1
|
Liu Y, Huang T, Wang L, Wang Y, Liu Y, Bai J, Wen X, Li Y, Long K, Zhang H. Traditional Chinese Medicine in the treatment of chronic atrophic gastritis, precancerous lesions and gastric cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118812. [PMID: 39260710 DOI: 10.1016/j.jep.2024.118812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/27/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic atrophic gastritis (CAG), precancerous lesions of gastric cancer (PLGC), and gastric cancer (GC), seriously threaten human health. Traditional Chinese medicine (TCM) has been employed in the treatment of chronic diseases for a long time and has shown remarkable efficacy. AIM OF THE STUDY Recently, there has been an increasing use of TCM in treating CAG, PLGC, and GC. The objective of this study is to compile a comprehensive overview of the existing research on the effects and molecular mechanisms of TCM, including formulas, single herbs, and active components. MATERIALS AND METHODS To obtain a comprehensive understanding of traditional use of TCM in treating these diseases, we reviewed ancient books and Chinese literature. In addition, keywords such as "TCM", "CAG", "PLGC", "GC", and "active ingredients" were used to collect modern research on TCM published in databases such as CNKI, Web of Science, and Pubmed up to April 2024. All collected information was then summarized and analyzed. RESULTS This study analyzed 174 articles, which covered the research progress of 20 TCM formulas, 14 single herbs, and 50 active ingredients in treating CAG, PLGC, and GC. Sources, effects, and molecular mechanisms of the TCM were summarized. CONCLUSIONS This article reviews the progress of TCM in the management of CAG, PLGC, and GC, which will provide a foundation for the clinical application and further development of TCM.
Collapse
Affiliation(s)
- Yuxi Liu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Tingting Huang
- Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China.
| | - Lu Wang
- Shaanxi University of Chinese Medicine, Middle section of Century Avenue, Xianyang, 712046, China.
| | - Yuan Wang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Yang Liu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Jingyi Bai
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Xinli Wen
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Ye Li
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Kaihua Long
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Hong Zhang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China; Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China; Shaanxi University of Chinese Medicine, Middle section of Century Avenue, Xianyang, 712046, China.
| |
Collapse
|
2
|
Jin JZ, Liang X, Liu SP, Wang RL, Zhang QW, Shen YF, Li XB. Association between autoimmune gastritis and gastric polyps: Clinical characteristics and risk factors. World J Gastrointest Oncol 2025; 17:92908. [DOI: 10.4251/wjgo.v17.i1.92908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 08/23/2024] [Accepted: 09/11/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The relationship between autoimmune gastritis (AIG) and gastric polyps (GPs) is not well understood.
AIM To explore the clinical characteristics and risk factors of AIG with GPs in patients.
METHODS This double center retrospective study included 530 patients diagnosed with AIG from July 2019 to July 2023. We collected clinical, biochemical, serological, and demographic data were of each patient. Logistic regression analyses, both multivariate and univariate, were conducted to pinpoint independent risk factors for GPs in patients with AIG patients. Receiver operating characteristic curves were utilized to establish the optimal cutoff values, sensitivity, and specificity of these risk factors for predicting GPs in patients with AIG.
RESULTS Patients with GPs had a higher median age than those without GPs [61 (52.25-69) years vs 58 (47-66) years, P = 0.006]. The gastrin-17 levels were significantly elevated in patients with GPs compared with those without GPs [91.9 (34.2-138.9) pmol/mL vs 60.9 (12.6-98.4) pmol/mL, P < 0.001]. Additionally, the positive rate of parietal cell antibody (PCA) antibody was higher in these patients than in those without GPs (88.6% vs 73.6%, P < 0.001). Multivariate and univariate analyses revealed that PCA positivity [odds ratio (OR) = 2.003, P = 0.017], pepsinogen II (OR = 1.053, P = 0.015), and enterochromaffin like cells hyperplasia (OR = 3.116, P < 0.001) were significant risk factors for GPs, while pepsinogen I was identified as a protective factor.
CONCLUSION PCA positivity and enterochromaffin like cells hyperplasia are significant risk factor for the development of GPs in patients with AIG. Elevated gastrin-17 levels may also play a role in this process. These findings suggest potential targets for further research and therapeutic intervention in managing GPs in patients with AIG.
Collapse
Affiliation(s)
- Jing-Zheng Jin
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Diseases, Shanghai 200001, China
| | - Xiao Liang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Diseases, Shanghai 200001, China
- Division of Gastroenterology and Hepatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shu-Peng Liu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Diseases, Shanghai 200001, China
- Division of Gastroenterology and Hepatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Rui-Lan Wang
- Division of Gastroenterology and Hepatology, Sichuan Armed Police Corps Hospital, Leshan 610041, Sichuan Province, China
| | - Qing-Wei Zhang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Diseases, Shanghai 200001, China
- Division of Gastroenterology and Hepatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yu-Feng Shen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Diseases, Shanghai 200001, China
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiao-Bo Li
- Department of Gastroenterology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai Institute of Digestive Diseases, Shanghai 200127, China
| |
Collapse
|
3
|
Liu S, Zhang T, Fang L, Hu L, Yin X, Tang X. Integrative pharmacological analysis of modified Zuojin formula: Inhibiting the HIF-1α-mediated glycolytic pathway in chronic atrophic gastritis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119136. [PMID: 39577677 DOI: 10.1016/j.jep.2024.119136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zuojin formula (ZJF) is a well-known herbal medicine in Pharmacopoeia of China, which is widely used for gastritis. Modified Zuojin formula (MZJF) was adapted based on traditional Chinese medicine (TCM) theories concerning gastric atrophy and dysplasia, along with extensive clinical experience, has been clinically employed to treat chronic atrophic gastritis (CAG). However, the underlying mechanisms by which MZJF intervenes in CAG remain to be fully elucidated. AIM OF THE STUDY The aim of this study was to evaluate the effects of MZJF intervention in CAG and explore its potential mechanisms. METHODS Four induction factors were used to establish a CAG rat model. HE and AB-PAS staining was utilized to assess the effects of MZJF in the intervention of CAG. The stomach weight index and gastric acid pH was used to assess the overall state of stomach. ELISA was used to assess the gastric mucosal inflammatory response. Using transmission electron microscopy to observe chief cells and parietal cells, we evaluated the improvement of ultrastructure by MZJF. Through network pharmacology analysis, the possible regulatory mechanism of MZJF in CAG was preliminarily explored. Binding interactions between MZJF components and predicted targets were explored using molecular docking. Subsequently, quantitative real-time PCR (qRT-PCR), Western blot, biochemical analysis and TUNEL staining were applied to validate the effect of MZJF on predicted pathway. RESULTS MZJF treatment ameliorated gastric mucosal pathology, inflammation, cellular ultrastructural damage and PG levels, halted the exacerbation of CAG in rats, along with a reduction in stomach weight index and gastric acid pH. A total of 79 compounds in MZJF targeting 203 CAG-related molecules were identified through network pharmacology. Enrichment analysis of the core targets was focused on the hypoxia inducible factor-1α (HIF-1α) signaling pathway. Molecular docking results identified HIF-1α as stable binding targets for MZJF primary components. Subsequently, PCR, WB, and biochemical results showed that MZJF suppressed the gene and protein expression levels of HIF-1α and its downstream molecules including glycolytic enzymes and transporters, modulated glucose, pyruvic acid and lactate levels in gastric mucosal tissue. Moreover, MZJF induced apoptosis of gastric epithelial cells, as evidenced by the upregulation of cleaved caspase-3, Bax, Bax/Bcl-2 and TUNEL positive cells ratio. CONCLUSIONS MZJF suppressed the HIF-1α-mediated glycolytic pathway, and promoted cell apoptosis, thereby halting the malignant transformation of CAG. The study provides a valuable reference point for applying TCM in preventing and treating CAG.
Collapse
Affiliation(s)
- Shan Liu
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Institute of Digestive Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Tai Zhang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University Health Science Center, Beijing, 100091, China; Peking University Health Science Center, Beijing, 100191, China.
| | - Lihui Fang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Lanshuo Hu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Xiaolan Yin
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361005, China.
| | - Xudong Tang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
4
|
Li J, Chen X, Mao C, Xiong M, Ma Z, Zhu J, Li X, Chen W, Ma H, Ye X. Epiberberine ameliorates MNNG-induced chronic atrophic gastritis by acting on the EGFR-IL33 axis. Int Immunopharmacol 2025; 145:113718. [PMID: 39642571 DOI: 10.1016/j.intimp.2024.113718] [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: 10/07/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
Chronic atrophic gastritis (CAG) is a prevalent form of chronic gastritis that presents with chronic inflammation of the gastric mucosa, localised gastric mucosal glandular atrophy and intestinal metaplasia. Despite the existence of diagnostic criteria, effective therapeutic strategies for this condition remain to be developed. The objective of this study was to examine the potential therapeutic benefits of epiberberine in mitigating MNNG-induced CAG and to elucidate the underlying mechanisms. MNNG was employed to establish a CAG mouse model and a GES-1 cell model, and EPI was observed to be efficacious in ameliorating the gastric mucosal injury and inflammatory infiltration induced by MNNG in the CAG model mice, a finding that was subsequently validated in the GES-1 model cells. Bioinformatics analysis indicated that EPI may exert a direct effect on EGFR, thereby regulating the expression of IL-33 and thereby achieving the therapeutic effect of CAG. This hypothesis was also validated by molecular docking prediction, CETSA, and overexpression of EGFR in GES-1 model cells, using EGFR agonists and inhibitors to further demonstrate that EPI may act as an antagonist supplement to EGFR for the treatment of CAG.
Collapse
Affiliation(s)
- Juan Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Xiantao Chen
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Changxia Mao
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Mengyuan Xiong
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jianyu Zhu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuegang Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China.
| | - Hang Ma
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
5
|
Cifuentes M, Verdejo HE, Castro PF, Corvalan AH, Ferreccio C, Quest AFG, Kogan MJ, Lavandero S. Low-Grade Chronic Inflammation: a Shared Mechanism for Chronic Diseases. Physiology (Bethesda) 2025; 40:0. [PMID: 39078396 DOI: 10.1152/physiol.00021.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/25/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
Inflammation is an important physiological response of the organism to restore homeostasis upon pathogenic or damaging stimuli. However, the persistence of the harmful trigger or a deficient resolution of the process can evolve into a state of low-grade, chronic inflammation. This condition is strongly associated with the development of several increasingly prevalent and serious chronic conditions, such as obesity, cancer, and cardiovascular diseases, elevating overall morbidity and mortality worldwide. The current pandemic of chronic diseases underscores the need to address chronic inflammation, its pathogenic mechanisms, and potential preventive measures to limit its current widespread impact. The present review discusses the current knowledge and research gaps regarding the association between low-grade chronic inflammation and chronic diseases, focusing on obesity, cardiovascular diseases, digestive diseases, and cancer. We examine the state of the art in selected aspects of the topic and propose future directions and approaches for the field.
Collapse
Affiliation(s)
- Mariana Cifuentes
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- OMEGA Laboratory, Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Hugo E Verdejo
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Division of Cardiovascular Diseases, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Pablo F Castro
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Division of Cardiovascular Diseases, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Alejandro H Corvalan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Hematology and Oncology, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Catterina Ferreccio
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Public Health, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomedicas (ICBM), Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Marcelo J Kogan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Department of Pharmacological & Toxicological Chemistry, Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomedicas (ICBM), Facultad Medicina, Universidad de Chile, Santiago, Chile
- Department of Biochemistry & Molecular Biology, Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
6
|
Rui Q, Li C, Rui Y, Zhang C, Xia C, Wang Q, Liu Y, Wang P. Human umbilical mesenchymal stem cells ameliorate atrophic gastritis in aging mice by participating in mitochondrial autophagy through Ndufs8 signaling. Stem Cell Res Ther 2024; 15:491. [PMID: 39707499 DOI: 10.1186/s13287-024-04094-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a chronic disease of the gastric mucosa characterized by a reduction or an absolute disappearance of the original gastric glands, possibly replaced by pseudopyloric fibrosis, intestinal metaplasia, or fibrosis. CAG develops progressively into intestinal epithelial metaplasia, dysplasia, and ultimately, gastric cancer. Epidemiological statistics have revealed a positive correlation between the incidence of CAG and age. Mesenchymal stem cells (MSCs) are a type of adult stem cells derived from mesoderm, with strong tissue repair capabilities. Therefore, the restoration of the gastric mucosa may serve as an efficacious strategy to ameliorate CAG and avert gastric cancer. However, the mechanisms by which MSCs inhibit the relentless progression of aging atrophic gastritis remain to be elucidated. This study endeavored to assess a novel approach utilizing MSCs to treat CAG and forestall carcinogenics. METHODS In this study, we selected mice with atrophic gastritis from naturally aging mice and administered human umbilical cord-derived mesenchymal stem cells (hUMSCs) via tail vein injection to evaluate the therapeutic effects of hUMSCs on age-related chronic atrophic gastritis. Initially, we employed methods such as ELISA, immunohistochemical analysis, and TUNEL assays to detect changes in the mice post-hUMSC injection. Proteomic and bioinformatics analyses were conducted to identify differentially expressed proteins, focusing on NADH: ubiquinone oxidoreductase core subunit S8 (Ndufs8). Co-culturing hUMSCs with Ndufs8 knockout gastric mucosal epithelial cells (GMECs), we utilized flow cytometry, Western blotting, real-time quantitative PCR, and immunofluorescence to investigate the mechanisms of action of hUMSCs. RESULTS We observed that hUMSCs are capable of migrating to and repairing damaged gastric mucosa. Initially, hUMSCs significantly enhanced the secretion of gastric proteins PG-1 and G17, while concurrently reducing inflammatory cytokines. Furthermore, hUMSCs mitigated gastric fibrosis and apoptosis in mucosal cells. Proteomic and bioinformatic analyses revealed alterations in the protein network involved in mitochondrial autophagy, with Ndufs8 playing a pivotal role. Upon knocking out Ndufs8 in GMECs, we noted mitochondrial damage and reduced autophagy, leading to an aged phenotype in GMECs. Co-culturing Ndufs8-knockout GMECs with hUMSCs demonstrated that hUMSCs could ameliorate mitochondrial dysfunction and restore the cell cycle in GMECs.
Collapse
Affiliation(s)
- Qiang Rui
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Chuyu Li
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Yiqi Rui
- Department of General Surgery,Geriatric Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Chuanzhuo Zhang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Cunbing Xia
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Qing Wang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yuanyuan Liu
- School of Medicine, Southeast University, Nanjing, 210096, China
| | - Peng Wang
- Department of General Surgery,Geriatric Hospital of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
7
|
Ren X, Suo B, Li C, Ping G, Ma L, Shi Y, Zhou K, Wang Y, Tian X, Zhou L, Song Z. Comparative analysis of the detection of antibiotic genotypic resistance with gastric mucosa, gastric fluid, and fecal samples in patients with Helicobacter pylori infection. J Clin Microbiol 2024:e0103424. [PMID: 39679670 DOI: 10.1128/jcm.01034-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Genotypic methods for detecting antibiotic resistance in Helicobacter pylori infection offer a rapid, convenient, and accurate approach for tailored therapy. However, existing studies predominantly examine single sample types and lack comparative analyses across different samples. This study comprehensively detects and compares genotypic resistance to clarithromycin and levofloxacin in gastric mucosa, gastric fluid, and fecal samples from the same patients. The study enrolled 183 participants, comprising 124 H. pylori-positive and 59 H. pylori-negative patients. All participants provided fecal samples and underwent gastroscopy for the collection of gastric mucosa and gastric fluid. Real-time PCR was employed to detect genotypic resistance to clarithromycin and levofloxacin in conjunction with bacterial culture and antibiotic susceptibility testing. Genotypic resistance detection rates for clarithromycin were 100% in gastric mucosa, 99.2% in gastric fluid, and 79.8% in fecal samples. For levofloxacin, detection rates were 97.6%, 96.8%, and 72.6%, respectively. The results showed that PCR detection for clarithromycin exhibited high sensitivity (0.94-0.95) and specificity (0.88-0.89) across all sample types. However, PCR detection for levofloxacin demonstrated slightly lower sensitivity (0.79-0.89) and specificity (0.79-0.83). The comparison of genotypic resistance results by PCR among the three sample types showed that gastric mucosa and gastric juice exhibited higher consistency, while the consistency between feces and both gastric mucosa and gastric juice was lower. This study confirmed good consistency between genotypic and phenotypic resistance in clarithromycin and levofloxacin. While both gastric mucosa and gastric fluid samples demonstrated high detection performance, the efficiency of detecting fecal samples was constrained by challenges in DNA extraction. IMPORTANCE This study, with a large sample size, comprehensively tested both Helicobacter pylori-negative and -positive patients, including rapid urease test, histopathological evaluation and staining, bacterial culture, susceptibility testing, and resistance gene mutation analysis. By simultaneously examining gastric mucosa, gastric juice, and fecal samples from the same individuals, we minimized confounding factors arising from different sample sources, ensuring the reliability of our results. This approach effectively delineated the differences and characteristics in detection performance among different sample types, offering crucial reference data for selecting appropriate detection samples and identifying areas for improvement. The findings revealed robust concordance between genotypic and phenotypic resistance, with both gastric mucosa and gastric juice samples demonstrating excellent detection performance. However, the efficiency of detecting resistance in fecal samples was hampered by challenges in DNA extraction.
Collapse
Affiliation(s)
- Xinlu Ren
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Baojun Suo
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Cailing Li
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Guangjie Ping
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Lingling Ma
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Kai Zhou
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Yuxin Wang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xueli Tian
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Liya Zhou
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Zhiqiang Song
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
8
|
Radadiya D, Desai M, Patel H, Velji-Ibrahim J, Spadaccini M, Srinivasan S, Khurana S, Thoguluva Chandrasekar V, Perisetti A, Repici A, Hassan C, Sharma P. Endoscopic submucosal dissection and endoscopic mucosal resection for Barrett's-associated neoplasia: a systematic review and meta-analysis of the published literature. Endoscopy 2024; 56:940-954. [PMID: 38942058 DOI: 10.1055/a-2357-6111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
BACKGROUND The role of endoscopic submucosal dissection (ESD) in the treatment of Barrett esophagus-associated neoplasia (BEN) has been evolving. We examined the efficacy and safety of ESD and endoscopic mucosal resection (EMR) for BEN. METHODS A database search was performed for studies reporting efficacy and safety outcomes of ESD and EMR for BEN. Pooled proportional and comparative meta-analyses were performed. RESULTS 47 studies (23 ESD, 19 EMR, 5 comparative) were included. The mean lesion sizes for ESD and EMR were 22.5 mm and 15.8 mm, respectively; most lesions were Paris type IIa. For ESD, pooled analysis showed rates of en bloc, R0, and curative resection, and local recurrence of 98%, 78%, 65%, and 2%, respectively. Complete eradication of dysplasia and intestinal metaplasia were achieved in 94% and 59% of cases, respectively. Pooled rates of perforation, intraprocedural bleeding, delayed bleeding, and stricture were 1%, 1%, 2%, and 10%, respectively. For EMR, pooled analysis showed rates of en bloc, R0, and curative resection, and local recurrence of 37%, 67%, 62%, and 6%, respectively. Complete eradication of dysplasia and intestinal metaplasia were achieved in 94% and 75% of cases. Pooled rates of perforation, intraprocedural bleeding, delayed bleeding, and stricture were 0.1%, 1%, 0.4%, and 8%, respectively. The mean procedure times for ESD and EMR were 113 and 22 minutes, respectively. Comparative analysis showed higher en bloc and R0 resection rates with ESD compared with EMR, with comparable adverse events. CONCLUSION ESD and EMR can both be employed to treat BEN depending on lesion type and size, and center expertise.
Collapse
Affiliation(s)
- Dhruvil Radadiya
- Gastroenterology, University of Kansas School of Medicine, Kansas City, United States
| | - Madhav Desai
- Gastroenterology, University of Minnesota Medical Center, Minneapolis, United States
- Gastroenterology and Hepatology, The University of Texas Health Science Center at Houston, Houston, United States
| | - Harsh Patel
- Gastroenterology, University of Kansas School of Medicine, Kansas City, United States
| | - Jena Velji-Ibrahim
- Internal Medicine, University of South Carolina School of Medicine, Greenville, United States
| | - Marco Spadaccini
- Endoscopy Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Sachin Srinivasan
- Gastroenterology, University of Kansas School of Medicine, Kansas City, United States
| | - Shruti Khurana
- Gastroenterology, University of Kansas School of Medicine, Kansas City, United States
| | | | - Abhilash Perisetti
- Gastroenterology, Kansas City VA Medical Center, Kansas City, United States
| | - Alessandro Repici
- Endoscopy Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Cesare Hassan
- Endoscopy Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Prateek Sharma
- Gastroenterology, University of Kansas School of Medicine, Kansas City, United States
- Gastroenterology, Kansas City VA Medical Center, Kansas City, United States
| |
Collapse
|
9
|
Triadafilopoulos G. Prevalence of Abnormalities at Tandem Endoscopy in Patients Referred for Colorectal Cancer Screening/Surveillance Colonoscopy. Cancers (Basel) 2024; 16:3998. [PMID: 39682185 DOI: 10.3390/cancers16233998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction: Performing a tandem endoscopy and colonoscopy in selected individuals has advantages, such as the early detection of benign and/or precancerous foregut diseases; it is efficient, and it may allow added therapies. It may also have disadvantages, such as generating anxiety from false-positive screening, possible harm from further testing, and unproven cost-effectiveness. Aims: We aimed to examine the prevalence of foregut endoscopic and histologic abnormalities in subjects referred for screening/surveillance colonoscopy who also underwent a tandem endoscopy. We wanted to (1) assess implications for cancer detection, intervention, and surveillance of precancerous foregut abnormalities, (2) identify benign foregut lesions, and (3) generate data on the utilities of this tandem approach. Patients and Methods: A retrospective cohort study of consecutive subjects referred for screening or surveillance colonoscopy who also underwent an endoscopy. Based on national screening guidelines, responses to prompting questions, personal or family history, or other risk factors, subjects were assigned to tandem endoscopy with biopsies (modified Seattle and Sydney protocols), under one anesthesia. Results: Of the 1004 patients referred for colonoscopy, 317 (32%) underwent tandem endoscopy. There were 214 women and 103 men. There were 237 Whites, 16 Asians, 40 Blacks, and 24 Hispanics. Median age was 59 (range 19-85). At endoscopy, we identified actionable benign (45%) peptic, inflammatory, and H. pylori-related abnormalities, and premalignant findings (i.e., intestinal metaplasia, 27%, dysplasia, 2%, and cancer 0.9%), comparable to the premalignant (40.3%) and malignant (0.6%) colonoscopy yield. Conclusions: When implemented based on national screening guidelines, tandem EGD and colonoscopy combines Barrett's esophagus and gastric cancer screening in one examination, and it has a high yield in a diverse US population.
Collapse
Affiliation(s)
- George Triadafilopoulos
- Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
10
|
Chen S, Xu L, Yan L, Zhang J, Zhou X, Wang J, Yan T, Wang J, He X, Ma H, Zhang X, Zhu S, Zhang Y, Xu C, Gao J, Ji X, Bai D, Chen Y, Chen H, Ke Y, Li L, Yu C, Mao X, Li T, Chen Y. A novel endoscopic artificial intelligence system to assist in the diagnosis of autoimmune gastritis: a multicenter study. Endoscopy 2024. [PMID: 39447610 DOI: 10.1055/a-2451-3071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
BACKGROUND Autoimmune gastritis (AIG), distinct from Helicobacter pylori-associated atrophic gastritis (HpAG), is underdiagnosed due to limited awareness. This multicenter study aimed to develop a novel endoscopic artificial intelligence (AI) system for assisting in AIG diagnosis. METHODS Patients diagnosed with AIG, HpAG, or nonatrophic gastritis (NAG), were retrospectively enrolled from six centers. Endoscopic images with relevant demographic and medical data were collected for development of the AI-assisted system based on a multi-site feature fusion model. The diagnostic performance of the AI model was evaluated in internal and external datasets. Endoscopists' performance with and without AI support was tested and compared using Mann-Whitney U test. Heatmap analysis was performed to interpret AI model outputs. RESULTS 18 828 endoscopy images from 1070 patients (294 AIG, 386 HpAG, 390 NAG) were collected. On testing datasets, AI identified AIG with 96.9 % sensitivity, 92.2 % specificity, and area under the receiver operating characteristic curve (AUROC) of 0.990 (internal), and 90.3 % sensitivity, 93.1 % specificity, and AUROC of 0.973 (external). The performance of AI (sensitivity 91.3 %) was comparable to that of experts (87.3 %) and significantly outperformed nonexperts (70.0 %; P = 0.01). With AI support, the overall performance of endoscopists was improved (sensitivity 90.3 % [95 %CI 86.0 %-93.2 %] vs. 78.7 % [95 %CI 73.6 %-83.2 %]; P = 0.008). Heatmap analysis revealed consistent focus of AI on atrophic areas. CONCLUSIONS This novel AI system demonstrated expert-level performance in identifying AIG and enhanced the diagnostic ability of endoscopists. Its application could be useful in guiding biopsy sampling and improving early detection of AIG.
Collapse
Affiliation(s)
- Shurong Chen
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Louzhe Xu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lingling Yan
- Department of Gastroenterology, Taizhou Hospital, Taizhou, China
| | - Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xuefeng Zhou
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Gastroenterology, the Second Hospital of Jiaxing, Jiaxing, China
| | - Jiayi Wang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Gastroenterology, CHC international hospital, Ningbo, China
| | - Tianlian Yan
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jinghua Wang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xinjue He
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Han Ma
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xuequn Zhang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shenghua Zhu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yizhen Zhang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chengfu Xu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianguo Gao
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Ji
- Department of Gastroenterology, the Second Hospital of Jiaxing, Jiaxing, China
| | - Dezhi Bai
- Department of Gastroenterology, the First People's hospital of Yuhang, Hangzhou, China
| | - Yuan Chen
- Department of Gastroenterology, the Third People's hospital of Zhoushan, Zhoushan, China
| | - Hongda Chen
- Department of Gastroenterology, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yini Ke
- Department of Rheumatology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lan Li
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chaohui Yu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xinli Mao
- Department of Gastroenterology, Taizhou Hospital, Taizhou, China
| | - Ting Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yi Chen
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Xu Y, Li GD, Wu CH, Zhong XQ. Nomogram prediction model for gastric cancer risk in chronic atrophic gastritis: Role of blood cell ratios. Shijie Huaren Xiaohua Zazhi 2024; 32:811-820. [DOI: 10.11569/wcjd.v32.i11.811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/20/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a precancerous condition for gastric cancer. Although endoscopy is the standard method for monitoring CAG, its invasive nature and high cost limit its use.
AIM To identify risk factors for gastric cancer in the CAG population, focusing on blood cell ratios, and develop a personalized prediction model using a nomogram.
METHODS A retrospective analysis was conducted on 314 GAG patients admitted to Hangzhou Normal University Affiliated Hospital from January 2018 to January 2024. Data collected included demographic, serological, and blood cell parameters. Independent risk factors were identified using multivariate logistic regression and a nomogram model was constructed with R. Model performance was assessed using the area under the ROC curve (AUC), the Hosmer-Lemeshow test, and decision curve analysis (DCA).
RESULTS Significant predictive factors for gastric cancer in the CAG population included male gender (odds ratio [OR] = 2.214, P < 0.05), Helicobacter pylori (H. pylori) infection (OR = 2.686, P < 0.05), gastrin 17 (G-17) (OR = 1.037, P < 0.05), hemoglobin-to-red blood cell distribution width ratio (HRR) (OR = 0.648, P < 0.05), and lymphocyte-to-monocyte ratio (LMR) (OR = 0.645, P < 0.05). The prediction model, with an AUC of 0.854, demonstrated good fit (Hosmer-Lemeshow test: χ2 = 6.062, P = 0.640). DCA indicated the potential generalizability of the model.
CONCLUSION The nomogram provides a noninvasive, convenient, and cost-effective tool for screening gastric cancer in CAG patients, showing excellent discrimination and calibration. Further large-scale, multicenter studies are necessary to validate its efficacy across diverse populations.
Collapse
Affiliation(s)
- Yang Xu
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
- Department of Gastroenterology and Hepatology, Hospital of Zhejiang People's Armed Police, Hangzhou 310051, Zhejiang Province, China
| | - Guo-Dong Li
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Chen-Han Wu
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Xue-Qing Zhong
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| |
Collapse
|
12
|
Jove A, Lin C, Hwang JH, Balasubramanian V, Fernandez-Becker NQ, Huang RJ. Serum Gastrin Levels Are Associated With Prevalent Neuroendocrine Tumors in Autoimmune Metaplastic Atrophic Gastritis. Am J Gastroenterol 2024:00000434-990000000-01469. [PMID: 39588964 DOI: 10.14309/ajg.0000000000003235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Autoimmune metaplastic atrophic gastritis (AMAG) is a precancerous condition that predisposes to gastric neuroendocrine tumors (gNETs). There exist no methods to stratify patients with AMAG for gNET risk. METHODS We identified a cohort of patients with AMAG within a university health system using histopathologic and serologic criteria. We analyzed features predictive of prevalent gNET. RESULTS We identified 181 patients with AMAG and 41 (22.7%) with prevalent gNET. Gastrin levels were elevated in gNET (1,859.8 vs 679.5 pg/mL, P < 0.001), and gastrin titers demonstrated good discrimination (c = 0.799, 95% CI 0.707-0.892) for gNET. DISCUSSION Gastrin levels differ significantly between patients with AMAG with and without gNET.
Collapse
Affiliation(s)
- Andre Jove
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Christina Lin
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
| | - Joo Ha Hwang
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
| | | | - Nielsen Q Fernandez-Becker
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
| | - Robert J Huang
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
| |
Collapse
|
13
|
Tan HJ, Tan EZY. Postprandial gastrin-17 level is a useful dynamic marker for atrophic gastritis. World J Gastrointest Endosc 2024; 16:623-626. [PMID: 39600554 PMCID: PMC11586718 DOI: 10.4253/wjge.v16.i11.623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Atrophic gastritis and intestinal metaplasia may progress to gastric malignancy. Non-invasive serum biomarkers have been extensively studied and proven to be useful as a screening tool to stratify risk and identify patients for endoscopy to detect early gastric cancer. These non-invasive biomarkers have been endorsed and recommended by many international consensus guidelines. In this letter, we reviewed the literature and evidence supporting the use of serum biomarkers as a dynamic test to monitor the status of atrophic gastritis.
Collapse
Affiliation(s)
- Huck-Joo Tan
- Department of Gastroenterology and Hepatology, Cengild GI Medical Center, Kuala Lumpur 50490, Malaysia
| | - Eunice Zhi-Yi Tan
- Department of Internal Medicine, University Hospital Southampton, Southampton SO16 6YD, United Kingdom
| |
Collapse
|
14
|
Yamaguchi N, Sakaguchi T, Wei JJ, Tazoe Y, Inamine T, Fukuda D, Ohnita K, Hirayama T, Isomoto H, Matsushima K, Tsukamoto K. The C/C Genotype of rs1231760 in RGS2 Is a Risk Factor for the Progression of H. pylori-Positive Atrophic Gastritis by Increasing RGS2 Expression. Diagnostics (Basel) 2024; 14:2563. [PMID: 39594230 PMCID: PMC11592620 DOI: 10.3390/diagnostics14222563] [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: 08/26/2024] [Revised: 10/26/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Chronic gastritis caused by Helicobacter pylori (H. pylori) infection can progress to gastric cancer through atrophic gastritis (AG). The risk of gastric cancer increases with the progression of AG. Therefore, investigating the risk factors for the progression of AG is important. METHODS Using the GTEx and GEO databases, we extracted thirty-four candidate genes involved in the progression of AG. Then, with in silico analysis using HaploReg v4.1 and JASPAR (Matrix ID: MA0113.3), we extracted rs1231760 of RGS2 as a key single-nucleotide polymorphism (SNP) that could be involved in the functional change in the candidate gene. A correlation analysis between the selected SNP and AG in 200 H. pylori-positive and 302 H. pylori-negative participants was conducted. For functional analysis of the SNP, a dual-luciferase assay using reporter plasmids with a major or minor allele sequence was carried out. RESULTS The frequency of the C/C genotype of rs1231760 was higher in the AG group than in the non-AG group (p = 0.0471). Functional analysis showed that the transcriptional activities were higher at the dexamethasone-stimulating C allele than at the others (p < 0.05). CONCLUSIONS The C/C genotype of rs1231760 in RGS2 could be a biomarker of high-risk H. pylori-positive AG because of an increase in RGS2 expression.
Collapse
Affiliation(s)
- Naoyuki Yamaguchi
- Department of Gastroenterology and Hepatology, Graduate School of Biological Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Takuki Sakaguchi
- Department of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, 36-1 Nishi-Cho, Yonago 683-8504, Japan
| | - Jing-Jing Wei
- Department of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, 36-1 Nishi-Cho, Yonago 683-8504, Japan
- Department of Endoscopy, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350004, China
| | - Yuna Tazoe
- Department of Pharmacotherapeutics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Tatsuo Inamine
- Department of Pharmacotherapeutics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Daisuke Fukuda
- Department of Gastroenterology and Hepatology, Graduate School of Biological Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
- Department of Surgical Oncology, Graduate School of Biological Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
- Fukuda Yutaka Surgical Clinic, 3-5 Hamaguchi-machi, Nagasaki 852-8107, Japan
| | - Ken Ohnita
- Department of Gastroenterology and Hepatology, Graduate School of Biological Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
- Shunkaikai Inoue Hospital, 6-12 Takara-machi, Nagasaki 850-0045, Japan
| | - Tatsuro Hirayama
- Department of Pharmacotherapeutics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Hajime Isomoto
- Department of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, 36-1 Nishi-Cho, Yonago 683-8504, Japan
| | - Kayoko Matsushima
- Department of Gastroenterology and Hepatology, Graduate School of Biological Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Kazuhiro Tsukamoto
- Department of Pharmacotherapeutics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| |
Collapse
|
15
|
Alruwaii ZI, Alsayed A, Albagashi J, Poveda J, Suliman WA, Al-Obaidy KI, Aljaroudi M, Montgomery E. Prevalence and Clinicopathological Features of Autoimmune Metaplastic Atrophic Gastritis in the Eastern Province of Saudi Arabia: A Regional Study. Int J Surg Pathol 2024:10668969241295348. [PMID: 39533765 DOI: 10.1177/10668969241295348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Introduction. Autoimmune metaplastic atrophic gastritis (AMAG, also termed autoimmune gastritis) is a chronic gastritis of autoimmune pathogenesis. Although its clinical and pathological features are well-documented in many countries, data from Middle Eastern populations remain scarce. This study examined the prevalence of AMAG in gastric specimens from the region, specifically from Saudi Arabia. Methods. We conducted a retrospective review of the pathology database of gastric specimens with a diagnosis of AMAG between 2020 and 2023. Detailed clinical, endoscopic, and pathological features of identified features were described. Result. Of the 978 gastric biopsies received, 17 patients were diagnosed with AMAG. The cohort comprised 11 women (64.7%) and 6 men (35.3%), presenting at a median age of 50 years (range: 32-85). Clinical manifestations varied widely, from abdominal pain (n = 6), dyspepsia (n = 2), symptomatic anemia with significant vitamin B12 deficiency (2 of 17) to asymptomatic/incidentally diagnosed patients (5 of 17). The tissue samples showed varying histological characteristics, with some showing lymphoplasmacytic infiltrate, mucosal atrophy, and hyperplasia of enterochromaffin-like cells. Conclusion. The observed prevalence of AMAG in our study aligns with global averages reported for other populations. The diverse clinical presentations highlight the need for awareness of findings in AMAG in gastric biopsies to ensure appropriate clinical management.
Collapse
Affiliation(s)
- Zainab I Alruwaii
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Anwar Alsayed
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Jafar Albagashi
- Division of Gastroenterology, Dammam Medical Complex, Dammam, Saudi Arabia
| | - Julio Poveda
- Department of Pathology, University of Miami Miller School of Medicine, Miami, USA
| | - Wael Al Suliman
- Division of Gastroenterology, Dammam Medical Complex, Dammam, Saudi Arabia
| | - Khaleel I Al-Obaidy
- Department of Pathology and Laboratory Medicine, Henry Ford Health, Detroit, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Mahdi Aljaroudi
- Division of Gastroenterology, Dammam Medical Complex, Dammam, Saudi Arabia
| | - Elizabeth Montgomery
- Department of Pathology, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
16
|
Rudolph JJ, Agyei O, Telvizian T, Ghaneie A. Gastric Neuroendocrine Tumors and Pernicious Anemia: A Case Report and Literature Review. Cureus 2024; 16:e73553. [PMID: 39669826 PMCID: PMC11637537 DOI: 10.7759/cureus.73553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Gastrointestinal neuroendocrine tumors (GI-NETs) are rare neoplasms, with the gastric (stomach) subtype (G-NETs) representing a significant clinical focus. Type 1 G-NETs are particularly noteworthy due to their relationship with autoimmune atrophic gastritis (AAG) and pernicious anemia (PA), conditions that impact vitamin B12 absorption. This report presents the case of a patient with a type 1 G-NET identified at the initial diagnosis of PA, demonstrating the connection between these conditions. In the literature review, we discuss the general mechanisms underlying PA, including its etiology, pathogenesis, clinical presentations, and diagnostic approaches. Emphasis is placed on the importance of recognizing and diagnosing this condition early, given the treatable nature of the associated gastric neuroendocrine dysregulation. Additionally, the report examines the broad spectrum of G-NETs, with a special emphasis on the characteristics of type 1 tumors. By considering recent developments in the field, we provide an overview of the current understanding of G-NET epidemiology, classification, clinical features, diagnosis, and management strategies.
Collapse
Affiliation(s)
| | - Obed Agyei
- Hematology and Medical Oncology, Lankenau Medical Center, Wynnewood, USA
| | - Talar Telvizian
- Hematology and Medical Oncology, Lankenau Medical Center, Wynnewood, USA
| | - Arezoo Ghaneie
- Hematology and Medical Oncology, Lankenau Medical Center, Wynnewood, USA
| |
Collapse
|
17
|
Nova-Camacho LM, De Burgos S, Ruiz Diaz I, Collins K. Comprehensive clinicopathologic features in autoimmune atrophic gastritis: Insights from a European cohort of 57 patients. Pathol Res Pract 2024; 263:155631. [PMID: 39357180 DOI: 10.1016/j.prp.2024.155631] [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: 08/22/2024] [Revised: 09/22/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
CONTEXT Autoimmune atrophic gastritis (AAG) is a frequently underdiagnosed disease due to its broad-spectrum clinical presentation. The diagnosis is based on histological confirmation of corpus-restricted metaplastic chronic atrophic gastritis. OBJECTIVE To thoroughly describe the histological features of a European cohort of AAG patients. DESIGN Clinical and pathological data of 57 out of 676 patients diagnosed with AAG were reviewed. RESULTS Thirty-nine patients were female and eighteen were male. The mean age was 62 years. Antibodies were identified in 32/42 patients (76 %). Vitamin B12 levels were low (< 200 pg/mL) in 37/54 patients (69 %). Serum gastrin levels was elevated (> 115 pg/mL) in all cases tested. Associated autoimmune/inflammatory conditions were identified in 20/57 patients (35 %). Histologically, deep chronic inflammation was present in 46/57 (81 %) patients. Complete destruction of oxyntic glands was observed in 45/57 (79 %) patients. Pyloric metaplasia was present in 54/57 (95 %) patients, intestinal metaplasia in 51/57 (89 %) patients, and pancreatic metaplasia in 20/57 (35 %) patients. Among ECL cell proliferation, linear hyperplasia was present in all 57/57 patients, micronodular hyperplasia in 55/57 patients, and adenomatoid hyperplasia in 10/57 patients. ECL cell dysplasia was identified in 5/57 patients, and neuroendocrine microtumor in 4/57 patients. CONCLUSIONS The diagnosing of AAG remains challenging due to the greater variability in symptoms than previously recognized. It is important to consider chronic AAG, especially with other concurrent autoimmune conditions. The importance of accurate diagnosis and surveillance is based on the potential development of type 1 gastric neuroendocrine tumor and increased risk of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Luiz M Nova-Camacho
- Department of Pathology, Donostia University Hospital, San Sebastian, Gipuzkoa 20014, Spain; Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32605, USA.
| | - Saul De Burgos
- Department of Pathology, Donostia University Hospital, San Sebastian, Gipuzkoa 20014, Spain
| | - Irune Ruiz Diaz
- Department of Pathology, Donostia University Hospital, San Sebastian, Gipuzkoa 20014, Spain
| | - Katrina Collins
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
18
|
Uchiyama K, Ando T, Kishimoto E, Nishimura T, Imamoto E, Takagi T, Ishikawa T, Naito Y, Itoh Y. Correlation of gastrointestinal symptom rating scale and frequency scale for the symptoms of gastroesophageal reflux disease with endoscopic findings. Scand J Gastroenterol 2024; 59:1220-1228. [PMID: 39301940 DOI: 10.1080/00365521.2024.2406537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Gastroesophageal Reflux Disease (GERD) is caused by the reflux of gastric contents into the esophagus and has a 13% global prevalence that is increasing. GERD symptoms negatively impact physical, social, and emotional quality of life. The Frequency Scale for the Symptoms of GERD (FSSG) and the Gastrointestinal Symptom Rating Scale (GSRS) determine the efficacy of treatment but may not correlate with endoscopically estimated esophageal mucosal injury severity. We aimed to probe the correlation between FSSG, GSRS, and esophageal injury severity to evaluate whether these scores can predict GERD severity. METHODS A total of 2962 patients who underwent physical examinations, including upper gastrointestinal endoscopy, at the Kyoto Kuramaguchi Medical Center, Japan, were enrolled in this study. Upper gastrointestinal endoscopy was used to diagnose fundic mucosal atrophy, reflux esophagitis based on the Los Angeles (LA) classification, gastroesophageal flap value function (GEFV) based on Hill's classification, and Barrett's esophagus. Endoscopic diagnoses were examined for correlations with FSSG and GSRS scores. RESULTS In reflux esophagitis, FSSG and GSRS scores correlated with LA-B and LA-C endoscopic diagnosis but not with LA-M and LA-A endoscopic findings. Multiple regression analysis results were similar. FSSG scores reflected advanced fundic gland mucosal atrophy, while GSRS scores associated with high grade of GEFV. CONCLUSIONS This is the first report to examine the correlation between FSSG and GSRS scores and endoscopic findings in a relatively large patient population. Our findings suggest that these scores can diagnose the severity of reflux esophagitis.
Collapse
Affiliation(s)
- Kazuhiko Uchiyama
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Etsuko Kishimoto
- Center for Health Promotion, Japanses Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | | | | | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
19
|
Jin G, Liu K, Guo Z, Dong Z. Precision therapy for cancer prevention by targeting carcinogenesis. Mol Carcinog 2024; 63:2045-2062. [PMID: 39140807 DOI: 10.1002/mc.23798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024]
Abstract
Cancer represents a major global public health burden, with new cases estimated to increase from 14 million in 2012 to 24 million by 2035. Primary prevention is an effective strategy to reduce the costs associated with cancer burden. For example, measures to ban tobacco consumption have dramatically decreased lung cancer incidence and vaccination against human papillomavirus can prevent cervical cancer development. Unfortunately, the etiological factors of many cancer types are not completely clear or are difficult to actively control; therefore, the primary prevention of such cancers is not practical. In this review, we update the progress on precision therapy by targeting the whole carcinogenesis process, especially for three high-risk groups: (1) those with chronic inflammation, (2) those with inherited germline mutations, and (3) those with precancerous lesions like polyps, gastritis, actinic keratosis or dysplasia. We believe that attenuating chronic inflammation, treating precancerous lesions, and removing high-risk tissues harboring germline mutations are precision methods for cancer prevention.
Collapse
Affiliation(s)
- Guoguo Jin
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiping Guo
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Xiang Z, Guan H, Zhao X, Xie Q, Hu X, Liu W, Sun X, Zhang S, Li M, Wang C. Characterization of active alkaloids and metabolites in rats after oral administration of Zuojin Pill using UHPLC-Q-TOF-MS combined with bioinformatics and molecular docking analyses. J Pharm Biomed Anal 2024; 249:116340. [PMID: 38986349 DOI: 10.1016/j.jpba.2024.116340] [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: 04/14/2024] [Revised: 06/18/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
Zuojin Pill (ZJP), a traditional Chinese medicine prescription composed of Rhizoma Coptidis and Euodiae Fructus in the ratio of 6:1 (w/w), has been widely used for the treatment of gastric disorders. However, an in-depth understanding of in vivo metabolism and distribution profiles of protoberberine alkaloids (PBAs) and indole alkaloids (IDAs) in ZJP is lacking. In this study, a method using ultra-high performance liquid chromatography coupled with quadruple time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) was developed to systematically screen the alkaloids and their metabolites in rat plasma and various tissues after oral administration of ZJP. Furthermore, bioinformatics and molecular docking analyses were conducted to elucidate the contribution of the alkaloids and metabolites enriched in the stomach to the therapeutic effect of ZJP on gastritis. A total of 33 compounds, including 7 prototype alkaloids and 26 metabolites, were chemically defined or tentatively identified in this work. The metabolic pathways of PBAs (hydroxylation, oxidation, reduction, demethylation, demethylenation, glucuronide conjugation, sulfate conjugation) and IDAs (hydroxylation, glucuronide conjugation) were revealed. Notably, 7 prototype alkaloids and 18 metabolites were detected in the stomach, indicating their propensity for gastric distribution. These alkaloids and metabolites showed strong affinities with the 7 hub targets associated with gastritis, such as CCR7, CXCR4, IL6, IFNG, CCL2, TNF, and PTPRC, and could be considered the potential active substances of ZJP for treating gastritis. In conclusion, this study clarified the gastric distribution propensity of PBAs and IDAs and their metabolites, as well as their favorable binding interactions with gastritis-related targets, which could provide essential data for the further study of the pharmacodynamic material basis and gastroprotective mechanism of ZJP.
Collapse
Affiliation(s)
- Zedong Xiang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Huida Guan
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Xiang Zhao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Qi Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Xianrun Hu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Wenkang Liu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Xin Sun
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Sitong Zhang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Manlin Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China.
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China.
| |
Collapse
|
21
|
Ma XZ, Zhou N, Luo X, Guo SQ, Mai P. Update understanding on diagnosis and histopathological examination of atrophic gastritis: A review. World J Gastrointest Oncol 2024; 16:4080-4091. [DOI: 10.4251/wjgo.v16.i10.4080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
Chronic atrophic gastritis (CAG) is a complex syndrome in which long-term chronic inflammatory stimulation causes gland atrophy in the gastric mucosa, reducing the stomach's ability to secrete gastric juice and pepsin, and interfering with its normal physiological function. Multiple pathogenic factors contribute to CAG incidence, the most common being Helicobacter pylori infection and the immune reactions resulting from gastric autoimmunity. Furthermore, CAG has a broad spectrum of clinical manifestations, including gastroenterology and extra-intestinal symptoms and signs, such as hematology, neurology, and oncology. Therefore, the initial CAG evaluation should involve the examination of clinical and serological indicators, as well as diagnosis confirmation via gastroscopy and histopathology if necessary. Depending on the severity and scope of atrophy affecting the gastric mucosa, a histologic staging system (Operative Link for Gastritis Assessment or Operative Link on Gastritis intestinal metaplasia) could also be employed. Moreover, chronic gastritis has a higher risk of progressing to gastric cancer (GC). In this regard, early diagnosis, treatment, and regular testing could reduce the risk of GC in CAG patients. However, the optimal interval for endoscopic monitoring in CAG patients remains uncertain, and it should ideally be tailored based on individual risk evaluations and shared decision-making processes. Although there have been many reports on CAG, the precise etiology and histopathological features of the disease, as well as the diagnosis of CAG patients, are yet to be fully elucidated. Consequently, this review offers a detailed account of CAG, including its key clinical aspects, aiming to enhance the overall understanding of the disease.
Collapse
Affiliation(s)
- Xiu-Zhen Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Department of Gastroenterology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China
| | - Ni Zhou
- Department of Gastroenterology, Xi'an International Medical Center, Xi’an 710000, Shaanxi Province, China
| | - Xiu Luo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Si-Qi Guo
- Department of Gastroenterology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Ping Mai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Department of Gastroenterology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
22
|
Tang Q, Wang R, Niu H, Li Y, Li Y, Hu Z, Liu X, Tao Y. Mapping network connection and direction among symptoms of depression and anxiety in patients with chronic gastritis. Psych J 2024; 13:824-834. [PMID: 38616130 PMCID: PMC11444727 DOI: 10.1002/pchj.757] [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: 12/27/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024]
Abstract
Regarding neurophysiological and developmental findings, anxiety and depression are usual comorbidities of gastritis patients. However, research related to anxiety and depression among chronic gastritis patients was conducted on the disease level while ignoring symptoms. Hence, we rendered the network approach to reveal the symptoms of anxiety and depression among chronic gastritis patients. Three hundred and sixty-nine chronic gastritis patients (female = 139, Mage = 55.87 years) were asked to complete the Self-Rating Anxiety Scale and Self-Rating Depression Scale. Three symptom networks and one directed acyclic graph (DAG) network were formed. First, in the anxiety network of chronic gastritis patients, dizziness was the most influential symptom. In the depression network of chronic gastritis patients, depressed affect and psychomotor retardation were the influential symptoms. Second, panic, easy fatiguability, weakness, palpitation, depressed affect, tachycardia, fatigue, and psychomotor agitation bridged the anxiety-depression network of chronic gastritis patients. Third, DAG networks showed that anxiousness and hopelessness could trigger other symptoms in the anxiety-depression networks of chronic gastritis patients. The current study provided insightful information on patients with chronic gastritis by examining the structures of symptoms.
Collapse
Affiliation(s)
- Qihui Tang
- Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Beijing Normal University, Beijing, China
| | - Rui Wang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Haiqun Niu
- School of Psychology, Nanjing Normal University, Nanjing, China
| | - Yifang Li
- Department of Chinese Medicine Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Yuting Li
- Department of Chinese Medicine Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
- School of Psychology, Shanghai University of Sport, Shanghai, China
| | - Zichao Hu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Xiangping Liu
- Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Beijing Normal University, Beijing, China
| | - Yanqiang Tao
- Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Beijing Normal University, Beijing, China
| |
Collapse
|
23
|
Burke E, Harkins P, Arumugasamy M. Incidence of Gastric Adenocarcinoma in Those With Gastric Atrophy: A Systematic Review. Cureus 2024; 16:e71768. [PMID: 39429990 PMCID: PMC11488155 DOI: 10.7759/cureus.71768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 10/22/2024] Open
Abstract
Gastric atrophy (GA), or atrophic gastritis, is a pre-neoplastic lesion of gastric cancer (GC). It is part of the Correa cascade, which culminates in intestinal-type gastric adenocarcinoma. The cascade posits that intestinal-type gastric adenocarcinoma develops along a defined pathway of pre-neoplastic stages. The cascade begins with chronic gastritis, most commonly caused by Helicobacter pylori (H. pylori) infection, and proceeds through GA, gastric intestinal metaplasia (GIM), both complete and incomplete, dysplasia, both low and high-grade, and culminating in intestinal-type gastric adenocarcinoma. Attempts in Europe have been made to identify patients at risk of developing GC and target them with surveillance oesophagogastroduodenoscopy (OGD). However, there remains uncertainty about GA's risk of developing into GC. This poses issues in terms of guiding the need for and determining intervals for surveillance OGDs, which are a costly form of surveillance. As such, we attempted to gather all available studies assessing the risk of GC developing from GA, which is the first step in the Correa cascade. This study was a comprehensive systematic review of published papers, reported per the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. This systematic review, which included a substantial 25,455 patients across 18 studies, found that the relative risk (RR) of GC in those with GA, using standardised incidence ratios as a measure of RR, was 15.1, with a 95% confidence interval ranging from 13.5 to 16.9. We conclude that GA does increase the risk of developing GC, and this risk may be higher than previously appreciated. Further large-scale studies are needed in Western cohorts of patients to precisely define this risk and guide the need for surveillance programs. These future studies must be standardised to account for H. pylori status, the topographical distribution of the GA, and the methods for assessing the degree of GA.
Collapse
Affiliation(s)
- Eoghan Burke
- Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, IRL
| | - Patricia Harkins
- Medicine, Royal College of Physicians of Ireland (RCPI), Dublin, IRL
| | | |
Collapse
|
24
|
Latorre G, Vargas JI, Shah SC, Ivanovic-Zuvic D, Achurra P, Fritzsche M, Leung JS, Ramos B, Jensen E, Uribe J, Montero I, Gandara V, Robles C, Bustamante M, Silva F, Dukes E, Corsi O, Martínez F, Binder V, Candia R, González R, Espino A, Agüero C, Sharp A, Torres J, Roa JC, Pizarro M, Corvalan AH, Rabkin CS, Camargo MC, Riquelme A. Implementation of the updated Sydney system biopsy protocol improves the diagnostic yield of gastric preneoplastic conditions: Results from a real-world study. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:793-803. [PMID: 37598725 PMCID: PMC10875143 DOI: 10.1016/j.gastrohep.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/30/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND The updated Sydney system biopsy protocol (USSBP) standardizes the sampling of gastric biopsies for the detection of preneoplastic conditions (e.g., gastric intestinal metaplasia [GIM]), but the real-world diagnostic yield is not well-described. AIM To determine whether regular application of USSBP is associated with higher detection of chronic atrophic gastritis (CAG), GIM and autoimmune gastritis (AIG). METHODS We performed a real-world retrospective study at an academic urban tertiary hospital in Chile. We manually reviewed medical records from consecutive patients undergoing esophagogastroduodenoscopy (EGD) from January to December 2017. Seven endoscopists who performed EGDs were categorized into two groups (USSBP 'regular' and USSBP 'infrequent') based on USSBP adherence, using minimum 20% adherence as the prespecified threshold. Multivariable logistic regression models were used to estimate the odds ratios (aOR) and 95% confidence intervals (CI) for the association between endoscopist groups and the likelihood of diagnosing CAG, GIM or AIG. RESULTS 1206 patients were included in the study (mean age: 58.5; 65.3% female). The USSBP regular group demonstrated a higher likelihood of detecting CAG (20% vs. 5.3%; aOR 4.03, 95%CI: 2.69-6.03), GIM (12.2% vs. 3.4%; aOR 3.91, 95%CI: 2.39-6.42) and AIG (2.9% vs. 0.8%; aOR 6.52, 95%CI: 1.87-22.74) compared to infrequent group. Detection of advanced-stage CAG (Operative Link for Gastritis Assessment stage III/IV) was significantly higher in the USSBP regular vs. infrequent group (aOR 5.84, 95%CI: 2.23-15.31). CONCLUSIONS Routine adherence to USSBP increases the detection rates of preneoplastic conditions, including CAG, GIM and AIG. Standardized implementation of USSBP should be considered in high gastric cancer risk populations.
Collapse
Affiliation(s)
- Gonzalo Latorre
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Ignacio Vargas
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Shailja C Shah
- Gastroenterology Section, Veterans Affairs, San Diego Healthcare System, San Diego, CA, USA; Division of Gastroenterology, University of California, San Diego, San Diego, CA, USA
| | - Danisa Ivanovic-Zuvic
- Department of Internal Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Achurra
- Department of Digestive Surgery, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Martín Fritzsche
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jai-Sen Leung
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bernardita Ramos
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Elisa Jensen
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javier Uribe
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Isabella Montero
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vicente Gandara
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Robles
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel Bustamante
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Silva
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eitan Dukes
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar Corsi
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisca Martínez
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victoria Binder
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roberto Candia
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Robinson González
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alberto Espino
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Agüero
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Allan Sharp
- Department of Digestive Surgery, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javiera Torres
- Department of Pathology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, Pontificia Universidad Católica de Chile, Santiago, Chile; Center for Cancer Prevention and Control, CECAN, Chile
| | - Margarita Pizarro
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H Corvalan
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDIS), Santiago, Chile
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Arnoldo Riquelme
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile; Center for Cancer Prevention and Control, CECAN, Chile.
| |
Collapse
|
25
|
Kubo T, Adachi Y, Sasaki Y, Adachi Y, Yoshida Y, Endo T, Ishii Y, Takahashi H, Goto A. Synchronous cancers of the stomach and esophagus in a patient with autoimmune gastritis and pernicious anemia: a case report and review of the literature. Int Cancer Conf J 2024; 13:367-373. [PMID: 39398913 PMCID: PMC11465013 DOI: 10.1007/s13691-024-00689-z] [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: 01/18/2024] [Accepted: 05/24/2024] [Indexed: 10/15/2024] Open
Abstract
Type-A gastritis/autoimmune gastritis (AIG) has gained renewed attention due to declining Helicobacter pylori infection rates and increasing eradication. AIG is associated with pernicious anemia (PA) and is prone to complicate various tumors, such as gastric cancer and neuroendocrine tumors. This report describes a case of AIG with PA in which gastric and esophageal cancers arose simultaneously. An 86-year-old woman had been diagnosed with PA and AIG 9 years earlier. As routine blood tests revealed high levels of carcinoembryonic antigen, she underwent esophagogastroduodenoscopy, which revealed a type 0-IIa lesion in the middle part of the stomach and a type 0-IIa + IIb lesion in the lower esophagus. Endoscopic submucosal dissection was performed on both lesions, since neither distant nor lymph node metastases were identified. Histological examination showed gastric tubular adenocarcinoma and esophageal squamous cell carcinoma. Immunohistology revealed that cells from neither cancer expressed gastrin, gastrin receptor, or p53. Whole-exome sequencing showed 8 gene mutations in the esophageal cancer and 6 mutations in 5 genes in the gastric cancer. The reason for the lack of p53 immunostaining was that TP53 was mutated in these cancers, although TP53 mutations differed. Thus, TP53 mutations may not be detected by immunostaining alone. When treating patients with AIG/PA, clinicians must be aware of the possibility of esophageal cancer coexisting with gastric cancer.
Collapse
Affiliation(s)
- Toshiyuki Kubo
- Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakaba-dai Hospital, 2-18 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, 062-0052 Japan
| | - Yasushi Adachi
- Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakaba-dai Hospital, 2-18 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, 062-0052 Japan
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-Ku, Sapporo, 060-8543 Japan
| | - Yasushi Sasaki
- Department of Biology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-Ku, Sapporo, 060-8543 Japan
| | - Yasuyo Adachi
- Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakaba-dai Hospital, 2-18 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, 062-0052 Japan
| | - Yukinari Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakaba-dai Hospital, 2-18 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, 062-0052 Japan
| | - Takao Endo
- Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakaba-dai Hospital, 2-18 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, 062-0052 Japan
| | - Yoshifumi Ishii
- Department of Pathology, Sapporo Shirakaba-dai Hospital, 2-18 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, 062-0052 Japan
| | - Hiroaki Takahashi
- Department of Gastroenterology and Hepatology, Keiyukai-daini Hospital, Hondori 13-chome kita7-1, Shiroishi-Ku, Sapporo, 003-0027 Japan
| | - Akira Goto
- Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakaba-dai Hospital, 2-18 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, 062-0052 Japan
| |
Collapse
|
26
|
Jannot AS, Girardeau Y, Chaussade S, Cerf-Bensussan N, Malamut G. Increased risk of gastric cancer in relation with pernicious anaemia in patients with primary antibody deficiency: A nationwide case control study. Dig Liver Dis 2024; 56:1760-1765. [PMID: 38853087 DOI: 10.1016/j.dld.2024.05.032] [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: 03/25/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND/AIM We aimed to assess gastrointestinal cancers risks in a large cohort of individuals with primary antibody deficiency (PAD) and their association with risk of autoimmune and inflammatory gastrointestinal diseases. METHODS Investigating a French national database of inpatient admissions between 2010 and 2018, we identified 12,748 patients with PAD and 38,244 control non-exposed individuals. We performed multiple exposed-non-exposed studies using conditional logistic regression. RESULTS In comparison with non-exposed patients, PAD patients had increased risk of in situ gastric carcinoma (Odds Ratio (OR) =10.5 [95 % CI 2.2; 50.5]), malignant gastric tumor (OR=3.2 [95 % CI 2.2; 4.4]) and colorectal cancer (OR=1.2 [95 % CI 1; 1.5]). PAD patients had also increased risk of pernicious anaemia (OR=8 |95 % CI 5.6; 11.5]), Crohn's disease (OR= 4.4 [95 % CI 3.5; 5.6]), ulcerative colitis (OR=2.9 [95 % CI 2.4; 3.6]) and coeliac disease (OR=13.3 [95 % CI 9.1; 19.5]). Within patients with gastric cancer, those with PAD had increased risk of pernicious anaemia (OR=8.4 [95 % CI 1.5; 215]; p = 0.01) but not of H. pylori infection. CONCLUSIONS Risk of gastric cancer is particularly high in PAD patients and notably risk of in situ gastric carcinoma in association with pernicious anaemia. It supports indication of early endoscopic screening in these patients.
Collapse
Affiliation(s)
- Anne-Sophie Jannot
- French National Rare Disease Registry (BNDMR), Greater Paris University Hospitals (AP-HP), Université Paris Cité, Paris, France; Université Paris Cité, HeKA, INRIA Paris, Inserm, Centre de Recherche des Cordeliers- Université Paris Cité, Paris, France
| | - Yannick Girardeau
- Department of Clinical Investigation and Clinical Epidemiology, AP-HP-Centre-Université Paris Cité, Hôpital Européen Georges Pompidou, Paris, France
| | - Stanislas Chaussade
- Department of Gastroenterology, AP-HP. Centre- Université Paris Cité, Hôpital Cochin, Paris, France
| | - Nadine Cerf-Bensussan
- Université de Paris, INSERM UMR 1163 and Imagine Institute, Laboratory of Intestinal Immunity, Université Paris Cité, Paris, France
| | - Georgia Malamut
- Department of Gastroenterology, AP-HP. Centre- Université Paris Cité, Hôpital Cochin, Paris, France; Université de Paris, INSERM UMR 1163 and Imagine Institute, Laboratory of Intestinal Immunity, Université Paris Cité, Paris, France.
| |
Collapse
|
27
|
Dave SR, Bagada JB, Desai JH, Jha S. The Study of Correlation between Symptoms of Gastritis and Endoscopic Biopsy. JOURNAL OF THE WEST AFRICAN COLLEGE OF SURGEONS 2024; 14:364-369. [PMID: 39309390 PMCID: PMC11412594 DOI: 10.4103/jwas.jwas_96_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/09/2023] [Indexed: 09/25/2024]
Abstract
Background Gastritis is a very common disorder that is widely distributed worldwide, representing one of the most prevalent pathological entities in gastroenterology and digestive endoscopy. This study aims to analyse the correlation between the endoscopic findings and the symptoms of gastritis. Upper gastro-intestinal (UGI) endoscopy is a widely used investigation for a variety of UGI symptoms namely dysphagia, dyspepsia, abdominal pain, etc., and when combined with biopsy, its diagnostic accuracy get increased manifold. Materials and Methods This is a study with 70 patients with symptoms of gastritis underwent UGI endoscopy and biopsy included in study. Biopsy was taken from gastric/duodenal mucosa and was sent for histopathology examination. Data of patient's age, gender, food habits, oral hygiene, etc., were then taken into consideration and findings of UGI endoscopy were correlated with these data and also correlated with histopathological report. Results Gastritis significantly affects the lifestyle of the majority of our participants as out of 70, 65 (95%) had gastritis in our study. Helicobacter pylori was present among 41.5 % of study participants. Other less common findings were esophagitis in 20 % and duodenitis in 7% of study participants. The mean age of participants were 46.76 ± 16.25 years. Conclusion Histopathology plays a major role in diagnosing H. pylori infection. So, histological examination as an adjunct to UGI endoscopy should be considered as best diagnostic tool rather than UGI endoscopy alone or clinical examination or radiological examination alone. It helps to treat the gastritis patients by diagnosing the underlying cause and management accordingly.
Collapse
Affiliation(s)
- Shravan R Dave
- Department of General Surgery, Pramukh Swami Medical College and Shree Krishna Hospital, Karamsad, Gujarat, India
| | - Jayeshkumar B Bagada
- Department of General Surgery, Pramukh Swami Medical College and Shree Krishna Hospital, Karamsad, Gujarat, India
| | - Jitesh H Desai
- Department of General Surgery, Pramukh Swami Medical College and Shree Krishna Hospital, Karamsad, Gujarat, India
| | - Shivangi Jha
- Department of General Surgery, Pramukh Swami Medical College and Shree Krishna Hospital, Karamsad, Gujarat, India
| |
Collapse
|
28
|
Zhou Y, Wang Q, Tang W, Ma Z, Yang Z, Li X, Chen W, Ma H, Ye X. Palmatine ameliorates N-methyl-N'-nitrosoguanidine-induced chronic atrophic gastritis through the STAT1/CXCL10 axis. FASEB J 2024; 38:e70037. [PMID: 39287361 DOI: 10.1096/fj.202401624r] [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: 07/15/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
Chronic atrophic gastritis (CAG) is a prevalent preneoplastic condition of the stomach. Palmatine (PAL), an isoquinoline alkaloid isolated from Rhizoma Coptidis (RC), has significant anti-inflammatory properties and is often used to treat gastrointestinal disorders. However, the mechanism of PAL on CAG remains unclear. In this study, N-methyl-N'-nitrosoguanidine (MNNG) was used to induce CAG inflammatory disease models in vivo and in vitro. The efficacy of five alkaloids in RC and the dose-dependent effects of the most effective PAL in CAG mice were evaluated in two animal experiments. RNA-seq and western blot revealed that PAL significantly improved IL-17, TNF, and NF-kappa B inflammation-related signaling pathways. Further hub gene prediction and experimental validation revealed that PAL modulated the STAT1/CXCL10 axis, thereby exerting attenuation of CAG through the regulation of IL-17, TNF-α, and p-p65 expression. In conclusion, PAL was proposed to mitigate MNNG-induced CAG, potentially through the inhibition of oxidative stress and inflammatory responses via the STAT1/CXCL10 axis. This approach is an effective complement to the use of PAL in the treatment of CAG.
Collapse
Affiliation(s)
- Yuan Zhou
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, China
| | - Qiaojiao Wang
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, China
| | - Wanyu Tang
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, China
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, China
| | - Zhipeng Yang
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xuegang Li
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Hang Ma
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, China
| |
Collapse
|
29
|
Jiang Q, Yu Y, Ren Y, Li S, He X. A review of deep learning methods for gastrointestinal diseases classification applied in computer-aided diagnosis system. Med Biol Eng Comput 2024:10.1007/s11517-024-03203-y. [PMID: 39343842 DOI: 10.1007/s11517-024-03203-y] [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: 05/22/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024]
Abstract
Recent advancements in deep learning have significantly improved the intelligent classification of gastrointestinal (GI) diseases, particularly in aiding clinical diagnosis. This paper seeks to review a computer-aided diagnosis (CAD) system for GI diseases, aligning with the actual clinical diagnostic process. It offers a comprehensive survey of deep learning (DL) techniques tailored for classifying GI diseases, addressing challenges inherent in complex scenes, clinical constraints, and technical obstacles encountered in GI imaging. Firstly, the esophagus, stomach, small intestine, and large intestine were located to determine the organs where the lesions were located. Secondly, location detection and classification of a single disease are performed on the premise that the organ's location corresponding to the image is known. Finally, comprehensive classification for multiple diseases is carried out. The results of single and multi-classification are compared to achieve more accurate classification outcomes, and a more effective computer-aided diagnosis system for gastrointestinal diseases was further constructed.
Collapse
Affiliation(s)
- Qianru Jiang
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, 310023, Zhejiang, P.R. China
| | - Yulin Yu
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, 310023, Zhejiang, P.R. China
| | - Yipei Ren
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, 310023, Zhejiang, P.R. China
| | - Sheng Li
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, 310023, Zhejiang, P.R. China
| | - Xiongxiong He
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, 310023, Zhejiang, P.R. China.
| |
Collapse
|
30
|
Qian ST, Zhao HY, Xie FF, Liu QS, Cai DL. Streptococcus anginosus in the development and treatment of precancerous lesions of gastric cancer. World J Gastrointest Oncol 2024; 16:3771-3780. [PMID: 39350992 PMCID: PMC11438778 DOI: 10.4251/wjgo.v16.i9.3771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/22/2024] [Accepted: 06/06/2024] [Indexed: 09/09/2024] Open
Abstract
The microbiota is strongly association with cancer. Studies have shown significant differences in the gastric microbiota between patients with gastric cancer (GC) patients and noncancer patients, suggesting that the microbiota may play a role in the development of GC. Although Helicobacter pylori (H. pylori) infection is widely recognized as a primary risk factor for GC, recent studies based on microbiota sequencing technology have revealed that non-H. pylori microbes also have a significant impact on GC. A recent study discovered that Streptococcus anginosus (S. anginosus) is more prevalent in the gastric mucosa of patients with GC than in that of those without GC. S. anginosus infection can spontaneously induce chronic gastritis, mural cell atrophy, mucoid chemotaxis, and heterotrophic hyperplasia, which promote the development of precancerous lesions of GC (PLGC). S. anginosus also disrupts the gastric barrier function, promotes the proliferation of GC cells, and inhibits apoptosis. However, S. anginosus is underrepresented in the literature. Recent reports suggest that it may cause precancerous lesions, indicating its emerging pathogenicity. Modern novel molecular diagnostic techniques, such as polymerase chain reaction, genetic testing, and Ultrasensitive Chromosomal Aneuploidy Detection, can be used to gastric precancerous lesions via microbial markers. Therefore, we present a concise summary of the relationship between S. anginosus and PLGC. Our aim was to further investigate new methods of preventing and treating PLGC by exploring the pathogenicity of S. anginosus on PLGC.
Collapse
Affiliation(s)
- Su-Ting Qian
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Hao-Yu Zhao
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Fei-Fei Xie
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Qing-Sheng Liu
- Science and Education Section, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Dan-Li Cai
- Intensive Care Unit, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 311122, Zhejiang Province, China
| |
Collapse
|
31
|
Dinis-Ribeiro M, Shah S, El-Serag H, Banks M, Uedo N, Tajiri H, Coelho LG, Libanio D, Lahner E, Rollan A, Fang JY, Moreira L, Bornschein J, Malfertheiner P, Kuipers EJ, El-Omar EM. The road to a world-unified approach to the management of patients with gastric intestinal metaplasia: a review of current guidelines. Gut 2024; 73:1607-1617. [PMID: 39122364 DOI: 10.1136/gutjnl-2024-333029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/14/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE During the last decade, the management of gastric intestinal metaplasia (GIM) has been addressed by several distinct international evidence-based guidelines. In this review, we aimed to synthesise these guidelines and provide clinicians with a global perspective of the current recommendations for managing patients with GIM, as well as highlight evidence gaps that need to be addressed with future research. DESIGN We conducted a systematic review of the literature for guidelines and consensus statements published between January 2010 and February 2023 that address the diagnosis and management of GIM. RESULTS From 426 manuscripts identified, 16 guidelines were assessed. There was consistency across guidelines regarding the purpose of endoscopic surveillance of GIM, which is to identify prevalent neoplastic lesions and stage gastric preneoplastic conditions. The guidelines also agreed that only patients with high-risk GIM phenotypes (eg, corpus-extended GIM, OLGIM stages III/IV, incomplete GIM subtype), persistent refractory Helicobacter pylori infection or first-degree family history of gastric cancer should undergo regular-interval endoscopic surveillance. In contrast, low-risk phenotypes, which comprise most patients with GIM, do not require surveillance. Not all guidelines are aligned on histological staging systems. If surveillance is indicated, most guidelines recommend a 3-year interval, but there is some variability. All guidelines recommend H. pylori eradication as the only non-endoscopic intervention for gastric cancer prevention, while some offer additional recommendations regarding lifestyle modifications. While most guidelines allude to the importance of high-quality endoscopy for endoscopic surveillance, few detail important metrics apart from stating that a systematic gastric biopsy protocol should be followed. Notably, most guidelines comment on the role of endoscopy for gastric cancer screening and detection of gastric precancerous conditions, but with high heterogeneity, limited guidance regarding implementation, and lack of robust evidence. CONCLUSION Despite heterogeneous populations and practices, international guidelines are generally aligned on the importance of GIM as a precancerous condition and the need for a risk-stratified approach to endoscopic surveillance, as well as H. pylori eradication when present. There is room for harmonisation of guidelines regarding (1) which populations merit index endoscopic screening for gastric cancer and GIM detection/staging; (2) objective metrics for high-quality endoscopy; (3) consensus on the need for histological staging and (4) non-endoscopic interventions for gastric cancer prevention apart from H. pylori eradication alone. Robust studies, ideally in the form of randomised trials, are needed to bridge the ample evidence gaps that exist.
Collapse
Affiliation(s)
- Mario Dinis-Ribeiro
- Department of Gastroenterology, Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- MEDCIDS (Department of Community Medicine, Health Information, and Decision), University of Porto, Porto, Portugal
| | - Shailja Shah
- Division of Gastroenterology, University of California and Jennifer Moreno Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Hashem El-Serag
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Matthew Banks
- University College London Hospital, University College London Hospitals NHS Foundation Trust, London, UK
| | - Noriya Uedo
- Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hisao Tajiri
- Endoscopy, The Jikei University School of Medicine, Tokyo, Japan
| | - Luiz Gonzaga Coelho
- Instituto Alfa de Gastrenterologia, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Diogo Libanio
- Department of Gastroenterology, Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- MEDCIDS (Department of Community Medicine, Health Information, and Decision), University of Porto, Porto, Portugal
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Antonio Rollan
- Facultad de Medicina Clinica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Leticia Moreira
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jan Bornschein
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Ernst J Kuipers
- Medical Department II, LMU University Clinic, München, Germany
| | - Emad M El-Omar
- UNSW Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
32
|
Huang M, Luo S, Yang J, Xiong H, Lu X, Ma X, Zeng J, Efferth T. Optimized therapeutic potential of Sijunzi-similar formulae for chronic atrophic gastritis via Bayesian network meta-analysis. EXCLI JOURNAL 2024; 23:1185-1207. [PMID: 39421026 PMCID: PMC11484511 DOI: 10.17179/excli2024-7618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024]
Abstract
Chronic atrophic gastritis (CAG) is considered as a significant risk factor for triggering gastric cancer incidence, if not effectively treated. Sijunzi decoction (SD) is a well-known classic formula for treating gastric disorders, and Sijunzi-similar formulae (SF) derived from SD have also been highly regarded by Chinese clinical practitioners for their effectiveness in treating chronic atrophic gastritis. Currently, there is a lack of meta-analysis for these formulae, leaving unclear which exhibits optimal efficacy. Therefore, we employed Bayesian network meta-analysis (BNMA) to evaluate the efficacy and safety of SF as an intervention for CAG and to establish a scientific foundation for the clinical utilization of SF. The result of meta-analysis demonstrated that the combination of SF and basic therapy outperformed basic therapy alone in terms of clinical efficacy rate, eradication rate of H. pylori, and incidence of adverse events. As indicated by the SUCRA value, Chaishao Liujunzi decoction (CLD) demonstrated superior efficacy in enhancing clinical effectiveness and ameliorating H. pylori infection, and it also showed remarkable effectiveness in minimizing the occurrence of adverse events. Comprehensive analysis of therapeutic efficacy suggests that CLD is most likely the optimal choice among these six formulations, holding potential value for optimizing clinical treatment strategies. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Meilan Huang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shiman Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiayue Yang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Huiling Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- TCM Regulating Metabolic Disease Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| |
Collapse
|
33
|
Chey WD, Howden CW, Moss SF, Morgan DR, Greer KB, Grover S, Shah SC. ACG Clinical Guideline: Treatment of Helicobacter pylori Infection. Am J Gastroenterol 2024; 119:1730-1753. [PMID: 39626064 DOI: 10.14309/ajg.0000000000002968] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/18/2024] [Indexed: 12/20/2024]
Abstract
ABSTRACT Helicobacter pylori is a prevalent, global infectious disease that causes dyspepsia, peptic ulcer disease, and gastric cancer. The American College of Gastroenterology commissioned this clinical practice guideline (CPG) to inform the evidence-based management of patients with H. pylori infection in North America. This CPG used Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) methodology to systematically analyze 11 Population, Intervention, Comparison, and Outcome questions and generate recommendations. Where evidence was insufficient or the topic did not lend itself to GRADE, expert consensus was used to create 6 key concepts. For treatment-naive patients with H. pylori infection, bismuth quadruple therapy (BQT) for 14 days is the preferred regimen when antibiotic susceptibility is unknown. Rifabutin triple therapy or potassium-competitive acid blocker dual therapy for 14 days is a suitable empiric alternative in patients without penicillin allergy. In treatment-experienced patients with persistent H. pylori infection, "optimized" BQT for 14 days is preferred for those who have not been treated with optimized BQT previously and for whom antibiotic susceptibility is unknown. In patients previously treated with optimized BQT, rifabutin triple therapy for 14 days is a suitable empiric alternative. Salvage regimens containing clarithromycin or levofloxacin should only be used if antibiotic susceptibility is confirmed. The CPG also addresses who to test, the need for universal post-treatment test-of-cure, and the current evidence regarding antibiotic susceptibility testing and its role in guiding the choice of initial and salvage treatment. The CPG concludes with a discussion of proposed research priorities to address knowledge gaps and inform future management recommendations in patients with H. pylori infection from North America.
Collapse
Affiliation(s)
- William D Chey
- Division of Gastroenterology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Colin W Howden
- University of Tennessee College of Medicine, Memphis, Tennessee, USA
| | - Steven F Moss
- Division of Gastroenterology, Department of Medicine, Providence VA Medical Center, Rhode Island Hospital & Brown University, Providence, Rhode Island, USA
| | - Douglas R Morgan
- Division of Gastroenterology and Hepatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Katarina B Greer
- Louis Stokes Cleveland Veteran Affairs Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Shilpa Grover
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Shailja C Shah
- Division of Gastroenterology, VA San Diego Healthcare System, Division of Gastroenterology, University of California San Diego, San Diego, California, USA
| |
Collapse
|
34
|
Niu W, Liu L, Dong Z, Bu X, Yao F, Wang J, Wu X, Chen C, Mao T, Wu Y, Yuan L, Wan X, Zhou H. A deep learning model based on magnifying endoscopy with narrow-band imaging to evaluate intestinal metaplasia grading and OLGIM staging: A multicenter study. Dig Liver Dis 2024; 56:1565-1571. [PMID: 38402085 DOI: 10.1016/j.dld.2024.02.001] [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: 11/05/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND AND PURPOSE Patients with stage III or IV of operative link for gastric intestinal metaplasia assessment (OLGIM) are at a higher risk of gastric cancer (GC). We aimed to construct a deep learning (DL) model based on magnifying endoscopy with narrow-band imaging (ME-NBI) to evaluate OLGIM staging. METHODS This study included 4473 ME-NBI images obtained from 803 patients at three endoscopy centres. The endoscopic expert marked intestinal metaplasia (IM) regions on endoscopic images of the target biopsy sites. Faster Region-Convolutional Neural Network model was used to grade IM lesions and predict OLGIM staging. RESULTS The diagnostic performance of the model for IM grading in internal and external validation sets, as measured by the area under the curve (AUC), was 0.872 and 0.803, respectively. The accuracy of this model in predicting the high-risk stage of OLGIM was 84.0%, which was not statistically different from that of three junior (71.3%, p = 0.148) and three senior endoscopists (75.3%, p = 0.317) specially trained in endoscopic images corresponding to pathological IM grade, but higher than that of three untrained junior endoscopists (64.0%, p = 0.023). CONCLUSION This DL model can assist endoscopists in predicting OLGIM staging using ME-NBI without biopsy, thereby facilitating screening high-risk patients for GC.
Collapse
Affiliation(s)
- Wenlu Niu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leheng Liu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhixia Dong
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiongzhu Bu
- School of Mechanical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Fanghao Yao
- School of Mechanical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Jing Wang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowan Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Congying Chen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiancheng Mao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulun Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Yuan
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinjian Wan
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Hui Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
35
|
Granot M, Beinvogl BC, Schvimer M, Goldsmith JD, Matar M, Ben Tov A, Feler AY, Nachum N, Morgenstern S, Mayer C, Shamir R, Weiss B, Shouval DS. Clinical characteristics and outcomes of pediatric patients with autoimmune gastritis. J Pediatr Gastroenterol Nutr 2024; 79:501-509. [PMID: 39010761 DOI: 10.1002/jpn3.12318] [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: 02/22/2024] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVES Autoimmune gastritis (AIG) is a rare chronic inflammatory disorder with potential long-term sequelae including gastric neoplasia. There is limited data on the natural history of pediatric AIG. We aimed to characterize the clinical course and outcomes of children with AIG. METHODS This was a multicenter retrospective study that included pediatric patients diagnosed with AIG between January 1, 2000 and December 31, 2021. Diagnosis of AIG was based on the demonstration of histological corpus-predominant atrophic gastritis, with or without positive antiparietal cell (APCA) or anti-intrinsic factor (IF) antibodies. Demographic, clinical, laboratory, endoscopic, and histologic data were retrieved, along with follow-up data. RESULTS Thirty-three patients, (23 females [69.7%], median age 12.0 [interquartile range 7.0-15.0] years at diagnosis) were identified. Twenty-two patients (66.7%) had positive APCA and/or anti-IF serology. The most common presenting manifestation was iron deficiency anemia (75%), and accompanying autoimmune disorders were significantly more common in patients with positive serology (62% vs. 18%, p < 0.05). Pseudo-pyloric or intestinal-type metaplasia was present at diagnosis in eight patients (24%), and 11 additional patients (33%) developed metaplasia during a median follow-up time of 27 (17.5-48.3) months. One patient developed a type 1 gastric neuroendocrine tumor. Helicobacter pylori was identified in only one patient, while two patients had prior eradication. Endoscopic and histologic improvements weren't identified in any patients. CONCLUSIONS AIG should be considered in patients with autoimmunity and resistant iron-deficiency anemia. H. pylori infection may not be associated with pediatric AIG. The development of neuroendocrine tumor in one patient, and the high rates of metaplasia, highlight the importance of surveillance.
Collapse
Affiliation(s)
- Maya Granot
- Pediatric Gastroenterology and Nutrition Unit, Edmond and Lily Safra Children's Hospital, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Beate C Beinvogl
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Michael Schvimer
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pathology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Jeffrey D Goldsmith
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Manar Matar
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Amir Ben Tov
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Gastroenterology Institute, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Anat Y Feler
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Gastroenterology Institute, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Nurit Nachum
- Pediatric Gastroenterology and Nutrition Unit, Edmond and Lily Safra Children's Hospital, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sara Morgenstern
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pathology, Rabin Medical Center, Petah Tikva, Israel
| | - Chen Mayer
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pathology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Raanan Shamir
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Batia Weiss
- Pediatric Gastroenterology and Nutrition Unit, Edmond and Lily Safra Children's Hospital, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dror S Shouval
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| |
Collapse
|
36
|
Bazin T, Nozeret K, Julié C, Lamarque D, Touati E. Protein Biomarkers of Gastric Preneoplasia and Cancer Lesions in Blood: A Comprehensive Review. Cancers (Basel) 2024; 16:3019. [PMID: 39272877 PMCID: PMC11394471 DOI: 10.3390/cancers16173019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Gastric cancer (GC) is a major cause of cancer-related mortality worldwide. It is often associated with a bad prognosis because of its asymptomatic phenotype until advanced stages, highlighting the need for its prevention and early detection. GC development is preceded by the emergence of gastric preneoplasia lesions (GPNLs), namely atrophic gastritis (AG), intestinal metaplasia (IM), and dysplasia (DYS). GC is currently diagnosed by endoscopy, which is invasive and costly and has limited effectiveness for the detection of GPNLs. Therefore, the discovery of non-invasive biomarkers in liquid biopsies, such as blood samples, in order to identify the presence of gastric preneoplasia and/or cancer lesions at asymptomatic stages is of paramount interest. This comprehensive review provides an overview of recently identified plasma/serum proteins and their diagnostic performance for the prediction of GPNLs and early cancer lesions. Autoantibodies appear to be promising biomarkers for AG, IM and early gastric cancer detection, along with inflammation and immunity-related proteins and antibodies against H. pylori virulence factors. There is a lack of specific protein biomarkers with which to detect DYS. Despite the need for further investigation and validation, some emerging candidates could pave the way for the development of reliable, non-invasive diagnostic tests for the detection and prevention of GC.
Collapse
Affiliation(s)
- Thomas Bazin
- Department of Gastroenterology and Nutritional Support, Center for Intestinal Failure, Reference Centre of Rare Disease MarDI, Assistance Publique-Hôpitaux de Paris (AP-HP) Beaujon Hospital, University Paris Cité, F-92110 Clichy, France
- Infection & Inflammation, Unité Mixte de Recherche (UMR) 1173, Inserm, Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ)/Université Paris Saclay, F-78180 Montigny-le-Bretonneux, France
| | - Karine Nozeret
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| | - Catherine Julié
- Department of Anatomical Pathology, Université Paris Saclay/Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Ambroise Paré, F-92100 Boulogne-Billancourt, France
| | - Dominique Lamarque
- Infection & Inflammation, Unité Mixte de Recherche (UMR) 1173, Inserm, Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ)/Université Paris Saclay, F-78180 Montigny-le-Bretonneux, France
- Department of Gastroenterology, Université Paris Saclay/Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Ambroise Paré, F-92100 Boulogne Billancourt, France
| | - Eliette Touati
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| |
Collapse
|
37
|
Wang YM, Sun JH, Sun RX, Liu XY, Li JF, Li RZ, Du YR, Zhou XZ. Treating chronic atrophic gastritis: identifying sub-population based on real-world TCM electronic medical records. Front Pharmacol 2024; 15:1444733. [PMID: 39170704 PMCID: PMC11335612 DOI: 10.3389/fphar.2024.1444733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Background and Objective Chronic atrophic gastritis (CAG) is a complex chronic disease caused by multiple factors that frequently occurs disease in the clinic. The worldwide prevalence of CAG is high. Interestingly, clinical CAG patients often present with a variety of symptom phenotypes, which makes it more difficult for clinicians to treat. Therefore, there is an urgent need to improve our understanding of the complexity of the clinical CAG population, obtain more accurate disease subtypes, and explore the relationship between clinical symptoms and medication. Therefore, based on the integrated platform of complex networks and clinical research, we classified the collected patients with CAG according to their different clinical characteristics and conducted correlation analysis on the classification results to identify more accurate disease subtypes to aid in personalized clinical treatment. Method Traditional Chinese medicine (TCM) offers an empirical understanding of the clinical subtypes of complicated disorders since TCM therapy is tailored to the patient's symptom profile. We gathered 6,253 TCM clinical electronic medical records (EMRs) from CAG patients and manually annotated, extracted, and preprocessed the data. A shared symptom-patient similarity network (PSN) was created. CAG patient subgroups were established, and their clinical features were determined through enrichment analysis employing community identification methods. Different clinical features of relevant subgroups were correlated based on effectiveness to identify symptom-botanical botanical drugs correspondence. Moreover, network pharmacology was employed to identify possible biological relationships between screened symptoms and medications and to identify various clinical and molecular aspects of the key subtypes using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Results 5,132 patients were included in the study: 2,699 males (52.60%) and 2,433 females (47.41%). The population was divided into 176 modules. We selected the first 3 modules (M29, M3, and M0) to illustrate the characteristic phenotypes and genotypes of CAG disease subtypes. The M29 subgroup was characterized by gastric fullness disease and internal syndrome of turbidity and poison. The M3 subgroup was characterized by epigastric pain and disharmony between the liver and stomach. The M0 subgroup was characterized by epigastric pain and dampness-heat syndrome. In symptom analysis, The top symptoms for symptom improvement in all three subgroups were stomach pain, bloating, insomnia, poor appetite, and heartburn. However, the three groups were different. The M29 subgroup was more likely to have stomach distention, anorexia, and palpitations. Citrus medica, Solanum nigrum, Jiangcan, Shan ci mushrooms, and Dillon were the most popular botanical drugs. The M3 subgroup has a higher incidence of yellow urine, a bitter tongue, and stomachaches. Smilax glabra, Cyperus rotundus, Angelica sinensis, Conioselinum anthriscoides, and Paeonia lactiflora were the botanical drugs used. Vomiting, nausea, stomach pain, and appetite loss are common in the M0 subgroup. The primary medications are Scutellaria baicalensis, Smilax glabra, Picrorhiza kurroa, Lilium lancifolium, and Artemisia scoparia. Through GO and KEGG pathway analysis, We found that in the M29 subgroup, Citrus medica, Solanum nigrum, Jiangcan, Shan ci mushrooms, and Dillon may exert their therapeutic effects on the symptoms of gastric distension, anorexia, and palpitations by modulating apoptosis and NF-κB signaling pathways. In the M3 subgroup, Smilax glabra, Cyperus rotundus, Angelica sinensis, Conioselinum anthriscoides, and Paeonia lactiflora may be treated by NF-κB and JAK-STAT signaling pathway for the treatment of stomach pain, bitter mouth, and yellow urine. In the M0 subgroup, Scutellaria baicalensis, Smilax glabra, Picrorhiza kurroa, Lilium lancifolium, and Artemisia scoparia may exert their therapeutic effects on poor appetite, stomach pain, vomiting, and nausea through the PI3K-Akt signaling pathway. Conclusion Based on PSN identification and community detection analysis, CAG population division can provide useful recommendations for clinical CAG treatment. This method is useful for CAG illness classification and genotyping investigations and can be used for other complicated chronic diseases.
Collapse
Affiliation(s)
- Yu-man Wang
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Jian-hui Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, China
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, China
| | - Run-xue Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, China
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, China
| | - Xiao-yu Liu
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Jing-fan Li
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Run-ze Li
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Yan-ru Du
- Hebei Hospital of Traditional Chinese Medicine, Hebei, China
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, China
- Hebei Provincial Key Laboratory of Integrated Traditional and Western Medicine Research on Gastroenterology, Hebei, China
| | - Xue-zhong Zhou
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
38
|
Massironi S, Gallo C, Lahner E, Sciola V, Cavalcoli F, Lenti MV, Zilli A, Dottori L, De Rossi G, Miceli E, Annibale B, Vecchi M, Cantù P, Di Sabatino A, Invernizzi P, Danese S. Occurrence and characteristics of endoscopic gastric polyps in patients with autoimmune gastritis (AGAPE study): A multicentric cross-sectional study. Dig Liver Dis 2024:S1590-8658(24)00887-9. [PMID: 39112216 DOI: 10.1016/j.dld.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/22/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Autoimmune gastritis (AIG) leads to increased gastrin (G) levels due to hypo-achlorhydria, providing proliferative stimuli on the gastric mucosa. AIMS To evaluate the incidence and characteristics of gastric polyps in AIG patients across six tertiary centers in Italy. METHODS A multicentric, cross-sectional study enrolled patients with AIG diagnosed from January 2000 to June 2023, who underwent at least one endoscopy. Data on demographics, clinical history, biochemical profiles, and endoscopic and histopathological findings were systematically collected. RESULTS Among 612 AIG patients followed for a median of 4 years, 222 (36.3 %) developed at least one gastric polyp. Of these, 214 were non-endocrine lesions detected in 162 patients, including 151 inflammatory (70.5 %), 29 adenomatous (13.6 %), 18 fundic gland polyps (8.4 %), 13 adenocarcinomas (6.1 %), and one MALT lymphoma. Additionally, 108 patients had gastric neuroendocrine neoplasms (gNENs), with 48 also having non-endocrine polyps. Older age and higher gastrin and chromogranin A levels were associated with polyp occurrence. No differences in OLGA/OLGIM stages or Helicobacter pylori status were noted among patients with and without lesions. CONCLUSION This large multicentric study underscores the substantial occurrence of gastric polyps in AIG patients, including notable rates of gNENs and adenocarcinomas, emphasizing the importance of proactive endoscopic surveillance and histopathological examination for effective management.
Collapse
Affiliation(s)
- Sara Massironi
- Division of Gastroenterology IRCCS San Gerardo dei Tintori Monza, MB, Italy and Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Camilla Gallo
- Division of Gastroenterology IRCCS San Gerardo dei Tintori Monza, MB, Italy and Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Edith Lahner
- Sapienza University of Rome, Dept Medical-surgical sciences and translational medicine, Rome, Italy
| | - Valentina Sciola
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Gastroenterology and Endoscopy Unit, Milan, Italy
| | - Federica Cavalcoli
- Gastroenterology and Digestive Endoscopy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessandra Zilli
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy, and Vita-Salute San Raffaele University, Milan, Italy
| | - Ludovica Dottori
- Sapienza University of Rome, Dept Medical-surgical sciences and translational medicine, Rome, Italy
| | - Gaia De Rossi
- Sapienza University of Rome, Dept Medical-surgical sciences and translational medicine, Rome, Italy
| | - Emanuela Miceli
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Bruno Annibale
- Sapienza University of Rome, Dept Medical-surgical sciences and translational medicine, Rome, Italy
| | - Maurizio Vecchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Gastroenterology and Endoscopy Unit, Milan, Italy
| | - Paolo Cantù
- Gastroenterology and Digestive Endoscopy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology IRCCS San Gerardo dei Tintori Monza, MB, Italy and Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy, and Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
39
|
Mülder DT, Hahn AI, Huang RJ, Zhou MJ, Blake B, Omofuma O, Murphy JD, Gutiérrez-Torres DS, Zauber AG, O'Mahony JF, Camargo MC, Ladabaum U, Yeh JM, Hur C, Lansdorp-Vogelaar I, Meester R, Laszkowska M. Prevalence of Gastric Precursor Lesions in Countries With Differential Gastric Cancer Burden: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol 2024; 22:1605-1617.e46. [PMID: 38438000 PMCID: PMC11272442 DOI: 10.1016/j.cgh.2024.02.023] [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: 02/09/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND & AIMS The prevalence of precursor lesions for gastric cancer (GC) and the differential burden between countries of varying GC risk is not well-understood. We conducted a systematic review and meta-analysis to estimate the global prevalence of precursor lesions. METHODS We estimated the prevalence of atrophic gastritis (AG), gastric intestinal metaplasia (IM), and dysplasia in regions with low, medium, and high GC incidence. Because IM is an advanced manifestation of AG, we assessed the prevalence of less advanced precursors, regardless of the presence of more advanced lesions. Prevalence was sub-stratified by Helicobacter pylori infection, symptomatology, and period (<2000, 2000-2010, and >2010). RESULTS Among the 582 articles that underwent full-text review, 166 studies met inclusion criteria. The global prevalence estimates of AG, IM, and dysplasia were 25.4%, 16.2%, and 2.0%, respectively, on the basis of 126 studies that reported the prevalence of less advanced precursors, regardless of the presence of more advanced lesions. The prevalence of all precursor lesions was higher in high and medium compared with low GC incidence countries (P < .01). Prevalence of AG and IM was significantly higher among H pylori-infected individuals (P < .01) but not statistically different between symptomatic and asymptomatic individuals (P > .17). All precursors demonstrated a secular decrease in prevalence over time. CONCLUSIONS Gastric precursor lesions have differences in prevalence in regions with differential GC incidence and are associated with H pylori infection. Because of the substantial prevalence of precursor lesions in both symptomatic and asymptomatic individuals, symptomatic evaluation may not be sufficient to identify individuals at risk. These estimates provide important insights for tailoring GC prevention strategies.
Collapse
Affiliation(s)
- Duco T Mülder
- Department of Public Health, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne I Hahn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Margaret J Zhou
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Benjamin Blake
- Weill Cornell Medical College of Cornell University, New York, New York
| | - Omonefe Omofuma
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - John D Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | | | - Ann G Zauber
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James F O'Mahony
- Department of Public Health, Erasmus Medical Center, Rotterdam, Netherlands; School of Economics, University College Dublin, Dublin, Ireland
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Uri Ladabaum
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Jennifer M Yeh
- Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts
| | - Chin Hur
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | | | - Reinier Meester
- Department of Public Health, Erasmus Medical Center, Rotterdam, Netherlands; Health Economics & Outcomes Research, Freenome Holdings Inc, San Francisco, California
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Department of Subspecialty Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
40
|
Tao X, Zhu Y, Dong Z, Huang L, Shang R, Du H, Wang J, Zeng X, Wang W, Wang J, Li Y, Deng Y, Wu L, Yu H. An artificial intelligence system for chronic atrophic gastritis diagnosis and risk stratification under white light endoscopy. Dig Liver Dis 2024; 56:1319-1326. [PMID: 38246825 DOI: 10.1016/j.dld.2024.01.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/06/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND AND AIMS The diagnosis and stratification of gastric atrophy (GA) predict patients' gastric cancer progression risk and determine endoscopy surveillance interval. We aimed to construct an artificial intelligence (AI) system for GA endoscopic identification and risk stratification based on the Kimura-Takemoto classification. METHODS We constructed the system using two trained models and verified its performance. First, we retrospectively collected 869 images and 119 videos to compare its performance with that of endoscopists in identifying GA. Then, we included original image cases of 102 patients to validate the system for stratifying GA and comparing it with endoscopists with different experiences. RESULTS The sensitivity of model 1 was higher than that of endoscopists (92.72% vs. 76.85 %) at image level and also higher than that of experts (94.87% vs. 85.90 %) at video level. The system outperformed experts in stratifying GA (overall accuracy: 81.37 %, 73.04 %, p = 0.045). The accuracy of this system in classifying non-GA, mild GA, moderate GA, and severe GA was 80.00 %, 77.42 %, 83.33 %, and 85.71 %, comparable to that of experts and better than that of seniors and novices. CONCLUSIONS We established an expert-level system for GA endoscopic identification and risk stratification. It has great potential for endoscopic assessment and surveillance determinations.
Collapse
Affiliation(s)
- Xiao Tao
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yijie Zhu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China; Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, PR China
| | - Zehua Dong
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Li Huang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Renduo Shang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Hongliu Du
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Junxiao Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xiaoquan Zeng
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Wen Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jiamin Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yanxia Li
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yunchao Deng
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Lianlian Wu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China.
| | - Honggang Yu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China.
| |
Collapse
|
41
|
Li Y, Hahn AI, Laszkowska M, Jiang F, Zauber AG, Leung WK. Global burden of young-onset gastric cancer: a systematic trend analysis of the global burden of disease study 2019. Gastric Cancer 2024; 27:684-700. [PMID: 38570392 PMCID: PMC11193827 DOI: 10.1007/s10120-024-01494-6] [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: 10/03/2023] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND While gastric cancer is generally declining globally, the temporal trend of young-onset (< 40 years) gastric cancer remains uncertain. We performed this analysis to determine the temporal trends of young-onset gastric cancer compared to late-onset cancer (≥ 40 years). METHODS We extracted cross-sectional data from the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2019. The burden of gastric cancer from 1990 to 2019 was assessed through indicators including incidence and mortality rates, which were classified at global, national, and regional levels, and according to socio-demographic indexes (SDI) and age or sex groups. Joinpoint regression analysis was used to identify specific years with significant changes. The correlation between AAPC with countries' average SDI was tested by Pearson's Test. RESULTS The global incidence rate of young-onset gastric cancer decreased from 2.20 (per 100,000) in 1990 to 1.65 in 2019 (AAPC: - 0.95; 95% confidence interval [CI] - 1.25 to - 0.65; P < 0.001). Late-onset cancer incidence also decreased from 59.53 (per 100,000) in 1990 to 41.26 in 2019 (AAPC: - 1.23; 95% CI - 1.39 to - 1.06, P < 0.001). Despite an overall decreasing trend, the incidence rate of young-onset cancer demonstrated a significant increase from 2015 to 2019 (annual percentage change [APC]: 1.39; 95% CI 0.06 to 2.74; P = 0.041), whereas no upward trend was observed in late-onset cancer. Mortality rates of young- and late-onset cancer both exhibited a significant decline during this period (AAPC: - 1.82; 95% CI - 2.15 to - 1.56; P < 0.001 and AAPC: - 1.69, 95% CI - 1.79 to - 1.59; P < 0.001). The male-to-female rate ratio for incidence and mortality in both age groups have been increasing since 1990. While countries with high SDI have had a greater decline in the incidence of late-onset gastric cancer (slope of AAPC change: - 0.20, P = 0.004), it was not observed in young-onset cancer (slope of AAPC change: - 0.11, P = 0.13). CONCLUSIONS The global incidence and mortality rates of both young- and late-onset gastric cancer have decreased since 1990. However, the incidence rate of young-onset cancer has demonstrated a small but significant upward trend since 2015. There was disparity in the decline in young-onset gastric cancer among male and high SDI countries. These findings could help to inform future strategies in preventing gastric cancer in younger individuals.
Collapse
Affiliation(s)
- Yunhao Li
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, 102 Pokfulam Road, Hong Kong, China
| | - Anne I Hahn
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Department of Subspecialty Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Fang Jiang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, 102 Pokfulam Road, Hong Kong, China
| | - Ann G Zauber
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Wai K Leung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, 102 Pokfulam Road, Hong Kong, China.
| |
Collapse
|
42
|
Lee S, Yun JM, Park JH, Kwon H. Association between Chronic Atrophic Gastritis and Bone Mineral Density among Women Older than 40 Years of Age in Korea. Korean J Fam Med 2024; 45:199-206. [PMID: 38356173 PMCID: PMC11273168 DOI: 10.4082/kjfm.22.0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis causes hypochlorhydria, hypergastrinemia, and malabsorption of nutrients, leading to lower bone mineral density. The few studies that investigated the association between chronic atrophic gastritis and bone mineral density have reported inconsistent findings. As such, the present study assessed the association between chronic atrophic gastritis and bone mineral density among a large sample of women >40 years of age in Korea. METHODS Data from 8,748 women >40 years of age who underwent esophagogastroduodenoscopy and bone densitometry were analyzed. Chronic atrophic gastritis was diagnosed using esophagogastroduodenoscopy. Bone mineral density of the lumbar vertebrae (L), femur neck, and femur total, measured using dual-energy X-ray absorptiometry, were the primary outcome variables. Low bone mineral density, which could be diagnosed as osteoporosis or osteopenia, was defined and analyzed as a secondary outcome. Linear regression was used to calculate adjusted mean values of bone mineral density. The association between low bone mineral density and chronic atrophic gastritis was analyzed using multiple logistic regression. RESULTS The adjusted mean bone mineral density for L1-L4 was 1.063±0.003, femur neck (0.826±0.002), and femur total (0.890±0.002) were significantly lower in patients with chronic atrophic gastritis than others (1.073±0.002, 0.836±0.001, 0.898±0.002, respectively; all P<0.01). Women with chronic atrophic gastritis exhibited an increased likelihood for osteopenia or osteoporosis, even after adjusting for age and other confounding factors (odds ratio, 1.25; 95% confidence interval, 1.13-1.40; P<0.01). However, subgroup analysis revealed statistical significance only in postmenopausal women (odds ratio, 1.27; P<0.001). CONCLUSION Chronic atrophic gastritis was associated with lower bone mineral density and a higher risk for osteopenia or osteoporosis among postmenopausal women.
Collapse
Affiliation(s)
- Seulki Lee
- Department of Family Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jae Moon Yun
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jin-Ho Park
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuktae Kwon
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Matsubayashi CO, Cheng S, Hulchafo I, Zhang Y, Tada T, Buxbaum JL, Ochiai K. Artificial intelligence for gastric cancer in endoscopy: From diagnostic reasoning to market. Dig Liver Dis 2024; 56:1156-1163. [PMID: 38763796 DOI: 10.1016/j.dld.2024.04.019] [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: 10/19/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Recognition of gastric conditions during endoscopy exams, including gastric cancer, usually requires specialized training and a long learning curve. Besides that, the interobserver variability is frequently high due to the different morphological characteristics of the lesions and grades of mucosal inflammation. In this sense, artificial intelligence tools based on deep learning models have been developed to support physicians to detect, classify, and predict gastric lesions more efficiently. Even though a growing number of studies exists in the literature, there are multiple challenges to bring a model to practice in this field, such as the need for more robust validation studies and regulatory hurdles. Therefore, the aim of this review is to provide a comprehensive assessment of the current use of artificial intelligence applied to endoscopic imaging to evaluate gastric precancerous and cancerous lesions and the barriers to widespread implementation of this technology in clinical routine.
Collapse
Affiliation(s)
- Carolina Ogawa Matsubayashi
- Endoscopy Unit, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, São Paulo, Brasil; AI Medical Service Inc., Tokyo, Japan.
| | - Shuyan Cheng
- Department of Population Health Science, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ismael Hulchafo
- Columbia University School of Nursing, New York, NY 10032, USA
| | - Yifan Zhang
- Department of Population Health Science, Weill Cornell Medical College, New York, NY 10065, USA
| | - Tomohiro Tada
- AI Medical Service Inc., Tokyo, Japan; Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - James L Buxbaum
- Division of Gastrointestinal and Liver Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Kentaro Ochiai
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
44
|
Jia J, Zhao H, Li F, Zheng Q, Wang G, Li D, Liu Y. Research on drug treatment and the novel signaling pathway of chronic atrophic gastritis. Biomed Pharmacother 2024; 176:116912. [PMID: 38850667 DOI: 10.1016/j.biopha.2024.116912] [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: 04/16/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a global digestive system disease and one of the important causes of gastric cancer. The incidence of CAG has been increasing yearly worldwide. PURPOSE This article reviews the latest research on the common causes and future therapeutic targets of CAG as well as the pharmacological effects of corresponding clinical drugs. We provide a detailed theoretical basis for further research on possible methods for the treatment of CAG and reversal of the CAG process. RESULTS CAG often develops from chronic gastritis, and its main pathological manifestation is atrophy of the gastric mucosa, which can develop into gastric cancer. The drug treatment of CAG can be divided into agents that regulate gastric acid secretion, eradicate Helicobacter. pylori (H. pylori), protect gastric mucous membrane, or inhibit inflammatory factors according to their mechanism of action. Although there are limited specific drugs for the treatment of CAG, progress is being made in defining the pathogenesis and therapeutic targets of the disease. Growing evidence shows that NF-κB, PI3K/AKT, Wnt/ β-catenin, MAPK, Toll-like receptors (TLRs), Hedgehog, and VEGF signaling pathways play an important role in the development of CAG.
Collapse
Affiliation(s)
- Jinhao Jia
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Huijie Zhao
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Special Administrative Region of China
| | - Qiusheng Zheng
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832003, PR China
| | - Guoli Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832003, PR China.
| | - Ying Liu
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China.
| |
Collapse
|
45
|
Cicek B, Oztas E. What Are Clinically Significant Endoscopic Findings? Clin Gastroenterol Hepatol 2024; 22:1550. [PMID: 37714397 DOI: 10.1016/j.cgh.2023.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/17/2023]
Affiliation(s)
- Bahattin Cicek
- Department of Gastroenterology, Acibadem Maslak Hospital, Istanbul, Turkey
| | - Erkin Oztas
- Department of Gastroenterology, Acibadem Maslak Hospital, Istanbul, Turkey
| |
Collapse
|
46
|
Choi E, Galicia Garcia G, Kishore Anna K, Albuja Altamirano MF, Yip L, Oh J, Lee JH. Vitamin B12 Deficiency Manifesting As Pancytopenia, Lymphadenopathy, and Fever: A Clinical Mimic of Hematologic Malignancies. Cureus 2024; 16:e64676. [PMID: 39149666 PMCID: PMC11326803 DOI: 10.7759/cureus.64676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
Pancytopenia is a complex medical condition characterized by decreased levels of red blood cells (RBCs), white blood cells (WBCs), and platelets (PLTs). It can arise from impaired production, peripheral destruction, or a combination of both. The causes of pancytopenia range from reversible factors like infections and medication reactions to irreversible conditions. Vitamin B12 deficiency is a notable reversible cause that can take years to manifest in adults due to stored reserves. However, deficiencies caused by impaired absorption, especially due to the lack of intrinsic factors (IFs), can lead to rapid deterioration within two to five years. A healthy 39-year-old male with an athletic lifestyle presented with symptoms such as dizziness, nausea, vomiting, palpitations, and fainting over a few days. These symptoms were preceded by weeks of persistent body aches, headaches, weakness, daily fevers, chills, and night sweats. Vital signs were stable. The physical examination revealed conjunctival pallor and lymphadenopathy in the submandibular and superficial cervical regions. Initial blood tests showed normocytic anemia (Hgb 4.9, MCV 80), leukopenia (2.99), thrombocytopenia (142), and elevated liver enzymes (AST 199, ALT 96, and total bilirubin of 2.04). The peripheral smear showed tear-drop cells and hypochromic cells. The initial impression was hematologic malignancy, including but not limited to leukemia, lymphoma, or myelofibrosis given clinical findings such as B-symptoms like night sweats, neck lymphadenopathy, and subjective daily fever, along with pancytopenia. The patient received a bolus of normal saline and a transfusion of two units of packed RBCs. CT scans of the chest, abdomen, and pelvis showed no adenopathy or splenomegaly. Although initial clinical assessment pointed toward a potential hematologic malignancy, comprehensive testing, including SPEP, reticulocyte count/fraction, serum folate, and serum vitamin B12, revealed only severe vitamin B12 deficiency, with a level of less than 150, with the presence of IF antibodies. Treatment involved intensive in-patient vitamin B12 injections followed by a detailed outpatient regimen. The patient completed a daily dose of vitamin B12 injections for seven consecutive days, followed by weekly injections for the next four weeks. Subsequent laboratory results demonstrated an increase in WBC count to 8.39, Hgb level to 13.2, and PLT count of 249, indicating a continued positive response to the vitamin B12 replacement therapy. In summary, pancytopenia poses a diagnostic challenge that demands careful evaluation of patient data and comprehensive testing. Vitamin B12 deficiency, which encompasses pernicious anemia (PA), is among the reversible factors to consider. This aspect holds significance before opting for more invasive measures like a bone marrow biopsy. Nutritional deficiencies need to be considered first as differentials in pancytopenia, even in the absence of typical signs of vitamin B12 deficiency (like macrocytosis and hypersegmented neutrophils) and in the presence of compelling clinical indications pointing to a hematologic malignancy.
Collapse
Affiliation(s)
- Eunhee Choi
- Internal Medicine, NYC Health and Hospital/Lincoln, New York, USA
| | | | | | | | - Laverne Yip
- Internal Medicine, NYC Health and Hospital/Lincoln, New York, USA
| | - Jaha Oh
- Nephrology, New York Presbyterian/Weill Cornell Medical Center, New York, USA
| | - Jung-Hyun Lee
- Neurology, Maimonides Medical Center, New York, USA
- Neurology, State University of New York Downstate Health Sciences University, New York, USA
- Neurology, Kings County Hospital Center, New York, USA
| |
Collapse
|
47
|
Zhang N, Chen P, Liang X, Sun J, Liu Q, Guan S, Wang Q. Luteolin targets the AGE-RAGE signaling to mitigate inflammation and ferroptosis in chronic atrophic gastritis. Aging (Albany NY) 2024; 16:10918-10930. [PMID: 38917486 PMCID: PMC11272119 DOI: 10.18632/aging.205969] [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: 01/23/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
Chronic atrophic gastritis (CAG) is a chronic inflammatory disease and precancerous lesion in stomach cancer. Abnormal activation cellular ferroptosis further damages gastric tissue, which is susceptible to inflammation. Luteolin has powerful anti-inflammatory and regulatory potential for cellular ferroptosis. We aimed to clarify the involvement of luteolin in inflammation and ferroptosis during CAG. Luteolin targets were searched to identify intersecting genes in the chronic atrophic gastritis disease database. The AGE-RAGE pathway is a potential target of luteolin for the treatment of chronic atrophic gastritis and a binding site between luteolin and RAGE was predicted through a computer simulation of molecular docking. We established a CAG rat model using N-methyl-N-nitro-N-nitroguanidine. The therapeutic effect of luteolin on CAG was detected using western blotting, qPCR, hematoxylin and eosin staining, lipid oxidation (MDA), and Fe2+ assays. Luteolin inhibited the AGE-RAGE signaling pathway and reduced the inflammatory response in gastric tissues. Additionally, luteolin downregulated the concentration of (MDA) and Fe2+, and CAG downregulated the expression levels of ACSL4 and NOX1 and upregulated the expression levels of FIH1 and GPX4 ferroptosis-related proteins, thus inhibiting the ferroptosis of gastric tissue cells, which had a therapeutic effect on CAG.
Collapse
Affiliation(s)
- Nailin Zhang
- Clinical Research Base Office, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
- Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
| | - Pingping Chen
- Department of Pharmacology, Hebei University of Chinese Medicine, Hebei, China
| | - Xiaoyan Liang
- Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Sun
- Hebei Key Laboratory of Turbidity Toxin Syndrome, Hebei University of Chinese Medicine, Hebei, China
| | - Qiquan Liu
- Department of Spleen and Stomach Diseases, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
| | - Shengjiang Guan
- Pharmaceutical Department, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
| | - Qiao Wang
- Pharmacological Analysis Teaching and Research Department, Hebei Medical University, Hebei, China
| |
Collapse
|
48
|
Yu Y, Shangguan X, Yu R, Wu Y, Xu E, Tan C. Unveiling early stage autoimmune gastritis: novel endoscopic insights from two case reports. Front Immunol 2024; 15:1416292. [PMID: 38953024 PMCID: PMC11215040 DOI: 10.3389/fimmu.2024.1416292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024] Open
Abstract
The predominant characteristic of autoimmune gastritis (AIG) is corpus-dominant advanced atrophy, which is mostly observed in the middle to late stages. More reports are needed on the endoscopic features of the early stage. In this report, we present two cases of early-stage AIG in which endoscopic examinations showed no atrophy of the gastric mucosa but displayed a transition of collecting venules from a regular to an irregular arrangement. In addition, yellowish-white cobblestone-like elevations were observed in the fundic gland region. Histologically, the observed manifestations included pseudohypertrophy and protrusion of parietal cells into the lumen, possibly along with hyperplasia of G cells, lymphocytic infiltration and potentially pseudopyloric gland metaplasia. Serologically, the anti-parietal cell antibody returned positive results, whereas the anti-intrinsic factor antibody yielded negative results. In this study, we summarized some endoscopic features of two patients, aiming to provide clues for endoscopists to detect early-stage AIG.
Collapse
Affiliation(s)
- Yunfeng Yu
- Digestive Endoscopy Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Huanan, China
| | - Xueli Shangguan
- Digestive Endoscopy Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Huanan, China
| | - Rong Yu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Huanan, China
| | - Yangpeng Wu
- Digestive Endoscopy Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - En Xu
- Digestive Endoscopy Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chuanchuan Tan
- Digestive Endoscopy Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
49
|
do Carmo GC, Cavalcante RM, de Aquino TMF. Gastric cancer: an overview. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e2024S116. [PMID: 38865536 PMCID: PMC11164282 DOI: 10.1590/1806-9282.2024s116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 06/14/2024]
|
50
|
Yoo JW, Laszkowska M, Mendelsohn RB. The Role of Screening and Early Detection in Upper Gastrointestinal Cancers. Hematol Oncol Clin North Am 2024; 38:693-710. [PMID: 38431494 DOI: 10.1016/j.hoc.2024.01.007] [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: 03/05/2024]
Abstract
Upper gastrointestinal cancers are among the leading causes of cancer deaths worldwide with exceptionally poor prognosis, which is largely attributable to frequently delayed diagnosis. Although effective screening is critical for early detection, the highly variable incidence of upper gastrointestinal cancers presents challenges, rendering universal screening programs suboptimal in most populations globally. Optimal strategies in regions of modest incidence, such as the United States, require a targeted approach, focused on high-risk individuals based on demographic, familial, and clinicopathologic risk factors. Assessment of underlying precancerous lesions has key implications for risk stratification and informing clinical decisions to improve patient outcomes.
Collapse
Affiliation(s)
- Jin Woo Yoo
- Gastroenterology, Hepatology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Monika Laszkowska
- Gastroenterology, Hepatology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Robin B Mendelsohn
- Gastroenterology, Hepatology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|