1
|
Lim SH, Kim N, Choi Y, Choi JM, Han YM, Kwak MS, Chung GE, Seo JY, Baek SM, Yoon H, Park YS, Lee DH. Assessing Serum Pepsinogen and Helicobacter pylori Tests for Detecting Diffuse-Type Gastric Cancer: Insights from a Large-Scale and Propensity-Score-Matched Study in Republic of Korea. Cancers (Basel) 2025; 17:955. [PMID: 40149291 PMCID: PMC11940262 DOI: 10.3390/cancers17060955] [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: 01/27/2025] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
The incidence of cancer and its associated mortality have increased over the past several decades [...].
Collapse
Affiliation(s)
- Seon Hee Lim
- Departments of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Healthcare Research Institute, Seoul 06236, Republic of Korea; (S.H.L.); (J.M.C.); (Y.M.H.); (M.-S.K.); (G.E.C.); (J.Y.S.)
| | - Nayoung Kim
- Department of Internal Medicine, Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea; (Y.C.); (S.M.B.); (H.Y.); (Y.S.P.); (D.H.L.)
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yonghoon Choi
- Department of Internal Medicine, Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea; (Y.C.); (S.M.B.); (H.Y.); (Y.S.P.); (D.H.L.)
| | - Ji Min Choi
- Departments of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Healthcare Research Institute, Seoul 06236, Republic of Korea; (S.H.L.); (J.M.C.); (Y.M.H.); (M.-S.K.); (G.E.C.); (J.Y.S.)
| | - Yoo Min Han
- Departments of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Healthcare Research Institute, Seoul 06236, Republic of Korea; (S.H.L.); (J.M.C.); (Y.M.H.); (M.-S.K.); (G.E.C.); (J.Y.S.)
| | - Min-Sun Kwak
- Departments of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Healthcare Research Institute, Seoul 06236, Republic of Korea; (S.H.L.); (J.M.C.); (Y.M.H.); (M.-S.K.); (G.E.C.); (J.Y.S.)
| | - Goh Eun Chung
- Departments of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Healthcare Research Institute, Seoul 06236, Republic of Korea; (S.H.L.); (J.M.C.); (Y.M.H.); (M.-S.K.); (G.E.C.); (J.Y.S.)
| | - Ji Yeon Seo
- Departments of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Healthcare Research Institute, Seoul 06236, Republic of Korea; (S.H.L.); (J.M.C.); (Y.M.H.); (M.-S.K.); (G.E.C.); (J.Y.S.)
| | - Sung Min Baek
- Department of Internal Medicine, Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea; (Y.C.); (S.M.B.); (H.Y.); (Y.S.P.); (D.H.L.)
| | - Hyuk Yoon
- Department of Internal Medicine, Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea; (Y.C.); (S.M.B.); (H.Y.); (Y.S.P.); (D.H.L.)
| | - Young Soo Park
- Department of Internal Medicine, Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea; (Y.C.); (S.M.B.); (H.Y.); (Y.S.P.); (D.H.L.)
| | - Dong Ho Lee
- Department of Internal Medicine, Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea; (Y.C.); (S.M.B.); (H.Y.); (Y.S.P.); (D.H.L.)
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
2
|
Guo W, Li Z, Mao W, Liu X, Yang Y, Yu J, Yang H, Gao R. Assessing omeprazole and flunixin meglumine co-administration in treating equine gastric ulcer syndrome in Mongolian horses. Equine Vet J 2025. [PMID: 39967295 DOI: 10.1111/evj.14477] [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: 12/03/2023] [Accepted: 12/22/2024] [Indexed: 02/20/2025]
Abstract
BACKGROUND Flunixin meglumine (FM) is commonly used in painful conditions in horses; however, it may contribute to equine gastric ulcer syndrome (EGUS). Some veterinarians combine omeprazole (OME) and FM to reduce EGUS risk. However, the efficacy and safety of this combination in Mongolian horses with chronic lameness remain unknown. OBJECTIVES To investigate the changes in lameness and EGUS scores in Mongolian horses through the comparison of three treatment strategies: FM, FM + OME and placebo treated control (CON) and to assess the effects of these treatments on gastric fluid pH, serum total protein, albumin and oxidative stress markers (MPO, SOD, CAT). STUDY DESIGN In vivo experiments. METHODS Eighteen Mongolian horses with initial American Association of Equine Practitioners lameness scores ≥3 of 5 were selected and equally divided into the placebo (CON), FM (1.1 mg/kg IV q. 24 h) and FM + OME (4 mg/kg PO q. 24 h) treatment groups in a randomised block design. During 15 days of treatment, weekly gastroscopy and physiological and biochemical tests were performed. Stomach tissues were harvested from two horses from each group for histopathological examination with haematoxylin and eosin, Masson's trichrome and periodic acid-Schiff (PAS) staining. RESULTS FM (median 1.0, interquartile range 0.0-1.0; p < 0.001) and FM + OME (1.0, 1.0-1.0; p < 0.001) significantly decreased lameness scores compared with CON (3.0, 3.0-4.0). Compared with CON (EGGD: 0.0, 0.0-1.0, p < 0.001; PG1: mean 231.9 ± standard deviation 25.2 ng/mL, p < 0.001) or FM + OME (EGGD: 0.8, 1.0-1.3, p = 0.003; PG1: 207.08 ± 34.85 ng/mL, p < 0.001), FM significantly increased equine gastric glandular disease (EGGD) grade (3.0, 2.0-3.3) and pepsinogen 1 (PG1) content (372.2 ± 33.2 ng/mL, p < 0.001). Compared with CON (total protein: 70.1 ± 2.9 g/L; albumin: 37.0 ± 3.0 g/L; Gastrin-17: 482.5 ± 48.1 pg/mL), FM significantly reduced total protein (62.8 ± 2.9 g/L, p = 0.003), albumin (31.5 ± 2.3 g/L, p = 0.01) and Gastrin-17 (GT-17) content (284.6 ± 57.2 pg/mL, p < 0.001). Compared with FM (EGGD: 3.0, 2.0-3.3; pH: 2.4 ± 0.3), FM + OME significantly decreased the EGGD grade (0.8, 1.0-1.3; p = 0.003) and significantly increased gastric fluid pH (7.4 ± 0.2; p < 0.001). FM + OME (207.1 ± 34.9 ng/mL) significantly decreased PG1 content compared with FM (372.24 ± 33.25 ng/mL; p < 0.001). Histopathology revealed that 15 days of FM treatment led to gastric lesions in horses, which were mitigated by combining with OME. MAIN LIMITATIONS Individual differences among horses were large, but the sample size was small and sampling was infrequent. CONCLUSIONS Compared with FM alone, use of FM + OME did not impact the reduction in lameness scores with therapy, but reduced the occurrence of EGGD in Mongolian horses. When used to manage chronic lameness, FM + OME might protect against FM-induced EGGD by increasing the gastric pH and decreasing serum PG1 content.
Collapse
Affiliation(s)
- Wenrui Guo
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhengyi Li
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Wei Mao
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xinyu Liu
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Ying Yang
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Jiahui Yu
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Huidi Yang
- Basic Medical School, Inner Mongolia Medical University, Hohhot, China
| | - Ruifeng Gao
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
3
|
Si YT, Xiong XS, Wang JT, Yuan Q, Li YT, Tang JW, Li YN, Zhang XY, Li ZK, Lai JX, Umar Z, Yang WX, Li F, Wang L, Gu B. Identification of chronic non-atrophic gastritis and intestinal metaplasia stages in the Correa's cascade through machine learning analyses of SERS spectral signature of non-invasively-collected human gastric fluid samples. Biosens Bioelectron 2024; 262:116530. [PMID: 38943854 DOI: 10.1016/j.bios.2024.116530] [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: 02/04/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024]
Abstract
The progression of gastric cancer involves a complex multi-stage process, with gastroscopy and biopsy being the standard procedures for diagnosing gastric diseases. This study introduces an innovative non-invasive approach to differentiate gastric disease stage using gastric fluid samples through machine-learning-assisted surface-enhanced Raman spectroscopy (SERS). This method effectively identifies different stages of gastric lesions. The XGBoost algorithm demonstrates the highest accuracy of 96.88% and 91.67%, respectively, in distinguishing chronic non-atrophic gastritis from intestinal metaplasia and different subtypes of gastritis (mild, moderate, and severe). Through blinded testing validation, the model can achieve more than 80% accuracy. These findings offer new possibilities for rapid, cost-effective, and minimally invasive diagnosis of gastric diseases.
Collapse
Affiliation(s)
- Yu-Ting Si
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xue-Song Xiong
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Jin-Ting Wang
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Quan Yuan
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yu-Ting Li
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Jia-Wei Tang
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Division of Microbiology and Immunology, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Yong-Nian Li
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xin-Yu Zhang
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zheng-Kang Li
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jin-Xin Lai
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zeeshan Umar
- Marshall Laboratory of Biomedical Engineering, School of Medicine, Shenzhen University, Guangdong Province, China
| | - Wei-Xuan Yang
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Fen Li
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China.
| | - Liang Wang
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Division of Microbiology and Immunology, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia; The Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
| | - Bing Gu
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Wang F, Pang R, Zhao X, Zhou B, Tian Y, Ma Y, Rong L. Plasma metabolomics and lipidomics reveal potential novel biomarkers in early gastric cancer: An explorative study. Int J Biol Markers 2024; 39:226-238. [PMID: 38859802 DOI: 10.1177/03936155241258780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
BACKGROUND Early identification and therapy can significantly improve the outcome for gastric cancer. However, there is still no perfect biomarker available for the detection of early gastric cancer. This study aimed to investigate the alterations in the plasma metabolites of early gastric cancer using metabolomics and lipidomics based on high-performance liquid chromatography-mass spectrometry (HPLC-MS), which detected potential biomarkers that could be used for clinical diagnosis. METHODS To investigate the changes in metabolomics and lipidomics, a total of 30 plasma samples were collected, consisting of 15 patients with early gastric cancer and 15 healthy controls. Extensive HPLC-MS-based untargeted metabolomic and lipidomic investigations were conducted. Differential metabolites and metabolic pathways were uncovered through the utilization of statistical analysis and bioinformatics analysis. Candidate biomarker screening was performed using support vector machine-based multivariate receiver operating characteristic analysis. RESULTS A disturbance was observed in a combined total of 19 metabolites and 67 lipids of the early gastric cancer patients. The analysis of KEGG pathways showed that the early gastric cancer patients experienced disruptions in the arginine biosynthesis pathway, the pathway for alanine, aspartate, and glutamate metabolism, as well as the pathway for glyoxylate and dicarboxylate metabolism. Plasma metabolomics and lipidomics have identified multiple biomarker panels that can effectively differentiate early gastric cancer patients from healthy controls, exhibiting an area under the curve exceeding 0.9. CONCLUSION These metabolites and lipids could potentially serve as biomarkers for the screening of early gastric cancer, thereby optimizing the strategy for the detection of early gastric cancer. The disrupted pathways implicated in early gastric cancer provide new clues for additional understanding of gastric cancer's pathogenesis. Nonetheless, large-scale clinical data are required to prove our findings.
Collapse
Affiliation(s)
- Feng Wang
- Department of Endoscopy Center, Peking University First Hospital, Beijing, China
| | - Ruifang Pang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xudong Zhao
- Department of Endoscopy Center, Peking University First Hospital, Beijing, China
| | - Bin Zhou
- Department of Endoscopy Center, Peking University First Hospital, Beijing, China
| | - Yuan Tian
- Department of Endoscopy Center, Peking University First Hospital, Beijing, China
| | - Yongchen Ma
- Department of Endoscopy Center, Peking University First Hospital, Beijing, China
| | - Long Rong
- Department of Endoscopy Center, Peking University First Hospital, Beijing, China
| |
Collapse
|
5
|
Hirabayashi M, Wilunda C, Murai U, Yamaji T, Iwasaki M, Inoue M, Tsugane S, Sawada N. Association between fish and shellfish consumption, n-3 polyunsaturated fatty acids, and gastric cancer risk: the Japan Public Health Center-based Prospective Study. Eur J Nutr 2024; 63:1529-1544. [PMID: 38703225 PMCID: PMC11329692 DOI: 10.1007/s00394-024-03343-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/30/2024] [Indexed: 05/06/2024]
Abstract
PURPOSE Fish and shellfish consumption is suggested to be a cancer-protective factor. However, studies investigating this association for gastric cancer, especially considering Helicobacter pylori (H. pylori) and atrophic gastritis (AG), are limited. We investigated gastric cancer risk associated with fish, shellfish, and n-3 polyunsaturated fatty acids (n-3 PUFAs) consumption among Japanese adults. METHODS 90,504 subjects enrolled in the Japan Public Health Center-based Prospective Study (JPHC Study) were followed until December 2013. Dietary intake data were collected using a food frequency questionnaire. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated for gastric cancer risk associated with fish and shellfish consumption and marine n-3 PUFAs (sum of eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA)) using Cox proportional hazards models. Among those with avaliable data, we conducted a subgroup analysis taking H. pylori infection and AG status into consideration. RESULTS There were 2,701 gastric cancer cases during an average of 15 years of follow-up. We observed an increased gastric cancer risk for salted fish consumption for men [HR for fifth quintile versus first quintile 1.43 (95% CI 1.18-1.75)] and for women [HR 1.33 (95% CI 1.00-1.77)]. We observed a weak risk reduction trend for women as the intake of marine n-3 PUFAs increased (p-trend:0.07). When we included H. pylori infection and atrophic gastritis status in the analysis, the associations diminished. CONCLUSION Our results suggest that salted fish increases gastric cancer risk for men and women, while marine n-3 PUFAs marginally decreases this risk among women in Japan.
Collapse
Affiliation(s)
- Mayo Hirabayashi
- Division of Prevention, Institute for Cancer Control, National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Calistus Wilunda
- Division of Cohort Research, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
- African Population and Health Research Center, Nairobi, Kenya
| | - Utako Murai
- Division of Cohort Research, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Taiki Yamaji
- Division of Epidemiology, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Motoki Iwasaki
- Division of Cohort Research, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
- Division of Epidemiology, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Manami Inoue
- Division of Prevention, Institute for Cancer Control, National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
- Division of Cohort Research, Institute for Cancer Control, National Cancer Center, Tokyo, Japan.
| | - Shoichiro Tsugane
- Division of Cohort Research, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
- Graduate School of Public Health, International University of Health and Welfare, Tokyo, Japan
| | - Norie Sawada
- Division of Cohort Research, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| |
Collapse
|
6
|
Huang Y, Yang H, Ding R, Wang L, Li J, Li W, Qin X, Xu Y, Qian J. Reference Ranges and Comparison of Pepsinogen by Chemiluminescence Immunoassay and Enzyme-Linked Immunosorbent Assay in Chinese Population. Cancer Manag Res 2024; 16:921-931. [PMID: 39099764 PMCID: PMC11296369 DOI: 10.2147/cmar.s459568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Objective Serum pepsinogen (PG) is a good indicator of atrophic changes in the gastric mucosa. Gastric mucosal atrophy is a high-risk factor for gastric cancer. Serological testing for PG combined with endoscopy can help to improve gastric cancer screening. In this study, we established the reference ranges of serum PG-I, PG-II, and the PG-I/II ratio (PGR) in the Chinese population by chemiluminescence immunoassay (CLIA) and enzyme-linked immunosorbent assay (ELISA). Besides, in the real world, doctors are often confused by the results of different testing platforms. Thus, a comparison of methods CLIA and ELISA was performed. Methods 2904 individuals were enrolled from six regions in China as part of the Chinese Adult Digestive Diseases Surveillance (2016) program. The individuals completed questionnaires and volunteered to undergo examinations, including gastroscopy, urea breath test, abdominal ultrasound examination and routine serologic tests. Serum was collected to measure PGs (including PG-I, PG-II and PGR) by CLIA and ELISA. Participants who were found obvious abnormalities or absent from the examinations were excluded. Ultimately, 747 healthy individuals were enrolled in this study. The Kolmogorov-Smirnov test was used to assess the distribution of variables. The Kruskal-Wallis H or Mann-Whitney U-tests were used to compare different sex, age, and geographical groups. The 95% reference ranges of PGs obtained by the two methods were established according to document CLSI-EP28-A3, with covariates of sex, age, and region. Spearman correlation analysis, linear regression analysis and allowable total error (ATE) zone analysis were utilized for comparing the two methods. Results On overall, the 95% reference ranges of PG-I, PG-II, and PGR measured by CLIA were 23.00-110.64 ng/mL, 2.50-19.13 ng/mL, and 3.87-13.30, respectively. Meanwhile, the reference ranges of PG-I, PG-II, and PGR measured by ELISA were 36.93-205.06 ng/mL, 1.65-17.96 ng/mL, and 7.50-33.60, respectively. Both PG-I and PG-II levels measured by the two platforms were found to be influenced by sex and age. PGR measured by CLIA was influenced by age but not by sex, while PGR measured by ELISA was not affected by either age or sex. Regional factors did not significantly impact the PG results, except for PG-I detected by ELISA. Ultimately, reference ranges for PGs were established based on age and sex stratification. Additionally, the Spearman correlation analysis revealed that the correlation coefficients for PG-I, PG-II, and PGR detected by the two methods were 0.899, 0.887, and 0.777, respectively, indicating a strong correlation between the two methods. The regression equation for the PG levels detected by two methods was obtained through linear regression analysis. The ATE analysis provided a visual depiction of the consistency between the two methods, clearly indicating the poor agreement between them. Conclusion This study established the reference ranges of PGs by strict and intact enrollment standard. In addition, the results indicated a strong linear relationship between the two methods, yet with a clear bias, which was valuable for laboratory interpretation.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Laboratory Medicine, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Rui Ding
- Department of Laboratory Medicine, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Li Wang
- Department of Epidemiology and Health Statistics, Institute of Basic Medicine, Peking Union Medical College, Beijing, People’s Republic of China
| | - Ji Li
- Department of Gastroenterology, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wenbo Li
- Department of Ultrasound, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Xuzhen Qin
- Department of Laboratory Medicine, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yingchun Xu
- Department of Laboratory Medicine, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Jiaming Qian
- Department of Gastroenterology, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
7
|
Yasuda K, Chinda D, Shimoyama T, Arai T, Akitaya K, Fujiwara S, Nomiya H, Sasaki Y, Komai K, Sawada Y, Saito Y, Chiba H, Sakuraba H, Fukuda S. Factors Predicting Effectiveness of Eradication Therapy for Helicobacter pylori-Associated Dyspepsia Symptoms. Life (Basel) 2024; 14:935. [PMID: 39202677 PMCID: PMC11355128 DOI: 10.3390/life14080935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Functional dyspepsia is distinguishable from Helicobacter pylori-associated dyspepsia. However, distinguishing H. pylori-associated dyspepsia from functional dyspepsia before H. pylori eradication is difficult. Therefore, in the present study, we aimed to investigate whether serum pepsinogen levels before H. pylori eradication are associated with the amelioration of dyspepsia after successful H. pylori eradication. Additionally, we examined the usefulness of serum pepsinogen levels and other factors in predicting dyspepsia outcomes. H. pylori eradication was effective in 14 patients (Responders) and ineffective in 19 patients (Non-responders). The pepsinogen I/II ratio in Responders (3.4 ± 1.2) and Non-responders (2.3 ± 1.0) differed significantly (p = 0.006). The optimal cut-off pepsinogen I/II value was 2.3. Multivariate logistic regression analysis showed that the adjusted odds ratio for Non-responders was 26.1 (95% confidence interval: 2.0-338.0, p = 0.012) for a pepsinogen I/II ratio ≤ 2.3 and 8.10 (95% confidence interval: 1.1-57.6, p = 0.037) for smoking habits. The pepsinogen I/II ratio and smoking habits were associated with the effects of H. pylori eradication on dyspeptic symptoms. Thus, the pepsinogen I/II ratio cut-off value can be used to identify patients likely to respond to H. pylori eradication after the resolution of dyspeptic symptoms.
Collapse
Affiliation(s)
- Kohei Yasuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
| | - Daisuke Chinda
- Division of Endoscopy, Hirosaki University Hospital, Hirosaki 036-8563, Japan
| | - Tadashi Shimoyama
- Department of Internal Medicine, Aomori General Health Examination Center, Aomori 030-0962, Japan;
| | - Tetsu Arai
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
| | - Kazuki Akitaya
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
| | - Sae Fujiwara
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
| | - Hiroki Nomiya
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
| | - Yoshio Sasaki
- Sasaki Clinic of Gastroenterology and Internal Medicine, Aomori 030-0914, Japan;
| | - Kazuo Komai
- Komai Clinic of Gastroenterology and Internal Medicine, Aomori 030-0947, Japan;
| | | | - Yoshiharu Saito
- Shinjo Clinic of Gastroenterology and Internal Medicine, Aomori 038-0042, Japan;
| | - Hironobu Chiba
- Chiba Clinic of Gastroenterology and Internal Medicine, Hirosaki 036-8316, Japan;
| | - Hirotake Sakuraba
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
| | - Shinsaku Fukuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
| | - the RINGO Study Group
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; (K.Y.); (T.A.); (K.A.); (S.F.); (H.N.); (H.S.); (S.F.)
- Division of Endoscopy, Hirosaki University Hospital, Hirosaki 036-8563, Japan
- Department of Internal Medicine, Aomori General Health Examination Center, Aomori 030-0962, Japan;
- Sasaki Clinic of Gastroenterology and Internal Medicine, Aomori 030-0914, Japan;
- Komai Clinic of Gastroenterology and Internal Medicine, Aomori 030-0947, Japan;
- Sawada Clinic of Internal Medicine, Hirosaki 036-8261, Japan;
- Shinjo Clinic of Gastroenterology and Internal Medicine, Aomori 038-0042, Japan;
- Chiba Clinic of Gastroenterology and Internal Medicine, Hirosaki 036-8316, Japan;
- Risk Investigation of Gastric Cancer and Observation after Eradication Study Group, Hirosaki 036-8562, Japan
| |
Collapse
|
8
|
Komene T, Kai K, Kinpara K, Sato T, Hokazono E, Shimosawa T, Osawa S, Seimiya M. Development of a plasma maltose assay method as a screening test for upper gastrointestinal disorders. Ann Clin Biochem 2024; 61:303-308. [PMID: 38111970 DOI: 10.1177/00045632231224218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
BACKGROUND AND OBJECTIVE The disaccharide loading test is a method to assess gastric mucosal damage. Since Trelan-G75, which is used for the sugar tolerance test, contains disaccharide maltose, if maltose is detected at a high sensitivity in the sample blood used in the sugar tolerance test, screening for upper gastrointestinal mucosal damage can be made simultaneously with the sugar tolerance test for the diagnosis of diabetes. METHODS Glucose-6-phosphate is generated by treating maltose with maltose phosphorylase, β-phosphoglucomutase, and glucose-1,6-bisphosphate. Then, change in the absorbance at 405 nm is measured by the enzymatic cycling method using Thio-NADP, β-NADPH, and Glucose-6-phosphate dehydrogenase. After evaluating the optimal condition for this method, it is mounted on an automatic biochemical analyzer, and samples after the sugar tolerance test were assayed. RESULTS Regarding the performance of this method, the repeatability was 10-50 μmol/L with a CV of ≤1.1%. Concerning the assay range, a curve passing the origin with a range of linearity up to 120 μmol/L was obtained. No effect of dyes or sugars in the blood was noted. As a result of application to patients with gastric mucosal disorders (those who had a health checkup), significant differences were observed depending on the stage of atrophic gastritis. DISCUSSION This method has a high sensitivity and a high precision and can be used for high-speed analysis on an automatic analyzer. It has the potential to be used as a screening test for gastric mucosal damage.
Collapse
Affiliation(s)
- Tetsuya Komene
- Department of Medical Technology and Sciences, School of Health Sciences at Narita, International University of Health and Welfare, Japan
| | - Kotoko Kai
- International University of Health and Welfare, Narita Hospital, Japan
| | - Kiyoko Kinpara
- International University of Health and Welfare, Narita Hospital, Japan
| | - Tomoaki Sato
- International University of Health and Welfare, Narita Hospital, Japan
| | - Eisaku Hokazono
- Division of Medical Sciences and Technology, Department of Health Sciences, Graduate School of Medicine, Kyushu University, Japan
| | - Tatsuo Shimosawa
- International University of Health and Welfare, Narita Hospital, Japan
| | - Susumu Osawa
- Research Center for Micro Blood Analysis, Leisure, Inc., Japan; Medical Laboratory Sciences, Health Sciences, School of Health Sciences at Narita, International University of Health and Welfare Graduate School, Japan
| | - Masanori Seimiya
- Department of Medical Technology and Sciences, School of Health Sciences at Narita, International University of Health and Welfare, Japan
| |
Collapse
|
9
|
Li Z, Du S, Wang X, Zhang L, Liu X, Fan Q, Yang H, Gao R. Clinical effects of a combination of phenylbutazone and omeprazole on chronic lameness in Mongolian horses. Equine Vet J 2024; 56:562-572. [PMID: 37337455 DOI: 10.1111/evj.13962] [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: 01/30/2023] [Accepted: 05/07/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Phenylbutazone (PBZ) is the most commonly used drug to treat symptoms of lameness in horses; however, it is associated with adverse effects such as gastric ulcer syndrome (EGUS). Interestingly, many practitioners prescribe omeprazole (OME) concurrently with PBZ to prevent the development of EGUS. However, the efficacy and safety of this practice in Mongolian horses with chronic lameness remain unknown. OBJECTIVES To evaluate the clinical effects of a combination of PBZ and OME on chronic lameness in Mongolian horses. STUDY DESIGN Randomised block experimental design. METHODS Eighteen Mongolian horses with lameness score was ≥3 points, were divided into three treatment groups, with six horses in each group: placebo (CON), PBZ (4.4 mg/kg PO q. 24 h), or PBZ plus OME (4 mg/kg PO q. 24 h; PBZ + OME) in a randomised block design based on the initial lameness score. The horses were treated for 15 days. During this period, weekly gastroscopy, and physiological and biochemical tests were performed. RESULTS Both PBZ (median 1.0, interquartile range [IQR]: 0.8-1.3; p = 0.01) and PBZ + OME (median 1.0, IQR: 1.0-1.0; p = 0.01) significantly decreased the lameness score compared with before administration. In addition, PBZ significantly increased the equine glandular gastric disease (EGGD) score (3.0 ± 0.6, p < 0.001), GT-17 content (293.4 ± 21.8 pg/mL, p < 0.001), and pepsinogen-1 (PG1) content (295.3 ± 38.3 ng/mL, p < 0.001) compared with CON or PBZ + OME. However, it significantly reduced the total protein (53.6 ± 1.5 g/L, p < 0.05) and albumin (25.5 ± 1.8 g/L, p < 0.05) contents. Nevertheless, compared with PBZ, PBZ + OME significantly decreased the EGGD score (0.3 ± 0.5, p < 0.001) and significantly increased the gastric fluid pH (7.3 ± 0.5, p < 0.001), total protein content (62.5 ± 4.6 g/L, p = 0.009), and albumin content (29.4 ± 1.1 g/L, p = 0.004). Meanwhile, they significantly diminished the gastrin 17 (GT-17) (162.0 ± 21.0 pg/mL, p < 0.001) and PG1 (182.4 ± 22.5 ng/mL, p < 0.001) contents. MAIN LIMITATIONS Individual differences in horses were larger, but the sample size was small. There was larger interval between observations for each index. CONCLUSIONS Compared with PBZ alone, PBZ + OME had no therapeutic effect on chronic lameness; however, it reduced the occurrence of EGGD in Mongolian horses. Horses may be protected against chronic lameness and PBZ-induced EGGD by increasing the pH value, decreasing serum PG1 and GT-17 content, and preventing the reduction of myeloperoxidase content.
Collapse
Affiliation(s)
- Zhengyi Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Shan Du
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaomin Wang
- Agriculture and Animal Husbandry Bureau, Karaqin Banner, Chifeng, China
| | - Lanxin Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xinyu Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Quanrong Fan
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Huidi Yang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Ruifeng Gao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
10
|
Fischbach W, Bornschein J, Hoffmann JC, Koletzko S, Link A, Macke L, Malfertheiner P, Schütte K, Selgrad DM, Suerbaum S, Schulz C. Update S2k-Guideline Helicobacter pylori and gastroduodenal ulcer disease of the German Society of Gastroenterology, Digestive and Metabolic Diseases (DGVS). ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:261-321. [PMID: 38364851 DOI: 10.1055/a-2181-2225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Affiliation(s)
| | - Jan Bornschein
- Translational Gastroenterology Unit John, John Radcliffe Hospital Oxford University Hospitals, Oxford, United Kingdom
| | - Jörg C Hoffmann
- Medizinische Klinik I, St. Marien- und St. Annastiftskrankenhaus, Ludwigshafen, Deutschland
| | - Sibylle Koletzko
- Kinderklinik und Kinderpoliklinik im Dr. von Haunerschen Kinderspital, LMU-Klinikum Munich, Munich, Deutschland
- Department of Paediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, 10-719 Olsztyn, Poland
| | - Alexander Link
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Universitätsklinikum Magdeburg, Magdeburg, Deutschland
| | - Lukas Macke
- Medizinische Klinik und Poliklinik II Campus Großhadern, Universitätsklinikum Munich, Munich, Deutschland
- Deutsches Zentrum für Infektionsforschung, Standort Munich, Munich, Deutschland
| | - Peter Malfertheiner
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Universitätsklinikum Magdeburg, Magdeburg, Deutschland
- Medizinische Klinik und Poliklinik II Campus Großhadern, Universitätsklinikum Munich, Munich, Deutschland
| | - Kerstin Schütte
- Klinik für Allgemeine Innere Medizin und Gastroenterologie, Niels-Stensen-Kliniken Marienhospital Osnabrück, Osnabrück, Deutschland
| | - Dieter-Michael Selgrad
- Medizinische Klinik Gastroenterologie und Onkologie, Klinikum Fürstenfeldbruck, Fürstenfeldbruck, Deutschland
- Klinik für Innere Medizin 1, Universitätsklinikum Regensburg, Regensburg, Deutschland
| | - Sebastian Suerbaum
- Universität Munich, Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Munich, Deutschland
- Nationales Referenzzentrum Helicobacter pylori, Pettenkoferstr. 9a, 80336 Munich, Deutschland
- Deutsches Zentrum für Infektionsforschung, Standort Munich, Munich, Deutschland
| | - Christian Schulz
- Medizinische Klinik und Poliklinik II Campus Großhadern, Universitätsklinikum Munich, Munich, Deutschland
- Deutsches Zentrum für Infektionsforschung, Standort Munich, Munich, Deutschland
| |
Collapse
|
11
|
Khangai A, Akada J, Saruuljavkhlan B, Gantuya B, Azzaya D, Oyuntsetseg K, Davaadorj D, Uchida T, Matsumoto T, Yamaoka Y. Utilization of an Automated Latex Agglutination Turbidity Assay for Assessing Gastric Mucosal Alteration during Helicobacter pylori Infection. Gut Liver 2024; 18:60-69. [PMID: 37720994 PMCID: PMC10791506 DOI: 10.5009/gnl220464] [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: 10/27/2022] [Revised: 01/21/2023] [Accepted: 02/07/2023] [Indexed: 09/19/2023] Open
Abstract
Background/Aims : A latex agglutination turbidity (LA) assay to test for serum antibodies has been approved in Japan and Korea for mass screening of Helicobacter pylori infection. In this study, we evaluated the LA assay for diagnosing H. pylori infection and predicting gastric mucosal changes in a Mongolian population. Methods : In total, 484 individuals were classified into H. pylori-positive (n=356) and H. pylori-negative (n=128) groups, as determined by histology and H. pylori culture. Results : The best cutoff, sensitivity, and specificity values for the LA assay were 18.35 U/mL, 74.2%, and 65.6%, respectively. The LA values in the atrophic gastritis group were statistically higher than those in the other groups (healthy, chronic gastritis, intestinal metaplasia, and gastric cancer, p<0.0001). The cutoff value to distinguish the atrophic gastritis group from the other four groups was 32.0 U/mL, and its area under the curve was 0.673, which was the highest among the E-plate, pepsinogen (PG) I, PG II, and PG I/II ratio tests in our data. The odds ratios for atrophic gastritis determined by the LA assay and PG I test in multiple logistic regression were 2.5 and 1.9, respectively, which were significantly higher than for the other tests. Conclusions : The LA assay can determine the risk of atrophic gastritis, which in turn is a considerable risk factor for gastric cancer. We propose using this assay in combination with the PG I/II ratio to avoid missing gastric cancer patients who have a low LA value (less than 32.0 U/mL).
Collapse
Affiliation(s)
- Ayush Khangai
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
- The Gastroenterology Center, The First Central Hospital of Mongolia, Ulaanbaatar, Mongolia
| | - Junko Akada
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Batsaikhan Saruuljavkhlan
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Boldbaatar Gantuya
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
- Endoscopy Unit, Mongolia-Japan Teaching Hospital, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Dashdorj Azzaya
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
- Endoscopy Unit, Mongolia-Japan Teaching Hospital, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Khasag Oyuntsetseg
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
- Endoscopy Unit, Mongolia-Japan Teaching Hospital, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Duger Davaadorj
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
- Endoscopy Unit, Mongolia-Japan Teaching Hospital, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Tomohisa Uchida
- Department of Advanced Medical Sciences, Oita University Faculty of Medicine, Oita, Japan
| | - Takashi Matsumoto
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
- Research Center for Global and Local Infectious Diseases (RCGLID), Oita University, Oita, Japan
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
Lee SY, Ahn YS, Moon HW. Comparison between the GastroPanel test and the serum pepsinogen assay interpreted with the ABC method-A prospective study. Helicobacter 2024; 29:e13056. [PMID: 38402559 DOI: 10.1111/hel.13056] [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: 10/30/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND AND AIMS This study aimed to validate Helicobacter pylori serological and pepsinogen (PG) assays for detecting infection and gastric neoplasm. METHODS Individuals who underwent serum Chorus H. pylori and HBI PG assays were included from May to September 2023. The GastroPanel test was performed using the same blood sample. HBI assay findings were interpreted with the ABC method using the criteria of corpus atrophy (PG I ≤ 70 ng/mL & I/II ≤3) and advanced corpus atrophy (PG I ≤ 30 ng/mL & I/II ≤2). RESULTS A total of 144 H. pylori-infected and 184 non-infected Koreans were analyzed. The Chorus test (sensitivity 97.2%, specificity 89.1%) showed higher area under the curve (0.993 vs. 0.972, p = 0.003) than the GastroPanel test (sensitivity 95.8%, specificity 86.4%). Using the GastroSoft application, the incidence of gastric neoplasms was highest in the corpus atrophy group (50%), followed by the low acid-output (25.8%), H. pylori infection (11.6%), and antral atrophy (9.1%) groups. There were no gastric neoplasms in the normal and high acid output groups. Using the ABC method, the incidence of gastric neoplasms was highest in the corpus atrophy groups (23.8% in Groups C and D), followed by Group B (12.3%) and Group A (2.4%). Corpus atrophy interpreted with the GastroSoft showed poor agreement (k = 0.225) with corpus atrophy interpreted with the ABC method, whereas it showed excellent agreement (k = 0.854) with advanced corpus atrophy. CONCLUSIONS Although the Chorus test was more accurate than the GastroPanel test, both assays discriminated high-risk individuals by detecting atrophy or infection. There were no gastric neoplasms in the normal or high acid-output groups (GastroSoft application), and gastric neoplasm incidence was lowest in Group A (ABC method). Corpus atrophy determined by GastroSoft application is more consistent with advanced corpus atrophy determined by the ABC method than is corpus atrophy.
Collapse
Affiliation(s)
- Sun-Young Lee
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Yeon-Sun Ahn
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Hee-Won Moon
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Wei Q, Bai J, Ji J, Zhang W. Correlation analysis between the development of congenital heart disease in 100 newborns and the risk factors of pregnant mothers. Minerva Pediatr (Torino) 2023; 75:921-923. [PMID: 37155218 DOI: 10.23736/s2724-5276.23.07269-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Affiliation(s)
- Qian Wei
- Department of Pediatrics, Affiliated Hospital of Hebei University, Baoding, China
| | - Jinxiu Bai
- Department of Pediatrics, Affiliated Hospital of Hebei University, Baoding, China
| | - Jinglu Ji
- Department of Pediatrics, Affiliated Hospital of Hebei University, Baoding, China
| | - Wei Zhang
- Department of Pediatrics, Affiliated Hospital of Hebei University, Baoding, China -
| |
Collapse
|
14
|
Sugano K, Moss SF, Kuipers EJ. Gastric Intestinal Metaplasia: Real Culprit or Innocent Bystander as a Precancerous Condition for Gastric Cancer? Gastroenterology 2023; 165:1352-1366.e1. [PMID: 37652306 DOI: 10.1053/j.gastro.2023.08.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023]
Abstract
Gastric intestinal metaplasia (GIM), which denotes conversion of gastric mucosa into an intestinal phenotype, can occur in all regions of the stomach, including cardiac, fundic, and pyloric mucosa. Since the earliest description of GIM, its association with gastric cancer of the differentiated (intestinal) type has been a well-recognized concern. Many epidemiologic studies have confirmed GIM to be significantly associated with subsequent gastric cancer development. Helicobacter pylori, the principal etiologic factor for gastric cancer, plays the most important role in predisposing to GIM. Although the role of GIM in the stepwise progression model of gastric carcinogenesis (the so-called "Correa cascade") has come into question recently, we review the scientific evidence that strongly supports this long-standing model and propose a new progression model that builds on the Correa cascade. Eradication of H pylori is the most important method for preventing gastric cancer globally, but the effect of eradication on established GIM, is limited, if any. Endoscopic surveillance for GIM may, therefore, be necessary, especially when there is extensive corpus GIM. Recent advances in image-enhanced endoscopy with integrated artificial intelligence have facilitated the identification of GIM and neoplastic lesions, which will impact preventive strategies in the near future.
Collapse
Affiliation(s)
| | - Steven F Moss
- Alpert Medical School of Brown University, Providence, Rhode Island
| | - Ernst J Kuipers
- Erasmus Medical Center, Rotterdam and Minister, Ministry of Health, Welfare, and Sport, Hague, The Netherlands
| |
Collapse
|
15
|
Joshi N, Bhat F, Bellad A, Sathe G, Jain A, Chavan S, Sirdeshmukh R, Pandey A. Urinary Proteomics for Discovery of Gastric Cancer Biomarkers to Enable Precision Clinical Oncology. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:361-371. [PMID: 37579183 PMCID: PMC10625469 DOI: 10.1089/omi.2023.0077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
For precision in clinical oncology practice, detection of tumor-derived peptides and proteins in urine offers an attractive and noninvasive alternative for diagnostic or screening purposes. In this study, we report comparative quantitative proteomic profiling of urine samples from patients with gastric cancer and healthy controls using tandem mass tags-based multiplexed mass spectrometry approach. We identified 1504 proteins, of which 246 were differentially expressed in gastric cancer cases. Notably, ephrin A1 (EFNA1), pepsinogen A3 (PGA3), sortilin 1 (SORT1), and vitronectin (VTN) were among the upregulated proteins, which are known to play crucial roles in the progression of gastric cancer. We also found other overexpressed proteins, including shisa family member 5 (SHISA5), mucin like 1 (MUCL1), and leukocyte cell derived chemotaxin 2 (LECT2), which had not previously been linked to gastric cancer. Using a novel approach for targeted proteomics, SureQuant, we validated changes in abundance of a subset of proteins discovered in this study. We confirmed the overexpression of vitronectin and sortilin 1 in an independent set of urine samples. Altogether, this study provides molecular candidates for biomarker development in gastric cancer, and the findings also support the promise of urinary proteomics for noninvasive diagnostics and personalized/precision medicine in the oncology clinic.
Collapse
Affiliation(s)
- Neha Joshi
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Firdous Bhat
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Anikha Bellad
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Gajanan Sathe
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Anu Jain
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sandip Chavan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ravi Sirdeshmukh
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Akhilesh Pandey
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Nagasaki N, Takigawa H, Ito M, Boda T, Kotachi T, Hayashi R, Yuge R, Urabe Y, Oka S, Tanaka S. Diagnostic performance of the normal range of gastrin calculated using strict criteria based on a combination of serum markers and pathological evaluation for detecting gastritis: a retrospective study. BMC Gastroenterol 2023; 23:167. [PMID: 37210509 DOI: 10.1186/s12876-023-02816-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 05/12/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND The ABC method, which combines the pepsinogen method and anti-Helicobacter pylori antibody titers, has been used for risk screening for gastric cancer in Japan. However, it has been reported that there are cases of gastritis and carcinogenesis risk even in group A, which is considered to be a low-risk group based on the ABC method. Currently, in group A, endoscopic examination is needed to strictly discriminate "patients without gastritis" (defined as true A patients) from those "with gastritis." A simple and minimally invasive diagnostic criterion for gastritis using serological markers is desirable. In this study, we aimed to identify the normal serum gastrin concentrations in normal stomach cases based on pathological diagnosis and investigate the usefulness of serum gastrin concentrations in diagnosing gastritis. METHODS Patients who underwent endoscopy and blood tests at Hiroshima University Hospital were enrolled in the study and categorized into the "pathologically-evaluated group" and "endoscopically-evaluated group," according to the evaluation method of atrophic gastritis. Initially, we measured serum gastrin concentrations in the normal stomach cases in the pathologically-evaluated group and calculated the normal range of serum gastrin concentrations. We used the upper limit of this normal range of serum gastrin concentrations and performed a validation study to determine its usefulness as a diagnostic marker for distinguishing between cases of gastritis and true A in the endoscopically-evaluated group. RESULTS The 95th percentile of serum gastrin concentrations in pathologically-evaluated normal stomach cases was 34.12-126.03 pg/mL. Using the upper limit of this normal range of serum gastrin concentrations, the sensitivity, specificity, positive predictive value, and negative predictive value for gastritis were 52.8%, 92.6%, 97.0%, and 31.0%, respectively. Additionally, the receiver operating characteristic (ROC) curve for the endoscopically-evaluated group showed an area under the ROC curve of 0.80. CONCLUSION The gastrin cut-off value of 126 pg/mL has a good positive predictive value (97.0%) for detecting gastritis positing its use as a marker for cases requiring endoscopy. However, the identification of patients with gastritis having normal serum gastrin concentrations due to insufficient sensitivity remains a challenge for the future.
Collapse
Affiliation(s)
- Naoko Nagasaki
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | - Hidehiko Takigawa
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8553, Japan.
| | - Masanori Ito
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Tomoyuki Boda
- Department of Internal Medicine, Hiroshima Memorial Hospital, Hiroshima, Japan
| | - Takahiro Kotachi
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8553, Japan
| | - Ryohei Hayashi
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8553, Japan
| | - Ryo Yuge
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8553, Japan
| | - Yuji Urabe
- Division of Regeneration and Medicine Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Shiro Oka
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shinji Tanaka
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8553, Japan
| |
Collapse
|
17
|
Aktualisierte S2k-Leitlinie Helicobacter
pylori und gastroduodenale Ulkuskrankheit der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) – Juli 2022 – AWMF-Registernummer: 021–001. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2023; 61:544-606. [PMID: 37146633 DOI: 10.1055/a-1975-0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
|
18
|
Malfertheiner P, Camargo MC, El-Omar E, Liou JM, Peek R, Schulz C, Smith SI, Suerbaum S. Helicobacter pylori infection. Nat Rev Dis Primers 2023; 9:19. [PMID: 37081005 PMCID: PMC11558793 DOI: 10.1038/s41572-023-00431-8] [Citation(s) in RCA: 299] [Impact Index Per Article: 149.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 04/22/2023]
Abstract
Helicobacter pylori infection causes chronic gastritis, which can progress to severe gastroduodenal pathologies, including peptic ulcer, gastric cancer and gastric mucosa-associated lymphoid tissue lymphoma. H. pylori is usually transmitted in childhood and persists for life if untreated. The infection affects around half of the population in the world but prevalence varies according to location and sanitation standards. H. pylori has unique properties to colonize gastric epithelium in an acidic environment. The pathophysiology of H. pylori infection is dependent on complex bacterial virulence mechanisms and their interaction with the host immune system and environmental factors, resulting in distinct gastritis phenotypes that determine possible progression to different gastroduodenal pathologies. The causative role of H. pylori infection in gastric cancer development presents the opportunity for preventive screen-and-treat strategies. Invasive, endoscopy-based and non-invasive methods, including breath, stool and serological tests, are used in the diagnosis of H. pylori infection. Their use depends on the specific individual patient history and local availability. H. pylori treatment consists of a strong acid suppressant in various combinations with antibiotics and/or bismuth. The dramatic increase in resistance to key antibiotics used in H. pylori eradication demands antibiotic susceptibility testing, surveillance of resistance and antibiotic stewardship.
Collapse
Affiliation(s)
- Peter Malfertheiner
- Medical Department II, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany.
- Medical Department Klinik of Gastroenterology, Hepatology and Infectiology, Otto-von-Guericke Universität, Magdeburg, Germany.
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Emad El-Omar
- Microbiome Research Centre, St George & Sutherland Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jyh-Ming Liou
- Department of Internal Medicine, National Taiwan University Cancer Center, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Richard Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christian Schulz
- Medical Department II, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- DZIF Deutsches Zentrum für Infektionsforschung, Partner Site Munich, Munich, Germany
| | - Stella I Smith
- Department of Molecular Biology and Biotechnology, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Sebastian Suerbaum
- DZIF Deutsches Zentrum für Infektionsforschung, Partner Site Munich, Munich, Germany
- Max von Pettenkofer Institute, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- National Reference Center for Helicobacter pylori, Munich, Germany
| |
Collapse
|
19
|
Miftahussurur M, Waskito LA, Syam AF, Nusi IA, Wibawa IDN, Rezkitha YAA, Fauzia KA, Siregar GA, Akil F, Waleleng BJ, Saudale AMJ, Abubakar A, Maulahela H, Richardo M, Rahman A, Namara YS, Sudarmo E, Adi P, Maimunah U, Setiawan PB, Doohan D, Uchida T, Dewayani A, Rejeki PS, Sugihartono T, Yamaoka Y. Serum pepsinogen level as a biomarker for atrophy, reflux esophagitis, and gastric cancer screening in Indonesia. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2022; 27:90. [PMID: 36685023 PMCID: PMC9854938 DOI: 10.4103/jrms.jrms_983_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 07/16/2022] [Accepted: 08/08/2022] [Indexed: 12/24/2022]
Abstract
Background Chronic dyspepsia's symptoms are frequently seen in primary to tertiary healthcare in Indonesia. This study aimed to describe the potential usability of pepsinogen (PG) values in determining gastric mucosal conditions, including superficial gastritis and atrophic gastritis. Materials and Methods We recruited 646 adult dyspeptic patients and then analyzed PG values (including PGI, PGII, and PGI/II ratio) with endoscopic findings, gastric mucosal damages, and Helicobacter pylori infection. The gastric mucosal damage and H. pylori infection were evaluated using histological examination based on the updated Sydney system. Results Among 646 enrolled patients, 308 (47.2%), 212 (32.8%), 91 (14.1%), 34 (5.2%), and 1 (0.2%) patient were diagnosed with normal mucosa, gastritis, reflux esophagitis, peptic ulcer disease, and gastric cancer, respectively. Significant differences in PGI, PGII, and PGI/II ratio values were observed among ethnic groups (all P < 0.01). The PGI and PGII levels were significantly higher and PGI/II was significantly lower in H. pylori-infected patients than in uninfected ones (all P < 0.001). The optimal cutoff value for PGII and PGI/II was 12.45 ng/mL with an area under the curve (AUC) value of 0.755 (0.702-0.811), sensitivity 59.3%, and specificity 77.1%; and 4.75 with AUC value of 0.821 (0.763-0.855), sensitivity 81.5%, and specificity 78.7%, respectively, to determine moderate-severe atrophy. Conclusion Serum PG levels, a useful biomarker, represent the endoscopic findings, especially for reflux esophagitis. In addition, the benefits of PG values detecting atrophic gastritis were limited to moderate-severe atrophic gastritis. This usefulness requires careful attention for several ethnic groups in Indonesia.
Collapse
Affiliation(s)
- Muhammad Miftahussurur
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Langgeng Agung Waskito
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Ari Fahrial Syam
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Iswan Abbas Nusi
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - I Dewa Nyoman Wibawa
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine University of Udayana, Denpasar, Indonesia
| | - Yudith Annisa Ayu Rezkitha
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Faculty of Medicine, University of Muhammadiyah Surabaya, Surabaya, Indonesia
| | - Kartika Afrida Fauzia
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Gontar Alamsyah Siregar
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, University of Sumatera Utara, Medan, Indonesia
| | - Fardah Akil
- Center of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Bradley Jimmy Waleleng
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, University of Sam Ratulangi, Prof. Dr. RD Kandou Hospital, Manado, Indonesia
| | | | - Azzaki Abubakar
- Division of Gastroenterohepatology, Department of Internal Medicine, Dr. Zainoel Abidin General Hospital, Banda Aceh, Indonesia
| | - Hasan Maulahela
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Marselino Richardo
- Department of Internal Medicine, Merauke City General Hospital, Merauke, Indonesia
| | - Abdul Rahman
- Department of Internal Medicine, Kolaka General Hospital, Kolaka, Indonesia
| | - Yoma Sari Namara
- Department of Internal Medicine, Anutapura General Hospital, Palu, Indonesia
| | - Eko Sudarmo
- Department of Internal Medicine, Dr. Hasan Busori General Hospital, Ternate, Indonesia
| | - Pangestu Adi
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Ummi Maimunah
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Poernomo Boedi Setiawan
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Dalla Doohan
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Tomohisa Uchida
- Department of Molecular Pathology, Oita University Faculty of Medicine, Yufu, Japan
| | - Astri Dewayani
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Infectious Disease Control, Oita University Faculty of Medicine, Yufu, Japan
| | - Purwo Sri Rejeki
- Department of Physiology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Titong Sugihartono
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Yoshio Yamaoka
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, Texas, United States
- Global Oita Medical Advanced Research Center for Health, Yufu, Japan
| |
Collapse
|
20
|
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
|
21
|
Haque E, Esmail A, Muhsen I, Salah H, Abdelrahim M. Recent Trends and Advancements in the Diagnosis and Management of Gastric Cancer. Cancers (Basel) 2022; 14:5615. [PMID: 36428707 PMCID: PMC9688354 DOI: 10.3390/cancers14225615] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Gastric cancer is an enigmatic malignancy that has recently been shown to be increasing in incidence globally. There has been recent progress in emerging technologies for the diagnosis and treatment of the disease. Improvements in non-invasive diagnostic techniques with serological tests and biomarkers have led to decreased use of invasive procedures such as endoscopy. A multidisciplinary approach is used to treat gastric cancer, with recent significant advancements in systemic therapies used in combination with cytotoxic chemotherapies. New therapeutic targets have been identified and clinical trials are taking place to assess their efficacy and safety. In this review, we provide an overview of the current and emerging treatment strategies and diagnostic techniques for gastric cancer.
Collapse
Affiliation(s)
- Emaan Haque
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdullah Esmail
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
| | - Ibrahim Muhsen
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haneen Salah
- Department of Pathology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
- Cockrell Center for Advanced Therapeutic Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
22
|
Razuka-Ebela D, Polaka I, Daugule I, Parshutin S, Santare D, Ebela I, Rudzite D, Vangravs R, Herrero R, Young Park J, Leja M. Lifestyle and dietary factors associated with serologically detected gastric atrophy in a Caucasian population in the GISTAR study. Eur J Cancer Prev 2022; 31:442-450. [PMID: 35131967 DOI: 10.1097/cej.0000000000000723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To identify dietary and lifestyle factors associated with decreased pepsinogen levels indicative of gastric atrophy. METHODS Participants aged 40 to 64 from the "Multicentric randomized study of H. pylori eradication and pepsinogen testing for prevention of gastric cancer mortality (GISTAR study)" in Latvia tested for serum pepsinogen, as well as for Helicobacter pylori infection by 13 C-urea breath test or serology were included. Data on sex, age, education, employment, diet, smoking, alcohol and proton pump inhibitor use were obtained by survey and compared for participants with and without serologically detected gastric atrophy defined as pepsinogen I/pepsinogen II ≤ 2 and pepsinogen I ≤ 30 ng/mL. RESULTS Of 3001 participants (median age 53, interquartile range, 11.0, 36.9% male) 52.8% had H. pylori and 7.7% had serologically detected gastric atrophy. In multivariate analysis, increasing age, consumption of alcohol, coffee, and onions were positively, while H. pylori , former smoking, pickled product and proton pump inhibitor use were inversely associated with gastric atrophy. Pepsinogen values were higher in smokers and those with H. pylori . Pepsinogen ratio was lower in those with H. pylori . When stratifying by H. pylori presence, significantly higher pepsinogen levels remained for smokers without H. pylori . CONCLUSION Several dietary factors and smoking were associated with serologically detected gastric atrophy. Pepsinogen levels differed by smoking and H. pylori status, which may affect the serologic detection of gastric atrophy. There seems to be a complicated interaction between multiple factors. A prospective study including atrophy determined by both serology and histology is necessary.
Collapse
Affiliation(s)
- Danute Razuka-Ebela
- Faculty of Medicine
- Institute of Clinical and Preventive Medicine, University of Latvia
| | - Inese Polaka
- Institute of Clinical and Preventive Medicine, University of Latvia
| | - Ilva Daugule
- Faculty of Medicine
- Institute of Clinical and Preventive Medicine, University of Latvia
| | - Sergei Parshutin
- Institute of Clinical and Preventive Medicine, University of Latvia
| | - Daiga Santare
- Faculty of Medicine
- Institute of Clinical and Preventive Medicine, University of Latvia
| | | | - Dace Rudzite
- Institute of Clinical and Preventive Medicine, University of Latvia
- Riga East University Hospital, Riga, Latvia
| | - Reinis Vangravs
- Institute of Clinical and Preventive Medicine, University of Latvia
| | - Rolando Herrero
- International Agency for Research on Cancer, Lyon, France
- Agencia Costarricense de Investigaciones Biomedicas, Fundación INCIENSA, Costa Rica
| | - Jin Young Park
- International Agency for Research on Cancer, Lyon, France
| | - Marcis Leja
- Faculty of Medicine
- Institute of Clinical and Preventive Medicine, University of Latvia
| |
Collapse
|
23
|
Demiryas S, Orhan A. Advancements in Gastrointestinal System Biomarkers. Biomark Med 2022. [DOI: 10.2174/9789815040463122010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The requirement for diagnostic surgical operations of gastrointestinal system
diseases significantly decreases with the help of proper diagnostic tools. These
modalities are also beneficial for identifying postoperative complications, allowing us
to diagnose them in earlier stages, and increasing the postoperative survival rates.
Biomarkers are considered an integral part of diagnostic examinations. Developments
of biomarkers used for diagnosing and treating abdominal diseases are essential for
improving our capabilities in non-invasive monitoring. In this chapter, we review both
the novel and the routine biomarkers in the diagnosis and follow-up of gastrointestinal
system diseases.
Collapse
Affiliation(s)
- Suleyman Demiryas
- Department of General Surgery, Cerrahpaşa School of Medicine, IIstanbul University –
Cerrahpaşa, Istanbul, Turkey
| | - Anıl Orhan
- Department of General Surgery, Haseki Training and Research Hospital, University of Health
Sciences, Istanbul, Turkey
| |
Collapse
|
24
|
Nagasaki N, Ito M, Boda T, Kotachi T, Takigawa H, Oka S, Tanaka S. Identification of Helicobacter pylori-related gastric cancer risk using serological gastritis markers and endoscopic findings: a large-scale retrospective cohort study. BMC Gastroenterol 2022; 22:299. [PMID: 35725370 PMCID: PMC9210732 DOI: 10.1186/s12876-022-02381-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/14/2022] [Indexed: 11/26/2022] Open
Abstract
Background Gastric cancer remains a severe public health problem worldwide, particularly in Japan. Recent studies have demonstrated that serum markers are beneficial for risk stratification in gastric cancer development. We aimed to evaluate the usefulness of serum markers either alone or in combination (serum markers plus endoscopy) for effective risk stratification of gastric cancer development. Methods We enrolled 22,736 patients aged 20–95 years who underwent blood sampling and endoscopic examination at Hiroshima University Hospital in Japan between 1990 and 2014. The serum pepsinogen (PG) levels and anti-Helicobacter pylori antibody (Hp-Ab) titers were evaluated in each patient. The enrolled patients were matched with the database of the Hiroshima Prefecture Regional Cancer Registry. We processed the medical records and excluded patients with possible confounding factors for PG levels, such as proton pump inhibitor use, prior successful eradication therapy, post-gastrectomy, severe hepatorenal dysfunction, Zollinger–Ellison syndrome, and autoimmune gastritis. Among the remaining 5131 patients, we reviewed records of endoscopic examinations and selected 1507 patients (mean age, 62.5 years; 985 men and 522 women) who underwent endoscopic examination more than three months after blood sampling.
First, based on the ABC method, patients were classified as follows: High PG levels and negative Hp-Ab, group A, high PG levels and positive Hp-Ab, group B, low PG levels and positive Hp-Ab, group C, and low PG levels and negative Hp-Ab, group D. Group A was further classified into two subgroups using endoscopic findings: true A without atrophic gastritis and pseudo A with atrophic gastritis. All patients underwent annual endoscopy follow-up. Results Among the 1,507 patients (mean age, 62.5 years; 985 men), 24 were diagnosed with newly developed gastric cancer. No significant difference in cancer development was found between group A (PG negative and Hp-Ab negative) and the other groups. Remarkably, no true A group subjects developed gastric cancer. Conclusions The combination of serum markers and endoscopic findings is essential for the risk evaluation of gastric cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02381-z.
Collapse
Affiliation(s)
- Naoko Nagasaki
- Department of Gastroenterology and Metabolism, Hiroshima University Hospital, Hiroshima, Japan
| | - Masanori Ito
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Tomoyuki Boda
- Department of Internal Medicine, Hiroshima Memorial Hospital, Hiroshima, Japan
| | - Takahiro Kotachi
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Hidehiko Takigawa
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Shiro Oka
- Department of Gastroenterology and Metabolism, Hiroshima University Hospital, Hiroshima, Japan
| | - Shinji Tanaka
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| |
Collapse
|
25
|
Razuka-Ebela D, Polaka I, Daugule I, Parshutin S, Santare D, Ebela I, Rudzite D, Vangravs R, Herrero R, Young Park J, Leja M. Factors Associated with False Negative Results in Serum Pepsinogen Testing for Precancerous Gastric Lesions in a European Population in the GISTAR Study. Diagnostics (Basel) 2022; 12:1166. [PMID: 35626319 PMCID: PMC9139962 DOI: 10.3390/diagnostics12051166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023] Open
Abstract
The accuracy of plasma pepsinogen (Pg) as a marker for precancerous gastric lesions (PGL) has shown variable results. We aimed to identify factors associated with false negative (FN) cases in Pg testing and to adjust cut-off values for these factors in order to improve Pg yield. Plasma Pg was measured and upper endoscopy with biopsy was performed within the "Multicentric randomized study of Helicobacter pylori eradication and pepsinogen testing for prevention of gastric cancer mortality: the GISTAR study". A multivariable logistic model was built for FN and multiple factors. Values of Pg were compared and sensitivity and specificity were calculated using pre-existing Pg cut-offs for factors showing strong associations with FN. New cut-offs were calculated for factors that showed substantially lower sensitivity. Of 1210 participants, 364 (30.1%) had histologically confirmed PGL, of which 160 (44.0%) were FN. Current smokers, men, and H. pylori positives were more likely FN. Smoking in H. pylori negatives was associated with a higher Pg I/II ratio and substantially lower sensitivity of Pg testing than in other groups. Adjusting Pg cut-offs for current smokers by H. pylori presence improved sensitivity for detecting PGL in this group. Our study suggests that adjusting Pg cut-offs for current smokers by H. pylori status could improve Pg test performance.
Collapse
Affiliation(s)
- Danute Razuka-Ebela
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Inese Polaka
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Ilva Daugule
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Sergei Parshutin
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Daiga Santare
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Inguna Ebela
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
| | - Dace Rudzite
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
- Riga East University Hospital, Hipokrāta iela 2, LV-1038 Riga, Latvia
| | - Reinis Vangravs
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Rolando Herrero
- International Agency for Research on Cancer, 150 Cours Albert Thomas, CEDEX 08, 69372 Lyon, France; (R.H.); (J.Y.P.)
- Agencia Costarricense de Investigaciones Biomedicas, Fundación INCIENSA, Avenida 9a Calles 64-68, San Jose 2250, Costa Rica
| | - Jin Young Park
- International Agency for Research on Cancer, 150 Cours Albert Thomas, CEDEX 08, 69372 Lyon, France; (R.H.); (J.Y.P.)
| | - Marcis Leja
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| |
Collapse
|
26
|
Wilunda C, Yamaji T, Iwasaki M, Inoue M, Tsugane S, Sawada N. Meat consumption and gastric cancer risk: the Japan Public Health Center-based Prospective Study. Am J Clin Nutr 2022; 115:652-661. [PMID: 34788357 DOI: 10.1093/ajcn/nqab367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/01/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The association of meat consumption with gastric cancer is inconclusive. OBJECTIVES We examined the association of meat consumption with gastric cancer risk among Japanese men and women. METHODS This cohort study included 42,328 male and 48,176 female participants of the Japan Public Health Center-based Prospective Study, who were aged 45 to 74 y at recruitment. Dietary intake data were collected from 1 January 1995 to 31 December 1999 using a validated food-frequency questionnaire. HRs and 95% CIs for gastric cancer were estimated using Cox proportional hazards regression models. RESULTS During a mean follow-up of 15 y, 1868 male and 833 female incident gastric cancer cases were identified. Intake of total and subtypes of meat was not associated with total gastric cancer. However, higher chicken consumption was associated with reduced distal gastric cancer risk in women (HR for quintile 5 vs. quintile 1: 0.75; 95% CI: 0.56, 0.99; P-trend = 0.027), with a similar but nonsignificant risk reduction among women with Helicobacter pylori (HR: 0.59; 95% CI: 0.29, 1.20; P-trend = 0.06) in subgroup analysis. CONCLUSIONS Meat consumption was not associated with total gastric cancer risk.
Collapse
Affiliation(s)
- Calistus Wilunda
- Epidemiology and Prevention Group, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Taiki Yamaji
- Epidemiology and Prevention Group, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Motoki Iwasaki
- Epidemiology and Prevention Group, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Manami Inoue
- Epidemiology and Prevention Group, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Institute for Cancer Control, National Cancer Center, Tokyo, Japan.,National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | - Norie Sawada
- Epidemiology and Prevention Group, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| |
Collapse
|
27
|
Xiang W, Wang R, Bai D, Yu TH, Chen XZ. Helicobacter Pylori Related Gastric Cancer Screening and Cost-Effectiveness Analysis: A Hospital-Based Cross-Sectional Study (SIGES). Nutr Cancer 2022; 74:2769-2778. [PMID: 35876250 DOI: 10.1080/01635581.2021.2022168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Wen Xiang
- Department of Gastrointestinal Surgery & Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Wang
- Department of Gastroenterology, Nursing Section, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Bai
- Department of Gastrointestinal Surgery & Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Tian-Hang Yu
- Department of Gastrointestinal Surgery & Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Xin-Zu Chen
- Department of Gastrointestinal Surgery & Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastrointestinal and Hernia Surgery, the Second People’s Hospital of Yibin City, West China Yibin Hospital, Sichuan University, Yibin, China
- Department of General Surgery, the First People’s Hospital of Longquanyi District, West China Longquan Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
28
|
Fujiwara SI, Konno M, Watanabe S, Toita N, Takahashi M. Serum pepsinogen cut-off values in Helicobacter pylori-infected children. Pediatr Int 2022; 64:e15247. [PMID: 36257610 DOI: 10.1111/ped.15247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the standard values for gender- and age-stratified serum pepsinogen (sPG) in Helicobacter pylori (H. pylori) non-infected children and to determine the optimal cut-off values of sPG for predicting H. pylori-infected gastritis in children. METHODS A prospective study for determination of sPG levels was performed in children with epigastric pain who underwent esophagogastroduodenoscopy over the past 16 years. After excluding subjects diagnosed with inflammatory bowel diseases, eosinophilic gastrointestinal disorders, or immunoglobulin A vasculitis, the diagnosis of H. pylori infection was defined by positive tissue culture or concordant-positive results for histology and the rapid urease test. RESULTS A total of 405 subjects were diagnosed as being H. pylori-infected (79) or non-infected (326). In the H. pylori non-infected group, there were no significant differences in sPG levels among age groups; males had higher sPG I and sPG II levels than females. In the H. pylori-infected group, sPG I and sPG II levels were significantly higher and the sPG I/II ratio was lower than those in the non-infected group. In receiver operating characteristics analyses in diagnosing H. pylori infection, the areas under the curves for sPG I, sPG II and sPG I/II ratio were 0.896, 0.980, and 0.946, respectively. The optimal cut-off value of sPG II of ≥9.0 ng/mL was considered positive for H. pylori infection (sensitivity: 92.4%, specificity: 93.9%). CONCLUSIONS The optimal cut-off value of sPG II of ≥9.0 ng/mL may be a good predictor of H. pylori-infected gastritis in children.
Collapse
Affiliation(s)
- Shin-Ichi Fujiwara
- Department of Pediatrics, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Mutsuko Konno
- Department of Pediatrics, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Satoshi Watanabe
- Department of Pediatrics, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Nariaki Toita
- Department of Pediatrics, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Michiko Takahashi
- Department of Pediatrics, Sapporo Kosei General Hospital, Sapporo, Japan
| |
Collapse
|
29
|
Walker EV, Girgis S, Yuan Y, Goodman KJ. Community-driven research in the canadian arctic: dietary exposure to methylmercury and gastric health outcomes. Int J Circumpolar Health 2021; 80:1889879. [PMID: 33646085 PMCID: PMC7928021 DOI: 10.1080/22423982.2021.1889879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/24/2021] [Accepted: 02/09/2021] [Indexed: 10/31/2022] Open
Abstract
Indigenous Arctic Canadians have a higher prevalence of gastric neoplasms relative to North Americans of European ancestry. We investigated the hypothesis that low-dose methylmercury exposure from eating fish/whale increases the risk of gastric cancer in Arctic communities. We used intermediate endpoints from an established model of gastric carcinogenesis: intestinal metaplasia, atrophy, and severe chronic gastritis. During 2008-2012, we obtained gastric biopsies from participants of community-driven projects in 3 communities. In 2016, we collected hair samples to measure methylmercury levels and interviewed them about diet. In cross-sectional analysis, logistic regression estimated odds ratios for the estimated effect of hair-methylmercury concentration on the prevalence of each pathology outcome stratified by selenium intake. Among 80 participants, prevalence of intestinal metaplasia, atrophy and severe chronic gastritis was 17, 29 and 38%, respectively. Adjusted Odds of severe chronic gastritis and atrophy were highest at hair-methylmercury concentrations ≥1μg/g when estimated selenium intake was 0, and approached 0 for all methylmercury levels as estimated selenium intake increased. Gastric pathology increased with methylmercury exposure when selenium intake was low. Though limited by small numbers, these findings suggest selenium ingested by eating fish/whale may counter harmful effects of methylmercury exposure in Arctic populations.
Collapse
Affiliation(s)
- Emily V. Walker
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Safwat Girgis
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Yan Yuan
- Department of Public Health Sciences, University of Alberta, Edmonton, Canada
| | - Karen J. Goodman
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Public Health Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
30
|
Baek SM, Kim N, Kwon YJ, Lee HS, Kim HY, Lee J, Yoon H, Shin CM, Park YS, Lee DH. Role of Serum Pepsinogen II and Helicobacter pylori Status in the Detection of Diffuse-Type Early Gastric Cancer in Young Individuals in South Korea. Gut Liver 2021; 14:439-449. [PMID: 31533397 PMCID: PMC7366145 DOI: 10.5009/gnl19091] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/25/2019] [Accepted: 07/12/2019] [Indexed: 12/24/2022] Open
Abstract
Background/Aims The utility of serum pepsinogen (sPG) I and the sPGI/II ratio as biomarkers for screening individuals with gastric cancer (GC) has not been established in Korea. The aim of this study was to define the role of sPG, especially sPGII, in GC screening. Methods This study enrolled 2,940 subjects, including patients with GC (n=1,124) or gastric dysplasia (n=353) and controls (n=1,463). Tests to determine sPG levels and Helicobacter pylori (HP) infection status were performed. Area under the curve and receiver operating characteristic curve were calculated to identify the optimal cutoff values for sPG. The usefulness of sPG levels for the detection of GC and gastric dysplasia was validated by multivariate logistic regression. Results The sPGI/II ratio was associated with the risk of gastric dysplasia and advanced-stage intestinal-type GC (IGC). In contrast, sPGII was associated with the risk of early-stage diffuse-type GC (DGC). Significantly higher risk was indicated by an sPGI/II ratio <3 for gastric dysplasia and advanced-stage IGC and by sPGII levels ≥20 µg/L for early-stage DGC. Positive HP status showed a stronger association with DGC than with IGC. When sPGII level and HP status were combined, the prevalence of DGC was higher in the ≥20 µg/L sPGII and HP-positive group. Age younger than 40 years was strongly related to early-stage DGC, especially in females (odds ratio, 21.00; p=0.006). Conclusions sPGII ≥20 ng/mL and positive HP status suggest a risk of early-stage DGC, particularly in young adult females in South Korea.
Collapse
Affiliation(s)
- Sung Min Baek
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University, Seongnam, Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
| | - Young Jae Kwon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyun Young Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jaebong Lee
- Division of Statistics in Medical Research Collaborating Center, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University, Seongnam, Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
31
|
Han XL, Yi CL, Ma JD, He Y, Wu LM, Wang YF, Yang HJ, Liang DY, Shi JF. Clinical Value of Pepsinogen in the Screening, Prevention, and Diagnosis of Gastric Cancer. Lab Med 2021; 53:71-77. [PMID: 34508270 DOI: 10.1093/labmed/lmab035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To compare the levels of serum pepsinogen (PG) in patients with gastric cancer (GC), patients with atrophic gastritis (AG), and healthy donors. Also, we explored the clinical value of PG detection for the diagnosis and treatment of GC. METHODS The PG level in peripheral blood from patients and heathy donors was determined using an Abbott automatic chemiluminescence instrument. The study included 117 patients with GC confirmed by gastroscopy and histopathology, of whom 13 patients had cancer at stage I, 47 at stage II, 41 at stage III, and 16 at stage IV. The AG group included 122 patients, and the control group had 120 healthy donors. The relationship between serum PG levels and the occurrence and development of GC, as well as the evaluation of the clinical value of diagnostic tests based on serum PG detection, were investigated by receiver operating characteristic (ROC) curve analyses. RESULTS Pepsinogen I (PGI) levels gradually decreased from the control group, the AG group, and the GC group. PGI exhibited high diagnostic value for GC (area under the curve [AUC], 0.834; cutoff, 51.2 ng/mL, sensitivity, 81.7%; specificity, 68.4%), PGII (AUC, 0.587; cutoff value, 13.05 ng/mL; sensitivity, 65.8%; specificity, 53.8%), and PGR (AUC, 0.752; cutoff, 5.65; sensitivity, 54.2%; specificity, 87.2%). The occurrence of GC was negatively correlated with serum levels of PGI (B = -0.054; OR = 0.947; 95% confidence interval [CI], 0.925-0.970; P <.001) and PGR (B = -0.420; OR = 0.657; 95% CI, 0.499-0.864; P = .003). CONCLUSIONS The combined detection of PGI, PGII, and PGR has important clinical value for the screening, prevention, and diagnosis of GC and could allow for earlier detection, diagnosis, and treatment of GC.
Collapse
Affiliation(s)
- Xiao-Lei Han
- Department of Clinical Laboratory, Anting Hospital, Jiading District, Shanghai, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chang-Lin Yi
- Department of Clinical Laboratory, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Jin-Dan Ma
- Department of Clinical Laboratory, Anting Hospital, Jiading District, Shanghai, China
| | - Yanhong He
- Department of Clinical Laboratory, Anting Hospital, Jiading District, Shanghai, China
| | - La-Mei Wu
- Department of Clinical Laboratory, Anting Hospital, Jiading District, Shanghai, China
| | - Yun-Feng Wang
- Digestive Internal Medicine, Kunshan Branch of Shanghai Cancer Hospital, Shanghai, China
| | - Hui-Jian Yang
- Department of Clinical Laboratory, Dongfang Hospital affiliated to Tongji University, Shanghai, China
| | - Dong-Yu Liang
- Department of Clinical Laboratory, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jin-Fang Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
32
|
Yang TW, Wang CC, Hung WC, Liu YH, Sung WW, Tsai MC. Improvement in the Mortality-to-Incidence Ratios for Gastric Cancer in Developed Countries With High Health Expenditures. Front Public Health 2021; 9:713895. [PMID: 34485236 PMCID: PMC8415830 DOI: 10.3389/fpubh.2021.713895] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/22/2021] [Indexed: 01/13/2023] Open
Abstract
The mortality-to-incidence ratio (MIR) is widely used to evaluate the efficacy of cancer management outcomes for individual countries. However, the association among health care expenditure, human development index (HDI), and changes in MIR over time (δMIR) remains unknown. We aimed to elucidate the significance between these indicators and gastric cancer outcomes in different countries. Among the regions, Asia had the highest number of new gastric cancer cases, gastric cancer-related deaths, age-standardized ratio of incidence, and mortality. Chile had the highest age-standardized ratio (ASR) for gastric cancer incidence and the highest ASR for mortality. Moreover, MIR was highest in Africa (0.91) and lowest in North America (0.43). Of note, MIR was negatively associated with HDI, current health expenditure (CHE) per capita, and CHE/GDP % and δMIR was positively associated with CHE/GDP % in countries with very high HDI. However, δMIR showed no significant associations with these indicators in the countries analyzed. In conclusion, increased HDI, CHE per capita, and CHE/GDP are associated with improved gastric cancer outcomes. In addition, the δMIR could be an indicator that can be used to evaluate the improvement in cancer management outcomes over time.
Collapse
Affiliation(s)
- Tzu-Wei Yang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chi-Chih Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-Cheng Hung
- Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Hsiang Liu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Wei Sung
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Urology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Chang Tsai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
33
|
Trivanovic D, Plestina S, Honovic L, Dobrila-Dintinjana R, Vlasic Tanaskovic J, Vrbanec D. Gastric cancer detection using the serum pepsinogen test method. TUMORI JOURNAL 2021; 108:386-391. [PMID: 33993805 DOI: 10.1177/03008916211014961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Gastric cancer (GC) is the eighth most common cause of cancer deaths in Croatia and one of the most common causes of cancer deaths worldwide. A reliable diagnostic tool for the early detection of GC is essential. OBJECTIVE We previously suggested a pepsinogen test method to reduce the mortality from GC by allowing early detection. Here, we report an updated analysis from a prospective single-center clinical study to evaluate the sensitivity and specificity of the pepsinogen test method and to determine whether this test can be used as a part of routine laboratory assessment of high-risk patients. METHODS We present mature data of the pepsinogen test method in the Croatian population after a median follow-up of 36 months. Statistical analyses were performed using a Mann-Whitney U test, multiple logistic regression, and receiver operating characteristics (ROC) to evaluate the predictive power of the assayed biomarkers. RESULTS Of the 116 patients, 25 patients had GC and 91 demonstrated a nonmalignant pathology based on tissue biopsy. Cutoff values were pepsinogen I ⩽70 and pepsinogen I/II ratio ⩽3.0. Using ROC curve analysis, the accuracy, sensitivity, specificity, positive predictive value, and negative predictive value were determined to be 87.22%, 78.12%, 90.10%, 71.43%, and 92.86%, respectively, for the diagnosis of GC. The area under the curve was 0.700 (95% confidence interval 0.57-0.83). CONCLUSION Pepsinogen tests are valuable for screening a population in need of further diagnosis and could help to avoid unnecessary invasive endoscopic procedures.
Collapse
Affiliation(s)
- Dragan Trivanovic
- Department of Oncology and Medical Faculty, General Hospital Pula, Pula, Croatia
| | - Stjepko Plestina
- Department of Oncology and Radiotherapy, Clinical Hospital Zagreb, Zagreb, Croatia
| | - Lorena Honovic
- Department of Clinical Chemistry and Medical Faculty, General Hospital Pula, Pula, Croatia
| | | | | | - Damir Vrbanec
- Department of Oncology and Medical Faculty, General Hospital Pula, Pula, Croatia
| |
Collapse
|
34
|
Coelho MCF, Ribeiro HG, Gomes CGDO, Marinho FP, Barbosa AJA, Coelho LGV. HELICOBACTER PYLORI CHRONIC GASTRITIS ON PATIENTS WITH PREMALIGNANT CONDITIONS: OLGA AND OLGIM EVALUATION AND SERUM BIOMARKERS PERFORMANCE. ARQUIVOS DE GASTROENTEROLOGIA 2021; 58:39-47. [PMID: 33909795 DOI: 10.1590/s0004-2803.202100000-08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND H. pylori chronic atrophic gastritis is a premalignant lesion, and its staging, according to OLGA and OLGIM systems aims to identify patients at increased risk of developing gastric cancer and optimize their follow-up. GastroPanel®, serum biomarkers panel including pepsinogen I (PGI), pepsinogen II (PGII), Gastrin 17 (G17) and anti- H. pylori antibodies is a noninvasive test for adenocarcinoma risk assessment in chronic H. pylori gastritis patients. OBJECTIVE Prospective study to evaluate the concordance between OLGA and OLGIM grading systems, as well as to evaluate GastroPanel´s performance in patients with premalignant lesions secondary to H. pylori chronic gastritis in Brazil. METHODS Patients with H. pylori chronic gastritis with premalignant lesions confirmed by histology were recruited from the gastrointestinal clinic of a University Hospital. All participants underwent endoscopic examination with biopsies which were reported according to updated Sydney system and premalignant lesions grading systems (OLGA and OLGIM). Blood samples were collected for biomarkers serological analysis (GastroPanel®, Biohit, Helsinki, Finland). The cut off values used to define high risk patients were those recommended by the manufacturer: PGI ≤30 µm/L and PGI/PGII ≤3. RESULTS 41 patients were recruited: 28 women, 13 men, mean age 67.3 (47-89, SD: 9.6) years. By OLGA system, were obtained: OLGA 0 (n=1), OLGA I (n=7), OLGA II (n=17), OLGA III (n=9), and OLGA IV (n=7). By OLGIM system, were obtained: OLGIM 0 (n=14), OLGIM I (n=5), OLGIM II (n=10), OLGIM III (n=10), and OLGIM IV (n=2). Regarding histological staging among patients staged as low risk (OLGA/OLGIM 0, I and II) and high risk (OLGA/OLGIM III and IV) for gastric cancer development, the concordance rate found between both classifications was 85.4%. Considering high risk patients, those patients thus included in at least one of the systems the final distribution of our sample considered 24 low-risk and 17 high-risk patients for the development of gastric cancer. To determine by GastroPanel® whether the patient would be at low or high risk of developing gastric cancer, PGI showed a sensitivity, specificity and accuracy of 0.47 (95%CI: 0.26-0.69), 0.67 (95%CI: 0.47-0.82), and 0.58 (95%CI: 0.43-0.72), respectively, while PGI/PGII showed sensitivity, specificity and accuracy of 0.06 (95%CI: 0.01-0.27), 0.83 (95%CI: 0.64-0.93) and 0.51 (95%CI: 0.36-0.66), respectively. CONCLUSION The histological classifications OLGA and OLGIM presented a substantial concordance rate among themselves. Simultaneous use of both histological classification systems increased the identification's rate of high-risk patients. Biomarker analysis was not effective to distinguish low to high risk patients in the studied population. Further studies are needed to validate its use in clinical practice in Brazil.
Collapse
Affiliation(s)
- Maria Clara Freitas Coelho
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Belo Horizonte, MG, Brasil.,Faculdade de Ciências Médicas de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Henrique Gomes Ribeiro
- Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Celio Geraldo de Oliveira Gomes
- Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Frederico Passos Marinho
- Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Alfredo J A Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Belo Horizonte, MG, Brasil.,Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Luiz Gonzaga Vaz Coelho
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Belo Horizonte, MG, Brasil.,Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| |
Collapse
|
35
|
Ghidini M, Petrillo A, Botticelli A, Trapani D, Parisi A, La Salvia A, Sajjadi E, Piciotti R, Fusco N, Khakoo S. How to Best Exploit Immunotherapeutics in Advanced Gastric Cancer: Between Biomarkers and Novel Cell-Based Approaches. J Clin Med 2021; 10:1412. [PMID: 33915839 PMCID: PMC8037391 DOI: 10.3390/jcm10071412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Despite extensive research efforts, advanced gastric cancer still has a dismal prognosis with conventional treatment options. Immune checkpoint inhibitors have revolutionized the treatment landscape for many solid tumors. Amongst gastric cancer subtypes, tumors with microsatellite instability and Epstein Barr Virus positive tumors provide the strongest rationale for responding to immunotherapy. Various predictive biomarkers such as mismatch repair status, programmed death ligand 1 expression, tumor mutational burden, assessment of tumor infiltrating lymphocytes and circulating biomarkers have been evaluated. However, results have been inconsistent due to different methodologies and thresholds used. Clinical implementation therefore remains a challenge. The role of immune checkpoint inhibitors in gastric cancer is emerging with data from monotherapy in the heavily pre-treated population already available and studies in earlier disease settings with different combinatorial approaches in progress. Immune checkpoint inhibitor combinations with chemotherapy (CT), anti-angiogenics, tyrosine kinase inhibitors, anti-Her2 directed therapy, poly (ADP-ribose) polymerase inhibitors or dual checkpoint inhibitor strategies are being explored. Moreover, novel strategies including vaccines and CAR T cell therapy are also being trialed. Here we provide an update on predictive biomarkers for response to immunotherapy with an overview of their strengths and limitations. We discuss clinical trials that have been reported and trials in progress whilst providing an account of future steps needed to improve outcome in this lethal disease.
Collapse
Affiliation(s)
- Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University, 00189 Rome, Italy;
- Medical Oncology (B), Policlinico Umberto I, 00161 Rome, Italy
| | - Dario Trapani
- Division of Early Drug Development for innovative therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy;
| | - Alessandro Parisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy
| | - Anna La Salvia
- Department of Oncology, University Hospital 12 De Octubre, 28041 Madrid, Spain;
| | - Elham Sajjadi
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Roberto Piciotti
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Shelize Khakoo
- Department of Medicine, Royal Marsden Hospital, London and Surrey, Sutton SM25PT, UK;
| |
Collapse
|
36
|
Yao Y, Ding Y, Bai Y, Zhou Q, Lee H, Li X, Teng L. Identification of Serum Circulating MicroRNAs as Novel Diagnostic Biomarkers of Gastric Cancer. Front Genet 2021; 11:591515. [PMID: 33597967 PMCID: PMC7882724 DOI: 10.3389/fgene.2020.591515] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-associated deaths worldwide. Due to the lack of typical symptoms and effective biomarkers for non-invasive screening, most patients develop advanced-stage GC by the time of diagnosis. Circulating microRNA (miRNA)-based panels have been reported as a promising tool for the screening of certain types of cancers. In this study, we performed differential expression analysis of miRNA profiles of plasma samples obtained from gastric cancer and non-cancer patients using two independent Gene Expression Omnibus (GEO) datasets: GSE113486 and GSE124158. We identified three miRNAs, hsa-miR-320a, hsa-miR-1260b, and hsa-miR-6515-5p, to distinguish gastric cancer cases from non-cancer controls. The three miRNAs showed an area under the curve (AUC) over 0.95 with high specificity (>93.0%) and sensitivity (>85.0%) in both the discovery datasets. In addition, we further validated these three miRNAs in two external datasets: GSE106817 [sensitivity: hsa-miR-320a (99.1%), hsa-miR-1260b (97.4%), and hsa-miR-6515-5p (92.2%); specificity: hsa-miR-320a (88.8%), hsa-miR-1260b (89.6%), and hsa-miR-6515-5p (88.7%); and AUC: hsa-miR-320a (96.3%), hsa-miR-1260b (97.4%), and hsa-miR-6515-5p (94.6%)] and GSE112264 [sensitivity: hsa-miR-320a (100.0%), hsa-miR-1260b (98.0%), and hsa-miR-6515.5p (98.0%); specificity: hsa-miR-320a (100.0%), hsa-miR-1260b (100.0%), and hsa-miR-6515.5p (92.7%); and AUC: hsa-miR-320a (1.000), hsa-miR-1260b (1.000), and hsa-miR-6515-5p (0.988)]. On the basis of these findings, the three miRNAs can be used as potential biomarkers for gastric cancer screening, which can provide patients with a higher chance of curative resection and longer survival.
Collapse
Affiliation(s)
- Yunjin Yao
- Department of Surgical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yuntong Bai
- Biomedical Engineering Department of Tulane University, New Orleans, LA, United States
| | - Quan Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hyun Lee
- Brown University, Warren Alpert School of Medicine, Providence, RI, United States
| | - Xiawei Li
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
37
|
Fan X, Qin X, Zhang Y, Li Z, Zhou T, Zhang J, You W, Li W, Pan K. Screening for gastric cancer in China: Advances, challenges and visions. Chin J Cancer Res 2021; 33:168-180. [PMID: 34158737 PMCID: PMC8181866 DOI: 10.21147/j.issn.1000-9604.2021.02.05] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is one of the major cancers in China and all over the world. Most GCs are diagnosed at an advanced stage with unfavorable prognosis. Along with some other countries, China has developed the government-funded national screening programs for GC and other major cancers. GC screening has been shown to effectively decrease the incidence of and mortality from GC in countries adopting nationwide screening programs (Japan and Korea) and in studies based on selected Chinese populations. The screening of GC relies mostly on gastroendoscopy, the accuracy, reliability and safety of which have been indicated by previous studies. However, considering its invasive screening approach, requirements on skilled endoscopists and pathologists, and a high cost, developing noninvasive methods to amend endoscopic screening would be highly needed. Numerous studies have examined biomarkers for GC screening and the combination of biomarkers involving pepsinogen, gastrin, and Helicobacter pylori antibodies has been proposed for risk stratification, seeking to narrow down the high-risk populations for further endoscopy. Despite all the achievements of endoscopic screening, evidence on appropriate screening age, intervals for repeated screening, novel biomarkers promoting precision prevention, and health economics need to be accumulated to inform policymakers on endoscopic screening in China. With the guide of Health China 2030 Planning Outline, we have golden opportunities to promote prevention and control of GC. In this review, we summarize the characteristics of screening programs in China and other East Asian countries and introduce the past and current approaches and strategies for GC screening, aiming for featuring the latest advances and key challenges, and illustrating future visions of GC screening.
Collapse
Affiliation(s)
- Xiaohan Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiangxiang Qin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhexuan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Tong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jingying Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Weicheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & 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 & Institute, Beijing 100142, China
| | - Kaifeng Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
38
|
Jiang W, Sun G, Zhang Y, Hu Z, Wen X, Men S, Xing Z, Zhang S, Huang B, Wang C. Simultaneous determination of gastric cancer biomarkers pepsinogen PGI/PGII using element tagged immunoassay coupled with inductively coupled plasma mass spectrometry detection. J Clin Lab Anal 2020; 34:e23287. [PMID: 32147885 PMCID: PMC7370713 DOI: 10.1002/jcla.23287] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES In this study, a new immunoassay for the simultaneous determination of pepsinogen I (PGI) and pepsinogen II (PGII) in serum based on element labeling strategy coupled with inductively coupled plasma mass spectrometry (ICP-MS) detection was proposed. METHODS The sandwich-type immunoassay was used to simultaneously detect PGI and PGII in serum. PGI and PGII were captured by anti-PGI and anti-PGII antibody immobilized on the magnetic beads and then banded with Eu3+ labeled anti-PGI detection antibody and Sm3+ labeled anti-PGII detection antibody, followed by ICP-MS detection. RESULTS The linear correlation coefficient (R2 ) of PGI and PGII standard curves was .9938 and .9911, with the dynamic range of 0-200 ng/mL and 0-60 ng/mL, respectively. The limit of detection for PGI and PGII was 1.8 ng/mL and 0.3 ng/mL, respectively. The average recovery was 101.41% ± 6.74% for PGI and 101.47% ± 4.20% for PGII. Good correlations were obtained between the proposed method and CLIA (r = .9588 for PGI, r = .9853 for PGII). CONCLUSIONS We established a mass spectrometry-based immunoassay for the simultaneous detection of PGI and PGII in a single analysis. The element tagged immunoassay coupled with ICP-MS detection has high sensitivity, accuracy, and specificity in clinical serum sample analysis.
Collapse
Affiliation(s)
- Wencan Jiang
- Department of Clinical Laboratory MedicineChinese People's Liberation Army General Hospital & Postgraduate Medical SchoolBeijingChina
| | - Gongwei Sun
- Beijing Key Laboratory for Microanalytical Methods and InstrumentationDepartment of ChemistryTsinghua UniversityBeijingChina
| | - Yuqing Zhang
- Beijing Key Laboratory for Microanalytical Methods and InstrumentationDepartment of ChemistryTsinghua UniversityBeijingChina
| | - Zhian Hu
- Beijing Key Laboratory for Microanalytical Methods and InstrumentationDepartment of ChemistryTsinghua UniversityBeijingChina
| | - Xinyu Wen
- Department of Clinical Laboratory MedicineChinese People's Liberation Army General Hospital & Postgraduate Medical SchoolBeijingChina
| | - Shasha Men
- Department of Clinical Laboratory MedicineChinese People's Liberation Army General Hospital & Postgraduate Medical SchoolBeijingChina
| | - Zhi Xing
- Beijing Key Laboratory for Microanalytical Methods and InstrumentationDepartment of ChemistryTsinghua UniversityBeijingChina
| | - Sichun Zhang
- Beijing Key Laboratory for Microanalytical Methods and InstrumentationDepartment of ChemistryTsinghua UniversityBeijingChina
| | - Biao Huang
- College of Life Science and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Chengbin Wang
- Department of Clinical Laboratory MedicineChinese People's Liberation Army General Hospital & Postgraduate Medical SchoolBeijingChina
| |
Collapse
|
39
|
Kishino T, Oyama T, Tomori A, Takahashi A, Shinohara T. Usefulness and Limitations of a Serum Screening System to Predict the Risk of Gastric Cancer. Intern Med 2020; 59:1473-1480. [PMID: 32188803 PMCID: PMC7364258 DOI: 10.2169/internalmedicine.3521-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective The aim of the present study was to evaluate the effectiveness and limitations of a serum screening system for predicting the risk of gastric cancer. Methods Serum pepsinogen I (PG I)/pepsinogen II (PG II) and Helicobacter pylori (HP) antibody levels were measured. Subjects were classified into four groupsaccording to their serological status (the ABC classification system). The grade of atrophic gastritis was assessed endoscopically. We evaluated gastric cancer detection rates according to the ABC classification system and the endoscopic grade of atrophy. Patients Individuals who underwent esophagogastroduodenoscopy (EGD) in a health check were prospectively enrolled in the present study. Results According to the ABC classification system, the gastric cancer detection rates in groups A, B, C, and D were 0.07% (4/6,105), 0.5% (8/1,739), 0.8% (16/2,010), and 1.1% (3/281), respectively. The gastric cancer detection rates in subjects with no atrophy, closed type (C-type) atrophy, and open type (O-type) atrophy were 0% (0/4,567), 0.2% (4/2,581), and 0.9% (27/2,987), respectively. In group A (HP(-)/PG(-)), the proportions of subjects with no atrophy, C-type atrophy, and O-type atrophy were 71.2%, 22.8%, and 6.0%, respectively. In group A, the gastric cancer detection rates in subjects with no atrophy, C-type atrophy, and O-type atrophy were 0%, 0.07%, and 0.8%, respectively. Conclusion The ABC classification system is useful for predicting the risk of gastric cancer. However, this system was limited in group A, which included individuals with a high risk of developing gastric cancer. An endoscopic diagnosis of atrophy may be more effective than the ABC classification system for predicting the risk of gastric cancer.
Collapse
Affiliation(s)
- Takaaki Kishino
- Department of Endoscopy, Saku Central Hospital Advanced Care Center, Japan
- Department of Gastroenterology and Hepatology, Center for Digestive and Liver Diseases, Nara City Hospital, Japan
| | - Tsuneo Oyama
- Department of Endoscopy, Saku Central Hospital Advanced Care Center, Japan
| | - Akihisa Tomori
- Department of Gastroenterology, Saku Central Hospital Advanced Care Center, Japan
| | - Akiko Takahashi
- Department of Endoscopy, Saku Central Hospital Advanced Care Center, Japan
| | - Tomoaki Shinohara
- Department of Gastroenterology, Saku Central Hospital Advanced Care Center, Japan
| |
Collapse
|
40
|
Identification of salivary volatile organic compounds as potential markers of stomach and colorectal cancer: A pilot study. J Oral Biosci 2020; 62:212-221. [PMID: 32474113 DOI: 10.1016/j.job.2020.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The purpose of the pilot study was to determine the potential diagnostic capabilities for the analysis of oxygen-containing salivary volatile organic compounds (VOCs) in stomach and colorectal cancer. METHODS Saliva samples of 11 patients with stomach cancer, 18 patients with colorectal cancer, and 16 healthy volunteers were analyzed through capillary gas chromatography. The levels of lipid peroxidation products and catalase activity were determined in all samples. To assess saliva diagnostic potential, we constructed a Classification and Regression Tree (CART). RESULTS It was shown that the use of a combination of saliva VOCs (acetaldehyde, acetone, propanol-2, and ethanol) allowed classification into Cancer/Control groups with a sensitivity and specificity of 95.7 and 90.9%, respectively. To clarify the location of the tumor, it was necessary to add a methanol level; in this case, the sensitivity for detecting stomach and colorectal cancer was 80.0% and 92.3%, respectively, while the specificity in both cases was 100%. When the lipid peroxidation product content was added to the VOC indicators, they were selected as the main factors for constructing the decision tree. For classification into Cancer/Control groups, only the triene conjugate and Schiff base content in saliva was sufficient. The combination of VOCs in saliva and lipid peroxidation indices improved the sensitivity and specificity for classification to 100%. CONCLUSION Preliminary data were obtained on the sensitivity and specificity of the diagnosis of stomach and colorectal cancer, which confirmed the promise of further studies on saliva VOCs for the purpose of clinical laboratory diagnostics.
Collapse
|
41
|
Miftahussurur M, Waskito LA, Aftab H, Vilaichone RK, Subsomwong P, Nusi IA, Syam AF, Ratanachu-ek T, Doohan D, Siregar G, Rezkitha YAA, Fauzia KA, Mahachai V, Yamaoka Y. Serum pepsinogens as a gastric cancer and gastritis biomarker in South and Southeast Asian populations. PLoS One 2020; 15:e0230064. [PMID: 32271765 PMCID: PMC7145115 DOI: 10.1371/journal.pone.0230064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/20/2020] [Indexed: 12/19/2022] Open
Abstract
Serum pepsinogens have been widely acknowledged as gastric mucosal biomarkers; however, a multicountry report on the benefits of pepsinogens as biomarkers has not yet been published. We analyzed 1,206 sera and gastric mucosal samples collected from Bangladesh, Bhutan, Indonesia, Myanmar, Nepal and Thailand then assessed the association between gastric mucosal changes and Helicobacter pylori infection. The new cutoff values for serum pepsinogen values were evaluated using a receiver operating characteristic analysis. The participants with H. pylori infection had significantly lower pepsinogen I and higher pepsinogen II values, but a lower pepsinogen I/II ratio than participants without the infection (all P < .001). The pepsinogen I and pepsinogen I/II values were significantly higher and lower, respectively, in individuals with atrophic gastritis than in those without (both P < .001). Among uninfected individuals, only the pepsinogen I/II ratio was significantly lower in atrophic individuals. Pepsinogen I/II ratio also were significantly different between disease among H. pylori-positive and H. pylori-negative individuals, suggesting the pepsinogen I/II ratio is a robust biomarker for determining both chronic and atrophic gastritis. The cutoffs for detecting chronic and atrophic gastritis for the pepsinogen I/II ratio were 4.65 and 4.95, respectively. In conclusion, pepsinogen levels are useful biomarker for both chronic gastritis and atrophic gastritis, but they should be used with caution. Population-based validation is necessary to determine the best cutoff values. Among all pepsinogen values, the pepsinogen I/II ratio was the most reliable gastric mucosal-change biomarker.
Collapse
Affiliation(s)
- Muhammad Miftahussurur
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine-Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Langgeng Agung Waskito
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Hafeza Aftab
- Department of Gastroenterology, Dhaka Medical College and Hospital, Dhaka, Bangladesh
| | - Ratha-korn Vilaichone
- Gastroenterology Unit, Digestive Diseases Research Center (DRC), Thammasat University Hospital, Pathumthani, Thailand
- Department of Medicine, Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
| | - Phawinee Subsomwong
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Iswan Abbas Nusi
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine-Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Ari Fahrial Syam
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | | | - Dalla Doohan
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Gontar Siregar
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine, University of Sumatera Utara, Medan, Indonesia
| | - Yudith Annisa Ayu Rezkitha
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Faculty of Medicine, University of Muhammadiyah Surabaya, Surabaya, Indonesia
| | - Kartika Afrida Fauzia
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | | | - Yoshio Yamaoka
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine-Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX, United States of America
- Global Oita Medical Advanced Research Center for Health, Yufu, Japan
| |
Collapse
|
42
|
Cha JH, Jang JS. Clinical correlation between serum pepsinogen level and gastric atrophy in gastric neoplasm. Korean J Intern Med 2020; 35:550-558. [PMID: 30400679 PMCID: PMC7214368 DOI: 10.3904/kjim.2018.282] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIMS The relationship between the serum pepsinogen (sPG) level and changes in gastric mucosa has been well studied. Here, we evaluated the usefulness of sPG (I, II, I/II ratio) and intragastric pH as a biomarker of severe gastric atrophy in gastric neoplastic lesions. METHODS A total of 186 consecutive Korean patients with gastric neoplastic lesions underwent endoscopic submucosal dissection (ESD) in this study. The serologic atrophy group had sPG I level ≤ 70 ng/mL and an sPG I/II ratio ≤ 3.0. Before ESD, overnight fasting venous blood and gastric juice samples were collected to measure the sPG level and intragastric pH. The degree of gastric atrophy was estimated by endoscopy, and the rapid urease test was performed to investigate Helicobacter pylori infection. RESULTS Patients who met the criteria of serologic atrophy showed more severe endoscopic atrophic changes (61% vs. 18%, p = 0.000). Older patients and those with more atrophic changes at the gastric upper body demonstrated both a lower sPG I level and a lower PG I/II ratio and more severe endoscopic atrophy. The sPG I/II ratio was the lowest in low grade dysplasia than in high grade dysplasia and early gastric cancer (EGC) (p = 0.015). In addition, patients who tested negative for serologic atrophy and H. pylori showed the lowest intragastric pH (p = 0.000). CONCLUSION A low sPG I level and a low I/II ratio were correlated with the severity of gastric atrophy in gastric neoplastic lesions, thus indicating it to be a sensitive biomarker of gastric precancerous lesions or EGC.
Collapse
Affiliation(s)
- Jae Hwang Cha
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Jin Seok Jang
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
- Correspondence to Jin Seok Jang, M.D. Department of Internal Medicine, Dong-A University Medical Center, 26 Daesingongwon-ro, Seo-gu, Busan 49201, Korea Tel: +82-51-240-2609 Fax: +82-51-253-2087 E-mail:
| |
Collapse
|
43
|
Ning P, Sun L, Dong N, Yuan Y. PGC-MG7 combination could be used as a follow-up panel for monitoring dynamical progression of gastric precancerous diseases. Chin J Cancer Res 2020; 32:89-95. [PMID: 32194308 PMCID: PMC7072012 DOI: 10.21147/j.issn.1000-9604.2020.01.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Objective The aim of this study was to investigate the value of the combined expression of the gastric mucosal differentiation protein pepsinogen C (PGC) and gastric cancer (GC)-associated antigen MG7 for the diagnosis of GC and prediction of the development from precancerous conditions to GC. Methods The gastric mucosal biopsies of 285 subjects enrolled from a region with a high incidence of GC were obtained and histopathologically examined. Subjects testing negative for GC (n=208) were followed up from 1998 to 2015. The levels of PGC and MG7 in the biopsies were determined by immunohistochemistry. Results PGC was positive in 91.4% of the non-atrophic gastritis, 26.5% of the atrophic gastritis, and 0% of the GC. MG7 was positive in 15.0% of the non-atrophic gastritis, 82.4% of the atrophic gastritis, and 94.8% of the GC. The non-atrophic gastritis group was predominantly “PGC+MG7−”. The atrophic gastritis and GC groups were predominantly “PGC−MG7+”. The rate of GC in subjects with “PGC−MG7+” staining was 113.4-fold higher [95% confidence interval (95% CI): 15.3−869.4, P<0.001] than that in subjects with other staining patterns. The sensitivity and specificity of the “PGC−MG7+” pattern were 92.2% and 78.8% for the detection of GC and 77.2% and 97.9% for GC and precancerous disease, respectively. In the follow-up cohort of non-GC subjects, the risk of developing GC was higher in those with the “PGC−MG7+” staining pattern. Conclusions Our data suggest that the “PGC−MG7+” pattern can be employed as a useful follow-up panel for detecting individuals with a high risk of GC, and the dynamic assessment of the follow-up panel needs multi-centre large-scale validation in the future.
Collapse
Affiliation(s)
- Peifang Ning
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang 110001, China.,Pathologic Department of Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang 110042, China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang 110001, China
| | - Nannan Dong
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
44
|
Zeng W, Zhang S, Yang L, Wei W, Gao J, Guo N, Wu F. Serum miR-101-3p combined with pepsinogen contributes to the early diagnosis of gastric cancer. BMC MEDICAL GENETICS 2020; 21:28. [PMID: 32041551 PMCID: PMC7011268 DOI: 10.1186/s12881-020-0967-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 02/03/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND This study aimed to explore the diagnostic value of serum miR-101-3p combined with pepsinogen (PG) on early diagnosis of gastric cancer (GC). METHODS A total of 61 atrophic gastritis (AG) and 86 GC patients, and 50 healthy volunteers were enrolled. The serum expression of miR-101-3p was measured by qRT-PCR. The serum content of carcinoembryonic antigen (CEA) was measured by Electrochemiluminescence immunoassay. The serum contents of PGI and PGII were measured by Enzyme linked immunosorbent assay. The diagnostic value of serum markers on AG and GC was analyzed by receiver operating characteristic (ROC) analysis. RESULTS The expression of miR-101-3p, the content of PGI and the ratio of PGI/II were significantly decreased, and the content of PGII was significantly increased in AG patients compared with those in normal controls. The changes of the above serum indicators were more obvious in GC patients than those in AG patients. The content of CEA was significantly higher in GC patients than that in AG patients. In addition, the expression of miR-101-3p was negatively associated with the submucosal infiltration in GC patients. MiR-101-3p exhibited high diagnostic value on AG (AUC 0.8493, sensitivity 80.33%, specificity 80%) and GC (AUC 0.8749, sensitivity 72.09%, specificity 86.49%). MiR-101-3p + PGI + PGI/II (AUC 0.856, sensitivity 80.23%, specificity 77.05%) exhibited a high diagnostic value in distinguishing between AG and GC. CONCLUSIONS MiR-101-3p was a potential diagnostic marker for AG and GC. MiR-101-3p + PGI + PGI/II was effective in distinguishing between AG and GC.
Collapse
Affiliation(s)
- Weiwei Zeng
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China.
| | - Shuxiang Zhang
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Lei Yang
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Wenchao Wei
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Jie Gao
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Ni Guo
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Fengting Wu
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| |
Collapse
|
45
|
Plasma proteomics-based identification of novel biomarkers in early gastric cancer. Clin Biochem 2020; 76:5-10. [PMID: 31765635 DOI: 10.1016/j.clinbiochem.2019.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/27/2019] [Accepted: 11/02/2019] [Indexed: 12/18/2022]
|
46
|
Ye DM, Xu G, Ma W, Li Y, Luo W, Xiao Y, Liu Y, Zhang Z. Significant function and research progress of biomarkers in gastric cancer. Oncol Lett 2020; 19:17-29. [PMID: 31897111 PMCID: PMC6924079 DOI: 10.3892/ol.2019.11078] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the most common gastrointestinal tumor types, and the incidence and mortality rates are higher in men compared with women. Various studies have revealed that gastric cancer is a spectrum of tumor types, which have biological and genetic diversity. It has proven to be difficult to improve the overall survival and disease-free survival of patients with gastric cancer through the use of traditional surgery and chemoradiation, as gastric cancer is usually identified at an advanced stage. In consequence, the outcome is frequently poor. Thus, novel biomarkers and anticancer targets are required to improve the outcome. As the identification of biomarkers has increased due to advances in research and the greater availability of bioinformatics and functional genomics, the potential therapeutic regimens available have also increased concurrently. These advances have also improved the ability to predict responses to chemotherapy, targeted therapy and immunotherapy, whilst other biomarkers predict post-treatment survival and recurrence based on their expression. This review focuses closely on the important functions of biomarkers in the timely diagnosis and treatment of gastric cancer, in addition to the advances in the study of certain novel markers in gastric cancer.
Collapse
Affiliation(s)
- Dong Mei Ye
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Gaosheng Xu
- Department of Surgery, Yueyang Maternal and Child Health Hospital, Yueyang, Hunan 414000, P.R. China
| | - Wei Ma
- Department of Surgery, Yueyang Maternal and Child Health Hospital, Yueyang, Hunan 414000, P.R. China
| | - Yuxuan Li
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Weiru Luo
- Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yiyang Xiao
- Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yong Liu
- Department of Pathology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiwei Zhang
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
47
|
De Marchi D, Panozzo MP, Antico A, Tozzoli R. Valutazione di un nuovo metodo immunometrico per la misura dei marcatori biochimico/sierologici di gastrite atrofica. LA RIVISTA ITALIANA DELLA MEDICINA DI LABORATORIO 2020; 15. [DOI: 10.23736/s1825-859x.19.00036-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
48
|
Bornschein J, Quante M, Fassan M. Gastric Cancer; Epidemiology and Diagnosis. ENCYCLOPEDIA OF GASTROENTEROLOGY 2020:553-564. [DOI: 10.1016/b978-0-12-801238-3.65678-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
49
|
Mezmale L, Isajevs S, Bogdanova I, Polaka I, Krigere A, Rudzite D, Rudule A, Kikuste I, Parshutin S, Tazhedinov A, Mushinskiy D, Sametayev D, Belikhina T, Igissinov N, Park JY, Herrero R, Leja M. Prevalence of Atrophic Gastritis in Kazakhstan and the Accuracy of Pepsinogen Tests to Detect Gastric Mucosal Atrophy. Asian Pac J Cancer Prev 2019; 20:3825-3829. [PMID: 31870128 PMCID: PMC7173362 DOI: 10.31557/apjcp.2019.20.12.3825] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Atrophic gastritis is considered precursor condition for gastric cancer. There is so far limited evidence on the performance of pepsinogens for atrophy detection in Central Asia. The aim of our study was to detect the prevalence of atrophic gastritis in the asymptomatic adult population in Kazakhstan as well as address the accuracy of pepsinogen testing in atrophy detection. METHODS Healthy individuals aged 40-64 were included. Upper endoscopy and pepsinogens (PG) evaluation were performed. PG were analysed in plasma by latex agglutination. Cut off values were used to define decreased PG values (PGR ≤ 3 and PG I ≤ 70 ng/mL); severely decreased PG values (PGR ≤ 2 and PG I ≤ 30 ng/mL). Biopsies were analyzed and obtained according to the updated Sydney System. PG test sensitivity, specificity and overall accuracy were assessed using the histological diagnosis as the "gold standard". RESULTS Altogether 157 individuals - female 40,1% and male 59,9% were included. Histologically, moderate to severe corpus atrophy, was present only in 1,3% cases. From all study subjects, 26,8% had decreased plasma PG values with cut-off values PGR ≤ 3 and PG I ≤ 70 ng/mL. The sensitivity of the PG test with this cut-off values was 50,0%, specificity 73,5%, overall accuracy 73,2% for detection of moderate to severe atrophy in the corpus. The sensitivity of PG test with cut-off values PGR ≤ 2 and PG I ≤30 ng/mL was 50,0%, specificity 90,9% and overall accuracy 90,4%. CONCLUSIONS The prevalence of gastric mucosal atrophy was low in the Kazakh population. Serological PG test screening nevertheless can play an important role in the diagnosis of gastric precancerous lesions. However, the diagnostic accuracy of the PG test is mainly dependent on the cut-off values for positive results.
Collapse
Affiliation(s)
- Linda Mezmale
- Institute of Clinical and Preventive Medicine,
- Faculty of Medicine, University of Latvia,
| | - Sergejs Isajevs
- Institute of Clinical and Preventive Medicine,
- Academic Histology Laboratory,
| | - Inga Bogdanova
- Institute of Clinical and Preventive Medicine,
- Academic Histology Laboratory,
| | | | | | | | - Aiga Rudule
- Institute of Clinical and Preventive Medicine,
| | - Ilze Kikuste
- Institute of Clinical and Preventive Medicine,
- Faculty of Medicine, University of Latvia,
- Digestive Disease Centre GASTRO, Riga, Latvia,
| | | | | | | | | | | | - Nurbek Igissinov
- Astana Medical University,
- Central Asian Cancer Institute, Nur-Sultan, Kazakhstan,
- International High School of Medicine,
- Eurasian Institute For Cancer Research, Bishkek, Kyrgyzstan,
| | - Jin Young Park
- Prevention and Implementation Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, Lyon, France.
| | - Rolando Herrero
- Prevention and Implementation Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, Lyon, France.
| | - Marcis Leja
- Institute of Clinical and Preventive Medicine,
- Faculty of Medicine, University of Latvia,
- Digestive Disease Centre GASTRO, Riga, Latvia,
| |
Collapse
|
50
|
Xu Y, Miremadi A, Link A, Malfertheiner P, Fitzgerald RC, Bornschein J. Feasibility of combined screening for upper gastrointestinal adenocarcinoma risk by serology and Cytosponge testing: the SUGAR study. J Clin Pathol 2019; 72:825-829. [PMID: 31235543 PMCID: PMC6874497 DOI: 10.1136/jclinpath-2019-205700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/18/2019] [Indexed: 01/11/2023]
Abstract
AIMS Aim was to assess the feasibility of serum markers to identify individuals at risk for gastro-oesophageal adenocarcinoma to reduce the number of individuals requiring invasive assessment by endoscopy. METHODS Blood samples from 56 patients with Barrett's oesophagus and 202 non-Barrett controls who previously took part in a trial assessing the accuracy of the Cytosponge for Barrett's oesophagus were assessed for serum pepsinogen (PG) 1 and 2, gastrin-17, trefoil factor 3 (TFF3) and Helicobacter pylori infection. RESULTS PG1 was pathological (<50 ng/mL) in 26 patients (10.1%), none of whom had Barrett's oesophagus (p<0.001). Smoking and drinking had no influence on these results. Pathological PG1 was associated with stomach pain (p=0.029), disruption of sleep (p=0.027) and disruption of diet by symptoms (p=0.019). Serum TFF3 was not associated with any clinical parameter. CONCLUSIONS Assessment of serum PG1 could be combined with a test for Barrett's oesophagus to identify additional patients requiring endoscopy.
Collapse
Affiliation(s)
- Yiwang Xu
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Ahmad Miremadi
- Histopathology Department, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Alexander Link
- Dept. of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Peter Malfertheiner
- Dept. of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Jan Bornschein
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK
| |
Collapse
|