standardised mortality for five racial-ethnic groups, in 3110 USA counties over 20 years, to describe spatial-temporal variations in stomach cancer mortality and disparities between racial-ethnic groups. Methods Redistribution methods for insufficient cause of death codes and validated small area estimation methods were applied to death registration data from the US National Vital Statistics System and population data from the US National Center for Health Statistics to estimate annual stomach cancer mortality rates. Estimates were stratified by county and racial-ethnic group (non-Latino and non-Hispanic [NL] American Indian or Alaska Native [AIAN], NL Asian or Pacific Islander [Asian], NL Black [Black], Latino or Hispanic [Latino], and NL White [White]) from 2000 to 2019. Estimates were corrected for misreporting of racial-ethnic group on death certificates using published misclassification ratios. We masked (ie, did not display) estimates for county and racial-ethnic group combinations with a mean annual population of less than 1000; thus, we report estimates for 3079 (of 3110) counties for the total population, and 474, 667, 1488, 1478, and 3051 counties for the AIAN, Asian, Black, Latino, and White populations, respectively. Findings Between 2000 and 2019, national age-standardised stomach cancer mortality was lowest among the White population in every year. Nationally, stomach cancer mortality declined for all racial-ethnic groups across this time period, with the most rapid declines occurring among the Asian (percent decline 48.3% [45.1-51.1]) and Black populations (42.6% [40.2-44.6]). Mortality among the other racial-ethnic groups declined more moderately, decreasing by 36.7% (35.3-38.1), 35.1% (32.2-37.7), and 31.6% (23.9-38.0) among the White, Latino, and AIAN populations, respectively. Similar patterns were observed at the county level, although with wide geographic variation. In 2019, a majority of counties had higher mortality rates among minoritised racial-ethnic populations compared to the White population: 81.1% (377 of 465 counties with unmasked estimates for both racial-ethnic groups) among the AIAN population, 88.2% (1295 of 1469) among the Latino population, 99.4% (663 of 667) among the Asian population, and 99.9% (1484 of 1486) among the Black population. However, the size of these disparities ranged widely across counties, with the largest range from 0.3 to 17.1 among the AIAN population. Interpretation Stomach cancer mortality has decreased substantially across populations and geographies in the USA. However, disparities in stomach cancer mortality among racial-ethnic groups are widespread and have persisted over the last two decades. Local-level data are crucial to understanding the scope of this unequal burden among minoritised groups in the USA. Funding National Institute on Minority Health and Health Disparities; National Heart, Lung, and Blood Institute; National Cancer Institute; National Institute on Aging; National Institute of Arthritis and Musculoskeletal and Skin Diseases; Office of Disease Prevention; and Office of Behavioral and Social Sciences Research, National Institutes of Health (contract #75N94019C00016).
Collapse
|
8
|
Lu M, Liu D, Li Y. Long intergenic non-protein coding RNA 467 inhibition elevates microRNA-27b-3p to repress malignant behaviors of gastric cancer cells via reducing STAT3. Cell Death Dis 2022; 8:100. [PMID: 35249109 PMCID: PMC8898310 DOI: 10.1038/s41420-022-00875-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 12/31/2021] [Accepted: 01/21/2022] [Indexed: 12/18/2022]
Abstract
Emerging evidence indicated that long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) exert critical effects on tumorigenesis of multiple malignancies, including gastric cancer (GC). We aim to explore the effects of long intergenic non-protein coding RNA 467 (LINC00467) and miR-27b-3p on GC. GC cells were initially cultured. LINC00467, miR-27b-3p, and signal transducer and activator of transcription 3 (STAT3) expression in GC were detected. The altered LINC00467 and/or miR-27b-3p were transfected into screened cells. Then, the biological activities of GC cells and the tumor growth in vivo were examined. The binding relationships among LINC00467, miR-27b-3p, and STAT3 were confirmed. It was indicated that LINC00467 was increased while miR-27b-3p was decreased in GC tissues and cells. Inhibition of LINC00467 hindered GC cell malignancy and blocked tumor development by upregulating miR-27b-3p. LINC00467 sponged miR-27b-3p and STAT3 was targeted by miR-27b-3p. It was discovered that LINC00467 reduction upregulates miR-27b-3p to repress malignant GC cell growth via inhibiting STAT3. This research may deepen the insight of molecular mechanisms on GC.
Collapse
|
9
|
Man J, Ni Y, Yang X, Zhang T, Yuan Z, Chen H, Chen X, Lu M, Ye W. Healthy Lifestyle Factors, Cancer Family History, and Gastric Cancer Risk: A Population-Based Case-Control Study in China. Front Nutr 2022; 8:774530. [PMID: 35004808 PMCID: PMC8727865 DOI: 10.3389/fnut.2021.774530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022] Open
Abstract
Background: We aimed to explore the relationship between lifestyle factors, cancer family history, and gastric cancer risk. Methods: We examined the association between lifestyle factors, cancer family history, and gastric cancer risk based on a population-based case-control study in Taixing, China, with 870 cases and 1928 controls. A lifestyle score was constructed considering body shape, smoking, alcohol drinking, tooth brushing habit, and food storage method. Unconditional logistic regression models were used to calculate odd ratios (ORs) and 95% confidence intervals (CIs). Results: Compared with participants with a lifestyle score of 0, subjects with a lifestyle score of 1 (OR 0.59, 95%CI 0.43–0.83), 2 (OR 0.42, 95%CI 0.30–0.59), 3 (OR 0.29, 95%CI 0.20–0.41), 4 (OR 0.20, 95%CI 0.13–0.32), or 5 (OR 0.10, 95%CI 0.04–0.22) had a lower risk of gastric cancer (P for trend < 0.001). Overall, 34% of gastric cancer cases (95%CI 27–41%) can be attributed to non-compliance with ≥3 healthy lifestyle. Family history of early-onset cancer is closely related to the occurrence of gastric cancer, with an OR ranging from 1.77 to 3.27. Regardless of family history, a good lifestyle is associated with a reduced risk of gastric cancer, with an OR value between 0.38 and 0.70. Conclusions: The early-onset cancer family history is closely related to the occurrence of gastric cancer and a good lifestyle is associated with a reduced risk of gastric cancer regardless of family history. Our results provide a basis for identifying and providing behavior guidance of high-risk groups of gastric cancer.
Collapse
Affiliation(s)
- Jinyu Man
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.,Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Yingchun Ni
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaorong Yang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China.,Clinical Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Tongchao Zhang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China.,Clinical Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Ziyu Yuan
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Hui Chen
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China.,Clinical Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Human Phenome Institute, Fudan University, Shanghai, China
| | - Ming Lu
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.,Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China.,Clinical Research Center, Qilu Hospital of Shandong University, Jinan, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Weimin Ye
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology and Health Statistics & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
10
|
Maubach G, Vieth M, Boccellato F, Naumann M. Helicobacter pylori-induced NF-κB: trailblazer for gastric pathophysiology. Trends Mol Med 2022; 28:210-222. [PMID: 35012886 DOI: 10.1016/j.molmed.2021.12.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023]
Abstract
NF-κB signaling pathways, induced by a variety of triggers, play a key role in regulating the expression of genes involved in the immune response and cellular responses to stress. The human pathogen Helicobacter pylori induces classical and alternative NF-κB signaling pathways via its effector ADP-L-glycero-β-D-manno-heptose (ADP-heptose). We review H. pylori- and NF-κB-dependent alterations in cellular processes and associated maladaptation leading to deleterious gastric pathophysiology that have implications for the diagnosis and treatment of gastric diseases. Therapeutic options for gastric cancer (GC) include clinically relevant small molecule inhibitors of NF-κB and epigenetic therapy approaches. In this context, gastric organoid biobanks originated from patient material, represent a valuable platform for translational applications to predict patient responses to chemotherapy, with a view to personalized medicine.
Collapse
Affiliation(s)
- Gunter Maubach
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Friedrich Alexander University, Erlangen-Nuremberg, 95445 Bayreuth, Germany
| | - Francesco Boccellato
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, OX37DQ Oxford, UK
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
11
|
Tran TT, Lee J, Gunathilake M, Cho H, Kim J. Influence of Fasting Glucose Level on Gastric Cancer Incidence in a Prospective Cohort Study. Cancer Epidemiol Biomarkers Prev 2021; 31:254-261. [PMID: 34758969 DOI: 10.1158/1055-9965.epi-21-0670] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/11/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND High fasting glucose has been indicated in relation to a higher risk of gastric cancer, but the majority of studies have focused on diabetes (fasting glucose ≥126 mg/dL). Here, we investigated whether fasting glucose levels, including prediabetic and diabetic levels, influence gastric cancer incidence. METHODS A prospective study was conducted with 41,837 participants aged 16 and older who underwent health examinations at the National Cancer Center in South Korea from August 2002 to December 2014. Participants were followed up until December 2017 to identify incident gastric cancer cases. A fasting glucose test was performed based on venous blood samples taken from participants after 8 hours of fasting. We used the Cox proportional hazards regression model to explore the association of fasting glucose levels with gastric cancer incidence. RESULTS We identified 263 incident gastric cancer cases during the follow-up period. A significant association of high fasting glucose with gastric cancer incidence was found for postmenopausal women [hazard ratio (HR) = 1.88; 95% confidence interval (CI) = 1.11-3.20]. There was also a significant association between high fasting glucose and gastric cancer incidence among all participants who were nonsmokers (HR = 1.89; 95% CI = 1.21-2.95), had a BMI < 25 kg/m2 (HR = 1.45; 95% CI = 1.00-2.12), and did not have a first-degree family history of gastric cancer (HR = 1.45; 95% CI = 1.06-1.99). CONCLUSIONS Our findings support that high fasting glucose is a risk factor for gastric cancer development in postmenopausal women. IMPACT Our results provide evidence for future planning and management regarding cancer prevention.
Collapse
Affiliation(s)
- Tao Thi Tran
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Jeonghee Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Madhawa Gunathilake
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Hyunsoon Cho
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Jeongseon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea.
| |
Collapse
|
12
|
Performance evaluation of four prediction models for risk stratification in gastric cancer screening among a high-risk population in China. Gastric Cancer 2021; 24:1194-1202. [PMID: 34152518 DOI: 10.1007/s10120-021-01204-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/13/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Early detection of gastric cancer (GC) is a critical step for decreasing mortality. The aim of this study was to evaluate the performance of four prediction models for risk stratification in the screening of GC and precancerous lesions among a large, high-risk population in China. DESIGN This study was a retrospective analysis of data from the Provincial Gastric Cancer Screening Program (Zhejiang, China) spanning the period between October 2016 and April 2019, in which 97,541 individuals from the urban areas of 10 cities in Zhejiang province, China participated in this program. Demographic and clinical characteristics data were collected, and serum pepsinogens I and II, gastrin-17, and anti-H. pylori IgG antibody were detected. Participants were asked to voluntarily undergo gastroscopy. The performance of the ABC method, new ABC method, Tu's prediction model, and Li's prediction model, which stratified participants into low-, medium- and high-risk subgroups, were evaluated using the area under the receiver-operating characteristic (ROC) curve (AUC) and Youden index. RESULTS Among the participants, 6005 (3447 males and 2558 females, mean age of 58.35 years), voluntarily underwent gastroscopy. Overall, 72 (1.20%) GC cases (30 early and 42 advanced) and 2006 cases with precancerous lesions (270 atrophic gastritis, 1634 intestinal metaplasia, and 102 dysplasia/intraepithelial neoplasia) were identified. Notably, Li's prediction model achieved the greatest AUC and Youden index values (0.708 and 0.319, respectively) for predicting GC, and exhibited the greatest ability to detect precancerous lesions, especially intestinal metaplasia. CONCLUSION Li's prediction model performs the best for risk stratification in the screening, detection, and diagnosis of GC and precancerous lesions, whereas the overall performance of the other three models is similar ( www.chictr.org.cn , ChiCTR2100043363).
Collapse
|
13
|
Liu H, Dong Z. Cancer Etiology and Prevention Principle: "1 + X". Cancer Res 2021; 81:5377-5395. [PMID: 34470778 DOI: 10.1158/0008-5472.can-21-1862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/16/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022]
Abstract
Cancer was previously thought to be an inevitable aspect of human health with no effective treatments. However, the results of in-depth cancer research suggest that most types of cancer may be preventable. Therefore, a comprehensive understanding of the disparities in cancer burden caused by different risk factors is essential to inform and improve cancer prevention and control. Here, we propose the cancer etiology and prevention principle "1 + X," where 1 denotes the primary risk factor for a cancer and X represents the secondary contributing risk factors for the cancer. We elaborate upon the "1 + X" principle with respect to risk factors for several different cancer types. The "1 + X" principle can be used for precise prevention of cancer by eliminating the main cause of a cancer and minimizing the contributing factors at the same time.
Collapse
Affiliation(s)
- Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Characteristics of 1270 Chinese sibling pairs with cancer. BMC Cancer 2021; 21:1027. [PMID: 34525964 PMCID: PMC8442325 DOI: 10.1186/s12885-021-08737-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/28/2021] [Indexed: 12/24/2022] Open
Abstract
Background Previous research found that the cancer history of an individual’s sibling may be a better indicator than that of the parents. We aim to provide recommendations for opportunistic screening for individuals whose sibling had been diagnosed with cancer. Methods During the physical examination in Cancer Hospital, Chinese Academy of Medical Sciences, 43,300 people were asked if they have at least two siblings who developed cancer. Results A total of 1270 sibling-pairs from 766 families developed cancer, including 367 pairs of brothers (Bro-pairs), 368 pairs of sisters (Sis-pairs), and 535 pairs of brother-and-sister (BroSis-pairs). The mean ages at diagnosis of cancer for the three groups were from 58 to 62 years. More than half of Bro-pairs (55.3%) or Sis-pairs (51.1%) had cancer from the same systemic origin, and more than a quarter of Bro-pairs (28.1%) and Sis-pairs (37.2%) developed the same type of cancer. However, only 36.0% of BroSis-pairs developed cancers from the same systemic origin, and 18.9% developed the same type of cancer. In Bro-pairs and BroSis-pairs, lung cancer and digestive system cancer were the most common cancers, while in Sis-pairs, breast cancer, lung cancer, cervical cancer, liver cancer and thyroid cancer were the most common ones. Conclusions If an individual’s sibling is diagnosed with cancer, the individual should consider participating in opportunistic screening annually, especially for lung cancer and digestive system cancers for both sexes. For sisters, breast cancer, cervical cancer and thyroid cancer should be screened early. Additionally, genetic services are essential for individuals who have siblings with cancer.
Collapse
|
15
|
Li HQ, Xue H, Yuan H, Wan GY, Zhang XY. Preferences of first-degree relatives of gastric cancer patients for gastric cancer screening: a discrete choice experiment. BMC Cancer 2021; 21:959. [PMID: 34445987 PMCID: PMC8393792 DOI: 10.1186/s12885-021-08677-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND It is very necessary to implement gastric cancer screening in China to reduce the mortality of gastric cancer, but there are no national screening guidelines and programs. Understanding of individual preferences is conducive to formulating more acceptable screening strategies, and discrete choice experiments can quantify individual preferences. In addition, the first-degree relatives of gastric cancer patients are at high risk for gastric cancer. Compared with those without a family history of gastric cancer, the risk of gastric cancer in first-degree relatives of gastric cancer patients is increased by 60%. Therefore, a discrete choice experiment was carried out to quantitatively analyse the preferences of first-degree relatives of gastric cancer patients for gastric cancer screening to serve as a reference for the development of gastric cancer screening strategies. METHODS A questionnaire was designed based on a discrete choice experiment, and 342 first-degree relatives of gastric cancer patients were investigated. In STATA 15.0 software, the data were statistically analysed using a mixed logit model. RESULTS The five attributes included in our study had a significant influence on the preferences of first-degree relatives of gastric cancer patients for gastric cancer screening (P < 0.05). Participants most preferred the sensitivity of the screening program to be 95% (coefficient = 1.424, P < 0.01) with a willingness to pay 2501.902 Yuan (95% CI, 738.074-4265.729). In addition, the participants' sex and screening experiences affected their preferences. An increase in sensitivity 35 to 95% had the greatest impact on the participants' willingness to choose a gastric cancer screening program. CONCLUSION The formulation of gastric cancer screening strategies should be rooted in people's preferences. The influence of sex differences and screening experiences on the preferences of people undergoing screening should be considered, and screening strategies should be formulated according to local conditions to help them play a greater role.
Collapse
Affiliation(s)
- Hui-Qin Li
- Department of Fundamental Nursing, School of Nursing, Jilin University, 965 Xinjiang Street, Changchun, 130021, Jilin Province, P. R. China
| | - Hui Xue
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin Province, P. R. China
| | - Hua Yuan
- Department of Fundamental Nursing, School of Nursing, Jilin University, 965 Xinjiang Street, Changchun, 130021, Jilin Province, P. R. China
| | - Guang-Ying Wan
- Department of Fundamental Nursing, School of Nursing, Jilin University, 965 Xinjiang Street, Changchun, 130021, Jilin Province, P. R. China
| | - Xiu-Ying Zhang
- Department of Fundamental Nursing, School of Nursing, Jilin University, 965 Xinjiang Street, Changchun, 130021, Jilin Province, P. R. China.
| |
Collapse
|
16
|
Family History and Gastric Cancer Risk: A Pooled Investigation in the Stomach Cancer Pooling (STOP) Project Consortium. Cancers (Basel) 2021; 13:cancers13153844. [PMID: 34359744 PMCID: PMC8345354 DOI: 10.3390/cancers13153844] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Research is still required to establish the relationship between family history (FH) and gastric cancer (GC) in relation to different histological types and anatomical sites. The present work aimed to examine the influence of first-degree FH on the risk of GC, also according to the GC location and histological type, including 5946 cases and 12,776 controls from 17 studies of 11 countries in three continents participating in the Stomach Cancer Pooling (StoP) Project consortium. This analysis confirms the effect of FH on the risk of GC, reporting an approximately doubled risk, and provides further quantification of the risk of GC according to the subsite and histotype. Abstract Although there is a clear relationship between family history (FH) and the risk of gastric cancer (GC), quantification is still needed in relation to different histological types and anatomical sites, and in strata of covariates. The objective was to analyze the risk of GC according to first-degree FH in a uniquely large epidemiological consortium of GC. This investigation includes 5946 cases and 12,776 controls from 17 studies of the Stomach Cancer Pooling (StoP) Project consortium. Summary odds ratios (OR) and the corresponding 95% confidence intervals (CIs) were calculated by pooling study-specific ORs using fixed-effect model meta-analysis techniques. Stratified analyses were carried out by sex, age, tumor location and histological type, smoking habit, socioeconomic status, alcohol intake and fruit consumption. The pooled OR for GC was 1.84 (95% CI: 1.64–2.04; I2 = 6.1%, P heterogeneity = 0.383) in subjects with vs. those without first-degree relatives with GC. No significant differences were observed among subgroups of sex, age, geographic area or study period. Associations tended to be stronger for non-cardia (OR = 1.82; 95% CI: 1.59–2.05 for subjects with FH) than for cardia GC (OR = 1.38; 95% CI: 0.98–1.77), and for the intestinal (OR = 1.92; 95% CI: 1.62–2.23) than for the diffuse histotype (OR = 1.62; 95% CI: 1.28–1.96). This analysis confirms the effect of FH on the risk of GC, reporting an approximately doubled risk, and provides further quantification of the risk of GC according to the subsite and histotype. Considering these findings, accounting for the presence of FH to carry out correct prevention and diagnosis measures is of the utmost importance.
Collapse
|
17
|
Repetto O, De Re V, Giuffrida P, Lenti MV, Magris R, Venerito M, Steffan A, Di Sabatino A, Cannizzaro R. Proteomics signature of autoimmune atrophic gastritis: towards a link with gastric cancer. Gastric Cancer 2021; 24:666-679. [PMID: 33620602 PMCID: PMC8064991 DOI: 10.1007/s10120-020-01148-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Autoimmune atrophic gastritis (AAG) is a chronic disease that can progress to gastric cancer (GC). To better understand AAG pathology, this proteomics study investigated gastric proteins whose expression levels are altered in this disease and also in GC. METHODS Using two-dimensional difference gel electrophoresis (2D-DIGE), we compared protein maps of gastric corpus biopsies from AAG patients and controls. Differentially abundant spots (|fold change|≥ 1.5, P < 0.01) were selected and identified by LC-MS/MS. The spots were further assessed in gastric antrum biopsies from AAG patients (without and with Helicobacter pylori infection) and from GC patients and unaffected first-degree relatives of GC patients. RESULTS 2D-DIGE identified 67 differentially abundant spots, with 28 more and 39 less abundant in AAG-corpus than controls. LC-MS/MS identified these as 53 distinct proteins. The most significant (adjusted P < 0.01) biological process associated with the less abundant proteins was "tricarboxylic acid cycle". Of the 67 spots, 57 were similarly differentially abundant in AAG-antrum biopsies irrespective of H. pylori infection status. The differential abundance was also observed in GC biopsies for 14 of 28 more abundant and 35 of 39 less abundant spots, and in normal gastric biopsies of relatives of GC patients for 6 and 25 spots, respectively. Immunoblotting confirmed the different expression levels of two more abundant proteins (PDIA3, GSTP gene products) and four less abundant proteins (ATP5F1A, PGA3, SDHB, PGC). CONCLUSION This study identified a proteomics signature of AAG. Many differential proteins were shared by GC and may be involved in the progression of AAG to GC.
Collapse
Affiliation(s)
- Ombretta Repetto
- Facility of Bio-Proteomics, Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, PN Italy
| | - Valli De Re
- Facility of Bio-Proteomics, Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, PN Italy
| | - Paolo Giuffrida
- Department of Internal Medicine, San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine, San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Raffaella Magris
- Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, PN Italy
| | - Marino Venerito
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, PN Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Renato Cannizzaro
- Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, PN Italy
| |
Collapse
|
18
|
Huang Q, Read M, Gold JS, Zou XP. Unraveling the identity of gastric cardiac cancer. J Dig Dis 2020; 21:674-686. [PMID: 32975049 DOI: 10.1111/1751-2980.12945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/11/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
The classification of gastric cardiac carcinoma (GCC) is controversial. It is currently grouped with esophageal adenocarcinoma (EAC) as an adenocarcinoma of the gastroesophageal junction (GEJ). Recently, diagnostic criteria for adenocarcinoma in the GEJ were established and GCC was separated from EAC. We viewed published evidence to clarify the GCC entity for better patient management. GCC arises in the cardiac mucosa located from 3 cm below and 2 cm above the GEJ line. Compared with EAC, GCC is more like gastric cancer and affects a higher proportion of female patients, younger patients, those with a lower propensity for reflux disease, a wider histopathologic spectrum, and more complex genomic profiles. Although GCC pathogenesis mechanisms remain unknown, the two-etiology proposal is appealing: in high-risk regions, the Correa pathway with Helicobacter pylori infection, chronic inflammation, low acid and intestinal metaplasia, dysplasia and carcinoma may apply, while in low-risk regions the sequence from reflux toxin-induced mucosal injury and high acid, to intestinal metaplasia, dysplasia and carcinoma may occur. In early GCC a minimal risk of nodal metastasis argues for a role of endoscopic therapy, whereas in advanced GCC, gastric cancer staging rules and treatment strategy appear to be more appropriate than the esophageal cancer staging scheme and therapy for better prognosis stratification and treatment. In this brief review we share recent insights into the epidemiology, histopathology and genetics of GCC and hope that this will stimulate further investigations in order to improve the clinical management of patients with GCC.
Collapse
Affiliation(s)
- Qin Huang
- Department of Pathology, Nanjing Drum Tower Hospital affiliated to Nanjing University Medical School, Nanjing, Jiangsu Province, China.,Department of Pathology and Laboratory Medicine, Veterans Affairs Boston Healthcare System, Harvard Medical School/Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Matthew Read
- Department of Surgery, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Jason S Gold
- Department of Surgery, Veterans Affairs Boston Healthcare System, Harvard Medical School/Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Xiao Ping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital affiliated to Nanjing University Medical School, Nanjing, Jiangsu Province, China
| |
Collapse
|
19
|
Wu R, Yang C, Ji L, Fan ZN, Tao YW, Zhan Q. Prevalence of gastric cancer precursors in gastroscopy-screened adults by family history of gastric cancer and of cancers other than gastric. BMC Cancer 2020; 20:1110. [PMID: 33198658 PMCID: PMC7670663 DOI: 10.1186/s12885-020-07612-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background People are at a high risk of gastric cancer if their first-degree relatives suffered from atrophic gastritis (AG), intestinal metaplasia (IM), intraepithelial neoplasia (IEN), dysplasia (DYS), or gastric cancer (GC). This study was performed to analyse the association between FDR-GC and GC precursors. Methods A cross-sectional study was performed to screen the prevalence of GC precursors from November 2016 to September 2019. A total of 1329 participants with FDR-GC, 193 participants with a family history of non-gastric cancer in FDRs (FDR-nGC), and 860 participants without a family history of cancer in FDRs (FDR-nC) were recruited in this study. The logistic regression model was used in this study. Results The prevalence of normal, Non-AG, AG/IM, IEN/DYS, and GC was 31.91, 44.21, 13.81, 8.73, and 1.34%, respectively. The prevalence of IEN/DYS was higher in people with FDR-GC and FDR-nGC (FDR-GC: odds ratio (OR) = 1.655; 95%CI, 1.153–2.376; FDR-nGC: OR = 1.984; 95%CI, 1.122–3.506) than those with FDR-nC. The younger the age at which FDRs were diagnosed with GC, the more likely the participants were to develop AG/IM (Ptrend = 0.019). The risk of precursors to GC was higher in participants whose FDR-GC was the mother than in those whose FDR-GC was the father or sibling (OR, non-AG: 1.312 vs. 1.007, 1.274; AG/IM: 1.430 vs. 1.296, 1.378; IEN/DYS: 1.988 vs. 1.573, 1.542). There was no statistically significant difference in non-AG (OR = 1.700; 95%CI, 0.940–3.074), AG/IM (OR = 1.291; 95%CI, 0.579–2.877), and IEN/DYS (OR = 1.265; 95%CI, 0.517–3.096) between participants with one or more FDR-GC. Conclusion People with FDR-GC and FDR-nGC are at a high risk of IEN/DYS. When an FDR was diagnosed at a younger age, the risk of AG/IM was higher. The risk of GC precursors was higher in people whose FDR-GC was the mother.
Collapse
Affiliation(s)
- Rui Wu
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital to Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China.,Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Cheng Yang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital to Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Lin Ji
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital to Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Zhi-Ning Fan
- Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yu-Wen Tao
- Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Qiang Zhan
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital to Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
20
|
A Medical Decision Support System to Assess Risk Factors for Gastric Cancer Based on Fuzzy Cognitive Map. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:1016284. [PMID: 33082836 PMCID: PMC7556058 DOI: 10.1155/2020/1016284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/19/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC), one of the most common cancers around the world, is a multifactorial disease and there are many risk factors for this disease. Assessing the risk of GC is essential for choosing an appropriate healthcare strategy. There have been very few studies conducted on the development of risk assessment systems for GC. This study is aimed at providing a medical decision support system based on soft computing using fuzzy cognitive maps (FCMs) which will help healthcare professionals to decide on an appropriate individual healthcare strategy based on the risk level of the disease. FCMs are considered as one of the strongest artificial intelligence techniques for complex system modeling. In this system, an FCM based on Nonlinear Hebbian Learning (NHL) algorithm is used. The data used in this study are collected from the medical records of 560 patients referring to Imam Reza Hospital in Tabriz City. 27 effective features in gastric cancer were selected using the opinions of three experts. The prediction accuracy of the proposed method is 95.83%. The results show that the proposed method is more accurate than other decision-making algorithms, such as decision trees, Naïve Bayes, and ANN. From the perspective of healthcare professionals, the proposed medical decision support system is simple, comprehensive, and more effective than previous models for assessing the risk of GC and can help them to predict the risk factors for GC in the clinical setting.
Collapse
|
21
|
Choi IJ, Kim CG, Lee JY, Kim YI, Kook MC, Park B, Joo J. Family History of Gastric Cancer and Helicobacter pylori Treatment. N Engl J Med 2020; 382:427-436. [PMID: 31995688 DOI: 10.1056/nejmoa1909666] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Helicobacter pylori infection and a family history of gastric cancer are the main risk factors for gastric cancer. Whether treatment to eradicate H. pylori can reduce the risk of gastric cancer in persons with a family history of gastric cancer in first-degree relatives is unknown. METHODS In this single-center, double-blind, placebo-controlled trial, we screened 3100 first-degree relatives of patients with gastric cancer. We randomly assigned 1838 participants with H. pylori infection to receive either eradication therapy (lansoprazole [30 mg], amoxicillin [1000 mg], and clarithromycin [500 mg], each taken twice daily for 7 days) or placebo. The primary outcome was development of gastric cancer. A prespecified secondary outcome was development of gastric cancer according to H. pylori eradication status, assessed during the follow-up period. RESULTS A total of 1676 participants were included in the modified intention-to-treat population for the analysis of the primary outcome (832 in the treatment group and 844 in the placebo group). During a median follow-up of 9.2 years, gastric cancer developed in 10 participants (1.2%) in the treatment group and in 23 (2.7%) in the placebo group (hazard ratio, 0.45; 95% confidence interval [CI], 0.21 to 0.94; P = 0.03 by log-rank test). Among the 10 participants in the treatment group in whom gastric cancer developed, 5 (50.0%) had persistent H. pylori infection. Gastric cancer developed in 0.8% of participants (5 of 608) in whom H. pylori infection was eradicated and in 2.9% of participants (28 of 979) who had persistent infection (hazard ratio, 0.27; 95% CI, 0.10 to 0.70). Adverse events were mild and were more common in the treatment group than in the placebo group (53.0% vs. 19.1%; P<0.001). CONCLUSIONS Among persons with H. pylori infection who had a family history of gastric cancer in first-degree relatives, H. pylori eradication treatment reduced the risk of gastric cancer. (Funded by the National Cancer Center, South Korea; ClinicalTrials.gov number, NCT01678027.).
Collapse
Affiliation(s)
- Il Ju Choi
- From the Center for Gastric Cancer (I.J.C., C.G.K., J.Y.L., Y.-I.K., M.-C.K.), the Division of Cancer Epidemiology and Management, Research Institute (I.J.C., Y.-I.K., B.P., J.J.), and the Biostatistics Collaboration Team, Research Core Center, Research Institute (B.P.) - all at the National Cancer Center, Goyang, South Korea
| | - Chan Gyoo Kim
- From the Center for Gastric Cancer (I.J.C., C.G.K., J.Y.L., Y.-I.K., M.-C.K.), the Division of Cancer Epidemiology and Management, Research Institute (I.J.C., Y.-I.K., B.P., J.J.), and the Biostatistics Collaboration Team, Research Core Center, Research Institute (B.P.) - all at the National Cancer Center, Goyang, South Korea
| | - Jong Yeul Lee
- From the Center for Gastric Cancer (I.J.C., C.G.K., J.Y.L., Y.-I.K., M.-C.K.), the Division of Cancer Epidemiology and Management, Research Institute (I.J.C., Y.-I.K., B.P., J.J.), and the Biostatistics Collaboration Team, Research Core Center, Research Institute (B.P.) - all at the National Cancer Center, Goyang, South Korea
| | - Young-Il Kim
- From the Center for Gastric Cancer (I.J.C., C.G.K., J.Y.L., Y.-I.K., M.-C.K.), the Division of Cancer Epidemiology and Management, Research Institute (I.J.C., Y.-I.K., B.P., J.J.), and the Biostatistics Collaboration Team, Research Core Center, Research Institute (B.P.) - all at the National Cancer Center, Goyang, South Korea
| | - Myeong-Cherl Kook
- From the Center for Gastric Cancer (I.J.C., C.G.K., J.Y.L., Y.-I.K., M.-C.K.), the Division of Cancer Epidemiology and Management, Research Institute (I.J.C., Y.-I.K., B.P., J.J.), and the Biostatistics Collaboration Team, Research Core Center, Research Institute (B.P.) - all at the National Cancer Center, Goyang, South Korea
| | - Boram Park
- From the Center for Gastric Cancer (I.J.C., C.G.K., J.Y.L., Y.-I.K., M.-C.K.), the Division of Cancer Epidemiology and Management, Research Institute (I.J.C., Y.-I.K., B.P., J.J.), and the Biostatistics Collaboration Team, Research Core Center, Research Institute (B.P.) - all at the National Cancer Center, Goyang, South Korea
| | - Jungnam Joo
- From the Center for Gastric Cancer (I.J.C., C.G.K., J.Y.L., Y.-I.K., M.-C.K.), the Division of Cancer Epidemiology and Management, Research Institute (I.J.C., Y.-I.K., B.P., J.J.), and the Biostatistics Collaboration Team, Research Core Center, Research Institute (B.P.) - all at the National Cancer Center, Goyang, South Korea
| |
Collapse
|
22
|
Ji L, Liu Z, Zhou B, Cai Y, An F, Wang L, Lv Z, Xia M, Yang J, Yuan J, Wang H, Zhou Z, Yang S, Hu L, Zhan Q. Community-Based Pilot Study of a Screening Program for Gastric Cancer in a Chinese Population. Cancer Prev Res (Phila) 2019; 13:73-82. [PMID: 31796467 DOI: 10.1158/1940-6207.capr-19-0372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/26/2019] [Accepted: 11/26/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Lin Ji
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zengchao Liu
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Bin Zhou
- Department of Biotechnology, Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Ying Cai
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Fangmei An
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Lei Wang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhifa Lv
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Min Xia
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jianbo Yang
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Jianfen Yuan
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Hui Wang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhiyi Zhou
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Shudong Yang
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Lei Hu
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Qiang Zhan
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China.
| |
Collapse
|
23
|
Cai Q, Zhu C, Yuan Y, Feng Q, Feng Y, Hao Y, Li J, Zhang K, Ye G, Ye L, Lv N, Zhang S, Liu C, Li M, Liu Q, Li R, Pan J, Yang X, Zhu X, Li Y, Lao B, Ling A, Chen H, Li X, Xu P, Zhou J, Liu B, Du Z, Du Y, Li Z. Development and validation of a prediction rule for estimating gastric cancer risk in the Chinese high-risk population: a nationwide multicentre study. Gut 2019; 68:1576-1587. [PMID: 30926654 PMCID: PMC6709770 DOI: 10.1136/gutjnl-2018-317556] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To develop a gastric cancer (GC) risk prediction rule as an initial prescreening tool to identify individuals with a high risk prior to gastroscopy. DESIGN This was a nationwide multicentre cross-sectional study. Individuals aged 40-80 years who went to hospitals for a GC screening gastroscopy were recruited. Serum pepsinogen (PG) I, PG II, gastrin-17 (G-17) and anti-Helicobacter pylori IgG antibody concentrations were tested prior to endoscopy. Eligible participants (n=14 929) were randomly assigned into the derivation and validation cohorts, with a ratio of 2:1. Risk factors for GC were identified by univariate and multivariate analyses and an optimal prediction rule was then settled. RESULTS The novel GC risk prediction rule comprised seven variables (age, sex, PG I/II ratio, G-17 level, H. pylori infection, pickled food and fried food), with scores ranging from 0 to 25. The observed prevalence rates of GC in the derivation cohort at low-risk (≤11), medium-risk (12-16) or high-risk (17-25) group were 1.2%, 4.4% and 12.3%, respectively (p<0.001).When gastroscopy was used for individuals with medium risk and high risk, 70.8% of total GC cases and 70.3% of early GC cases were detected. While endoscopy requirements could be reduced by 66.7% according to the low-risk proportion. The prediction rule owns a good discrimination, with an area under curve of 0.76, or calibration (p<0.001). CONCLUSIONS The developed and validated prediction rule showed good performance on identifying individuals at a higher risk in a Chinese high-risk population. Future studies are needed to validate its efficacy in a larger population.
Collapse
Affiliation(s)
- Quancai Cai
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chunping Zhu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department, China Medical University, Shenyang, China
| | - Qi Feng
- Department of Gastroenterology, Mianyang Central Hospital, Mianyang, China
| | - Yichao Feng
- Department of Gastroenterology, Affiliated Hospital of Yan’an University, Yanan, China
| | - Yingxia Hao
- Department of Gastroenterology, Baoding First Central Hospital, Baoding, China
| | - Jichang Li
- Department of Gastroenterology, Baoji Central Hospital, Baoji, China
| | - Kaiguang Zhang
- Department of Gastroenterology, Anhui Provincial Hospital, Hefei, China
| | - Guoliang Ye
- Department of Gastroenterology, Affiliated Hospital of Ningbo University, Ningbo, China
| | - Liping Ye
- Department of Gastroenterology, Taizhou Hospital, Taizhou, China
| | - Nonghua Lv
- Department of Gastroenterology, First Affiliated Hospital of Nanchang University, NanChang, China
| | - Shengsheng Zhang
- Department of Gastroenterology, Beijing Traditional Chinese Medicine Hospital of Capital Medical University, Beijing, China
| | - Chengxia Liu
- Department of Gastroenterology, Affiliated Hospital of Binzhou Medical College, Binzhou, China
| | - Mingquan Li
- Department of Gastroenterology, Yan’an People’s Hospital, Yan’an, China
| | - Qi Liu
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Rongzhou Li
- Department of Gastroenterology, Ruian People’s Hospital, Rui’an, China
| | - Jie Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
| | - Xiaocui Yang
- Department of Gastroenterology, Ankang Central Hospital, Ankang, China
| | - Xuqing Zhu
- Department of Gastroenterology, Taizhou Municipal Hospital, Taizhou, China
| | - Yumei Li
- Department of Gastroenterology, Zhejiang General Team Hospital of Chinese People’s Armed Police Force, Hangzhou, China
| | - Bo Lao
- Department of Gastroenterology, Ningbo Yinzhou District Second Hospital, Ningbo, China
| | - Ansheng Ling
- Department of Gastroenterology, Anqing First People’s Hospital, Anqing, China
| | - Honghui Chen
- Department of Gastroenterology, Second Affiliated Hospital of South China University, Hengyang, China
| | - Xiuling Li
- Department of Gastroenterology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Ping Xu
- Department of Gastroenterology, Shanghai Songjiang Distict Central Hospital, Shanghai, China
| | - Jianfeng Zhou
- Department of Gastroenterology, Ningbo Beilun District Xiaogang Hospital, Ningbo, China
| | - Baozhen Liu
- Department of Gastroenterology, Binzhou People’s Hospital, Binzhou, China
| | - Zhiqiang Du
- Department of Gastroenterology, Jianyang People’s Hospital, Jianyang, China
| | - Yiqi Du
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
24
|
Youn Nam S, Park BJ, Nam JH, Ryu KH, Kook MC, Kim J, Lee WK. Association of current Helicobacter pylori infection and metabolic factors with gastric cancer in 35,519 subjects: A cross-sectional study. United European Gastroenterol J 2018; 7:287-296. [PMID: 31080613 DOI: 10.1177/2050640618819402] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/18/2018] [Indexed: 12/13/2022] Open
Abstract
Background The effect of current infection of Helicobacter pylori on gastric cancer has rarely been studied in a large population. We investigated the association of current H. pylori infection and metabolic factors with gastric cancer in a large population. Methods Persons who made their first visit to the National Cancer Center for a health examination, including endoscopy and H. pylori testing using gastric tissue between 2003 and 2013, were included. The association of H. pylori with gastric cancer was estimated using odds ratios (ORs) and 95% confidence intervals (CIs). Results Among 35,519 people, 113 gastric cancer and 158 gastric dysplasia cases were detected. In the adjusted analysis, gastric cancer was associated with current H. pylori infection (OR, 2.39; 95% CI, 1.53-3.74), age (OR, 1.06; 95% CI 1.04-1.08), first-degree relatives with gastric cancer (OR, 2.08; 95% CI, 1.30-3.32) and hyperglycaemia (OR, 1.66; 95% CI, 1.04-2.65), whereas it was inversely associated with high-density lipoprotein (HDL) (OR, 0.49; 95% CI, 0.22-0.94). In the subanalysis, gastric cancer was associated with first-degree relatives with gastric cancer (OR, 3.23; 95% CI, 1.39-7.50) in the absence of H. pylori, whereas it was associated with hyperglycaemia (OR, 1.98; 95% CI, 1.16-3.39) in the presence of H. pylori. Conclusions Gastric cancer was associated with current H. pylori infection, hyperglycaemia, and low HDL levels in a large population.
Collapse
Affiliation(s)
- Su Youn Nam
- Gastroenterology, Center for Gastric Cancer, Kyungpook National University Hospital, Daegu, Korea.,Department of Internal Medicine, Center for Cancer Prevention and Detection, Goyang, Korea
| | - Bum Joon Park
- Department of Internal Medicine, Center for Cancer Prevention and Detection, Goyang, Korea
| | - Ji Hyung Nam
- Department of Internal Medicine, Center for Cancer Prevention and Detection, Goyang, Korea
| | - Kum Hei Ryu
- Department of Internal Medicine, Center for Cancer Prevention and Detection, Goyang, Korea
| | | | - Jeongseon Kim
- Molecular Epidemiology Branch, National Cancer Center, Goyang, Korea
| | - Won Kee Lee
- Biostatistics, Kyungpook National University Hospital and School of Medicine, Daegu, Korea
| |
Collapse
|