1
|
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
|
2
|
Vavallo M, Cingolani S, Cozza G, Schiavone FP, Dottori L, Palumbo C, Lahner E. Autoimmune Gastritis and Hypochlorhydria: Known Concepts from a New Perspective. Int J Mol Sci 2024; 25:6818. [PMID: 38999928 PMCID: PMC11241626 DOI: 10.3390/ijms25136818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Autoimmune atrophic gastritis is an immune-mediated disease resulting in autoimmune destruction of the specialized acid-producing gastric parietal cells. As a consequence, in autoimmune atrophic gastritis, gastric acid secretion is irreversibly impaired, and the resulting hypochlorhydria leads to the main clinical manifestations and is linked, directly or indirectly, to the long-term neoplastic complications of this disease. In the last few years, autoimmune atrophic gastritis has gained growing interest leading to the acquisition of new knowledge on different aspects of this disorder. Although reliable serological biomarkers are available and gastrointestinal endoscopy techniques have substantially evolved, the diagnosis of autoimmune atrophic gastritis is still affected by a considerable delay and relies on histopathological assessment of gastric biopsies. One of the reasons for the diagnostic delay is that the clinical presentations of autoimmune atrophic gastritis giving rise to clinical suspicion are very different, ranging from hematological to neurological-psychiatric up to gastrointestinal and less commonly to gynecological-obstetric symptoms or signs. Therefore, patients with autoimmune atrophic gastritis often seek advice from physicians of other medical specialties than gastroenterologists, thus underlining the need for increased awareness of this disease in a broad medical and scientific community.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Edith Lahner
- Gastroenterology Unit, Sant’Andrea University Hospital, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy (G.C.); (F.P.S.)
| |
Collapse
|
3
|
Liao X, Lin R, Zhang Z, Tian D, Liu Z, Chen S, Xu G, Su M. Genome-wide DNA methylation and transcriptomic patterns of precancerous gastric cardia lesions. J Natl Cancer Inst 2024; 116:681-693. [PMID: 38258659 DOI: 10.1093/jnci/djad244] [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: 06/27/2023] [Revised: 10/25/2023] [Accepted: 11/15/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Intestinal metaplasia (IM) and intraepithelial neoplasia (IEN) are considered precursors of gastric cardia cancer (GCC). Here, we investigated the histopathologic and molecular profiles of precancerous gastric cardia lesions (PGCLs) and biomarkers for risk stratification of gastric cardia IM. METHODS We conducted a hospital-based evaluation (n = 4578) for PGCL profiles in high-incidence and non-high-incidence regions for GCC in China. We next performed 850K methylation arrays (n = 42) and RNA-seq (n = 44) in tissues with PGCLs. We then examined the protein expression of candidate biomarker using immunohistochemistry. RESULTS Of the 4578 participants, 791 were diagnosed with PGCLs (600 IM, 62 IM with IEN, and 129 IEN). We found that individuals from high-incidence regions (26.7%) were more likely to develop PGCLs than those from non-high-incidence areas (13.5%). DNA methylation and gene expression alterations, indicated by differentially methylated probes (DMPs) and differentially expressed genes (DEGs), exhibited a progressive increase from type I IM (DMP = 210, DEG = 24), type II IM (DMP = 3402, DEG = 129), to type III IM (DMP = 3735, DEG = 328), peaking in IEN (DMP = 47 373, DEG = 2278). Three DEGs with aberrant promoter methylation were identified, shared exclusively by type III IM and IEN. Of these DEGs, we found that OLFM4 expression appears in IMs and increases remarkably in IENs (P < .001). CONCLUSIONS We highlight that type III IM and IEN share similar epigenetic and transcriptional features in gastric cardia and propose biomarkers with potential utility in risk prediction.
Collapse
Affiliation(s)
- Xiaoqi Liao
- Department of Pathology, Shantou University Medical College, Shantou, People's Republic of China
| | - Runhua Lin
- Department of Pathology, Shantou University Medical College, Shantou, People's Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, People's Republic of China
| | - Zhihua Zhang
- Department of Pathology, Shantou University Medical College, Shantou, People's Republic of China
| | - Dongping Tian
- Department of Pathology, Shantou University Medical College, Shantou, People's Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, People's Republic of China
| | - Zhaohui Liu
- Department of Gastroenterology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Songqin Chen
- Department of Pathology, Jieyang People's Hospital, Jieyang, People's Republic of China
| | - Guohua Xu
- Department of Gastroenterology, Huiyang Sanhe Hospital, Huizhou, People's Republic of China
| | - Min Su
- Department of Pathology, Shantou University Medical College, Shantou, People's Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, People's Republic of China
| |
Collapse
|
4
|
Ushiku T, Lauwers GY. Pathology and Clinical Relevance of Gastric Epithelial Dysplasia. Gastroenterol Clin North Am 2024; 53:39-55. [PMID: 38280750 DOI: 10.1016/j.gtc.2023.11.003] [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: 01/29/2024]
Abstract
Gastric dysplasia is defined as an unequivocally neoplastic epithelium. Dysplastic lesions are characterized by cellular atypia reflective of abnormal differentiation and disorganized glandular architecture. The last few years have been marked by a refinement of the prognosis and risk of progression of gastric dysplasia and the recognition of novel morphologic patterns of dysplasia. Determination of the correct diagnosis and grade of dysplasia are critical steps since it will be predicting the risk of malignant transformation and help tailor appropriate surveillance strategy. This review describes the morphologic characteristics of conventional dysplasia and nonconventional gastric dysplasia that have been more recently characterized.
Collapse
Affiliation(s)
- Tetsuo Ushiku
- Department of Pathology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Gregory Y Lauwers
- Department of Pathology, Gastrointestinal Pathology Section, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; Departments of Pathology and Oncologic Sciences, Tampa, FL, USA.
| |
Collapse
|
5
|
Rugge M, Genta RM, Malfertheiner P, Dinis-Ribeiro M, El-Serag H, Graham DY, Kuipers EJ, Leung WK, Park JY, Rokkas T, Schulz C, El-Omar EM. RE.GA.IN.: the Real-world Gastritis Initiative-updating the updates. Gut 2024; 73:407-441. [PMID: 38383142 DOI: 10.1136/gutjnl-2023-331164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 02/23/2024]
Abstract
At the end of the last century, a far-sighted 'working party' held in Sydney, Australia addressed the clinicopathological issues related to gastric inflammatory diseases. A few years later, an international conference held in Houston, Texas, USA critically updated the seminal Sydney classification. In line with these initiatives, Kyoto Global Consensus Report, flanked by the Maastricht-Florence conferences, added new clinical evidence to the gastritis clinicopathological puzzle.The most relevant topics related to the gastric inflammatory diseases have been addressed by the Real-world Gastritis Initiative (RE.GA.IN.), from disease definitions to the clinical diagnosis and prognosis. This paper reports the conclusions of the RE.GA.IN. consensus process, which culminated in Venice in November 2022 after more than 8 months of intense global scientific deliberations. A forum of gastritis scholars from five continents participated in the multidisciplinary RE.GA.IN. consensus. After lively debates on the most controversial aspects of the gastritis spectrum, the RE.GA.IN. Faculty amalgamated complementary knowledge to distil patient-centred, evidence-based statements to assist health professionals in their real-world clinical practice. The sections of this report focus on: the epidemiology of gastritis; Helicobacter pylori as dominant aetiology of environmental gastritis and as the most important determinant of the gastric oncogenetic field; the evolving knowledge on gastric autoimmunity; the clinicopathological relevance of gastric microbiota; the new diagnostic horizons of endoscopy; and the clinical priority of histologically reporting gastritis in terms of staging. The ultimate goal of RE.GA.IN. was and remains the promotion of further improvement in the clinical management of patients with gastritis.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Azienda Zero, Veneto Tumour Registry, Padua, Italy
| | - Robert M Genta
- Gastrointestinal Pathology, Inform Diagnostics Research Institute, Dallas, Texas, USA
- Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Malfertheiner
- Medizinische Klinik und Poliklinik II, Ludwig Maximilian Universität Klinikum München, Munich, Germany
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | - Mario Dinis-Ribeiro
- Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Hashem El-Serag
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Houston VA Health Services Research & Development Center of Excellence, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - David Y Graham
- Department of Medicine, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Ernst J Kuipers
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Jin Young Park
- International Agency for Research on Cancer, Lyon, France
| | - Theodore Rokkas
- Gastroenterology, Henry Dunant Hospital Center, Athens, Greece
| | | | - Emad M El-Omar
- Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Dilaghi E, Dottori L, Pivetta G, Dalla Bella M, Esposito G, Ligato I, Pilozzi E, Annibale B, Lahner E. Incidence and Predictors of Gastric Neoplastic Lesions in Corpus-Restricted Atrophic Gastritis: A Single-Center Cohort Study. Am J Gastroenterol 2023; 118:2157-2165. [PMID: 37207305 DOI: 10.14309/ajg.0000000000002327] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/09/2023] [Indexed: 05/21/2023]
Abstract
INTRODUCTION Corpus-restricted atrophic gastritis is a chronic inflammatory disorder leading to possible development of type 1 neuroendocrine tumors (T1gNET), intraepithelial neoplasia (IEN), and gastric cancer (GC). We aimed to assess occurrence and predictors of gastric neoplastic lesions in patients with corpus-restricted atrophic gastritis at long-term follow-up. METHODS A prospective single-center cohort of patients with corpus-restricted atrophic gastritis adhering to endoscopic-histological surveillance was considered. Follow-up gastroscopies were scheduled according to the management of epithelial precancerous conditions and lesions of the stomach guidelines. In case of new/worsening of known symptoms, gastroscopy was anticipated. Cox regression analyses and Kaplan-Meier survival curves were obtained. RESULTS Two hundred seventy-five patients with corpus-restricted atrophic gastritis (72.0% female, median age 61 [23-84] years) were included. At a median follow-up of 5 (1-17) years, the annual incidence rate person-year was 0.5%, 0.6%, 2.8%, and 3.9% for GC/high-grade IEN, low-grade IEN, T1gNET, and all gastric neoplastic lesions, respectively. All patients showed at baseline operative link for gastritis assessment (OLGA)-2, except 2 low-grade (LG) IEN patients and 1 T1gNET patient with OLGA-1. Age older than 60 years (hazard ratio [HR] 4.7), intestinal metaplasia without pseudopyloric metaplasia (HR 4.3), and pernicious anemia (HR 4.3) were associated with higher risk for GC/HG-IEN or LG-IEN development and shorter mean survival time for progression (13.4, 13.2, and 11.1, respectively, vs 14.7 years, P = 0.01). Pernicious anemia was an independent risk factor for T1gNET (HR 2.2) and associated with a shorter mean survival time for progression (11.7 vs 13.6 years, P = 0.04) as well as severe corpus atrophy (12.8 vs 13.6 years, P = 0.03). DISCUSSION Patients with corpus-restricted atrophic gastritis are at increased risk for GC and T1gNET despite low-risk OLGA scores, and those aged older than 60 years with corpus intestinal metaplasia or pernicious anemia seem to display a high-risk scenario.
Collapse
Affiliation(s)
- Emanuele Dilaghi
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Ludovica Dottori
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Giulia Pivetta
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Martina Dalla Bella
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Irene Ligato
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Italy
| |
Collapse
|
7
|
Guo GH, Zhang JJ, Ye SY, Ying XQ. Clinical value of serum Mg7-Ag detection combined with magnifying endoscopy with narrow-band imaging in diagnosis of high-risk gastric low-grade intraepithelial neoplasia. Shijie Huaren Xiaohua Zazhi 2023; 31:438-445. [DOI: 10.11569/wcjd.v31.i11.438] [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] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Human gastric cancer associated antigen (Mg7-Ag) can be highly expressed in gastric cancer cell lines, and is closely related to gastric cancer progression. Magnifying endoscopy with narrow-band imaging (ME-NBI) has high sensitivity and specificity in the diagnosis of early gastric cancer, especially for differentiated gastric cancer. This study assessed the clinical diagnostic value of serum Mg7-Ag detection combined with ME-NBI for high-risk low grade intraepithelial neoplasia (LGIN).
AIM To evaluate the clinical value of serum Mg7-Ag detection combined with ME-NBI in judging whether there is pathological upgrading of gastric LGIN after endoscopic dissection (ESD).
METHODS Patients who were diagnosed with LGIN at our hospital from January 2019 to December 2022 and underwent diagnostic ESD within 1 mo were selected and divided into a high-risk group (pathologically upgraded), a low-risk group (pathologically not upgraded), and an inflammation group (pathologically downgraded) according to the differences in pathological results after ESD and preoperative endoscopic biopsy. Serum Mg7-Ag levels were detected in all cases within 1 week before ESD, and gastroscopic ME-NBI screening was performed. The levels of serum Mg7-Ag, the number of Mg7-Ag (+) and Mg7-Ag (-) patients, and the number of ME-NBI (+) and ME-NBI (-) patients in the three groups were recorded. The sensitivity, specificity, accuracy, and positive likelihood ratio of serum Mg7-Ag and ME-NBI, alone and in combination, for predicting pathological upgrading after ESD of LGIN lesions were calculated and analyzed.
RESULTS A total of 125 patients with gastric LGIN who underwent diagnostic ESD were included in this study, including 37 in the high-risk group, 76 in the low-risk group, and 12 in the inflammation group. Serum Mg7-Ag levels were significantly different among the three groups (P < 0.05), and Mg7-Ag levels in the high-risk group were significantly higher than those in the low-risk group and the inflammation group (P < 0.05). The positive rate of ME-NBI in the inflammation group was significantly lower than those in the other two groups (P < 0.05), and the rate in the high-risk group was higher than that in the low-risk group (P < 0.05). There was no significant difference in the sensitivity, specificity, accuracy, and positive likelihood ratio of Mg7-Ag and ME-NB alone in predicting the pathological upgrading of LGIN (P > 0.05). The sensitivity of combined Mg7-Ag detection and ME-NBI in predicting high-risk LGIN was slightly lower than that of either of them alone; the specificity was 90.5%, the accuracy was 84.0%, and the positive likelihood ratio was 2.417, all of which were significantly higher than those of single tests (P < 0.05). When Mg7-Ag + ME-NBI results were negative, the accuracy and positive likelihood for cases in the inflammation group were significantly higher than those of single tests (P < 0.05).
CONCLUSION Combined serum Mg7-Ag detection and ME-NBI screening has important clinical value in judging whether gastric LGIN lesions will undergo pathological upgrading.
Collapse
|
8
|
Li C, Liu Z, Xu G, Wu S, Peng Y, Wu R, Zhao S, Liao X, Lin R. Aberrant DNA methylation and expression of EYA4 in gastric cardia intestinal metaplasia. Saudi J Gastroenterol 2022; 28:456-465. [PMID: 36453428 PMCID: PMC9843510 DOI: 10.4103/sjg.sjg_228_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Intestinal metaplasia (IM) of the gastric cardia is an important premalignant lesion. However, there is limited information concerning its epidemiological and molecular features. Herein, we aimed to provide an overview of the epidemiological data for gastric cardiac IM and evaluate the role of EYA transcriptional coactivator and phosphatase 4 (EYA4) as an epigenetic biomarker for gastric cardiac IM. METHODS The study was conducted in the context of the gastric cardiac precancerous lesion program in southern China, which included 718 non-cancer participants, who undertook endoscopic biopsy and pathological examination in three endoscopy centers, between November 2018 and November 2021. Pyrosequencing and immunohistochemistry were performed to examine the DNA methylation status and protein expression level of EYA4. RESULTS Gastric cardiac IM presented in 14.1% (101/718) of participants and was more common among older (>50 years; 22.0% [95% CI: 17.8-26.8]) than younger participants (≤50 years; 6.7% [95% CI: 4.5-9.9]; P < 0.001). IM was more common in male participants (16.9% [95% CI: 13.2-21.3] vs. 11.3% [95% CI: 8.3-15.1]; P = 0.04). Pyrosequencing revealed that IM tissues exhibited significantly higher DNA methylation levels in EYA4 gene than normal tissues (P = 0.016). Further, the protein expression level of EYA4 was reduced in IM and absent in intraepithelial neoplasia tissues compared to normal tissues (P < 0.001). CONCLUSIONS Detection rates of gastric cardiac IM increase with age and are higher in men. Our findings highlight the important role of promoter hypermethylation and downregulation of EYA4 in gastric cardiac IM development.
Collapse
Affiliation(s)
- Chenxi Li
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
| | - Zhaohui Liu
- Department of Gastroenterology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, P.R. China
| | - Guohua Xu
- Department of Gastroenterology, Huiyang Sanhe Hospital, Huizhou, P.R. China
| | - Shibin Wu
- Department of Gastroenterology, Huiyang Sanhe Hospital, Huizhou, P.R. China
| | - Yunhui Peng
- Department of Gastroenterology, Huiyang Sanhe Hospital, Huizhou, P.R. China
| | - Ruinuan Wu
- Department of Pathology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, P.R. China
| | - Shukun Zhao
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
| | - Xiaoqi Liao
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
| | - Runhua Lin
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou, P.R. China
- Address for correspondence: Dr. Runhua Lin, Department of Pathology, Shantou University Medical College, No. 22, Xinling Road, Shantou 515041, P.R. China. E-mail:
| |
Collapse
|
9
|
Norwood DA, Montalvan EE, Dominguez RL, Morgan DR. Gastric Cancer: Emerging Trends in Prevention, Diagnosis, and Treatment. Gastroenterol Clin North Am 2022; 51:501-518. [PMID: 36153107 DOI: 10.1016/j.gtc.2022.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gastric adenocarcinoma (GC) is the fourth leading cause of global cancer mortality, and the leading infection-associated cancer. Helicobacter pylori is the dominant risk factor for GC and classified as an IARC class I carcinogen. Surveillance of gastric premalignant conditions is now indicated in high-risk patients. Upper endoscopy is the gold standard for GC diagnosis, and image-enhanced endoscopy increases the detection of gastric premalignant conditions and early gastric cancer (EGC). Clinical staging is crucial for treatment approach, defining early gastric cancer, operable locoregional disease, and advanced GC. Endoscopic submucosal dissection is the treatment of choice for most EGC. Targeted therapies are rapidly evolving, based on biomarkers including MSI/dMMR, HER2, and PD-L1. These advancements in surveillance, diagnostic and therapeutic strategies are expected to improve GC survival rates in the near term.
Collapse
Affiliation(s)
- Dalton A Norwood
- UAB Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Western Honduras Gastric Cancer Prevention Initiative, Copan Region Ministry of Health, Sala de Endoscopia, Calle 1 S, Hospital Regional de Occidente, Santa Rosa de Copán 41101, Honduras
| | - Eleazar E Montalvan
- Western Honduras Gastric Cancer Prevention Initiative, Copan Region Ministry of Health, Sala de Endoscopia, Calle 1 S, Hospital Regional de Occidente, Santa Rosa de Copán 41101, Honduras; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ricardo L Dominguez
- Western Honduras Gastric Cancer Prevention Initiative, Copan Region Ministry of Health, Sala de Endoscopia, Calle 1 S, Hospital Regional de Occidente, Santa Rosa de Copán 41101, Honduras
| | - Douglas R Morgan
- UAB Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
10
|
Xu MQ, Sun K, Cao C, Yin HH, Wang XJ, Yin QH, Wang LJ, Tao L, Wang K, Li F, Zhang WJ. Age-related twin-peak prevalence profiles of H. pylori infection, gastritis, GIN and gastric cancer: Analyses of 70,534 patients with gastroscopic biopsies. PLoS One 2022; 17:e0265885. [PMID: 35862441 PMCID: PMC9302749 DOI: 10.1371/journal.pone.0265885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 03/10/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives H. pylori (Hp) infection has been indicated in the pathogenesis of gastric diseases including gastric cancer (GC). This study aimed at exploring the relationships between Hp infection and gastric diseases including GC in a large dataset of routine patients undergoing gastroscopy. Methods From November 2007 to December 2017, 70,534 first-time visiting patients aged 18–94 years with gastroscopic biopsies were histologically diagnosed and analyzed. Patients’ data were entered twice in an Excel spreadsheet database and analyzed using the SPSS (version 22.0) software package and statistical significance was defined as P<0.05 for all analyses. Results The first interesting observation was age-related twin-peak prevalence profiles (TPPs) for Hp infection, gastritis, and advanced diseases with different time spans (TS) between the first and second occurring peaks. Hp infection and gastritis had TPPs occurring at earlier ages than TPPs of gastric introepithelial neoplasia (GIN) and GC. More patients were clustered at the second occurring TPPs. The time spans (TS) from the first occurring peak of Hp infection to the first occurring peaks of other gastric diseases varied dramatically with 0–5 years for gastritis; 5–15 years for GINs, and 5–20 years for GC, respectively. The number of males with Hp infection and gastric diseases, excluding non-atrophic gastritis (NAG), was more than that of females (P<0.001). Conclusions We have first observed age-related twin-peak prevalence profiles for Hp infection, gastritis, GIN, and GC, respectively, among a large population of patients undergoing gastroscopy. The second prevalence peak of GC is at ages of 70–74 years indicating that many GC patients would be missed during screening because the cut-off age for screening is 69 years old in China.
Collapse
Affiliation(s)
- Meng Qing Xu
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Gastroenterology, Jinling Hospital, Nanjing, Jiangsu, China
| | - Ke Sun
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chong Cao
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Hui Hui Yin
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiao Jun Wang
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Qi Hang Yin
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Li Jie Wang
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lin Tao
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Kui Wang
- Department of Preventive Medicine, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Feng Li
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wen Jie Zhang
- Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- * E-mail: ,
| |
Collapse
|
11
|
Rugge M. Big Data on Gastric Dysplasia Support Gastric Cancer Prevention. Clin Gastroenterol Hepatol 2022; 20:1226-1228. [PMID: 35123082 DOI: 10.1016/j.cgh.2022.01.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Massimo Rugge
- Department of Medicine DIMED, Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy; Azienda Zero Padova, Veneto Tumor Cancer Registry, Padova, Italy; Baylor College of Medicine, Houston, Texas
| |
Collapse
|
12
|
Latorre G, Pizarro M, Ford J, Gándara V, Muñoz G, Araya J, Bellolio E, Villaseca MÁ, Fuentes-López E, Cortés P, Rollán A, Bufadel M, Araya R, Vargas J, Espino A, Sharp A, Agüero C, Donoso A, Bresky G, Pedrero P, Rueda C, Calvo A, Odagaki T, Moriyama T, Ishida T, Parra-Blanco A, Camargo M, González R, Corvalán A, Riquelme A. Evaluation of Trefoil Factor 3 as a Non-Invasive Biomarker of Gastric Intestinal Metaplasia and Gastric Cancer in a High-Risk Population. GASTROENTEROLOGIA Y HEPATOLOGIA 2022; 46:411-418. [PMID: 35580739 DOI: 10.1016/j.gastrohep.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/07/2022] [Accepted: 04/01/2022] [Indexed: 01/06/2023]
|
13
|
Zan X, Chen Z, Guo Q, Wang Y, Zhang Z, Ji R, Zheng Y, Zhang J, Wu Z, Li M, Wang X, Ye Y, Li X, An F, Xu C, Lu L, Fan P, Zhang J, Guan Q, Li Q, Liu M, Ren Q, Hu X, Lu H, Wang Y, Zhang H, Zhao Y, Gou X, Shu X, Wang J, Hu Z, Liu R, Yuan H, Liu J, Qiao L, Zhou Y. The Association of Trefoil Factors with Gastric Cancer and Premalignant Lesions: A Cross-sectional Population-based Cohort Study. Cancer Epidemiol Biomarkers Prev 2022; 31:625-632. [PMID: 35027436 DOI: 10.1158/1055-9965.epi-21-0760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/18/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A lack of research on the association of trefoil factors (TFFs) with gastric cancer (GC) and premalignant lesions (PMLs) in the general populations is an important obstacle to the application of TFFs for GC screening. We aimed to analyze the association of TFFs with GC and PMLs in a general population. METHODS We evaluated 3,986 adults residing in Wuwei, China. We collected baseline characteristics and GC risk factors, including TFFs, endoscopic diagnosis, and pathological information. Three logistic regression models were generated to analyze the association between TFFs and GC, as well as PMLs. Adjusted odds ratio (OR) and 95% confidence intervals (95% CI) were calculated to determine the strength of association. RESULTS Compared with pepsinogen (PG) and anti-Helicobacter pylori immunoglobulin G antibody (Hp-IgG), TFFs had significant association with GC and PMLs after adjusting for biomarkers and risk factors (P < 0.05). The ORs [95% CI] for TFF1 (1.67 [1.27-2.20]), TFF2 (2.66 [2.01-3.51]), and TFF3 (1.32 [1.00-1.74]) were larger than the ORs for PGI (0.79 [0.61-1.03]), PGI/II (1.00 [0.76-1.31]) and Hp-IgG (0.99 [0.73-1.35]) in the GC group. In intestinal metaplasia (IM) group, not only the TFF3 serum level was the highest, but also the OR (1.92 [1.64-2.25]) was the highest. CONCLUSIONS Trefoil factor were associated with risk of GC and PMLs. IMPACT Serum TFFs can improve the screening of high-risk populations for GC.
Collapse
Affiliation(s)
- Xiangyi Zan
- Department of Gastroenterology, The First Affiliated Hospital of Lanzhou University
| | - Zhaofeng Chen
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Qinghong Guo
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yuping Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Zhiyi Zhang
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital
| | - Rui Ji
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Ya Zheng
- Department of Gastroenterology, First Hospital of Lanzhou University
| | | | | | - Min Li
- School of Basic Medical Sciences, Lanzhou University
| | - Xiang Wang
- Department of Gastroenterology, Lanzhou University Second Hospital
| | - Yuwei Ye
- Department of Gastroenterology, First Hospital of Lanzhou University
| | | | | | | | | | | | - Jun Zhang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Quanlin Guan
- First Hospital of Lanzhou University, Lanzhou University
| | - Qiang Li
- The First Affiliated Hospital of Lanzhou University
| | - Min Liu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Qian Ren
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xiaobin Hu
- Lanzhou University School of Public Health
| | - Hong Lu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yuling Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Hongling Zhang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yue Zhao
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xi Gou
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xiaochuang Shu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Jun Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Zenan Hu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Rong Liu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Hao Yuan
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Jiankang Liu
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Liang Qiao
- Storr Liver Centre, the Westmead Institute for Medical Research, the University of Sydney at the Westmead
| | - Yongning Zhou
- Department of Gastroenterology, First Hospital of Lanzhou University
| |
Collapse
|
14
|
Nakajima T, Noguchi A, Tanaka S, Nishida T, Hatta H, Kakiuchi T, Takagi K, Minamisaka T, Fujinami H, Yoshizumi T, Imura J. The potential diagnostic significance of crypt differentiation in gastric dysplasia. Histopathology 2021; 80:529-537. [PMID: 34608656 DOI: 10.1111/his.14581] [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: 07/06/2021] [Revised: 09/03/2021] [Accepted: 10/03/2021] [Indexed: 11/29/2022]
Abstract
AIMS This study investigated the relationship between the differentiation of tumour cells into crypts, which is determined by cell differentiation into Paneth and neuroendocrine cells, and tumour infiltration in gastric dysplasia. METHODS AND RESULTS The lesions were endoscopically biopsied low-grade dysplasia (LGD), endoscopically resected high-grade dysplasia (HGD), or cancer with submucosal invasion. LGD (n = 32) displayed crypt differentiation across the entire width of the tumour in all cases. Crypt differentiation was identified as a characteristic of tumours with low biological malignancy. HGD (n = 40) included tumours with a mixture of areas with and without crypt differentiation (n = 25) and tumours with crypt differentiation throughout the entire width (n = 15). Of the cancers with submucosal invasion (n = 30), the morphological progression of HGD area with crypt differentiation, HGD area without crypt differentiation and invasive cancer without crypt differentiation was confirmed for 23 samples. In two lesions, invasive cancer without crypt differentiation developed from HGD without crypt differentiation throughout the tumour width. In five samples, well-differentiated tubular adenocarcinoma with crypt differentiation developed from HGD with crypt differentiation and invaded with lamina propria-like stroma. CONCLUSIONS Loss of crypt differentiation could be an objective indicator of infiltration in the progression of HGD to invasive cancer. The invasive potential of dysplasia depends on the presence or absence of crypt differentiation.
Collapse
Affiliation(s)
- Takahiko Nakajima
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Akira Noguchi
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Shinichi Tanaka
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takeshi Nishida
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Hideki Hatta
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Toshiko Kakiuchi
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Kohji Takagi
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takashi Minamisaka
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Haruka Fujinami
- Department of Gastroenterology and Hepatology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Tetsuya Yoshizumi
- Department of Surgery, Kouseiren Namerikawa Hospital, 119 Tokiwacho, Namerikawa, 936-8585, Japan
| | - Johji Imura
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
15
|
Gastritis: The clinico-pathological spectrum. Dig Liver Dis 2021; 53:1237-1246. [PMID: 33785282 DOI: 10.1016/j.dld.2021.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023]
Abstract
The inflammatory spectrum of gastric diseases includes different clinico-pathological entities, the etiology of which was recently established in the international Kyoto classification. A diagnosis of gastritis combines the information resulting form the gross examination (endoscopy) and histology (microscopy). It is important to consider the anatomical/functional heterogeneity of the gastric mucosa when obtaining representative mucosal biopsy samples. Gastritis includes self-limiting and non-self-limiting (long-standing) inflammatory diseases, and the latter are epidemiologically, biologically and clinically linked to the onset of gastric cancer (i.e. "inflammation-associated cancer"). Different biological models of inflammation-associated gastric oncogenesis have been proposed. Helicobacter pylori (H. pylori) gastritis is the most prevalent worldwide, and H. pylori is classified as a first-class carcinogen. On these bases, eradicating H. pylori is mandatory for the primary prevention of gastric cancer. Non-self-limiting gastritis may also be triggered by the immune-mediated destruction of gastric parietal cells, resulting in autoimmune gastritis. In both H. pylori-related and autoimmune gastritis, the non-self-limiting inflammation results in atrophy of the gastric mucosa, which is the main factor promoting gastric cancer. Long-term follow-up studies consistently demonstrate the prognostic impact of the histological staging of gastritis in gastric cancer secondary prevention strategies.
Collapse
|
16
|
Cho YS, Chung IK, Jung Y, Han SJ, Yang JK, Lee TH, Park SH, Kim SJ. Risk stratification of patients with gastric lesions indefinite for dysplasia. Korean J Intern Med 2021; 36:1074-1082. [PMID: 32898394 PMCID: PMC8435497 DOI: 10.3904/kjim.2018.285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 04/16/2020] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND/AIMS There are no definite guidelines for the management of gastric lesions diagnosed as indefinite for dysplasia (IND) by endoscopic forceps biopsy (EFB). Therefore, this study aimed to evaluate the clinical outcomes of gastric IND and predictive factors for gastric neoplasm. METHODS This study included 457 patients with a first diagnosis of gastric IND by EFB between January 2005 and December 2013. Patient characteristics and endoscopic and pathological data were reviewed and compared. RESULTS Of the 457 gastric IND patients, 128 (28%) were diagnosed with invasive carcinoma, 21 (4.6%) with high-grade dysplasia, 31 (6.8%) with low-grade dysplasia, and 277 (60.6%) as negative for dysplasia. Of lesions observed, 180 (39.4%) showed upgraded histology. Multivariate analysis revealed that surface erythema (odds ratio [OR], 2.804; 95% confidence interval [CI], 1.741 to 4.516), spontaneous bleeding (OR, 2.618; 95% CI, 1.298 to 5.279), lesion size ≥ 1 cm (OR, 5.762; 95% CI, 3.459 to 9.597), and depressed morphology (OR, 2.183; 95% CI, 1.155 to 4.124) were significant risk factors for high-grade dysplasia or adenocarcinoma. The ORs associated with 2 and ≥ 3 risk factors were 7.131 and 34.86, respectively. CONCLUSION Precautions should be taken in the management of gastric IND patients, especially when risk factors, including surface erythema, spontaneous bleeding, lesion size ≥ 1 cm, and depressed morphology are present. Considering the combined effect of the presence of multiple risk factors on the incidence of high-grade dysplasia or adenocarcinoma, endoscopic resection should be recommended if a gastric IND patient has at two or more of these factors.
Collapse
Affiliation(s)
- Young Sin Cho
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Il-Kwun Chung
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- Correspondence to Il-Kwun Chung, M.D. Division of Gastroenterology, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam-gu, Cheonan 31151, Korea Tel: +82-41-570-3892 Fax: +82-41-574-5762 E-mail:
| | - Yunho Jung
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Su Jung Han
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jae Kook Yang
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Tae Hoon Lee
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Sang-Heum Park
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Sun-Joo Kim
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| |
Collapse
|
17
|
Grillo F, Mastracci L, Saragoni L, Vanoli A, Limarzi F, Gullo I, Ferro J, Paudice M, Parente P, Fassan M. Neoplastic and pre-neoplastic lesions of the oesophagus and gastro-oesophageal junction. Pathologica 2021; 112:138-152. [PMID: 33179618 PMCID: PMC7931575 DOI: 10.32074/1591-951x-164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Oesophageal and gastro-oesophageal junction (GOJ) neoplasms, and their predisposing conditions, may be encountered by the practicing pathologist both as biopsy samples and as surgical specimens in daily practice. Changes in incidence of oesophageal squamous cell carcinomas (such as a decrease in western countries) and in oesophageal and GOJ adenocarcinomas (such as a sharp increase in western countries) are being reported globally. New modes of treatment have changed our histologic reports as specific aspects must be detailed such as in post endoscopic resections or with regards to post neo-adjuvant therapy tumour regression grades. The main aim of this overview is therefore to provide an up-to-date, easily available and clear diagnostic approach to neoplastic and pre-neoplastic conditions of the oesophagus and GOJ, based on the most recent available guidelines and literature.
Collapse
Affiliation(s)
- Federica Grillo
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, Italy.,Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, Genova, Italy
| | - Luca Mastracci
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, Italy.,Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, Genova, Italy
| | - Luca Saragoni
- UO Anatomia Patologica, Ospedale G.B. Morgagni-L. Pierantoni, Forlì, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, Pavia, Italy
| | - Francesco Limarzi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST/IRCCS), Meldola (FC), Italy
| | - Irene Gullo
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ) & Department of Pathology, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal and Instituto de Investigação e Inovação em Saúde (i3S) & Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal
| | - Jacopo Ferro
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, Italy
| | - Michele Paudice
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, Italy
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Italy
| |
Collapse
|
18
|
Aumpan N, Vilaichone RK, Pornthisarn B, Chonprasertsuk S, Siramolpiwat S, Bhanthumkomol P, Nunanan P, Issariyakulkarn N, Ratana-Amornpin S, Miftahussurur M, Mahachai V, Yamaoka Y. Predictors for regression and progression of intestinal metaplasia (IM): A large population-based study from low prevalence area of gastric cancer (IM-predictor trial). PLoS One 2021; 16:e0255601. [PMID: 34379655 PMCID: PMC8357097 DOI: 10.1371/journal.pone.0255601] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background Gastric intestinal metaplasia (IM) can lead to gastric cancer. Until now, there have been limited studies of predictors for regression and progression of IM. This study aimed to determine risk factors associated with regression or progression of IM for guiding proper management and prevention of gastric cancer. Methods 2,025 patients undergoing gastroscopy in Thammasat University Hospital, Thailand were enrolled during September 2017-August 2019. Patients’ data including baseline characteristics, laboratory results, and histopathology of gastric biopsies from University medical database were extensively reviewed. Results 2,025 patients had mean age of 61.3 years and 44.2% were males. Overall H. pylori prevalence was 47.5%. There were 1,551(76.6%) patients with chronic gastritis and 361(17.8%) with IM. Of 400 patients with chronic gastritis having follow-up endoscopy and repeated gastric biopsies, 104(26%) had persistent H. pylori infection and 27(26%) developed IM during mean follow-up time of 24 months. Persistent H. pylori infection was significantly associated with development of IM (OR 3.16, 95%CI 1.56–6.39, p = 0.001). Regression, persistence, and progression of IM were demonstrated in 57.3%, 39.2%, and 3.5% of patients, respectively. Age >65 years, persistent H. pylori infection, and diabetes mellitus were significantly associated with persistent IM or progression to dysplasia with OR 2.47(95%CI 1.33–4.61, p = 0.004), OR 2.64(95%CI 1.13–6.18, p = 0.025), and OR 2.54(95%CI 1.16–5.54, p = 0.019), respectively. Patients without H. pylori infection had more IM regression than patients with persistent infection (60.4%vs.39.4%, p = 0.035). Patients with persistent H. pylori infection significantly had higher IM progression to dysplasia (15.2%vs.2.1%; OR 11.15, 95%CI 1.18–105.24, p = 0.035) than noninfected. During 24 months of study, 30 patients (1.5%) were diagnosed with gastric cancer. Conclusion Regression of IM could be achieved by successful H. pylori eradication. Persistent H. pylori infection was significantly associated with development and progression of IM to dysplasia. Age >65 years and diabetes mellitus were also significant predictors for IM progression.
Collapse
Affiliation(s)
- Natsuda Aumpan
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Ratha-korn Vilaichone
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- * E-mail:
| | - Bubpha Pornthisarn
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Soonthorn Chonprasertsuk
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sith Siramolpiwat
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
| | - Patommatat Bhanthumkomol
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Pongjarat Nunanan
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Navapan Issariyakulkarn
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sarita Ratana-Amornpin
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Muhammad Miftahussurur
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Varocha Mahachai
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
19
|
Mannion A, Dzink-Fox J, Shen Z, Piazuelo MB, Wilson KT, Correa P, Peek RM, Camargo MC, Fox JG. Helicobacter pylori Antimicrobial Resistance and Gene Variants in High- and Low-Gastric-Cancer-Risk Populations. J Clin Microbiol 2021; 59:e03203-20. [PMID: 33692136 PMCID: PMC8091839 DOI: 10.1128/jcm.03203-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/28/2021] [Indexed: 02/07/2023] Open
Abstract
Colombia, South America has one of the world's highest burdens of Helicobacter pylori infection and gastric cancer. While multidrug antibiotic regimens can effectively eradicate H. pylori, treatment efficacy is being jeopardized by the emergence of antibiotic-resistant H. pylori strains. Moreover, the spectrum of and genetic mechanisms for antibiotic resistance in Colombia is underreported. In this study, 28 H. pylori strains isolated from gastric biopsy specimens from a high-gastric-cancer-risk (HGCR) population living in the Andes Mountains in Túquerres, Colombia and 31 strains from a low-gastric-cancer-risk (LGCR) population residing on the Pacific coast in Tumaco, Colombia were subjected to antibiotic susceptibility testing for amoxicillin, clarithromycin, levofloxacin, metronidazole, rifampin, and tetracycline. Resistance-associated genes were amplified by PCR for all isolates, and 29 isolates were whole-genome sequenced (WGS). No strains were resistant to amoxicillin, clarithromycin, or rifampin. One strain was resistant to tetracycline and had an A926G mutation in its 16S rRNA gene. Levofloxacin resistance was observed in 12/59 isolates and was significantly associated with N87I/K and/or D91G/Y mutations in gyrA Most isolates were resistant to metronidazole; this resistance was significantly higher in the LGCR (31/31) group compared to the HGCR (24/28) group. Truncations in rdxA and frxA were present in nearly all metronidazole-resistant strains. There was no association between phylogenetic relationship and resistance profiles based on WGS analysis. Our results indicate H. pylori isolates from Colombians exhibit multidrug antibiotic resistance. Continued surveillance of H. pylori antibiotic resistance in Colombia is warranted in order to establish appropriate eradication treatment regimens for this population.
Collapse
Affiliation(s)
- Anthony Mannion
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - JoAnn Dzink-Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | | | | | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
20
|
Piazuelo MB, Bravo LE, Mera RM, Camargo MC, Bravo JC, Delgado AG, Washington MK, Rosero A, Garcia LS, Realpe JL, Cifuentes SP, Morgan DR, Peek RM, Correa P, Wilson KT. The Colombian Chemoprevention Trial: 20-Year Follow-Up of a Cohort of Patients With Gastric Precancerous Lesions. Gastroenterology 2021; 160:1106-1117.e3. [PMID: 33220252 PMCID: PMC7956231 DOI: 10.1053/j.gastro.2020.11.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Helicobacter pylori eradication and endoscopic surveillance of gastric precancerous lesions are strategies to reduce gastric cancer (GC) risk. To our knowledge, this study is the longest prospective cohort of an H pylori eradication trial in a Hispanic population. METHODS A total of 800 adults with precancerous lesions were randomized to anti-H pylori treatment or placebo. Gastric biopsy samples taken at baseline and 3, 6, 12, 16, and 20 years were assessed by our Correa histopathology score. A generalized linear mixed model with a participant-level random intercept was used to estimate the effect of H pylori status on the score over time. Logistic regression models were used to estimate progression by baseline diagnosis and to estimate GC risk by intestinal metaplasia (IM) subtype and anatomic location. RESULTS Overall, 356 individuals completed 20 years of follow-up. Anti-H pylori therapy (intention-to-treat) reduced progression of the Correa score (odds ratio [OR], 0.59; 95% confidence interval [CI], 0.38-0.93). H pylori-negative status had a beneficial effect on the score over time (P = .036). Among individuals with IM (including indefinite for dysplasia) at baseline, incidence rates per 100 person-years were 1.09 (95% CI, 0.85-1.33) for low-grade/high-grade dysplasia and 0.14 (95% CI, 0.06-0.22) for GC. Incomplete-type (vs complete-type) IM at baseline presented higher GC risk (OR, 13.4; 95% CI, 1.8-103.8). Individuals with corpus (vs antrum-restricted) IM showed an OR of 2.1 (95% CI, 0.7-6.6) for GC. CONCLUSIONS In a high-GC-risk Hispanic population, anti-H pylori therapy had a long-term beneficial effect against histologic progression. Incomplete IM is a strong predictor of GC risk.
Collapse
Affiliation(s)
- M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Luis E. Bravo
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Valle del Cauca, Colombia
| | - Robertino M. Mera
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Juan C. Bravo
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Valle del Cauca, Colombia
| | - Alberto G. Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Luz S. Garcia
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Valle del Cauca, Colombia
| | | | | | - Douglas R. Morgan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Division of Gastroenterology, Department of Medicine, University of Alabama, Birmingham, AL, USA
| | - Richard M. Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pelayo Correa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA,Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
21
|
Abstract
Background: We aimed to provide insight into the actual frequencies of gastric adenoma types and their association with gastritis status and associated mucosal changes with a focus on Helicobacter infection and the operative link on gastritis assessment (OLGA)/operative link on gastric intestinal metaplasia assessment (OLGIM) staging. Methods: From the archive of the Institute of Pathology in Bayreuth, we collected a consecutive series of 1058 gastric adenomas diagnosed between 1987 and 2017. Clinicopathological parameters retrieved from diagnostic reports included adenoma type and localization, associated mucosal changes in antrum and corpus (i.e., type of gastritis, the extent of intestinal metaplasia and atrophy), gender, date of birth, and date of diagnosis. Results: Intestinal-type adenoma was the most frequent adenoma (89.1%), followed by foveolar-type adenoma (4.3%), pyloric gland adenoma (3.4%), adenomas associated with hereditary tumor syndromes (2.8%), and oxyntic gland adenoma (0.4%). Adenomas were found in the background of Helicobacter pylori (H. pylori) gastritis in 23.9%, Ex-H. pylori gastritis in 36.0%, autoimmune gastritis in 24.8%, chemical reactive gastritis in 7.4%, and others in 0.1%. More than 70% of patients with gastric adenomas had low-risk stages in OLGA and OLGIM. Conclusions: We found a higher frequency of foveolar-type adenoma than anticipated from the literature. It needs to be questioned whether OLGA/OLGIM staging can be applied to all patients.
Collapse
|
22
|
Koulis A, Busuttil RA, Boussioutas A. Premalignant lesions of the stomach and management of early neoplastic lesions. RESEARCH AND CLINICAL APPLICATIONS OF TARGETING GASTRIC NEOPLASMS 2021:185-216. [DOI: 10.1016/b978-0-323-85563-1.00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
23
|
Varga MG, Wood CR, Butt J, Ryan ME, You WC, Pan K, Waterboer T, Epplein M, Shaffer CL. Immunostimulatory membrane proteins potentiate H. pylori-induced carcinogenesis by enabling CagA translocation. Gut Microbes 2021; 13:1-13. [PMID: 33382363 PMCID: PMC7781638 DOI: 10.1080/19490976.2020.1862613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/04/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Infection with Helicobacter pylori is the single greatest risk factor for developing gastric adenocarcinoma. In prospective, population-based studies, seropositivity to the uncharacterized H. pylori proteins Hp0305 and Hp1564 was significantly associated with cancer risk in East Asia. However, the mechanism underlying this observation has not been elucidated. Here, we show that Hp0305 and Hp1564 act in concert with previously ascribed H. pylori virulence mechanisms to orchestrate cellular alterations that promote gastric carcinogenesis. In samples from 546 patients exhibiting premalignant gastric lesions, seropositivity to Hp0305 and Hp1564 was significantly associated with increased gastric atrophy across all stomach conditions. In vitro, depletion of Hp0305 and Hp1564 significantly reduced levels of gastric cell-associated bacteria and markedly impaired the ability of H. pylori to stimulate pro-inflammatory cytokine production. Remarkably, our studies revealed that Hp1564 is required for translocation of the oncoprotein CagA into gastric epithelial cells. Our data provide experimental insight into the molecular mechanisms governing novel H. pylori pathogenicity factors that are strongly associated with gastric disease and highlight the potential of Hp0305 and Hp1564 as robust molecular tools that can improve identification of individuals that are highly susceptible to gastric cancer. We demonstrate that Hp0305 and Hp1564 augment H. pylori-mediated inflammation and gastric cancer risk by promoting key bacteria-gastric cell interactions that facilitate delivery of oncogenic microbial cargo to target cells. Thus, therapeutically targeting microbial interactions driven by Hp0305/Hp1564 may enable focused H. pylori eradication strategies to prevent development of gastric malignancies in high-risk populations.
Collapse
Affiliation(s)
- Matthew G. Varga
- Department of Epidemiology, Lineberger Comprehensive Cancer Center and Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Cecily R. Wood
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Julia Butt
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Mackenzie E. Ryan
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Wei-Cheng You
- Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kaifeng Pan
- Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Meira Epplein
- Department of Population Health Sciences and Duke Cancer Institute, Cancer Control and Population Sciences Program, Duke University, Durham, NC, USA
| | - Carrie L. Shaffer
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
24
|
Huang H, Rong Y, Wang M, Guo Z, Yu Y, Long Z, Chen X, Wang H, Ding J, Yan L, Peng J. Analysis of gastroscopy results among healthy people undergoing a medical checkup: a retrospective study. BMC Gastroenterol 2020; 20:412. [PMID: 33297981 PMCID: PMC7724807 DOI: 10.1186/s12876-020-01557-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Background The association of upper gastrointestinal endoscopic findings with sex, age, and Helicobacter pylori infection in asymptomatic healthy people is unclear. The aim of this study was to retrospectively determine the associations of upper gastrointestinal endoscopic findings in asymptomatic healthy people with sex, age, and H. pylori infection.
Methods A retrospective study was conducted on 2923 patients from a health examination center in Xiangya Hospital between September 2015 and September 2019. Data on sex, age, H. pylori infection, and gastroscopy results were collected.
Results Among 2923 asymptomatic patients who underwent gastroscopy, 2911 (99.59%) had abnormal results. The top three results were chronic gastritis (95.11%), peptic ulcer (17.45%), and duodenitis (9.17%). Inflammation of the gastric mucosa in chronic gastritis was more severe in the H. pylori-positive group. The incidence of peptic ulcer decreased with increasing age and was higher in men, patients aged < 30 years, and H. pylori-positive patients. The incidence of polyps was higher in women (9.54%) than in men (5.94%), and the incidence in individuals aged ≥60 years (11.63%) was higher than that in those aged < 60 years (6.83%). The pathological results of gastric polyps depended on the location of the lesion. Conclusion The incidence of abnormal upper gastrointestinal endoscopic results is high in asymptomatic healthy people undergoing a check-up and is associated with sex, age, and H. pylori infection. Gastroscopy should be considered part of a routine health check.
Collapse
Affiliation(s)
- Haosu Huang
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yanting Rong
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Meng Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Zimeng Guo
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yanghua Yu
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Zhenpu Long
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Xiaoxiao Chen
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Hanyue Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Junjie Ding
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Lu Yan
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Jie Peng
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
25
|
Gastric cancer screening in low incidence populations: Position statement of AEG, SEED and SEAP. GASTROENTEROLOGIA Y HEPATOLOGIA 2020; 44:67-86. [PMID: 33252332 DOI: 10.1016/j.gastrohep.2020.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/06/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
This positioning document, sponsored by the Asociación Española de Gastroenterología, the Sociedad Española de Endoscopia Digestiva and the Sociedad Española de Anatomía Patológica, aims to establish recommendations for the screening of gastric cancer (GC) in low incidence populations, such as the Spanish. To establish the quality of the evidence and the levels of recommendation, we used the methodology based on the GRADE system (Grading of Recommendations Assessment, Development and Evaluation). We obtained a consensus among experts using a Delphi method. The document evaluates screening in the general population, individuals with relatives with GC and subjects with GC precursor lesions (GCPL). The goal of the interventions should be to reduce GC related mortality. We recommend the use of the OLGIM classification and determine the intestinal metaplasia (IM) subtype in the evaluation of GCPL. We do not recommend to establish endoscopic mass screening for GC or Helicobacter pylori. However, the document strongly recommends to treat H.pylori if the infection is detected, and the investigation and treatment in individuals with a family history of GC or with GCPL. Instead, we recommend against the use of serological tests to detect GCPL. Endoscopic screening is suggested only in individuals that meet familial GC criteria. As for individuals with GCPL, endoscopic surveillance is only suggested in extensive IM associated with additional risk factors (incomplete IM and/or a family history of GC), after resection of dysplastic lesions or in patients with dysplasia without visible lesion after a high quality gastroscopy with chromoendoscopy.
Collapse
|
26
|
Waters KM, Salimian KJ, Assarzadegan N, Hutchings D, Makhoul EP, Windon AL, Wong MT, Voltaggio L, Montgomery EA. Cell polarity (the 'four lines') distinguishes gastric dysplasia from epithelial changes in reactive gastropathy. Histopathology 2020; 78:453-458. [PMID: 32841414 DOI: 10.1111/his.14242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/19/2020] [Indexed: 12/24/2022]
Abstract
AIMS Gastric dysplasia is a risk factor for synchronous and subsequent gastric carcinoma. Distinguishing gastric dysplasia from reactive changes is subject to interobserver disagreement and is a frequent reason for expert consultation. We previously used assessment of surface cell polarity (the 'four lines') as a key feature to decrease equivocal diagnoses in Barrett oesophagus. In the current study, we examined for the presence or absence of the four lines in gastric dysplasia and reactive gastropathy. MATERIALS AND METHODS The study includes all (n = 91) in-house biopsies with at least gastric dysplasia from the surgical pathology archives of two academic institutions during a 5-year period from 2008 to 2012. A reactive gastropathy group (n = 60) was created for comparison. RESULTS The dysplasia/neoplasia group was comprised of 14 biopsies of gastric foveolar-type dysplasia, 59 of intestinal-type dysplasia, 14 with dysplasia in fundic gland polyps, three pyloric gland adenomas and one oxyntic gland adenoma. Loss of surface cell polarity was seen in all 88 dysplasia cases with evaluable surface epithelium. All 57 reactive gastropathy cases with evaluable surface epithelium showed intact surface cell polarity except in focal areas directly adjacent to erosions in 17 cases, where the thin wisp of residual surface mucin could not be appreciated on haematoxylin and eosin. CONCLUSION Surface cell polarity (the four lines) was lost in all gastric dysplasia biopsies with evaluable surface epithelium and maintained in all biopsies of reactive gastropathy. Caution should be taken in using this feature adjacent to erosions in reactive gastropathy.
Collapse
Affiliation(s)
- Kevin M Waters
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kevan J Salimian
- Department of Pathology, Johns Hopkins Hospital, Baltimore, MD, USA
| | | | | | - Elias P Makhoul
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Annika L Windon
- Department of Pathology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Mary T Wong
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | |
Collapse
|
27
|
Nam JH, Nam SY, Park BJ, Ryu KH. Effect of Helicobacter pylori infection and its eradication on gastric regenerating atypia in 22 133 subjects. J Gastroenterol Hepatol 2020; 35:1532-1539. [PMID: 32083327 DOI: 10.1111/jgh.15016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/06/2020] [Accepted: 02/18/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM The aim of this study was to identify factors affecting persistent gastric regenerating atypia and determine the effect of Helicobacter pylori eradication on the course of this lesion. METHODS In cross-sectional setting, comprehensive health check-up subjects who underwent both endoscopy and H. pylori test from 2001 to 2009 were included. The association between H. pylori and gastric regenerating atypia was evaluated. In cohort setting, patients with regenerating atypia who underwent H. pylori test from 2001 to 2013 were included. Factors affecting positive pathology (persistent regenerating atypia or new development of neoplasm) in patients with regenerating atypia at baseline were investigated. RESULTS In cross-sectional setting, regenerating atypia was observed in 1.1% (241/22 133). H. pylori infection was associated with gastric regenerating atypia (adjusted odds ratio, 1.47; 95% confidence interval [CI], 1.12-1.91). In cohort setting, 310 patients with regenerating atypia were finally eligible. Positive pathology rate during follow up was 16.1% (15/93) in the persistent infection group, 2.8% (3/106) in successful eradication group, and 4.5% (5/111) in baseline H. pylori-negative group. Persistent H. pylori infection increased the risk of positive pathology (adjusted risk ratio [RR], 7.18; 95% CI, 1.95-26.48) compared to H. pylori eradication group. Persistent H. pylori infection increased the risk of regenerative atypia (adjusted RR, 5.70; 95% CI, 1.46-22.17) and new neoplasm (adjusted RR, 10.74; 95% CI, 1.10-105.17) compared to baseline negative H. pylori. CONCLUSIONS H. pylori infection is an independent risk factor for gastric regenerating atypia. Eradication of H. pylori seems helpful for regression of regenerating atypia.
Collapse
Affiliation(s)
- Ji Hyung Nam
- Department of Internal Medicine, Center for Cancer Prevention and Detection, National Cancer Center, Goyang, South Korea.,Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, South Korea
| | - Su Youn Nam
- Department of Internal Medicine, Center for Cancer Prevention and Detection, National Cancer Center, Goyang, South Korea.,Center for Gastric Cancer, Kyungpook National University Hospital Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Bum Joon Park
- Department of Internal Medicine, Center for Cancer Prevention and Detection, National Cancer Center, Goyang, South Korea
| | - Kum Hei Ryu
- Department of Internal Medicine, Center for Cancer Prevention and Detection, National Cancer Center, Goyang, South Korea
| |
Collapse
|
28
|
Esposito G, Angeletti S, Cazzato M, Galli G, Conti L, Di Giulio E, Annibale B, Lahner E. Narrow band imaging characteristics of gastric polypoid lesions: a single-center prospective pilot study. Eur J Gastroenterol Hepatol 2020; 32:701-705. [PMID: 32356956 DOI: 10.1097/meg.0000000000001697] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Gastric polypoid lesions (GPL) are endoscopic findings whose histological nature is difficult to determine with white-light endoscopy. Hyperplastic polyps (HP), type-1 gastric carcinoids (T1-GC) and adenomas are the most frequent GPL needing different management. Narrow-band imaging (NBI) has high accuracy for gastric malignant lesions but few studies assessed whether GPL display specific NBI characteristics. We aimed to investigate the endoscopic NBI appearances of GPL. MATERIALS AND METHODS During gastroscopies, images of GPL were recorded, and lesions were removed for histological evaluation. Two endoscopists blindly reviewed the digital images and registered the endoscopic NBI appearances on a specific check-list. GPL were categorized in HP, adenomas and T1-GC using histology as gold standard. RESULTS Overall 52 GPL, observed in 40 patients [F55%; age 63 (36-85) years], were included: 29 (55.8%) HP; 18 (34.6%) T1-GC; 5 (9.6%) adenomas. The median size was seven (2-35) mm. A regular circular mucosal pattern was more frequently observed in HP and T1-GC compared to adenomas (P < 0.001). T1-GC showed a central erosion in 77.8% (P < 0.001 versus HP) with a clear demarcation line in 33.3%. Adenomas had tubule-villous mucosal pattern in 80% (P = 0.01 versus other lesions). CONCLUSION NBI analysis of the mucosal pattern seems to be effective to endoscopically discriminate between adenomas and HP while the main characteristic of T1-GC seems to be the presence of a central erosion, sometimes with demarcation line. The endoscopic NBI characterization of GPL may contribute to optimize the management of these lesions.
Collapse
Affiliation(s)
- Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Whary MT, Avenia JMR, Bravo LE, Lofgren JL, Lertpiriyapong K, Mera-Giler R, Piazuelo MB, Correa P, Peek RM, Wilson KT, Fox JG. Contrasting serum biomarker profiles in two Colombian populations with different risks for progression of premalignant gastric lesions during chronic Helicobacter pylori infection. Cancer Epidemiol 2020; 67:101726. [PMID: 32447242 DOI: 10.1016/j.canep.2020.101726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Colombians in coastal Tumaco have a lower incidence of Helicobacter pylori-associated gastric cancer compared to individuals from Tuquerres in the high Andes. This is despite nearly universal prevalence of H. pylori infection and chronic gastritis. METHODS H. pylori infection was confirmed by Steiner stain and serology using African and European-origin strains. Gastric histology and serum inflammatory biomarkers in dyspeptic Tumaco or Tuquerres patients were evaluated to predict progression of gastric lesions. RESULTS H. pylori infection was nearly universal by Steiner stain and serology. IgG response to European-origin H. pylori strains were greater than African-origin. High gastric cancer-risk Tuquerres patients, compared to low-risk Tumaco, had significant odds ratios for lesion progression associated with serum IL-5, trefoil factor 3 (TFF3), and low pepsinogen I/II ratio. Sensitivity and specificity for these parameters was 63.8% and 67.9%, respectively, with correctly classifying patients at 66.7%. Most odds ratios for 26 other biomarkers were significant for the town of residency, indicating an environmental impact on Tumaco patients associated with decreased lesion progression. CONCLUSION An IL-5 association with progression of gastric lesions is novel and could be evaluated in addition to TFF3 and pepsinogen I/II ratio as a non-invasive prognostic screen. Results suggest Tumaco patients were exposed to infectious diseases beyond H. pylori such as the documented high incidence of helminthiasis and toxoplasmosis. IMPACT Results support a prior recommendation to evaluate TFF3 and pepsinogen I/II together to predict aggressive gastric histology. Our data indicate IL-5 should be further evaluated as prognostic parameter.
Collapse
Affiliation(s)
- Mark T Whary
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Luis E Bravo
- Department of Pathology, Universidad Del Valle, Cali, Colombia
| | - Jennifer L Lofgren
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kvin Lertpiriyapong
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robertino Mera-Giler
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pelayo Correa
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard M Peek
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
30
|
Incidence Rate of Gastric Cancer Adenocarcinoma in Patients With Gastric Dysplasia: A Systematic Review and Meta-Analysis. J Clin Gastroenterol 2019; 53:703-710. [PMID: 31415022 DOI: 10.1097/mcg.0000000000001257] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several studies have reported the risk of progression to gastric adenocarcinoma (GAC) in patients with gastric dysplasia (GD); however, the findings are controversial. We performed a systematic review and meta-analysis to study the incidence rate of GAC among patients with GD. Using a comprehensive search strategy, we systematically searched online databases including PubMed, Scopus, EMBASE, Cochrane Library, and Web of Science databases for identifying all relevant original articles through inception until July 2018. Cochran Q and I tests were used to assess heterogeneities between included studies. The incidence rates of GAC and their corresponding 95% confidence intervals (CIs) were pooled using random-effect or fixed-effect models. Of the 1980 retrieved records, 30 eligible articles (61 studies) were included. The overall pooled incidence rate of GAC was 40.36 (95% CI, 27.08-55.71; I, 96.0%) cases per 1000 person-years in patients with GD. Subgroup analysis according to the type of GD indicated the highest incidence rate of GAC was 186.40 (95% CI, 106.63-285.60; I, 94.6%) per 1000 person-years among patients with high-grade dysplasia (HGD) lesions. Although the incidence rates of GAC in low-grade dysplasia (LGD) lesions and in nonclassified lesions were 11.25 (95% CI, 3.91-21.22; I, 89.3%), and 1.40 (95% CI, 0.00-9.71; I, 78.8%), respectively. Compared with patients with LGD lesions, progression rate from GD to GAC was roughly 16 times greater in patients with HGD lesions. As the majority of patients with GAC are diagnosed in an advanced stage our study suggests strict management of HGD lesions to prevent GAC.
Collapse
|
31
|
Koulis A, Buckle A, Boussioutas A. Premalignant lesions and gastric cancer: Current understanding. World J Gastrointest Oncol 2019; 11:665-678. [PMID: 31558972 PMCID: PMC6755108 DOI: 10.4251/wjgo.v11.i9.665] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/29/2019] [Accepted: 08/21/2019] [Indexed: 02/05/2023] Open
Abstract
Over the last two decades there has been a broad paradigm shift in our understanding of gastric cancer (GC) and its premalignant states from gross histological models to increasingly precise molecular descriptions. In this review we reflect upon the historic approaches to describing premalignant lesions and GC, highlight the current molecular landscape and how this could inform future risk assessment prevention strategies.
Collapse
Affiliation(s)
- Athanasios Koulis
- Upper Gastrointestinal Translational Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- the Sir Peter MacCallum Department of Surgical Oncology, the University of Melbourne, Melbourne 3010, Australia
| | - Andrew Buckle
- Upper Gastrointestinal Translational Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- the Sir Peter MacCallum Department of Surgical Oncology, the University of Melbourne, Melbourne 3010, Australia
| | - Alex Boussioutas
- Upper Gastrointestinal Translational Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- the Sir Peter MacCallum Department of Surgical Oncology, the University of Melbourne, Melbourne 3010, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, 3050, Australia
| |
Collapse
|
32
|
[Dysplasia in gastrointestinal mucosa: Interobserver variability and value of histological examination in the diagnosis of these lesions]. Ann Pathol 2019; 40:19-23. [PMID: 31255412 DOI: 10.1016/j.annpat.2019.04.007] [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: 01/10/2019] [Revised: 04/13/2019] [Accepted: 04/25/2019] [Indexed: 11/21/2022]
Abstract
Dysplasia is a preneoplastic lesion. Histological diagnosis is based on the presence of architectural and cytological modifications, and dedifferentiation, which the intensity is variable. Dysplasia is commonly graded as low and high grade. We achieved a retrospective study of 90 cases of gastrointestinal dysplasia collected in the Department of Pathology of Hassan II university hospital of Fez, during a period of 3 years. The cases were reviewed independently and blindly without clinical information by two pathologists. Their diagnoses were compared to the initial diagnosis. Interobserver concordance in the diagnosis of the degree of dysplasia was evaluated. The average age of our patients was 56 years oldwithout predominance of sex. The interobserver concordance in the diagnosis of digestive dysplasia between the three pathologists was moderate (with a Kappa estimated at 0.42). Concordant diagnosis for digestive low grade dysplasia was observed in 50 cases of the 55 cases (90%) and only 18 cases of high-grade dysplasia of 35 cases studied (50%). The agreement for the low-grade dysplasia is more significant. However, there is significant variation in the analysis of high-grade dysplasia. This work reveals a moderate reproducibility in the diagnosis of gastrointestinal dysplasia lesions and mostly well marked for high-grade dysplasia.
Collapse
|
33
|
Shibagaki K, Fukuyama C, Mikami H, Izumi D, Yamashita N, Mishiro T, Oshima N, Ishimura N, Sato S, Ishihara S, Nagase M, Araki A, Ishikawa N, Maruyama R, Kushima R, Kinoshita Y. Gastric foveolar-type adenomas endoscopically showing a raspberry-like appearance in the Helicobacter pylori -uninfected stomach. Endosc Int Open 2019; 7:E784-E791. [PMID: 31198840 PMCID: PMC6561766 DOI: 10.1055/a-0854-3818] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
Background and study aims Foveolar-type adenoma is described as a very rare tumor that occurs in individuals without Helicobacter pylori (HP) infection and diagnosed as adenocarcinoma in the Japanese Classification of Gastric Carcinoma (JCGC). However, we have frequently encountered patients with foveolar-type adenoma that endoscopically resembles a hyperplastic polyp, suggesting that it has just been overlooked to date. Here, we analyzed clinicopathological characteristics of a special subtype of foveolar-type adenoma showing specific endoscopic findings. Patients and methods From a total of 212 patients with gastric cancer resected during a 22-month period, we enrolled 14 (6.6 %) diagnosed with foveolar-type adenoma (adenocarcinoma in JCGC). HP infection status was determined by eradication history, HP serum IgG antibody level, urea breath test, and endoscopic and histological findings. All lesions were observed using white-light endoscopy and narrow-band imaging with magnification endoscopy (NBIME). Endoscopically resected lesions were histologically examined. Results None of 14 patients had a current or past history of HP infection. All lesions were visualized on non-atrophic gastric mucosa as small reddish protrusions with fine granular surface, showing a raspberry-like appearance. NBIME showed papillary or gyrus-like microstructures with irregular capillary. Lesions were histologically diagnosed as foveolar-type adenoma showing MUC5AC-positive gastric mucin phenotype. Ki-67 was overexpressed (median labeling index 69.9 %, range 28.4 - 92.1 %), though all lesions were an intraepithelial tumor without stromal invasion. p53 over-staining was not seen in any. Conclusions Raspberry-like lesions on non-atrophic gastric mucosa in HP-uninfected individuals should be evaluated for the possibility of a special subtype of foveolar-type adenoma.
Collapse
Affiliation(s)
- Kotaro Shibagaki
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Chika Fukuyama
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Hironobu Mikami
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Daisuke Izumi
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Noritsugu Yamashita
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Tsuyoshi Mishiro
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Naoki Oshima
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Norihisa Ishimura
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Shuichi Sato
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Shunji Ishihara
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Mamiko Nagase
- Department of Pathology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Asuka Araki
- Department of Pathology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Noriyoshi Ishikawa
- Department of Pathology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Riruke Maruyama
- Department of Pathology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Ryoji Kushima
- Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Yoshikazu Kinoshita
- Department of Gastroenterology, Faculty of Medicine, Shimane University, Izumo, Japan
| |
Collapse
|
34
|
Kim JM, Sohn JH, Cho MY, Kim WH, Chang HK, Jung ES, Kook MC, Jin SY, Chae YS, Park YS, Kang MS, Kim H, Lee JH, Park DY, Kim KM, Kim H, Suh YJ, Seol SY, Jung HY, Kim DH, Lee NR, Park SH, You JH. Inter-observer Reproducibility in the Pathologic Diagnosis of Gastric Intraepithelial Neoplasia and Early Carcinoma in Endoscopic Submucosal Dissection Specimens: A Multi-center Study. Cancer Res Treat 2019; 51:1568-1577. [PMID: 30971066 PMCID: PMC6790834 DOI: 10.4143/crt.2019.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/29/2019] [Indexed: 01/15/2023] Open
Abstract
PURPOSE The diagnostic criteria of gastric intraepithelial neoplasia (IEN) are controversial across the world. We investigated how many discrepancies occur in the pathologic diagnosis of IEN and early gastric carcinoma in endoscopic submucosal dissection (ESD) specimens, and evaluated the reasons of the discordance. Materials and Methods We retrospectively reviewed 1,202 ESD specimens that were originally diagnosed as gastric IEN and early carcinoma at 12 institutions. RESULTS The final consensus diagnosis of carcinoma were 756 cases, which were originally 692 carcinomas (91.5%), 43 high-grade dysplasias (5.7%), 20 low-grade dysplasias (2.6%), and 1 others (0.1%), respectively. High- and low-grade dysplasia were finally made in 63 and 342 cases, respectively. The diagnostic concordance with the consensus diagnosis was the highest for carcinoma (91.5%), followed by low-grade dysplasia (86.3%), others (63.4%) and high-grade dysplasia (50.8%). The general kappa value was 0.83, indicating excellent concordance. The kappa values of individual institutions ranged from 0.74 to 1 and correlated with the proportion of carcinoma cases. The cases revised to a final diagnosis of carcinoma exhibited both architectural abnormalities and cytologic atypia. The main differential points between low- and high-grade dysplasias were the glandular distribution and glandular shape. Additional features such as the glandular axis, surface maturation, nuclear stratification and nuclear polarity were also important. CONCLUSION The overall concordance of the diagnosis of gastric IEN and early carcinoma in ESD specimens was excellent. It correlated with the proportion of carcinoma cases, demonstrating that the diagnostic criteria for carcinoma are more reproducible than those for dysplasia.
Collapse
Affiliation(s)
- Joon Mee Kim
- Department of Pathology, Inha University School of Medicine, Incheon, Korea
| | - Jin Hee Sohn
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mee-Yon Cho
- Department of Pathology, Yonsei University Wonju Severance Christian Hospital, Wonju, Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Kyung Chang
- Department of Pathology, Kosin University College of Medicine, Busan, Korea
| | - Eun Sun Jung
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - So-Young Jin
- Department of Pathology, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Yang Seok Chae
- Department of Pathology, Korea University College of Medicine, Seoul, Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Seon Kang
- Department of Pathology, Inje University Busan Paik Hospital, Busan, Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hyuk Lee
- Department of Pathology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Do Youn Park
- Department of Pathology, Pusan National University College of Medicine, Busan, Korea
| | - Kyoung Mee Kim
- Department of Pathology, Samsung Medical Cancer, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hoguen Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ju Suh
- Department of Pathology, Department of Biomedical Sciences, Inha University School of Medicine, Incheon, Korea
| | - Sang Yong Seol
- Department of Pathology, Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Korea
| | - Hwoon-Yong Jung
- Department of Pathology, Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Asan Digestive Disease Research Institute, Seoul, Korea
| | - Deuck-Hwa Kim
- Department of Pathology, Department of Statistics, Inha University, Incheon, Korea
| | - Na Rae Lee
- Department of Pathology, National Evidence-based Healthcare Collaborating Agency, Seoul, Korea
| | - Seung-Hee Park
- Department of Pathology, National Evidence-based Healthcare Collaborating Agency, Seoul, Korea
| | - Ji Hye You
- Department of Pathology, National Evidence-based Healthcare Collaborating Agency, Seoul, Korea
| |
Collapse
|
35
|
Massironi S, Zilli A, Elvevi A, Invernizzi P. The changing face of chronic autoimmune atrophic gastritis: an updated comprehensive perspective. Autoimmun Rev 2019; 18:215-222. [PMID: 30639639 DOI: 10.1016/j.autrev.2018.08.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023]
|
36
|
Buzzelli JN, O'Connor L, Scurr M, Chung Nien Chin S, Catubig A, Ng GZ, Oshima M, Oshima H, Giraud AS, Sutton P, Judd LM, Menheniott TR. Overexpression of IL-11 promotes premalignant gastric epithelial hyperplasia in isolation from germline gp130-JAK-STAT driver mutations. Am J Physiol Gastrointest Liver Physiol 2019; 316:G251-G262. [PMID: 30520693 DOI: 10.1152/ajpgi.00304.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Expression of the cytokine IL-11 is elevated in human Helicobacter pylori infection and progressively increases with worsening gastric pathology. Additionally, IL-11 is required for tumor development in STAT3-dependent murine models of gastric cancer (GC) and, when administered acutely, causes resolving atrophic gastritis. However, it is unclear whether locally elevated IL-11 ligand expression can, in isolation from oncogenic gp130-JAK-STAT pathway mutations, initiate GC pathogenesis. Here we developed a transgenic mouse model of stomach-specific (keratin 19 promoter) IL-11 ligand overexpression. Keratin 19 promoter-IL-11 transgenic ( K19-IL11Tg) mice showed specific IL-11 overexpression in gastric corpus and antrum but not elsewhere in the gastrointestinal tract or in other tissues. K19-IL11Tg mice developed spontaneous premalignant disease of the gastric epithelium, progressing from atrophic gastritis to TFF2-positive metaplasia and severe epithelial hyperplasia, including adenoma-like lesions in a subset of older (1 yr old) animals. Although locally advanced, the hyperplastic lesions remained noninvasive. H. pylori infection in K19-IL11Tg mice accelerated some aspects of the premalignant phenotype. Finally, K19-IL11Tg mice had splenomegaly in association with elevated serum IL-11, with spleens showing an expanded myeloid compartment. Our results provide direct in vivo functional evidence that stomach-specific overexpression of IL-11, in isolation from germline gp130-JAK-STAT3 genetic drivers, is sufficient for premalignant progression. These findings have important functional implications for human GC, in which frequent IL-11 overexpression occurs in the reported absence of somatic mutations in gp130 signaling components. NEW & NOTEWORTHY We provide direct in vivo functional evidence that stomach-specific overexpression of the cytokine IL-11, in isolation from gp130-JAK-STAT3 pathway mutations, can trigger spontaneous atrophic gastritis progressing to locally advanced epithelial hyperplasia (but not dysplasia or carcinoma), which does not require, but may be accelerated by, concomitant Helicobacter pylori infection.
Collapse
Affiliation(s)
- Jon N Buzzelli
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Louise O'Connor
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Michelle Scurr
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Sharleen Chung Nien Chin
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Angelique Catubig
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Garrett Z Ng
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Masanobu Oshima
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University , Kanazawa , Japan
| | - Hiroko Oshima
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University , Kanazawa , Japan
| | - Andrew S Giraud
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia.,Department of Paediatrics, University of Melbourne, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Philip Sutton
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia.,Department of Paediatrics, University of Melbourne, The Royal Children's Hospital , Parkville, Victoria , Australia.,Faculty of Veterinary and Agricultural Science, University of Melbourne , Parkville, Victoria , Australia
| | - Louise M Judd
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia.,Department of Paediatrics, University of Melbourne, The Royal Children's Hospital , Parkville, Victoria , Australia
| | - Trevelyan R Menheniott
- Murdoch Children's Research Institute, The Royal Children's Hospital , Parkville, Victoria , Australia.,Department of Paediatrics, University of Melbourne, The Royal Children's Hospital , Parkville, Victoria , Australia
| |
Collapse
|
37
|
Rugge M, Meggio A, Pravadelli C, Barbareschi M, Fassan M, Gentilini M, Zorzi M, Pretis GD, Graham DY, Genta RM. Gastritis staging in the endoscopic follow-up for the secondary prevention of gastric cancer: a 5-year prospective study of 1755 patients. Gut 2019; 68:11-17. [PMID: 29306868 DOI: 10.1136/gutjnl-2017-314600] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 11/21/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Operative link on gastritis assessment (OLGA) staging for gastritis ranks the risk for gastric cancer (GC) in progressive stages (0-IV). This prospective study aimed at quantifying the cancer risk associated with each gastritis stage. DESIGN A cohort of 1755 consecutive patients with dyspepsia underwent initial (T-0) oesophagogastroduodenoscopy with mapped gastric biopsies, OLGA staging and assessment of Helicobacter pylori infection. Patients were followed for 55 months (median); patients with stages II III and IV underwent a second endoscopy/restaging (T-1), and those with stages 0 and I were followed clinically and through in-depth clinical and record checking. Endpoints were OLGA stage at T-1 and development of gastric epithelial neoplasia. RESULTS At T-0, 77.6% of patients had stage 0, 14.4% stage I, 5.1% stage II, 2.1% stage III and 0.85% stage IV. H. pylori infection was detected in 603 patients at T-0 and successfully eradicated in 602 of them; 220 had a documented history of H. pylori eradication; and 932 were H. pylori naïve-negative. Incident neoplastic lesions (prevalence=0.4%; low-grade intraepithelial neoplasia (IEN)=4; high-grade IEN=1; GC=2) developed exclusively in patients with stages III-IV. The risk for epithelial neoplasia was null in patients at stages 0, I and II (95% CI 0 to 0.4), 36.5 per 1000 person-years in patients at stage III (95% CI 13.7 to 97.4) and 63.1 per 1000 person-years in patients at stage IV (95% CI 20.3 to 195.6). CONCLUSIONS This prospective study confirms that OLGA staging reliably predicts the risk for development of gastric epithelial neoplasia. Although no neoplastic lesions arose in H. pylori-naïve patients, the H. pylori eradication in subjects with advanced stages (III-IV) did not abolish the risk for neoplastic progression.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), Pathology Unit, University of Padua, Padova, Italy.,Veneto Tumor Registry, Veneto Region, Padova, Italy
| | - Alberto Meggio
- Department of Gastroenterology, Trento and Rovereto Hospital, Trento, Italy
| | - Cecilia Pravadelli
- Department of Gastroenterology, Trento and Rovereto Hospital, Trento, Italy
| | | | - Matteo Fassan
- Department of Medicine (DIMED), Pathology Unit, University of Padua, Padova, Italy
| | | | - Manuel Zorzi
- Veneto Tumor Registry, Veneto Region, Padova, Italy
| | - Giovanni De Pretis
- Department of Gastroenterology, Trento and Rovereto Hospital, Trento, Italy
| | - David Y Graham
- Department of Medicine, Michael E DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas, USA
| | - Robert M Genta
- Miraca Life Sciences Research Institute, Irving, Texas, USA.,Departments of Pathology and Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
38
|
Del Moral-Hernández O, Castañón-Sánchez CA, Reyes-Navarrete S, Martínez-Carrillo DN, Betancourt-Linares R, Jiménez-Wences H, de la Peña S, Román-Román A, Hernández-Sotelo D, Fernández-Tilapa G. Multiple infections by EBV, HCMV and Helicobacter pylori are highly frequent in patients with chronic gastritis and gastric cancer from Southwest Mexico: An observational study. Medicine (Baltimore) 2019; 98:e14124. [PMID: 30653141 PMCID: PMC6370051 DOI: 10.1097/md.0000000000014124] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The chronic inflammation and damage to the gastric epithelium induced by Helicobacter pylori (H. pylori) are the main risk factors for gastric cancer development. Epstein-Barr virus (EBV) and human cytomegalovirus (HCMV) induce chronic inflammation and have been found in gastric tumors. The objectives this observational study were to determine the frequency of multiple infections by Helicobacter pylori, Epstein-Barr virus (EBV) and human cytomegalovirus (HCMV) and to relate the infection by EBV and HCMV with H. pylori vacA/cagA genotypes in patients with chronic gastritis or gastric cancer. DNA from H. pylori, EBV and HCMV was detected by PCR in biopsies from 106 Mexican patients with chronic gastritis and 32 from gastric cancer. The cagA status and the vacA genotypes of H. pylori were determined by PCR. In chronic gastritis and gastric cancer EBV was found in 69.8% and 87.5%, HCMV in 52.8% and 53.1%, and H. pylori in 48.1% and 40.6%, respectively. In chronic gastritis, 53% of H. pylori patients were EBV and 33% were both EBV/HCMV; in gastric cancer, 92.3% of H. pylori-infected individuals were EBV and 46.1% were EVB/HCMV. All the intestinal- and mixed-type tumors and the 83.3% of diffuse-type tumors were EBV. No significant differences were found between single infections or coinfections with the diagnosis or the cancer type. The H. pylori genotypes were not related to EBV or HCMV infection. The frequency of dual infections by H. pylori, EBV and HCMV is higher in patients from southwest Mexico than other populations. It is likely that these pathogens act synergistically to induce inflammation and gastric cancer.
Collapse
Affiliation(s)
- Oscar Del Moral-Hernández
- Laboratory of Virology and Epigenetics of Cancer, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero
| | | | | | | | | | - Hilda Jiménez-Wences
- Laboratory of Clinical Research, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero
| | - Sol de la Peña
- Postdoctoral Fellow CONACYT in Laboratory of Clinical Research, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero
| | - Adolfo Román-Román
- Laboratory of Bacteriology Research, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, Mexico
| | - Daniel Hernández-Sotelo
- Laboratory of Virology and Epigenetics of Cancer, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero
| | - Gloria Fernández-Tilapa
- Laboratory of Clinical Research, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero
| |
Collapse
|
39
|
Rugge M, Genta RM, Fassan M, Valentini E, Coati I, Guzzinati S, Savarino E, Zorzi M, Farinati F, Malfertheiner P. OLGA Gastritis Staging for the Prediction of Gastric Cancer Risk: A Long-term Follow-up Study of 7436 Patients. Am J Gastroenterol 2018; 113:1621-1628. [PMID: 30333540 DOI: 10.1038/s41395-018-0353-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/20/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Gastritis OLGA-staging ranks the risk for gastric cancer (GC) in progressive stages (0-IV). This long-term follow-up study quantifies the GC risk associated with each OLGA stage. METHODS Consecutive patients (7436) underwent esophagogastroscopy (T-0), with mapped gastric biopsies, OLGA staging, and H. pylori status assessment. Patients with neoplastic lesion (invasive or non-invasive) at the index endoscopy (and/or within 12 months) were excluded. All patients were followed-up (T-1) by combining different sources of clinical/pathological information (Regional Registries of: (i) esophagogastroduodenoscopies; (ii) pathology reports; (iii) cancer, (iv) mortality). The endpoint was histologically documented development of gastric epithelial neoplasia. RESULTS At T-0, the patients' distribution by OLGA stage was: Stage 0 = 80.8%; Stage I = 12.6%; Stage II = 4.3%; Stage III = 2.0%; Stage IV = 0.3%; H. pylori infection was detected in 25.9% of patients. At the end of the follow-up (mean/median = 6.3/6.6 years), 28 incident neoplasia were documented (overall prevalence = 0.60 per 103/person-years; low-grade intraepithelial neoplasia = 17/28; high-grade intraepithelial neoplasia = 4/28; GC = 7/28). By OLGA stage at the enrollment, the rate of incident neoplasia was: Stage 0 = 1 case; rate/103 person-years = 0.03; 95%CI: 0.004-0.19; Stage I = 2 cases; rate/103 person-years = 0.34; 95%CI: 0.09-1.36; Stage II = 3 cases; rate/103 person-years = 1.48; 95%CI: 0.48-4.58; Stage III = 17 cases; rate/103 person-years = 19.1; 95%CI: 11.9-30.7; Stage IV = 5 cases; rate/103 person-years = 41.2; 95%CI: 17.2-99.3. Multivariate analysis including gender, age, H. pylori status, and OLGA stage at enrollment only disclosed OLGA stage as predictor of neoplastic progression (OLGA stage III: HR = 712.4, 95%CI = 92.543-5484.5; OLGA stage IV: HR = 1450.7, 95%CI = 166.7-12626.0). CONCLUSIONS Among 7436 patients, OLGA stages at the enrollment correlated significantly with different risk for gastric neoplasia. Based on the obtained results, gastritis staging is a critical adjunct in endoscopy follow-up protocols aimed at GC secondary prevention.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany.,Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Robert M Genta
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany.,Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Elisa Valentini
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Irene Coati
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Stefano Guzzinati
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Edoardo Savarino
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Manuel Zorzi
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Fabio Farinati
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| |
Collapse
|
40
|
Wu S, Zhang Y, Zhang L, Ma JL, Zhou T, Li ZX, Liu WD, Li WQ, You WC, Pan KF. Methylation and Expression of Nonclustered Protocadherins Encoding Genes and Risk of Precancerous Gastric Lesions in a High-Risk Population. Cancer Prev Res (Phila) 2018; 11:717-726. [PMID: 30213786 DOI: 10.1158/1940-6207.capr-18-0119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/01/2018] [Accepted: 08/29/2018] [Indexed: 11/16/2022]
Abstract
Nonclustered protocadherins (PCDH) family is a group of cell-cell adhesion molecules. We have found differentially methylated genes in the nonclustered PCDHs family associated with Helicobacter pylori (H. pylori) infection in prior genome-wide methylation analysis. To further investigate the methylation and expression of nonclustered PCDHs encoding genes in H. pylori--related gastric carcinogenesis process, four candidate genes including PCDH7, PCDH10, PCDH17, and PCDH20 were selected, which were reported to be tumor suppressors for digestive cancers. A total of 747 participants with a spectrum of gastric lesions were enrolled from a high-risk population of gastric cancer. Promoter methylation levels of four genes were significantly higher in H. pylori-positive subjects than the negative group (all P < 0.001). Elevated methylation levels of PCDH10 and PCDH17 were observed with the increasing severity of gastric lesions (both P trend < 0.001). In the protein expression analysis, PCDH17 expression was inversely associated with gastric lesions; the OR [95% confidence interval (CI)] was 0.49 (0.26-0.95) for chronic atrophic gastritis (CAG), 0.31 (0.15-0.63) for intestinal metaplasia, and 0.38 (0.19-0.75) for indefinite dysplasia and dysplasia, compared with superficial gastritis. In addition, PCDH10 expression was significantly lower in CAG (OR, 0.40; 95% CI, 0.24-0.68). The inverse association between methylation and protein expression of PCDH10 and PCDH17 was further supported when we explored the methylation and mRNA expression in The Cancer Genome Atlas database (all P < 0.001). Our study found elevated promoter methylation and decreased expression of PCDH10 and PCDH17 in advanced gastric lesions, suggesting that elevated PCDH10 and PCDH17 methylation may be an early event in gastric carcinogenesis. Cancer Prev Res; 11(11); 717-26. ©2018 AACR.
Collapse
Affiliation(s)
- Si Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Yang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Lian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Jun-Ling Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Tong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Zhe-Xuan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Wei-Dong Liu
- Linqu Public Health Bureau, Linqu, Shandong, People's Republic of China
| | - Wen-Qing Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Wei-Cheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China.
| | - Kai-Feng Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China.
| |
Collapse
|
41
|
Epplein M, Butt J, Zhang Y, Hendrix LH, Abnet CC, Murphy G, Zheng W, Shu XO, Tsugane S, Qiao YL, Taylor PR, Shimazu T, Yoo KY, Park SK, Kim J, Jee SH, Waterboer T, Pawlita M, You WC, Pan KF. Validation of a Blood Biomarker for Identification of Individuals at High Risk for Gastric Cancer. Cancer Epidemiol Biomarkers Prev 2018; 27:1472-1479. [PMID: 30158280 DOI: 10.1158/1055-9965.epi-18-0582] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/11/2018] [Accepted: 08/23/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Helicobacter pylori is the leading cause of gastric cancer, yet the majority of infected individuals will not develop neoplasia. Previously, we developed and replicated serologic H. pylori biomarkers for gastric cancer risk among prospective cohorts in East Asia and now seek to validate the performance of these biomarkers in identifying individuals with premalignant lesions. METHODS This cross-sectional study included 1,402 individuals from Linqu County screened by upper endoscopy. H. pylori protein-specific antibody levels were assessed using multiplex serology. Multivariable-adjusted logistic regression models were used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for prevalent intestinal metaplasia, indefinite dysplasia, or dysplasia, compared with superficial or mild atrophic gastritis. RESULTS Compared with individuals seronegative to Omp and HP0305, individuals seropositive to both were seven times more likely to have precancerous lesions (OR, 7.43; 95% CI, 5.59-9.88). A classification model for precancerous lesions that includes age, smoking, and seropositivity to H. pylori, Omp, and HP0305 resulted in an area under the curve (AUC) of 0.751 (95% CI, 0.725-0.777), which is significantly better than the same model, including the established gastric cancer risk factor CagA (AUC, 0.718; 95% CI, 0.691-0.746, P difference = 0.0002). CONCLUSIONS The present study of prevalent precancerous gastric lesions provides support for two new serum biomarkers of gastric cancer risk, Omp and HP 0305. IMPACT Our results support further research into the serological biomarkers Omp and HP0305 as possible improvements over the established virulence marker CagA for identifying individuals with precancerous lesions in East Asia.
Collapse
Affiliation(s)
| | - Julia Butt
- German Cancer Research Center, Heidelberg, Germany
| | - Yang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | | | | | - Gwen Murphy
- National Cancer Institute, Rockville, Maryland
| | - Wei Zheng
- Vanderbilt University, Nashville, Tennessee
| | | | | | - You-Lin Qiao
- Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | - Sue K Park
- Seoul National University, Seoul, Republic of Korea
| | - Jeongseon Kim
- National Cancer Center of Korea, Gyeonggi-do, Republic of Korea
| | - Sun Ha Jee
- Yonsei University, Seoul, Republic of Korea
| | | | | | - Wei-Cheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Kai-Feng Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
42
|
Sobrino-Cossío S, Abdo Francis J, Emura F, Galvis-García E, Márquez Rocha M, Mateos-Pérez G, González-Sánchez C, Uedo N. Efficacy of narrow-band imaging for detecting intestinal metaplasia in adult patients with symptoms of dyspepsia. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO (ENGLISH EDITION) 2018. [DOI: 10.1016/j.rgmxen.2018.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
43
|
Mera RM, Bravo LE, Camargo MC, Bravo JC, Delgado AG, Romero-Gallo J, Yepez MC, Realpe JL, Schneider BG, Morgan DR, Peek RM, Correa P, Wilson KT, Piazuelo MB. Dynamics of Helicobacter pylori infection as a determinant of progression of gastric precancerous lesions: 16-year follow-up of an eradication trial. Gut 2018; 67:1239-1246. [PMID: 28647684 PMCID: PMC5742304 DOI: 10.1136/gutjnl-2016-311685] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/17/2017] [Accepted: 04/30/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate the long-term effect of cumulative time exposed to Helicobacter pylori infection on the progression of gastric lesions. DESIGN 795 adults with precancerous gastric lesions were randomised to receive anti-H. pylori treatment at baseline. Gastric biopsies were obtained at baseline and at 3, 6, 12 and 16 years. A total of 456 individuals attended the 16-year visit. Cumulative time of H. pylori exposure was calculated as the number of years infected during follow-up. Multivariable logistic regression models were used to estimate the risk of progression to a more advanced diagnosis (versus no change/regression) as well as gastric cancer risk by intestinal metaplasia (IM) subtype. For a more detailed analysis of progression, we also used a histopathology score assessing both severity and extension of the gastric lesions (range 1-6). The score difference between baseline and 16 years was modelled by generalised linear models. RESULTS Individuals who were continuously infected with H. pylori for 16 years had a higher probability of progression to a more advanced diagnosis than those who cleared the infection and remained negative after baseline (p=0.001). Incomplete-type IM was associated with higher risk of progression to cancer than complete-type (OR, 11.3; 95% CI 1.4 to 91.4). The average histopathology score increased by 0.20 units/year (95% CI 0.12 to 0.28) among individuals continuously infected with H. pylori. The effect of cumulative time of infection on progression in the histopathology score was significantly higher for individuals with atrophy (without IM) than for individuals with IM (p<0.001). CONCLUSIONS Long-term exposure to H. pylori infection was associated with progression of precancerous lesions. Individuals infected with H. pylori with these lesions may benefit from eradication, particularly those with atrophic gastritis without IM. Incomplete-type IM may be a useful marker for the identification of individuals at higher risk for cancer.
Collapse
Affiliation(s)
- Robertino M Mera
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Luis E Bravo
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Colombia
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Juan C Bravo
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Colombia
| | - Alberto G Delgado
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Judith Romero-Gallo
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maria C Yepez
- Centro de Estudios en Salud, Universidad de Nariño, Pasto, Colombia
| | - José L Realpe
- Centro de Estudios en Salud, Universidad de Nariño, Pasto, Colombia
| | - Barbara G Schneider
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Douglas R Morgan
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Richard M Peek
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Pelayo Correa
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Keith T Wilson
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - M Blanca Piazuelo
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
44
|
Sobrino-Cossío S, Abdo Francis J, Emura F, Galvis-García E, Márquez Rocha M, Mateos-Pérez G, González-Sánchez C, Uedo N. La eficacia de la imagen de banda estrecha para la detección de metaplasia intestinal en pacientes adultos con síntomas de dispepsia. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2018; 83:245-252. [PMID: 29449092 DOI: 10.1016/j.rgmx.2017.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/13/2017] [Accepted: 08/25/2017] [Indexed: 12/20/2022]
|
45
|
Fang JY, Du YQ, Liu WZ, Ren JL, Li YQ, Chen XY, Lv NH, Chen YX, Lv B. Chinese consensus on chronic gastritis (2017, Shanghai). J Dig Dis 2018; 19:182-203. [PMID: 29573173 DOI: 10.1111/1751-2980.12593] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
46
|
Metabolomic analysis reveals altered metabolic pathways in a rat model of gastric carcinogenesis. Oncotarget 2018; 7:60053-60073. [PMID: 27527852 PMCID: PMC5312368 DOI: 10.18632/oncotarget.11049] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 07/07/2016] [Indexed: 12/27/2022] Open
Abstract
Gastric cancer (GC) is one of the most malignant tumors with a poor prognosis. Alterations in metabolic pathways are inextricably linked to GC progression. However, the underlying molecular mechanisms remain elusive. We performed NMR-based metabolomic analysis of sera derived from a rat model of gastric carcinogenesis, revealed significantly altered metabolic pathways correlated with the progression of gastric carcinogenesis. Rats were histologically classified into four pathological groups (gastritis, GS; low-grade gastric dysplasia, LGD; high-grade gastric dysplasia, HGD; GC) and the normal control group (CON). The metabolic profiles of the five groups were clearly distinguished from each other. Furthermore, significant inter-metabolite correlations were extracted and used to reconstruct perturbed metabolic networks associated with the four pathological stages compared with the normal stage. Then, significantly altered metabolic pathways were identified by pathway analysis. Our results showed that oxidative stress-related metabolic pathways, choline phosphorylation and fatty acid degradation were continually disturbed during gastric carcinogenesis. Moreover, amino acid metabolism was perturbed dramatically in gastric dysplasia and GC. The GC stage showed more changed metabolite levels and more altered metabolic pathways. Two activated pathways (glycolysis; glycine, serine and threonine metabolism) substantially contributed to the metabolic alterations in GC. These results lay the basis for addressing the molecular mechanisms underlying gastric carcinogenesis and extend our understanding of GC progression.
Collapse
|
47
|
Rentien AL, Lévy M, Copie-Bergman C, Gagniere C, Dupuis J, Le Baleur Y, Belhadj K, Sobhani I, Haioun C, Delchier JC, Amiot A. Long-term course of precancerous lesions arising in patients with gastric MALT lymphoma. Dig Liver Dis 2018; 50:181-188. [PMID: 29102522 DOI: 10.1016/j.dld.2017.10.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS To evaluate the prevalence and the long-term course of gastric precancerous lesions in patients with GML. PATIENTS AND METHODS In this retrospective single-centre study, we included 179 patients with GML, 70 with gastric diffuse large B-cell lymphoma (GDLBCL) and 152 with Helicobacter pylori-associated gastritis (HpG), from January 1995 to January 2014. The presence of atrophic gastritis, intestinal metaplasia and neoplastic lesion has been assessed at baseline and during follow-up. RESULTS Atrophic gastritis was more frequent in the GML group whereas there was also a trend for intestinal metaplasia and gastric dysplasia. In patients with GML, atrophic gastritis, intestinal metaplasia and gastric dysplasia were more frequent in the GML area than in other part of the stomach. During follow-up, the prevalence of atrophic gastritis remained stable overtime whereas intestinal metaplasia and dysplasia tend to increase overtime. In multivariate analysis, the occurrence of dysplasia or carcinoma was associated with the presence of intestinal metaplasia at baseline and male gender. CONCLUSION GML is associated with gastric precancerous lesion to a higher extent than GDLBCL and HpG. Those precancerous lesions do not regress despite achievement of complete remission of GML and tend to increase overtime.
Collapse
Affiliation(s)
- Anne-Laure Rentien
- Department of Gastroenterology, Henri Mondor University Hospital, APHP, Creteil F-94010, France; Paris Est-Creteil University (UPEC), Creteil F-94010, France; EC2M3-EA7375 Unit, Creteil, France
| | - Michaël Lévy
- Department of Gastroenterology, Henri Mondor University Hospital, APHP, Creteil F-94010, France
| | - Christiane Copie-Bergman
- Paris Est-Creteil University (UPEC), Creteil F-94010, France; Department of Pathology, Henri Mondor University Hospital, APHP, Creteil F-94010, France; Unit UMR-S 955, INSERM, Creteil F-94010, France
| | - Charlotte Gagniere
- Department of Gastroenterology, Henri Mondor University Hospital, APHP, Creteil F-94010, France; Paris Est-Creteil University (UPEC), Creteil F-94010, France; EC2M3-EA7375 Unit, Creteil, France
| | - Jehan Dupuis
- Lymphoid Malignancies Unit, Henri Mondor University Hospital, APHP, Creteil F-94010, France
| | - Yann Le Baleur
- Department of Gastroenterology, Henri Mondor University Hospital, APHP, Creteil F-94010, France
| | - Karim Belhadj
- Lymphoid Malignancies Unit, Henri Mondor University Hospital, APHP, Creteil F-94010, France
| | - Iradj Sobhani
- Department of Gastroenterology, Henri Mondor University Hospital, APHP, Creteil F-94010, France
| | - Corinne Haioun
- Paris Est-Creteil University (UPEC), Creteil F-94010, France; Unit UMR-S 955, INSERM, Creteil F-94010, France; Lymphoid Malignancies Unit, Henri Mondor University Hospital, APHP, Creteil F-94010, France
| | - Jean-Charles Delchier
- Department of Gastroenterology, Henri Mondor University Hospital, APHP, Creteil F-94010, France; Paris Est-Creteil University (UPEC), Creteil F-94010, France
| | - Aurelien Amiot
- Department of Gastroenterology, Henri Mondor University Hospital, APHP, Creteil F-94010, France; Paris Est-Creteil University (UPEC), Creteil F-94010, France; EC2M3-EA7375 Unit, Creteil, France.
| |
Collapse
|
48
|
Genome-wide DNA methylation profiles altered by Helicobacter pylori in gastric mucosa and blood leukocyte DNA. Oncotarget 2018; 7:37132-37144. [PMID: 27206798 PMCID: PMC5095064 DOI: 10.18632/oncotarget.9469] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/24/2016] [Indexed: 01/28/2023] Open
Abstract
Purpose To investigate Helicobacter pylori (H.pylori) associated genome-wide aberrant methylation patterns in gastric mucosa and blood leukocyte DNA, a population-based study was conducted in Linqu County. Results A total of 3000 and 386 CpGs were differentially methylated after successful H.pylori eradication in gastric mucosa and blood leukocyte DNA respectively, and 17 were the same alteration trend in the both tissues. The differentially methylated CpGs were located more frequently in promoters or CpG islands for gastric mucosa and gene body or open sea for blood leukocyte DNA. In eradicated gastric mucosa, the hypermethylated CpGs were enriched across inflammatory pathways, while the hypomethylated CpGs in tube morphogenesis, development and so on. The final validation found lower SPI1, PRIC285 and S1PR4 methylation levels in H.pylori positive subjects by case-control comparison, and increased methylation levels in H.pylori eradicated gastric mucosa by self-comparison. The Cancer Genome Atlas (TCGA) database analysis suggested that the up-regulation of the three genes by hypomethylation might be associated with gastric carcinogenesis. Experimental Design Infinium HumanMethylation 450K BeadChip was used to compare methylation profiles prior to and after eradication treatment. The methylation levels of identified candidate differentially methylated genes before and after H.pylori eradication were further validated by two stages (Stage I: self-comparison of 16 subjects before and after anti-H.pylori treatment; Stage II: case-control comparison of 25 H.pylori positive and 25 negative subjects and self-comparison of 50 anti-H.pylori treated subjects). Conclusions Novel H.pylori associated aberrant methylated genes were identified across the whole genome both in gastric mucosa and blood leukocyte DNA.
Collapse
|
49
|
Neoplastic Lesions of Gastric Adenocarcinoma and Proximal Polyposis Syndrome (GAPPS) Are Gastric Phenotype. Am J Surg Pathol 2017; 42:1-8. [PMID: 29112017 DOI: 10.1097/pas.0000000000000924] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neoplastic lesions of gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS) are gastric phenotype. GAPPS was reported in 2011 as a new autosomal dominant gastric polyposis syndrome characterized by involvement of the gastric body/fundus with sparing of the antrum by multiple polyps, reported to be primarily fundic gland polyps (FGPs), with progression to dysplasia and adenocarcinoma of intestinal type. Our series consists of 51 endoscopic biopsies and 5 gastrectomy specimens from 25 patients belonging to a previously defined GAPPS family. Slides were reviewed and further stains performed. Endoscopy was abnormal in 15 of the 25 patients: carpeting polyposis of the gastric body and fundus in 14 and a gastric mass without polyposis in one. The most common polypoid lesion (seen in 12 patients) was a disorganized proliferation of specialized/oxyntic glands high up in the mucosa involving the attenuated foveolar region around the gastric pits, which we have termed "hyperproliferative aberrant pits". Well developed FGP were seen in 10 patients. Established neoplastic lesions seen in 9 patients were: (1) discrete gastric adenomas, (2) multifocal "flat" dysplasia in the setting of hyperproliferative aberrant pits +/- FGPs, (3) adenomatous tissue associated with adenocarcinoma. All cases of dysplasia were of gastric phenotype based on morphology and mucin immunohistochemistry. IN CONCLUSION (1) the spectrum of gastric pathology associated with GAPPS is wider than previously reported, (2) the earliest microscopic clue is the finding of hyperproliferative aberrant pits, and (3) the dysplasia is gastric phenotype and the subsequent adenocarcinoma may follow the gastric pathway of carcinogenesis.
Collapse
|
50
|
Rugge M, Genta RM, Di Mario F, El-Omar EM, El-Serag HB, Fassan M, Hunt RH, Kuipers EJ, Malfertheiner P, Sugano K, Graham DY. Gastric Cancer as Preventable Disease. Clin Gastroenterol Hepatol 2017; 15:1833-1843. [PMID: 28532700 DOI: 10.1016/j.cgh.2017.05.023] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023]
Abstract
Gastric cancer, 1 of the 5 most common causes of cancer death, is associated with a 5-year overall survival rate less than 30%. A minority of cancers occurs as part of syndromic diseases; more than 90% of adenocarcinomas are considered as the ultimate consequence of a longstanding mucosal inflammation. Helicobacter pylori infection is the leading etiology of non-self-limiting gastritis, which may result in atrophy of the gastric mucosa and impaired acid secretion. Gastric atrophy establishes a field of cancerization prone to further molecular and phenotypic changes, possibly resulting in cancer growth. This well-understood natural history provides the clinicopathologic rationale for primary and secondary cancer prevention strategies. A large body of evidence demonstrates that combined primary (H pylori eradication) and secondary (mainly endoscopy) prevention efforts may prevent or limit the progression of gastric oncogenesis. This approach, which is tailored to different country-specific gastric cancer incidence, socioeconomic, and cultural factors, requires that the complementary competences of gastroenterologists, oncologists, and pathologists be amalgamated into a common strategy of health policy.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), University of Padua, Padua, Italy; Veneto Tumor Registry, Veneto Region, Padua, Italy.
| | - Robert M Genta
- Miraca Life Sciences Research Institute, Irving, and Departments of Pathology and Medicine, Baylor College of Medicine, Houston, Texas
| | - Francesco Di Mario
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | - Emad M El-Omar
- St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Hashem B El-Serag
- Department of Medicine, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Richard H Hunt
- Division of Gastroenterology, Department of Medicine and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Ernst J Kuipers
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Kentaro Sugano
- Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - David Y Graham
- Department of Medicine, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|