1
|
Inoue-Choi M, Freedman ND, Etemadi A, Hashemian M, Brennan P, Roshandel G, Poustchi H, Boffetta P, Kamangar F, Amiriani T, Norouzi A, Dawsey S, Malekzadeh R, Abnet CC. One-carbon metabolism biomarkers and upper gastrointestinal cancer in the Golestan Cohort Study. Int J Cancer 2024; 155:1944-1957. [PMID: 39109892 PMCID: PMC11449644 DOI: 10.1002/ijc.35115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 10/04/2024]
Abstract
Incidence of esophageal and gastric cancer has been linked to low B-vitamin status. We conducted matched nested case-control studies of incident esophageal squamous cell carcinoma (ESCC; 340 case-control pairs) and gastric cancer (GC; 352 case-control pairs) within the Golestan Cohort Study. The primary exposure was plasma biomarkers: riboflavin and flavin mononucleotide (FMN) (vitamin B2), pyridoxal phosphate (PLP) (B6), cobalamin (B12), para-aminobenzoylglutamate (pABG) (folate), and total homocysteine (tHcy); and indicators for deficiency: 3-hydroxykyurenine-ratio (HK-r for vitamin B6) and methylmalonic acid (MMA for B12). We estimated odds ratios (ORs) and 95% confidence intervals (CIs) using conditional logistic regression adjusting for matching factors and potential confounders. High proportions of participants had low B-vitamin and high tHcy levels. None of the measured vitamin B levels was associated with the risk of ESCC and GC, but elevated level of MMA was marginally associated with ESCC (OR = 1.42, 95% CI = 0.99-2.04) and associated with GC (OR = 1.53, 95% CI = 1.05-2.22). Risk of GC was higher for the highest versus lowest quartile of HK-r (OR = 1.95, 95%CI = 1.19-3.21) and for elevated versus non-elevated HK-r level (OR = 1.59, 95% CI = 1.13-2.25). Risk of ESCC (OR = 2.81, 95% CI = 1.54-5.13) and gastric cancer (OR = 2.09, 95%CI = 1.17-3.73) was higher for the highest versus lowest quartile of tHcy. In conclusion, insufficient vitamin B12 was associated with higher risk of ESCC and GC, and insufficient vitamin B6 status was associated with higher risk of GC in this population with prevalent low plasma B-vitamin status. Higher level of tHcy, a global indicator of OCM function, was associated with higher risk of ESCC and GC.
Collapse
Affiliation(s)
- Maki Inoue-Choi
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Neal D Freedman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Arash Etemadi
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Maryam Hashemian
- Epidemiology and Community Health Branch, Division of Intramural Research, National Heart, Lung, Blood Institute, Bethesda, Maryland, USA
| | - Paul Brennan
- Genetic Section, Genomic Epidemiology, International Agency for Research on Cancer, Lyon, France
| | - Gholamreza Roshandel
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Golestan, Iran
| | - Hossein Poustchi
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Golestan, Iran
| | - Paolo Boffetta
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Farin Kamangar
- Department of Biology, School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, Maryland, USA
| | - Taghi Amiriani
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Golestan, Iran
| | - Alireza Norouzi
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Golestan, Iran
| | - Sandy Dawsey
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Reza Malekzadeh
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Christian C Abnet
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Li M, Gao N, Wang SL, Guo YF, Liu Z. Hotspots and trends of risk factors in gastric cancer: A visualization and bibliometric analysis. World J Gastrointest Oncol 2024; 16:2200-2218. [PMID: 38764808 PMCID: PMC11099465 DOI: 10.4251/wjgo.v16.i5.2200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/08/2024] [Accepted: 03/11/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The lack of specific symptoms of gastric cancer (GC) causes great challenges in its early diagnosis. Thus it is essential to identify the risk factors for early diagnosis and treatment of GC and to improve the survival rates. AIM To assist physicians in identifying changes in the output of publications and research hotspots related to risk factors for GC, constructing a list of key risk factors, and providing a reference for early identification of patients at high risk for GC. METHODS Research articles on risk factors for GC were searched in the Web of Science core collection, and relevant information was extracted after screening. The literature was analyzed using Microsoft Excel 2019, CiteSpace V, and VOSviewer 1.6.18. RESULTS A total of 2514 papers from 72 countries and 2507 research institutions were retrieved. China (n = 1061), National Cancer Center (n = 138), and Shoichiro Tsugane (n = 36) were the most productive country, institution, or author, respectively. The research hotspots in the study of risk factors for GC are summarized in four areas, namely: Helicobacter pylori (H. pylori) infection, single nucleotide polymorphism, bio-diagnostic markers, and GC risk prediction models. CONCLUSION In this study, we found that H. pylori infection is the most significant risk factor for GC; single-nucleotide polymorphism (SNP) is the most dominant genetic factor for GC; bio-diagnostic markers are the most promising diagnostic modality for GC. GC risk prediction models are the latest current research hotspot. We conclude that the most important risk factors for the development of GC are H. pylori infection, SNP, smoking, diet, and alcohol.
Collapse
Affiliation(s)
- Meng Li
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ning Gao
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shao-Li Wang
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yu-Feng Guo
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zhen Liu
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
3
|
Bhattacharjee A, Sahoo OS, Sarkar A, Bhattacharya S, Chowdhury R, Kar S, Mukherjee O. Infiltration to infection: key virulence players of Helicobacter pylori pathogenicity. Infection 2024; 52:345-384. [PMID: 38270780 DOI: 10.1007/s15010-023-02159-9] [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: 09/08/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
PURPOSE This study aims to comprehensively review the multifaceted factors underlying the successful colonization and infection process of Helicobacter pylori (H. pylori), a prominent Gram-negative pathogen in humans. The focus is on elucidating the functions, mechanisms, genetic regulation, and potential cross-interactions of these elements. METHODS Employing a literature review approach, this study examines the intricate interactions between H. pylori and its host. It delves into virulence factors like VacA, CagA, DupA, Urease, along with phase variable genes, such as babA, babC, hopZ, etc., giving insights about the bacterial perspective of the infection The association of these factors with the infection has also been added in the form of statistical data via Funnel and Forest plots, citing the potential of the virulence and also adding an aspect of geographical biasness to the virulence factors. The biochemical characteristics and clinical relevance of these factors and their effects on host cells are individually examined, both comprehensively and statistically. RESULTS H. pylori is a Gram-negative, spiral bacterium that successfully colonises the stomach of more than half of the world's population, causing peptic ulcers, gastric cancer, MALT lymphoma, and other gastro-duodenal disorders. The clinical outcomes of H. pylori infection are influenced by a complex interplay between virulence factors and phase variable genes produced by the infecting strain and the host genetic background. A meta-analysis of the prevalence of all the major virulence factors has also been appended. CONCLUSION This study illuminates the diverse elements contributing to H. pylori's colonization and infection. The interplay between virulence factors, phase variable genes, and host genetics determines the outcome of the infection. Despite biochemical insights into many factors, their comprehensive regulation remains an understudied area. By offering a panoramic view of these factors and their functions, this study enhances understanding of the bacterium's perspective, i.e. H. pylori's journey from infiltration to successful establishment within the host's stomach.
Collapse
Affiliation(s)
- Arghyadeep Bhattacharjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
- Department of Microbiology, Kingston College of Science, Beruanpukuria, Barasat, West Bengal, 700219, India
| | - Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
| | - Ahana Sarkar
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
| | - Saurabh Bhattacharya
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, P.O.B. 12272, 9112001, Jerusalem, Israel
| | - Rukhsana Chowdhury
- School of Biological Sciences, RKM Vivekananda Educational and Research Institute Narendrapur, Kolkata, India
| | - Samarjit Kar
- Department of Mathematics, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
| | - Oindrilla Mukherjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India.
| |
Collapse
|
4
|
Li ZX, Bronny K, Formichella L, Mejías-Luque R, Burrell T, Macke L, Lang U, Vasapolli R, Hysenaj O, Stallforth I, Vieth M, You WC, Zhang Y, Suerbaum S, Schulz C, Pan KF, Gerhard M. A multiserological line assay to potentially discriminate current from past Helicobacter pylori infection. Clin Microbiol Infect 2024; 30:114-121. [PMID: 37827383 DOI: 10.1016/j.cmi.2023.10.006] [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: 07/03/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
OBJECTIVES Early diagnosis is important in controlling Helicobacter pylori-induced gastritis and progression to gastric malignancy. Serological testing is an efficient non-invasive diagnostic method, but currently does not allow differentiation between active and past infections. To fill this diagnostic gap we investigated the diagnostic value of a panel of ten H. pylori-specific antibodies in individuals with different H. pylori infection status within a German population. METHODS We used the recomLine Helicobacter IgG 2.0 immunoblotting assay to analyse ten H. pylori-specific antibodies in serum samples collected from 1108 volunteers. From these, 788 samples were used to build exposure and infection status models and 320 samples for model validation. H. pylori infection status was verified by histological examination. We applied logistic regression to select antibodies correlated to infection status and developed, with independent validation, discriminating models and risk scores. Receiving operating characteristic analysis was performed to assess the accuracy of the discriminating models. RESULTS Antibody reactivity against cytotoxin-associated gene A (CagA), H. pylori chaperone (GroEL), and hook-associated protein 2 homologue (FliD) was independently associated with the risk of H. pylori exposure with ORs and 95% CIs of 99.24 (46.50-211.80), 46.17 (17.45-122.17), and 22.16 (8.46-55.04), respectively. A risk score comprising these three selected antibodies differentiated currently H. pylori infected or eradicated participants from negatives with an area under the curve of 0.976 (95% CI: 0.965-0.987) (Model 1). Seropositivity for vacuolating cytotoxin A (VacA), GroEL, FliD, H. pylori adhesin A (HpaA), and γ-glutamyl transpeptidase (gGT) was associated with a current infection with an area under the curve of 0.870 (95% CI: 0.837-0.903), which may help discriminate currently infected patients from eradicated ones (Model 2). DISCUSSION The recomLine assay is sensitive and specific in determining H. pylori infection and eradication status and thus represents a valuable tool in the management of H. pylori infection.
Collapse
Affiliation(s)
- Zhe-Xuan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China; PYLOTUM Key Joint Laboratory for Upper GI Cancer, Technische Universität München, Munich, Germany, Peking University Cancer Hospital & Institute, Beijing, China; Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany; Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Kathrin Bronny
- Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Luca Formichella
- Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Raquel Mejías-Luque
- PYLOTUM Key Joint Laboratory for Upper GI Cancer, Technische Universität München, Munich, Germany, Peking University Cancer Hospital & Institute, Beijing, China; Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Teresa Burrell
- Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Lukas Macke
- Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany; Medical Department II, University Hospital, LMU, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ulrich Lang
- Medical Department II, University Hospital, LMU, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Riccardo Vasapolli
- Medical Department II, University Hospital, LMU, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Olsi Hysenaj
- Medical Department II, University Hospital, LMU, Munich, Germany
| | - Ingrid Stallforth
- Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Michael Vieth
- PYLOTUM Key Joint Laboratory for Upper GI Cancer, Technische Universität München, Munich, Germany, Peking University Cancer Hospital & Institute, Beijing, China; Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Wei-Cheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China; PYLOTUM Key Joint Laboratory for Upper GI Cancer, Technische Universität München, Munich, Germany, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China; PYLOTUM Key Joint Laboratory for Upper GI Cancer, Technische Universität München, Munich, Germany, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sebastian Suerbaum
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Max von Pettenkofer Institute, Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany; National Reference Center for Helicobacter Pylori, Munich, Germany
| | - Christian Schulz
- Medical Department II, University Hospital, LMU, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Kai-Feng Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China; PYLOTUM Key Joint Laboratory for Upper GI Cancer, Technische Universität München, Munich, Germany, Peking University Cancer Hospital & Institute, Beijing, China
| | - Markus Gerhard
- PYLOTUM Key Joint Laboratory for Upper GI Cancer, Technische Universität München, Munich, Germany, Peking University Cancer Hospital & Institute, Beijing, China; Institute of Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
5
|
Hua Z, Xu L, Zhu J, Xiao L, Lu B, Wu J, Wu Z, Zhou Q, Zhang J. Helicobacter pylori infection altered gastric microbiota in patients with chronic gastritis. Front Cell Infect Microbiol 2023; 13:1221433. [PMID: 37662018 PMCID: PMC10470091 DOI: 10.3389/fcimb.2023.1221433] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Objective The present study aims to investigate the effect of Helicobacter pylori (Hp) infection on gastric mucosal microbiota in patients with chronic gastritis. Methods Here recruited a population of 193 patients with both chronic gastritis and positive rapid urease, including 124 patients with chronic atrophic gastritis (CAG) and 69 patients with chronic non-atrophic gastritis (nCAG). Immunoblotting was used to detect four serum Hp antibodies (UreA, UreB, VacA and CagA) to determine the types of virulent Hp-I and avirulent Hp-II infections. Gastric microbiota was profiled by 16S rRNA gene V3-V4 region, and R software was used to present the relationship between the microbial characteristics and the type of Hp infection. Results In the stomach of patients with Hp-positive gastritis, the dominant gastric bacterial genera included Ralstonia (23.94%), Helicobacter (20.28%), Pseudonocardia (9.99%), Mesorhizobium (9.21%), Bradyrhizobium (5.05%), and Labrys (4.75%). The proportion of Hp-I infection was significantly higher in CAG patients (91.1%) than in nCAG patients (71.0%) (P < 0.001). The gastric microbiota richness index (observed OTUs, Chao) was significantly lower in CAG patients than in nCAG patients (P <0.05). Compared with avirulent Hp-II infection, virulent Hp-I infection significantly decreased the Shannon index in CAG patients (P <0.05). In nCAG patients, Hp-I infected patients had lower abundances of several dominant gastric bacteria (Aliidiomarina, Reyranella, Halomonas, Pseudomonas, Acidovorax) than Hp-II infected patients. Meanwhile, in CAG patients, Hp-I infected patients occupied lower abundances of several dominant oral bacteria (Neisseria, Staphylococcus and Haemophilus) than Hp-II infected patients. In addition, bile reflux significantly promoted the colonization of dominant oral microbiota (Veillonella, Prevotella 7 and Rothia) in the stomach of CAG patients. There was no significant symbiotic relationship between Helicobacter bacteria and non-Helicobacter bacteria in the stomach of nCAG patients, while Helicobacter bacteria distinctly linked with the non-Helicobacter bacteria (Pseudolabrys, Ralstonia, Bradyrhizobium, Mesorhizobium and Variovorax) in CAG patients. Conclusions Virulent Hp infection alters the gastric microbiota, reduces microbial diversity, and enhances the symbiotic relationship between the Helicobacter bacteria and non-Helicobacter bacteria in patients with chronic gastritis. The data provides new evidence for treating Hp infection by improving the gastric microbiota.
Collapse
Affiliation(s)
- Zhaolai Hua
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
- Institute of Tumor Prevention and Control, People’s Hospital of Yangzhong City, Yangzhong, China
| | - Le Xu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiahui Zhu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ling Xiao
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Lu
- Department of Oncology, People’s Hospital of Yangzhong City, Yangzhong, China
| | - Jianping Wu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenfeng Wu
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qihai Zhou
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
| | - Junfeng Zhang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Yao P, Kartsonaki C, Butt J, Jeske R, de Martel C, Plummer M, Guo Y, Clark S, Walters RG, Chen Y, Avery D, Lv J, Yu C, Wang H, Hill M, Peto R, Li L, Waterboer T, Chen Z, Millwood IY, Yang L. Helicobacter pylori multiplex serology and risk of non-cardia and cardia gastric cancer: a case-cohort study and meta-analysis. Int J Epidemiol 2023; 52:1197-1208. [PMID: 36913255 PMCID: PMC10396410 DOI: 10.1093/ije/dyad007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/19/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Helicobacter pylori infection is a major cause of non-cardia gastric cancer (NCGC), but uncertainty remains about the associations between sero-positivity to different H. pylori antigens and risk of NCGC and cardia gastric cancer (CGC) in different populations. METHODS A case-cohort study in China included ∼500 each of incident NCGC and CGC cases and ∼2000 subcohort participants. Sero-positivity to 12 H. pylori antigens was measured in baseline plasma samples using a multiplex assay. Hazard ratios (HRs) of NCGC and CGC for each marker were estimated using Cox regression. These were further meta-analysed with studies using same assay. RESULTS In the subcohort, sero-positivity for 12 H. pylori antigens varied from 11.4% (HpaA) to 70.8% (CagA). Overall, 10 antigens showed significant associations with risk of NCGC (adjusted HRs: 1.33 to 4.15), and four antigens with CGC (HRs: 1.50 to 2.34). After simultaneous adjustment for other antigens, positive associations remained significant for NCGC (CagA, HP1564, HP0305) and CGC (CagA, HP1564, HyuA). Compared with CagA sero-positive only individuals, those who were positive for all three antigens had an adjusted HR of 5.59 (95% CI 4.68-6.66) for NCGC and 2.17 (95% CI 1.54-3.05) for CGC. In the meta-analysis of NCGC, the pooled relative risk for CagA was 2.96 (95% CI 2.58-3.41) [Europeans: 5.32 (95% CI 4.05-6.99); Asians: 2.41 (95% CI 2.05-2.83); Pheterogeneity<0.0001]. Similar pronounced population differences were also evident for GroEL, HP1564, HcpC and HP0305. In meta-analyses of CGC, two antigens (CagA, HP1564) were significantly associated with a higher risk in Asians but not Europeans. CONCLUSIONS Sero-positivity to several H. pylori antigens was significantly associated with an increased risk of NCGC and CGC, with varying effects between Asian and European populations.
Collapse
Affiliation(s)
- Pang Yao
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Julia Butt
- Infections and Cancer Epidemiology Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rima Jeske
- Infections and Cancer Epidemiology Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Catherine de Martel
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Martyn Plummer
- Department of Statistics, University of Warwick, Coventry, UK
| | - Yu Guo
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sarah Clark
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Center for Public Health and Epidemic Preparedness & Response, Peking University, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Center for Public Health and Epidemic Preparedness & Response, Peking University, Beijing, China
| | - Hao Wang
- NCDs Prevention and Control Department, Zhejiang CDC, Zhejiang, China
| | - Michael Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Center for Public Health and Epidemic Preparedness & Response, Peking University, Beijing, China
| | - Tim Waterboer
- Infections and Cancer Epidemiology Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Li S, Zhao W, Xia L, Kong L, Yang L. How Long Will It Take to Launch an Effective Helicobacter pylori Vaccine for Humans? Infect Drug Resist 2023; 16:3787-3805. [PMID: 37342435 PMCID: PMC10278649 DOI: 10.2147/idr.s412361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Helicobacter pylori infection often occurs in early childhood, and can last a lifetime if not treated with medication. H. pylori infection can also cause a variety of stomach diseases, which can only be treated with a combination of antibiotics. Combinations of antibiotics can cure H. pylori infection, but it is easy to relapse and develop drug resistance. Therefore, a vaccine is a promising strategy for prevention and therapy for the infection of H. pylori. After decades of research and development, there has been no appearance of any H. pylori vaccine reaching the market, unfortunately. This review summarizes the aspects of candidate antigens, immunoadjuvants, and delivery systems in the long journey of H. pylori vaccine research, and also introduces some clinical trials that have displayed encouraging or depressing results. Possible reasons for the inability of an H. pylori vaccine to be available over the counter are cautiously discussed and some propositions for the future of H. pylori vaccines are outlined.
Collapse
Affiliation(s)
- Songhui Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009People’s Republic of China
| | - Wenfeng Zhao
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009People’s Republic of China
| | - Lei Xia
- Bloomage Biotechnology Corporation Limited, Jinan, People’s Republic of China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009People’s Republic of China
| | - Lei Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009People’s Republic of China
| |
Collapse
|
8
|
El Hafa F, Wang T, Ndifor VM, Jin G. Association between Helicobacter pylori antibodies determined by multiplex serology and gastric cancer risk: A meta-analysis. Helicobacter 2022; 27:e12881. [PMID: 35212073 DOI: 10.1111/hel.12881] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/17/2022] [Accepted: 01/30/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Previous studies have reported the association between limited number of Helicobacter pylori (H. pylori) antigens and gastric cancer (GC) risk. The present study evaluated the association between serum antibodies against 15 different H. pylori proteins measured by using multiplex serology assay and GC risk. METHODS We searched PubMed databases, Embase, Web of Science, and Cochrane Library for relevant articles. A meta-analysis was used to pool studies and to estimate odds ratios (ORs) with 95% confidence intervals (95%CIs) of different H. pylori antigens associated with GC risk. Heterogeneity was investigated using Cochran's Q test and I-squared statistic. RESULTS Nine studies were identified, with a total of 3209 GC cases and 6964 controls. Five H. pylori virulence factors were significantly associated with non-cardia GC risk at p-value <0.0033 including: CagA (OR = 3.22, 95%CI: 2.10-4.94), HP0305 (OR = 1.72, 95%CI: 1.32-2.25), HyuA (OR = 1.42, 95%CI: 1.13-1.79), Omp (OR = 1.83, 95%CI: 1.30-2.58), and VacA (OR = 2.05, 95%CI: 1.67-2.52). However, none of the 15 antigens was associated with cardia GC risk. In subgroup analysis by ethnicity, we identified 7 antigens associated with the risk of non-cardia GC among East Asian while only two antigens were identified in European population. Nevertheless, CagA and GroEL showed a stronger association in Caucasian (CagA OR = 5.83, 95%CI: 3.31-10.26; GroEL OR = 3.66, 95%CI: 1.58-8.50) compared with East Asian (CagA OR = 2.20, 95% CI: 1.85-2.61; GroEL OR = 1.47, 95%CI: 1.29-1.68). CONCLUSIONS This study determined that H. pylori infection increases the risk of non-cardia GC with differential effects by its virulence factors and with different patterns among East Asian and European populations. These results advance the understanding of the effect of H. pylori on GC.
Collapse
Affiliation(s)
- Fadoua El Hafa
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Tianpei Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China.,Public Health Institute of Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Valerie Mbuhnwi Ndifor
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China.,Public Health Institute of Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Yang L, Kartsonaki C, Yao P, de Martel C, Plummer M, Chapman D, Guo Y, Clark S, Walters RG, Chen Y, Pei P, Lv J, Yu C, Jeske R, Waterboer T, Clifford GM, Franceschi S, Peto R, Hill M, Li L, Millwood IY, Chen Z. The relative and attributable risks of cardia and non-cardia gastric cancer associated with Helicobacter pylori infection in China: a case-cohort study. Lancet Public Health 2021; 6:e888-e896. [PMID: 34838195 PMCID: PMC8646857 DOI: 10.1016/s2468-2667(21)00164-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Helicobacter pylori infection is a major cause of non-cardia gastric cancer (NCGC), but its causal role in cardia gastric cancer (CGC) is unclear. Moreover, the reported magnitude of association with NCGC varies considerably, leading to uncertainty about population-based H pylori screening and eradication strategies in high-risk settings, particularly in China, where approximately half of all global gastric cancer cases occur. Our aim was to assess the associations of H pylori infection, both overall and for individual infection biomarkers, with the risks of NCGC and CGC in Chinese adults. METHODS A case-cohort study was done in adults from the prospective China Kadoorie Biobank study, aged 30-79 years from ten areas in China (Qingdao, Haikou, Harbin, Suzhou, Liuzhou, Henan, Sichuan, Hunan, Gansu, and Zhejiang), and included 500 incident NCGC cases, 437 incident CGC cases, and 500 subcohort participants who were cancer-free and alive within the first two years since enrolment in 2004-08. H pylori biomarkers were measured in stored baseline plasma samples using a sensitive immunoblot assay (HelicoBlot 2.1), with adapted criteria to define H pylori seropositivity. Cox regression was used to estimate adjusted hazard ratios (HRs) for NCGC and CGC associated with H pylori infection. These values were used to estimate the number of gastric cancer cases attributable to H pylori infection in China. FINDINGS Of the 512 715 adults enrolled in the China Kadoorie Biobank between June, 2004, and July, 2008, 500 incident NCGC cases, 437 incident CGC cases, and 500 subcohort participants were selected for analysis. The seroprevalence of H pylori was 94·4% (95% CI 92·4-96·4) in NGCG, 92·2% (89·7-94·7) in CGC, and 75·6% (71·8-79·4) in subcohort participants. H pylori infection was associated with adjusted HRs of 5·94 (95% CI 3·25-10·86) for NCGC and 3·06 (1·54-6·10) for CGC. Among the seven individual infection biomarkers, cytotoxin-associated antigen had the highest HRs for both NCGC (HR 4·41, 95% CI 2·60-7·50) and CGC (2·94, 1·53-5·68). In this population, 78·5% of NCGC and 62·1% of CGC cases could be attributable to H pylori infection. H pylori infection accounted for an estimated 339 955 cases of gastric cancer in China in 2018. INTERPRETATION Among Chinese adults, H pylori infection is common and is the cause of large numbers of gastric cancer cases. Population-based mass screening and the eradication of H pylori should be considered to reduce the burden of gastric cancer in high-risk settings. FUNDING Cancer Research UK, Wellcome Trust, UK Medical Research Council, British Heart Foundation, Kadoorie Charitable Foundation, National Key Research and Development Program of China, and National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Pang Yao
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Catherine de Martel
- Early Detection, Prevention, and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Martyn Plummer
- Department of Statistics, University of Warwick, Coventry, UK
| | - Daniel Chapman
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yu Guo
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China
| | - Sarah Clark
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Pei Pei
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| | - Rima Jeske
- Infections and Cancer Epidemiology Division, German Cancer Research Centre, Heidelberg, Germany
| | - Tim Waterboer
- Infections and Cancer Epidemiology Division, German Cancer Research Centre, Heidelberg, Germany
| | - Gary M Clifford
- Early Detection, Prevention, and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | | | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Michael Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| | - Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Jala I, Almanfaluthi ML, Laha T, Kanthawong S, Tangkawattana S, Saichua P, Suttiprapa S, Sripa B. Helicobacter pylori GroEL Seropositivity Is Associated with an Increased Risk of Opisthorchis viverrini-Associated Hepatobiliary Abnormalities and Cholangiocarcinoma. THE KOREAN JOURNAL OF PARASITOLOGY 2021; 59:363-368. [PMID: 34470087 PMCID: PMC8413853 DOI: 10.3347/kjp.2021.59.4.363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/02/2021] [Indexed: 11/23/2022]
Abstract
Despite the synergistic effect of Opisthorchis viverrini and Helicobacter pylori co-infection on pathogenesis of severe hepatobiliary abnormalities (HBA) including advanced periductal fibrosis and replace with cholangiocarcinoma (CCA) have been established, the immune response to H. pylori in O. viverrini infected population has never been explored. Hence, this study aimed to investigate the antibody responses to 2 immunogenic H. pylori proteins in O. viverrini-infected patients with HBA and CCA. The risk analysis by multinomial logistic regression revealed that GroEL seropositivity was associated with higher risks of hepatobiliary abnormalities and CCA with adjusted odds ratios (95% confidence intervals) of 2.11 (95% CI=1.20-3.71, P=0.008) and 2.13 (95% CI=1.21-3.75, P=0.009), respectively. These findings indicate that GroEL seropositivity might be a biomarker for early detection of O. viverrini associated HBA and CCA.
Collapse
Affiliation(s)
- Isabelle Jala
- Tropical Medicine Graduate Program (International Program), Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Muhammad Luthfi Almanfaluthi
- Tropical Medicine Graduate Program (International Program), Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sakawrat Kanthawong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sirikachorn Tangkawattana
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Department of Pathobiology, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Prasert Saichua
- Tropical Medicine Graduate Program (International Program), Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- Tropical Medicine Graduate Program (International Program), Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Banchob Sripa
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
11
|
Song L, Song M, Rabkin CS, Williams S, Chung Y, Van Duine J, Liao LM, Karthikeyan K, Gao W, Park JG, Tang Y, Lissowska J, Qiu J, LaBaer J, Camargo MC. Helicobacter pylori Immunoproteomic Profiles in Gastric Cancer. J Proteome Res 2020; 20:409-419. [PMID: 33108201 DOI: 10.1021/acs.jproteome.0c00466] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic Helicobacter pylori infection is the major risk factor for gastric cancer (GC). However, only some infected individuals develop this neoplasia. Previous H. pylori serology studies have been limited by investigating small numbers of candidate antigens. Therefore, we evaluated humoral responses to a nearly complete H. pylori immunoproteome (1527 proteins) among 50 GC cases and 50 controls using Nucleic Acid Programmable Protein Array (NAPPA). Seropositivity was defined as median normalized intensity ≥2 on NAPPA, and 53 anti-H. pylori antibodies had >10% seroprevalence. Anti-GroEL exhibited the greatest seroprevalence (77% overall), which agreed well with ELISA using whole-cell lysates of H. pylori cells. After an initial screen by H. pylori-NAPPA, we discovered and verified that 12 antibodies by ELISA in controls had ≥15% of samples with an optical reading value exceeding the 95th percentile of the GC group. ELISA-verified antibodies were validated blindly in an independent set of 100 case-control pairs. As expected, anti-CagA seropositivity was positively associated with GC (odds ratio, OR = 5.5; p < 0.05). After validation, six anti-H. pylori antibodies showed lower seropositivity in GC, with ORs ranging from 0.44 to 0.12 (p < 0.05): anti-HP1118/Ggt, anti-HP0516/HsIU, anti-HP0243/NapA, anti-HP1293/RpoA, anti-HP0371/FabE, and anti-HP0875/KatA. Among all combinations, a model with anti-Ggt, anti-HslU, anti-NapA, and anti-CagA had an area under the curve of 0.73 for discriminating GC vs. controls. This study represents the first comprehensive assessment of anti-H. pylori humoral profiles in GC. Decreased responses to multiple proteins in GC may reflect mucosal damage and decreased bacterial burden. The higher prevalence of specific anti-H. pylori antibodies in controls may suggest immune protection against GC development.
Collapse
Affiliation(s)
- Lusheng Song
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Minkyo Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland 20892-2590, United States
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland 20892-2590, United States
| | - Stacy Williams
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Yunro Chung
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States.,College of Health Solutions, Arizona State University, Phoenix, Arizona 85004, United States
| | - Jennifer Van Duine
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Linda M Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland 20892-2590, United States
| | - Kailash Karthikeyan
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Weimin Gao
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Jin G Park
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Yanyang Tang
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Jolanta Lissowska
- Division of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, 02-034 Warsaw, Poland
| | - Ji Qiu
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5001, United States
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland 20892-2590, United States
| |
Collapse
|
12
|
Seropositivity for Helicobacter pylori and hepatobiliary cancers in the PLCO study. Br J Cancer 2020; 123:909-911. [PMID: 32595210 PMCID: PMC7493958 DOI: 10.1038/s41416-020-0961-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 05/20/2020] [Accepted: 06/11/2020] [Indexed: 11/08/2022] Open
Abstract
Helicobacter has been suggested to play a possible role in hepatitis, gallstones, and hepatobiliary tumours. We assessed whether seropositivity to 15 H. pylori proteins was associated with subsequent incidence of 74 biliary tract and 105 liver cancer cases vs. 357 matched controls in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO). Odds ratios and 95% confidence intervals were computed by conditional logistic regression after adjustment for known hepatobiliary cancer risk factors. H. pylori seropositivity was not associated with either biliary tract (1.76, 0.90–3.46) or liver cancer (0.87, 0.46–1.65). CagA seropositivity was associated with both endpoints, although the latter association was not statistically significant (biliary tract: 2.16, 1.03–4.50; liver cancer: 1.96, 0.98–3.93) and neither association was statistically significant after correcting for multiple comparisons. Together, these results suggest possible associations between H. pylori and hepatobiliary cancer and suggest the value of future studies investigating the association. Trial registration number: NCT00339495.
Collapse
|
13
|
Butt J, Blot WJ, Shrubsole MJ, Waterboer T, Pawlita M, Epplein M. Differences in antibody levels to H. pylori virulence factors VacA and CagA among African Americans and whites in the Southeast USA. Cancer Causes Control 2020; 31:601-606. [PMID: 32222845 DOI: 10.1007/s10552-020-01295-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/13/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Helicobacter pylori (H. pylori) is the leading cause of gastric cancer. High antibody levels to H. pylori virulence factors Vacuolating cytotoxin A (VacA) and Cytotoxin-associated gene A (CagA) have been suggested as gastric cancer risk markers. In the USA, H. pylori sero-prevalence is twofold higher in African Americans compared to whites. We sought to assess whether African Americans also exhibit higher antibody levels to VacA and CagA. METHODS Antibody responses to H. pylori proteins were measured by multiplex serology in 686 African Americans and whites of the Southern Community Cohort Study. Among VacA- and CagA-seropositives, we analyzed the association of race with antibody level using logistic regression models to produce odds ratios (OR) and 95% confidence intervals (CI). RESULTS Sero-positive African Americans had significantly higher mean antibody levels to both VacA and CagA, which resulted in increased odds for the highest quartile of antibody levels compared to sero-positive whites (VacA, OR: 6.08; 95% CI 3.41, 10.86; CagA, OR: 3.77; 95% CI 1.61, 8.84). CONCLUSION Our findings support future studies to assess the association of differential antibody responses by race with risk of gastric cancer in the USA, which could then aid in developing targeted H. pylori eradication strategies.
Collapse
Affiliation(s)
- Julia Butt
- Cancer Control and Population Health Sciences Program, Duke Cancer Institute and Department of Population Health Sciences, Duke University, Durham, NC, USA. .,Infection and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| | - William J Blot
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Martha J Shrubsole
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tim Waterboer
- Infection and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Michael Pawlita
- Infection and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Meira Epplein
- Cancer Control and Population Health Sciences Program, Duke Cancer Institute and Department of Population Health Sciences, Duke University, Durham, NC, USA
| |
Collapse
|
14
|
Bioinformatics Analysis of Potential Key Genes in Trastuzumab-Resistant Gastric Cancer. DISEASE MARKERS 2019; 2019:1372571. [PMID: 31949544 PMCID: PMC6948351 DOI: 10.1155/2019/1372571] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 10/03/2019] [Accepted: 11/09/2019] [Indexed: 12/24/2022]
Abstract
Background This study was performed to identify genes related to acquired trastuzumab resistance in gastric cancer (GC) and to analyze their prognostic value. Methods The gene expression profile GSE77346 was downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were obtained by using GEO2R. Functional and pathway enrichment was analyzed by using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Search Tool for the Retrieval of Interacting Genes (STRING), Cytoscape, and MCODE were then used to construct the protein-protein interaction (PPI) network and identify hub genes. Finally, the relationship between hub genes and overall survival (OS) was analyzed by using the online Kaplan-Meier plotter tool. Results A total of 327 DEGs were screened and were mainly enriched in terms related to pathways in cancer, signaling pathways regulating stem cell pluripotency, HTLV-I infection, and ECM-receptor interactions. A PPI network was constructed, and 18 hub genes (including one upregulated gene and seventeen downregulated genes) were identified based on the degrees and MCODE scores of the PPI network. Finally, the expression of four hub genes (ERBB2, VIM, EGR1, and PSMB8) was found to be related to the prognosis of HER2-positive (HER2+) gastric cancer. However, the prognostic value of the other hub genes was controversial; interestingly, most of these genes were interferon- (IFN-) stimulated genes (ISGs). Conclusions Overall, we propose that the four hub genes may be potential targets in trastuzumab-resistant gastric cancer and that ISGs may play a key role in promoting trastuzumab resistance in GC.
Collapse
|
15
|
Wang R, Liu K, Chen XZ. Associations between gastric cancer risk and virus infection other than Epstein-Barr virus: The protocol of a systematic review and meta-analysis based on epidemiological studies. Medicine (Baltimore) 2019; 98:e16708. [PMID: 31393376 PMCID: PMC6709195 DOI: 10.1097/md.0000000000016708] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Gastric cancer is one of the infection associated malignancies. In addition to Helicobacter pylori and Epstein-Barr virus (EBV), other oncoviruses might play potential roles in the development of gastric cancer. Associations of oncoviruses other than EBV with gastric cancer risk are aimed to be comprehensively reviewed and assessed in this systematic review and meta-analysis, to identify any potentially causative oncovirus. It might be informative to identify or deny certain oncoviruses which are candidates of risk factor for gastric cancer. To our knowledge, there is no comprehensive review on oncoviruses other than EBV associated with gastric cancer risk. Positive findings might be helpful to suggest further mechanism investigation and high-risk subpopulation recommendation. METHODS PubMed database will be searched up to Dec 31, 2018. The studies, compared the positivity of any oncovirus other than EBV between cases with histologically proven gastric cancer and healthy or nonmalignant controls, are eligible. The detection of oncovirus either in tissue or blood is acceptable. Selection, quality assessment (Newcastle-Ottawa Scale), and data extraction of eligible studies will be performed by 2 independent reviewers. Pooled prevalence of any oncovirus will be combined by meta-analysis for rate. Pooled odds ratio between gastric cancer cases and controls will be estimated by meta-analysis. Heterogeneity and publication bias will be tested. In sensitivity analysis, the leave-one-out method and exclusion of low power studies will be applied where applicable. RESULTS This review was not submitted for any ethical approval due to the literature-based nature. The results will be published in a journal and presented at conferences for academic purposes.Registration number was CRD42015029703 in the PROSPERO International Prospective Register of Systematic Reviews. CONCLUSIONS To our knowledge, there is no comprehensive review on oncoviruses other than EBV associated with gastric cancer risk. Positive findings might be helpful to suggest further mechanism investigation and high-risk subpopulation recommendation.
Collapse
Affiliation(s)
- Rui Wang
- Nursing Section, Department of Gastroenterology
| | - Kai Liu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Xin-Zu Chen
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Varga MG, Cai H, Waterboer T, Murphy G, Shimazu T, Taylor PR, Qiao YL, Park SK, Yoo KY, Jee SH, Cho ER, Kim J, Abnet CC, Tsugane S, Cai Q, Zheng W, Pawlita M, Shu XO, Epplein M. Epstein-Barr Virus Antibody Titers Are Not Associated with Gastric Cancer Risk in East Asia. Dig Dis Sci 2018; 63:2765-2772. [PMID: 29948559 PMCID: PMC6139270 DOI: 10.1007/s10620-018-5154-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 05/30/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Epstein-Barr virus (EBV)-positive gastric cancers represent a distinct subtype of gastric cancers and account for nearly 10% of the gastric cancer burden, yet risk detection strategies for this cancer subtype are lacking. METHODS We conducted a nested case-control study where we assayed 4 EBV antigens [viral capsid antigen (VCA), early antigen (EA), Epstein-Barr nuclear antigen (EBNA), and BZLF1-encoded replication activator protein (ZEBRA)] in either sera or plasma from 1447 gastric cancer cases and 1797 controls obtained from seven prospective cohorts representing individuals from the high gastric cancer-risk countries of China, Japan, and Korea. RESULTS The prevalence of EBV sero-positivity was universal with the exception of one sero-negative individual, and the highest titers of the EBV antigens VCA (OR 0.95, 95% CI 0.78-1.17), EBNA (OR 0.88, 95% CI 0.72-1.08), EA (OR 0.97, 95% CI 0.79-1.19), and ZEBRA (OR 0.87, 95% CI 0.71-1.07) were not associated with risk of incident gastric cancer. When we stratified these data by H. pylori status, there was no change in the association. CONCLUSIONS Multiplex serology of the aforementioned EBV antigens in serum may not be a suitable biomarker for predicting gastric cancer risk in East Asian populations.
Collapse
Affiliation(s)
- Matthew G. Varga
- Department of Epidemiology, Gillings School of Global Public Health
and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel
Hill, 3207B Michael Hooker Research Center, Chapel Hill, NC 27599, USA,Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center and Ingram Cancer Center, Vanderbilt University Medical Center,
Nashville, TN 37203, USA
| | - Hui Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center and Ingram Cancer Center, Vanderbilt University Medical Center,
Nashville, TN 37203, USA
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, Research
Program in Infection, Inflammation, and Cancer, German Cancer Research Center
(DFKZ), 69120 Heidelberg, Germany
| | - Gwen Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Bethesda, MD 20892, USA
| | - Taichi Shimazu
- Epidemiology and Prevention Group, National Cancer Center, Tokyo
104-0045, Japan
| | - Phil R. Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Bethesda, MD 20892, USA
| | - You-Lin Qiao
- Department of Cancer Epidemiology, Chinese Academy of Medical
Sciences and Peking Union Medial College, Beijing 100021, China
| | - Sue K. Park
- Department of Biomedical Sciences, Cancer Research Institute, Seoul
National University College of Medicine, Seoul 110-799, Korea
| | - Keun-Young Yoo
- Department of Preventive Medicine, Seoul National University College
of Medicine, Seoul 110-799, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Institute for
Health Promotion, Yonsei University, Seoul 120-752, Korea
| | - Eo Rin Cho
- Department of Epidemiology and Health Promotion, Institute for
Health Promotion, Yonsei University, Seoul 120-752, Korea
| | - Jeongseon Kim
- Division of Cancer Epidemiology and Prevention, Research Institute,
National Cancer Center, Goyang 410-769, Korea
| | - Christian C. Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Bethesda, MD 20892, USA
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, National Cancer Center, Tokyo
104-0045, Japan
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center and Ingram Cancer Center, Vanderbilt University Medical Center,
Nashville, TN 37203, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center and Ingram Cancer Center, Vanderbilt University Medical Center,
Nashville, TN 37203, USA
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, Research
Program in Infection, Inflammation, and Cancer, German Cancer Research Center
(DFKZ), 69120 Heidelberg, Germany
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center and Ingram Cancer Center, Vanderbilt University Medical Center,
Nashville, TN 37203, USA
| | - Meira Epplein
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center and Ingram Cancer Center, Vanderbilt University Medical Center,
Nashville, TN 37203, USA,Department of Population Health Sciences, Duke University and
Cancer Control and Population Sciences Program, Duke Cancer Institute, Durham, NC
27705, USA
| |
Collapse
|
17
|
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: 2.3] [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
|
18
|
Murphy G, Abnet CC, Choo-Wosoba H, Vogtmann E, Weinstein SJ, Taylor PR, Männistö S, Albanes D, Dawsey SM, Rehfeld JF, Freedman ND. Serum gastrin and cholecystokinin are associated with subsequent development of gastric cancer in a prospective cohort of Finnish smokers. Int J Epidemiol 2018; 46:914-923. [PMID: 28369403 DOI: 10.1093/ije/dyx030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2017] [Indexed: 12/19/2022] Open
Abstract
Background Gastrin, which induces gastric acid secretion, and a structurally similar hormone, cholecystokinin (CCK)-a potent acid inhibitor, may each play a role in gastric cancer. However, few studies have investigated this hypothesis in humans. We therefore investigated whether serum gastrin or CCK concentrations at baseline were associated with the incidence of gastric non-cardia adenocarcinomas (GNCA), oesophagogastric junctional adenocarcinomas (EGJA) or gastric carcinoid tumours over 24 years of follow-up in a study nested within the all-male Alpha-Tocopherol, Beta-Carotene Cancer Prevention (ATBC) Study of Finnish smokers. Methods Totals of 283 incident GNCA, 96 EGJA and 10 gastric carcinoid cases, and 778 matched controls, were included in our analysis. Gastrin and CCK were measured using specific radioimmunoassays. Odds ratios (OR) and 95% confidence intervals (95% CI) were estimated by multivariable logistic regression with adjustment for all known or suspected confounding factors, including Helicobacter pylori seropositivity. Results Those with high gastrin (Q4 vs Q1), had an increased risk of GNCA (fully adjusted OR: 1.92; 95% CI: 1.21, 3.05) and gastric carcinoids, though the small number of carcinoid cases meant the fully adjusted model was unstable (age-adjusted continuous model OR: 4.67; 95% CI: 2.67, 8.15). CCK was associated with risk of GNCA only for those in Q3 relative to Q1 (OR: 0.56; 95% CI: 0.33, 0.96), and no significant trend was observed. Conclusions Our data suggest that high serum concentrations of gastrin may be associated independently with an increased risk of gastric cancer; the role of CCK in cancer risk is less clear.
Collapse
Affiliation(s)
- Gwen Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Hyoyoung Choo-Wosoba
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Emily Vogtmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Philip R Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Satu Männistö
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
19
|
Multiplex serology of Helicobacter pylori antigens in detection of current infection and atrophic gastritis - A simple and cost-efficient method. Microb Pathog 2018; 119:137-144. [DOI: 10.1016/j.micpath.2018.04.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
|
20
|
Li H, Zhang B, Hu X, Dong Y, Fan Q, Guo F, Ren X, Zhou H, Tian W, Zhao Y. Serum Helicobacter pylori FliD antibody and the risk of gastric cancer. Oncotarget 2017; 7:22397-408. [PMID: 26968951 PMCID: PMC5008368 DOI: 10.18632/oncotarget.7981] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 02/23/2016] [Indexed: 02/06/2023] Open
Abstract
FliD and CagA are important virulence factors of H. pylori. We aimed to evaluate the screening values of FliD and CagA for gastric cancer (GC). Serum samples were obtained from 232 cases and 266 controls in a case-control study. Unconditional multivariate logistic regression with odds ratios (ORs) and 95% confidence intervals (CIs) was used to analyze the relationships between FliD, CagA and GC. The sensitivities, specificities and receiver operating characteristic (ROC) curves were calculated. Finally, the combined screening values of FliD, FlaA, NapA and CagA were assessed based on discriminant analysis. In all subjects, the associations of FliD and CagA with GC were evident with ORs (95% CIs) of 7.6 (4.7-12.3) and 2.5 (1.6-3.8), respectively (*p<0.001). The areas under ROC curves (AUCs) for FliD and CagA were 0.800 and 0.653, respectively. The AUC for the combination of FliD, FlaA and NapA was 0.915, which represented an increase of 0.115 over that of FliD alone (*p<0.001). These findings indicate that the FliD antibody is associated with GC and could exhibit high validity as a biomarker in screening for GC patients. The combination of FliD, FlaA and NapA improved the screening validity.
Collapse
Affiliation(s)
- Hailin Li
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Bing Zhang
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Xiaomeng Hu
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Yingzi Dong
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Qing Fan
- Xiangfang Center for Disease Control and Prevention, Harbin, Heilongjiang Province, P. R. China
| | - Fang Guo
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Xiyun Ren
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Haibo Zhou
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Wenjing Tian
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Yashuang Zhao
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| |
Collapse
|
21
|
Performance of a Multiplex Serological Helicobacter pylori Assay on a Novel Microfluidic Assay Platform. Proteomes 2017; 5:proteomes5040024. [PMID: 28972560 PMCID: PMC5748559 DOI: 10.3390/proteomes5040024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/19/2017] [Accepted: 09/29/2017] [Indexed: 01/25/2023] Open
Abstract
Infection with Helicobacter pylori (H. pylori) occurs in 50% of the world population, and is associated with the development of ulcer and gastric cancer. Serological diagnostic tests indicate an H. pylori infection by detecting antibodies directed against H. pylori proteins. In addition to line blots, multiplex assay platforms provide smart solutions for the simultaneous analysis of antibody responses towards several H. pylori proteins. We used seven H. pylori proteins (FliD, gGT, GroEL, HpaA, CagA, VacA, and HP0231) and an H. pylori lysate for the development of a multiplex serological assay on a novel microfluidic platform. The reaction limited binding regime in the microfluidic channels allows for a short incubation time of 35 min. The developed assay showed very high sensitivity (99%) and specificity (100%). Besides sensitivity and specificity, the technical validation (intra-assay CV = 3.7 ± 1.2% and inter-assay CV = 5.5 ± 1.2%) demonstrates that our assay is also a robust tool for the analysis of the H. pylori-specific antibody response. The integration of the virulence factors CagA and VacA allow for the assessment of the risk for gastric cancer development. The short assay time and the performance of the platform shows the potential for implementation of such assays in a clinical setting.
Collapse
|
22
|
Fernández-de-Larrea N, Michel A, Romero B, Butt J, Pawlita M, Pérez-Gómez B, Castaño-Vinyals G, Moreno V, Martín V, Amiano P, Castilla J, Fernández-Tardón G, Dierssen-Sotos T, Clofent J, Alguacil J, Huerta JM, Jiménez-Moleón JJ, Barricarte A, Molinuevo A, Fernández-Villa T, Casabonne D, Sierra Á, Kogevinas M, de Sanjosé S, Pollán M, Del Campo R, Waterboer T, Aragonés N. Antibody reactivity against Helicobacter pylori proteins in a sample of the Spanish adult population in 2008-2013. Helicobacter 2017; 22. [PMID: 28737284 DOI: 10.1111/hel.12401] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Differences in Helicobacter pylori protein expression have been related to the risk of severe gastric diseases. In Spain, a marked geographic pattern in gastric cancer mortality has long been reported. OBJECTIVE To characterize antibody reactivity patterns against 16 H. pylori proteins, by age, sex, and region of birth, in a large sample of the Spanish adult population. MATERIALS AND METHODS Antibody reactivity was quantified by H. pylori multiplex serology in a sample from the control group of the multicase-control study MCC-Spain. For this analysis, 2555 population-based controls were included. Each participant was classified as seropositive or seronegative for each protein according to specific cutoffs. Overall H. pylori seroprevalence was defined as positivity against ≥4 proteins. Descriptive analyses by age, sex, and region of birth were performed for both seroprevalence and seroreactivity (continuous measure). Differences among groups were tested by logistic and linear regression models. RESULTS Overall H. pylori seroprevalence increased with age in both sexes. For ages 55-74, seroprevalence was lower in women than in men (84% vs 92%, P<.001). Region of birth explained 7% of the variability in seroprevalence. Among H. pylori seropositive subjects, proteins with the highest seroprevalence were GroEL, NapA, HP231, and Omp. Seropositivity for most of the proteins increased or remained stable with age, rising mainly for CagA, GroEL, and HyuA in women. A clear cohort effect was not observed. CONCLUSIONS This is the first study to describe the antibody patterns against 16 H. pylori proteins in the Spanish population. We found variability in the H. pylori antibody profiles according to both individual factors such as age and sex, and environmental factors such as the region of birth. The slightness of the reduction in seropositivity with decreasing age highlights the ongoing importance of this infection.
Collapse
Affiliation(s)
- Nerea Fernández-de-Larrea
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos III, Madrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Beatriz Romero
- Department of Microbiology, Ramón y Cajal University Hospital (IRYCIS), Madrid, Spain
| | - Julia Butt
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Beatriz Pérez-Gómez
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos III, Madrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Gemma Castaño-Vinyals
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Victor Moreno
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Cancer Prevention and Control Program, Catalan Institute of Oncology-IDIBELL, Hospitalet de Llobregat, Spain
| | - Vicente Martín
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,The Research Group in Gene, Environment and Health Interactions (GIGAS), University of León, León, Spain.,Faculty of Health Sciences, Area of Preventive Medicine and Public Health, Department of Biomedical Sciences, University of León, León, Spain
| | - Pilar Amiano
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Public Health Division of Gipuzkoa, BioDonostia Research Institute, San Sebastián, Spain
| | - Jesús Castilla
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Instituto de Salud Pública de Navarra - Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Guillermo Fernández-Tardón
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,IUOPA, University of Oviedo, Oviedo, Spain
| | - Trinidad Dierssen-Sotos
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,IDIVAL, University of Cantabria, Santander, Spain
| | - Juan Clofent
- Gastroenterology Department, Sagunto University Hospital, Sagunto, Spain.,Gastroenterology Department, La Fe University and Politecnic Hospital, Valencia, Spain
| | - Juan Alguacil
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Centro de Investigación en Recursos Naturales, Salud, y Medio Ambiente (RENSMA), Universidad de Huelva, Huelva, Spain
| | - José María Huerta
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | - José Juan Jiménez-Moleón
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada Health Research Institute (ibs.GRANADA), Granada, Spain.,Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| | - Aurelio Barricarte
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Instituto de Salud Pública de Navarra - Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Amaia Molinuevo
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Tania Fernández-Villa
- The Research Group in Gene, Environment and Health Interactions (GIGAS), University of León, León, Spain.,Faculty of Health Sciences, Area of Preventive Medicine and Public Health, Department of Biomedical Sciences, University of León, León, Spain
| | - Delphine Casabonne
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Cancer Epidemiology Research Program, Catalan Institute of Oncology-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Ángeles Sierra
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos III, Madrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Manolis Kogevinas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Silvia de Sanjosé
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Cancer Epidemiology Research Program, Catalan Institute of Oncology-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Marina Pollán
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos III, Madrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Rosa Del Campo
- Department of Microbiology, Ramón y Cajal University Hospital (IRYCIS), Madrid, Spain.,Spanish Network for Research in Infectious Diseases, (REIPI) Red Española de Investigación en Patología Infecciosa, Sevilla, Spain
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nuria Aragonés
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos III, Madrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health), Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
23
|
Fernández de Larrea-Baz N, Pérez-Gómez B, Michel A, Romero B, Lope V, Pawlita M, Fernández-Villa T, Moreno V, Martín V, Willhauck-Fleckenstein M, López-Abente G, Castilla J, Fernández-Tardón G, Dierssen-Sotos T, Santibáñez M, Peiró R, Jiménez-Moleón JJ, Navarro C, Castaño-Vinyals G, Kogevinas M, Pollán M, de Sanjosé S, Del Campo R, Waterboer T, Aragonés N. Helicobacter pylori serological biomarkers of gastric cancer risk in the MCC-Spain case-control Study. Cancer Epidemiol 2017; 50:76-84. [PMID: 28888185 DOI: 10.1016/j.canep.2017.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/10/2017] [Accepted: 08/04/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Helicobacter pylori infection is one of the main risk factors for non-cardia gastric cancer. However, only a minority of infected persons develop the disease. This study aims at identifying H. pylori related serological biomarkers of risk for gastric cancer. METHODS Incident gastric cancer cases and population controls (age, sex and region frequency-matched) from the MCC-Spain multicase-control Study were included. Seroreactivities against 16H. pylori proteins were determined using multiplex serology. Infection was defined as seropositivity against≥4 proteins. Relation of serological results to non-cardia and cardia gastric cancer was assessed using multivariable mixed logistic regression and principal components analysis. RESULTS Seroprevalence was 88% among 2071 controls, 95% among 202 non-cardia gastric cancer cases (OR=1.9 (95% CI: 1.0-3.6)) and 85% among 62 cardia cancer cases (OR=0.5 (95% CI: 0.3-1.1)). In infected subjects, seropositivity for UreA, HP231, NapA and Cagδ was associated with lower non-cardia gastric cancer risk, while seropositivity for CagA and VacA was associated with higher risk. Seropositivity for CagA and seronegativity for Cagδ maintained the association after additional adjustment by serostatus of significant proteins. We identified two antibody reactivity patterns: the "virulent-pattern", related to a threefold higher risk of non-cardia gastric cancer and the "non-virulent pattern", related to a 60% decreased risk (4th vs. first quartile). CONCLUSIONS In our population, people seropositive for H. pylori were characterized by two patterns of antibody reactivity against H. pylori proteins: 1) Combined high seroreactivity against several proteins, associated with a lower non-cardia gastric cancer risk, and 2) High seroreactivity against CagA and VacA, associated with an increased risk.
Collapse
Affiliation(s)
- Nerea Fernández de Larrea-Baz
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Carlos III Health Institute (ISCIII), Avda. Monforte de Lemos 5, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain.
| | - Beatriz Pérez-Gómez
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Carlos III Health Institute (ISCIII), Avda. Monforte de Lemos 5, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Beatriz Romero
- Department of Microbiology, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)-Ramón y Cajal Health Research Institute (IRYCIS), Ctra. de Colmenar Viejo km. 9,100, 28034, Madrid, Spain
| | - Virginia Lope
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Carlos III Health Institute (ISCIII), Avda. Monforte de Lemos 5, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Tania Fernández-Villa
- The Research Group in Gene - Environment and Health Interactions (GIIGAS), University of León, Spain; Department of Biomedical Sciences, Area of Preventive Medicine and Public Health, University of León, Campus de Vegazana s/n, 24071, León, Spain
| | - Victor Moreno
- Cancer Prevention and Control Program, Catalan Institute of Oncology, Avinguda de la Granvia de l'Hospitalet 199-203, 08908, Hospitalet de Llobregat, Barcelona, Spain; Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Gran Via de les Corts Catalanes, 585, 08007, Barcelona, Spain; Colorectal Cancer Group, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908, Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Vicente Martín
- The Research Group in Gene - Environment and Health Interactions (GIIGAS), University of León, Spain; Department of Biomedical Sciences, Area of Preventive Medicine and Public Health, University of León, Campus de Vegazana s/n, 24071, León, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Martina Willhauck-Fleckenstein
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Gonzalo López-Abente
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Carlos III Health Institute (ISCIII), Avda. Monforte de Lemos 5, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Jesús Castilla
- Instituto de Salud Pública de Navarra, IdiSNA-Navarra Institute for Health Research, C/Leyre, 15, 31003, Pamplona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Guillermo Fernández-Tardón
- University Institute of Oncology (IUOPA), University of Oviedo, Fernando Bongera. Building "Santiago Gascón", 1 st Floor, Campus of "El Cristo" B, 33006, Oviedo, Asturias, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Trinidad Dierssen-Sotos
- University of Cantabria - IDIVAL-Instituto de Investigación Marqués de Valdecilla-Marqués de Valdecilla Research Institute (IDIVAL), C/Cardenal Herrera Oria, s/n, 39011, Santander, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Miguel Santibáñez
- University of Cantabria - IDIVAL-Instituto de Investigación Marqués de Valdecilla-Marqués de Valdecilla Research Institute (IDIVAL), C/Cardenal Herrera Oria, s/n, 39011, Santander, Spain; Centro de Investigación en Recursos Naturales, Salud, y Medio Ambiente (RENSMA), University of Huelva, Campus de El Carmen, 21007, Huelva, Spain
| | - Rosana Peiró
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana FISABIO-Salud Pública - Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Avda. de Catalunya, 21, 46020, Valencia, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - José Juan Jiménez-Moleón
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)-Granada Health Research Institute (ibs.GRANADA), 18012, Granada, Spain; Department of Preventive Medicine and Public Health, University of Granada, Avda. de la Investigación, 11, Building A, 8th Floor, 18016, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Carmen Navarro
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Ronda de Levante, 11, 2ª planta, 30008, Murcia, Spain; Department of Health and Social Sciences, University of Murcia, Avda. Teniente Flomesta, 5, 30003, Murcia, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Gemma Castaño-Vinyals
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Doctor Aiguader, 88, 08003, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Dr. Aiguader, 88, 08003, Barcelona, Spain; Pompeu Fabra University (UPF), Plaça de la Mercè, 10-12, 08002, Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Manolis Kogevinas
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Doctor Aiguader, 88, 08003, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Dr. Aiguader, 88, 08003, Barcelona, Spain; Pompeu Fabra University (UPF), Plaça de la Mercè, 10-12, 08002, Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Marina Pollán
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Carlos III Health Institute (ISCIII), Avda. Monforte de Lemos 5, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Silvia de Sanjosé
- Cancer Epidemiology Research Program, Catalan Institute of Oncology-IDIBELL, Avinguda de la Granvia de l'Hospitalet 199-203, 08908, l'Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Rosa Del Campo
- Department of Microbiology, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)-Ramón y Cajal Health Research Institute (IRYCIS), Ctra. de Colmenar Viejo km. 9,100, 28034, Madrid, Spain; Red Española de Investigación en Patología Infecciosa (REIPI)-Spanish Network for Research in Infectious Diseases (REIPI), Spain
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Nuria Aragonés
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Carlos III Health Institute (ISCIII), Avda. Monforte de Lemos 5, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)-CIBER of Epidemiology and Public Health (CIBERESP), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| |
Collapse
|
24
|
Shen H, Zhong M, Wang W, Liao P, Yin X, Rotroff D, Knepper TC, Mcleod HL, Zhou C, Xie S, Li W, Xu B, He Y. EBV infection and MSI status significantly influence the clinical outcomes of gastric cancer patients. Clin Chim Acta 2017; 471:216-221. [PMID: 28601671 DOI: 10.1016/j.cca.2017.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Epstein-Barr virus (EBV) and microsatellite instability (MSI) are associated with the carcinogenesis of many kinds of tumors, including gastric cancer (GC). However, the impact of EBV and MSI status on the prognosis of stage II and III GC is still unclear. The aim of this study was to find out the prognostic value of EBV and MSI status in a population of GC patients from Southern China. METHODS Patients were genotyped for EBV infection based on the detection of EBV DNA from the formalin-fixed paraffin-embedded (FFPE) specimens. Sequentially, MSI status was measured by direct sequencing. Clinical characteristics and overall survival (OS) were analyzed in 202 GC patients. Additionally, the association of EBV and MSI status with chemotherapy-based toxicity was analyzed in 324 GC patients. RESULTS The survival analysis revealed EBV+ patients had a poorer OS than EBV- patients (HR=1.75, 95% CI: 1.08-2.82, FDR p=0.04). This survival advantage for EBV- patients was also found in patients <60y (FDR p=0.04) and patient with stage III disease (FDR p=0.04). CONCLUSIONS EBV infection and MSI status are associated with overall survival of gastric cancer patients. However, traditional chemotherapy showed no difference on outcome of patients in EBV and MSI subgroups.
Collapse
Affiliation(s)
- Hua Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China; Gastroenterology and Urology Department, Hunan Cancer hospital, Xiangya School of Medicine, Central South University, Changsha, China
| | - Meizuo Zhong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Weili Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Ping Liao
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Xianli Yin
- Gastroenterology and Urology Department, Hunan Cancer hospital, Xiangya School of Medicine, Central South University, Changsha, China
| | - Daniel Rotroff
- Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL, USA
| | - Todd C Knepper
- Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL, USA
| | - Howard L Mcleod
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China; Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL, USA
| | - Chengfang Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Shangchen Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Wei Li
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Biaobo Xu
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.
| | - Yijing He
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China; Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL, USA.
| |
Collapse
|
25
|
Validation of a Novel Immunoline Assay for Patient Stratification according to Virulence of the Infecting Helicobacter pylori Strain and Eradication Status. J Immunol Res 2017. [PMID: 28638837 PMCID: PMC5468576 DOI: 10.1155/2017/8394593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Helicobacter pylori infection shows a worldwide prevalence of around 50%. However, only a minority of infected individuals develop clinical symptoms or diseases. The presence of H. pylori virulence factors, such as CagA and VacA, has been associated with disease development, but assessment of virulence factor presence requires gastric biopsies. Here, we evaluate the H. pylori recomLine test for risk stratification of infected patients by comparing the test score and immune recognition of type I or type II strains defined by the virulence factors CagA, VacA, GroEL, UreA, HcpC, and gGT with patient's disease status according to histology. Moreover, the immune responses of eradicated individuals from two different populations were analysed. Their immune response frequencies and intensities against all antigens except CagA declined below the detection limit. CagA was particularly long lasting in both independent populations. An isolated CagA band often represents past eradication with a likelihood of 88.7%. In addition, a high recomLine score was significantly associated with high-grade gastritis, atrophy, intestinal metaplasia, and gastric cancer. Thus, the recomLine is a sensitive and specific noninvasive test for detecting serum responses against H. pylori in actively infected and eradicated individuals. Moreover, it allows stratifying patients according to their disease state.
Collapse
|
26
|
Fernández de Larrea-Baz N, Michel A, Romero B, Pérez-Gómez B, Moreno V, Martín V, Dierssen-Sotos T, Jiménez-Moleón JJ, Castilla J, Tardón A, Ruiz I, Peiró R, Tejada A, Chirlaque MD, Butt JA, Olmedo-Requena R, Gómez-Acebo I, Linares P, Boldo E, Castells A, Pawlita M, Castaño-Vinyals G, Kogevinas M, de Sanjosé S, Pollán M, Del Campo R, Waterboer T, Aragonés N. Helicobacter pylori Antibody Reactivities and Colorectal Cancer Risk in a Case-control Study in Spain. Front Microbiol 2017; 8:888. [PMID: 28611733 PMCID: PMC5447227 DOI: 10.3389/fmicb.2017.00888] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 05/02/2017] [Indexed: 12/19/2022] Open
Abstract
Background: Several studies have suggested that Helicobacter pylori (H. pylori) infection is a risk factor for colorectal cancer (CRC), while others have not confirmed this hypothesis. This work aimed to assess the relation of CRC with H. pylori seropositivity and with seropositivity to 16 H. pylori proteins, in the MultiCase-Control study, MCC-Spain. Methods: MCC-Spain is a multicase-control study carried out in Spain from 2008 to 2013. In total, 2,140 histologically-confirmed incident CRC cases and 4,098 population-based controls were recruited. Controls were frequency-matched by sex, age, and province. Epidemiological data were collected through a questionnaire fulfilled by face-to-face interviews and a self-administered food-frequency questionnaire. Seroreactivities against 16 H. pylori proteins were determined in 1,488 cases and 2,495 controls using H. pylori multiplex serology. H. pylori seropositivity was defined as positivity to ≥4 proteins. Multivariable logistic regression mixed models were used to estimate odds ratios (OR) and 95% confidence intervals (CI). Results:H. pylori seropositivity was not associated with increased CRC risk (OR = 0.91; 95% CI: 0.71–1.16). Among H. pylori seropositive subjects, seropositivity to Cagδ showed a lower CRC risk, and risk decreased with increasing number of proteins seropositive. Seropositivity to the most recognized virulence factors, CagA and VacA, was not associated with a higher CRC risk. No statistically significant heterogeneity was identified among tumor sites, although inverse relations were stronger for left colon cancer. An interaction with age and sex was found: H. pylori seropositivity was associated with a lower CRC risk in men younger than 65 and with a higher risk in older women. Conclusions: Our results suggest that neither H. pylori seropositivity, nor seropositivity to the virulence factor CagA are associated with a higher CRC risk. A possible effect modification by age and sex was identified.
Collapse
Affiliation(s)
- Nerea Fernández de Larrea-Baz
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos IIIMadrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ)Heidelberg, Germany
| | - Beatriz Romero
- Department of Microbiology, Ramón y Cajal University Hospital (IRYCIS)Madrid, Spain
| | - Beatriz Pérez-Gómez
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos IIIMadrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Cancer Epidemiology Research Group, Oncology and Hematology Area, IIS Puerta de Hierro (Puerta de Hierro Health Research Institute)Madrid, Spain
| | - Victor Moreno
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Cancer Prevention and Control Program, Catalan Institute of OncologyHospitalet de Llobregat, Spain.,Department of Clinical Sciences, Faculty of Medicine, University of BarcelonaBarcelona, Spain.,Colorectal Cancer Group, Bellvitge Biomedical Research Institute (IDIBELL)Hospitalet de Llobregat, Spain
| | - Vicente Martín
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,The Research Group in Gene - Environment and Health Interactions, University of LeónLeón, Spain.,Area of Preventive Medicine and Public Health, Faculty of Health Sciences, Department of Biomedical Sciences, University of LeónLeón, Spain
| | - Trinidad Dierssen-Sotos
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Division of Epidemiology and Computational Biology, School of Medicine, University of Cantabria-IDIVALSantander, Spain
| | - José J Jiménez-Moleón
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Granada Health Research Institute (ibs.GRANADA) - Instituto de Investigación Biosanitaria de GranadaGranada, Spain.,Department of Preventive Medicine and Public Health, University of GranadaGranada, Spain
| | - Jesús Castilla
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Instituto de Salud Pública de Navarra, IdiSNA-Navarra Institute for Health ResearchPamplona, Spain
| | - Adonina Tardón
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Molecular Epidemiology of Cancer Unit, Oncology Institute, Department of Medicine, University of OviedoOviedo, Spain
| | - Irune Ruiz
- Department of Pathology, Donostia University HospitalDonostia, Spain
| | - Rosana Peiró
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO) - Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana FISABIO-Salud PúblicaValencia, Spain
| | - Antonio Tejada
- Coloproctology Unit, Department of General Surgery, Huelva University Hospital ComplexHuelva, Spain
| | - María D Chirlaque
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Department of Epidemiology, Regional Health Council, IMIB-ArrixacaMurcia, Spain.,Department of Health and Social Sciences, University of MurciaMurcia, Spain
| | - Julia A Butt
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ)Heidelberg, Germany
| | - Rocío Olmedo-Requena
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Granada Health Research Institute (ibs.GRANADA) - Instituto de Investigación Biosanitaria de GranadaGranada, Spain.,Department of Preventive Medicine and Public Health, University of GranadaGranada, Spain
| | - Inés Gómez-Acebo
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Division of Epidemiology and Computational Biology, School of Medicine, University of Cantabria-IDIVALSantander, Spain
| | - Pedro Linares
- Department of Gastroenterology and Hepatology, Complejo Asistencial Universitario de LeónLeón, Spain
| | - Elena Boldo
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos IIIMadrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Cancer Epidemiology Research Group, Oncology and Hematology Area, IIS Puerta de Hierro (Puerta de Hierro Health Research Institute)Madrid, Spain
| | - Antoni Castells
- Gastroenterology Department, Hospital ClínicBarcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Barcelona, Spain.,CIBER Liver and Digestive Diseases - CIBER Enfermedades Hepáticas y Digestivas (CIBEREHD)Madrid, Spain.,Department of Gastroenterology, University of BarcelonaBarcelona, Spain
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ)Heidelberg, Germany
| | - Gemma Castaño-Vinyals
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,ISGlobal, Centre for Research in Environmental Epidemiology (CREAL)Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM)Barcelona, Spain.,Department of Experimental and Health Sciences, Universitat Pompeu FabraBarcelona, Spain
| | - Manolis Kogevinas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,ISGlobal, Centre for Research in Environmental Epidemiology (CREAL)Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM)Barcelona, Spain.,Department of Experimental and Health Sciences, Universitat Pompeu FabraBarcelona, Spain
| | - Silvia de Sanjosé
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Cancer Epidemiology and Research Program, Catalan Institute of Oncology-IDIBELLHospitalet de Llobregat, Spain
| | - Marina Pollán
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos IIIMadrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain.,Cancer Epidemiology Research Group, Oncology and Hematology Area, IIS Puerta de Hierro (Puerta de Hierro Health Research Institute)Madrid, Spain
| | - Rosa Del Campo
- Department of Microbiology, Ramón y Cajal University Hospital (IRYCIS)Madrid, Spain.,Spanish Network for Research in Infectious Diseases - Red Española de Investigación en Patología InfecciosaSevilla, Spain
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ)Heidelberg, Germany
| | - Nuria Aragonés
- Environmental and Cancer Epidemiology Area, National Center of Epidemiology, Instituto de Salud Carlos IIIMadrid, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER of Epidemiology and Public Health) - Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Madrid, Spain
| |
Collapse
|
27
|
Zepeda Gurrola RC, Fu Y, Rodríguez Luna IC, Benítez Cardoza CG, López López MDJ, López Vidal Y, Gutíerrez GRA, Rodríguez Pérez MA, Guo X. Novel protein interactions with an actin homolog (MreB) of Helicobacter pylori determined by bacterial two-hybrid system. Microbiol Res 2017; 201:39-45. [PMID: 28602400 DOI: 10.1016/j.micres.2017.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/24/2017] [Accepted: 04/22/2017] [Indexed: 02/06/2023]
Abstract
The bacterium Helicobacter pylori infects more than 50% of the world population and causes several gastroduodenal diseases, including gastric cancer. Nevertheless, we still need to explore some protein interactions that may be involved in pathogenesis. MreB, an actin homolog, showed some special characteristics in previous studies, indicating that it could have different functions. Protein functions could be realized via protein-protein interactions. In the present study, the MreB protein from H. pylori 26695 fused with two tags 10×His and GST in tandem was overexpressed and purified from Escherchia coli. The purified recombinant protein was used to perform a pull-down assay with H. pylori 26695 cell lysate. The pulled-down proteins were identified by mass spectrometry (MALDI-TOF), in which the known important proteins related to morphogenesis were absent but several proteins related to pathogenesis process were observed. The bacterial two-hybrid system was further used to evaluate the protein interactions and showed that new interactions of MreB respectively with VacA, UreB, HydB, HylB and AddA were confirmed but the interaction MreB-MreC was not validated. These results indicated that the protein MreB in H. pylori has a distinct interactome, does not participate in cell morphogenesis via MreB-MreC but could be related to pathogenesis.
Collapse
Affiliation(s)
| | - Yajuan Fu
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Cd. Reynosa Tamaulipas, Mexico
| | | | | | | | - Yolanda López Vidal
- Facultad de Medicina, División de Investigación, Universidad Nacional Autónoma de Mexico
| | - Germán Rubén Aguilar Gutíerrez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | - Mario A Rodríguez Pérez
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Cd. Reynosa Tamaulipas, Mexico
| | - Xianwu Guo
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Cd. Reynosa Tamaulipas, Mexico.
| |
Collapse
|
28
|
Seo JH, Youn HS. Eradication Therapy for Pediatric Helicobacter pyloriInfection. THE KOREAN JOURNAL OF HELICOBACTER AND UPPER GASTROINTESTINAL RESEARCH 2017. [DOI: 10.7704/kjhugr.2017.17.1.16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Ji-Hyun Seo
- Department of Pediatrics, Gyeongsang National University School of Medicine, Jinju, Korea
- Department of Pediatrics, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Hee-Shang Youn
- Department of Pediatrics, Gyeongsang National University School of Medicine, Jinju, Korea
- Department of Pediatrics, Gyeongsang Institute of Health Science, Jinju, Korea
| |
Collapse
|
29
|
Wang T, Zhang Y, Su H, Li Z, Zhang L, Ma J, Liu W, Zhou T, You W, Pan K. Helicobacter pylori antibody responses in association with eradication outcome and recurrence: a population-based intervention trial with 7.3-year follow-up in China. Chin J Cancer Res 2017; 29:127-136. [PMID: 28536491 PMCID: PMC5422414 DOI: 10.21147/j.issn.1000-9604.2017.02.05] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To identify serum biomarkers that may predict the short or long term outcomes of anti-Helicobacter pylori (H. pylori) treatment, a follow-up study was performed based on an intervention trial in Linqu County, China. METHODS A total of 529 subjects were selected randomly from 1,803 participants to evaluate total anti-H. pylori immunoglobulin G (IgG) and 10 specific antibody levels before and after treatment at 1-, 2- and 7.3-year. The outcomes of anti-H. pylori treatment were also parallelly assessed by13C-urea breath test at 45-d after treatment and 7.3-year at the end of follow-up. RESULTS We found the medians of anti-H. pylori IgG titers were consistently below cut-off value through 7.3 years in eradicated group, however, the medians declined in recurrence group to 1.2 at 1-year after treatment and slightly increased to 2.0 at 7.3-year. While the medians were significantly higher (>3.0 at 2- and 7.3-year) among subjects who failed the eradication or received placebo. For specific antibody responses, baseline seropositivities of FliD and HpaA were reversely associated with eradication failure [for FliD, odds ratio (OR)=0.44, 95% confidence interval (95% CI): 0.27-0.73; for HpaA, OR=0.32, 95% CI: 0.17-0.60]. The subjects with multiple positive specific antibodies at baseline were more likely to be successfully eradicated in a linear fashion (Ptrend=0.006). CONCLUSIONS Our study suggested that total anti-H. pylori IgG level may serve as a potential monitor of long-term impact on anti-H. pylori treatment, and priority forH. pylori treatment may be endowed to the subjects with multiple seropositive antibodies at baseline, especially for FliD and HapA.
Collapse
Affiliation(s)
- Tianyi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Huijuan Su
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhexuan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Junling Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Weidong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Tong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Weicheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Kaifeng Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
30
|
Su YL, Huang HL, Huang BS, Chen PC, Chen CS, Wang HL, Lin PH, Chieh MS, Wu JJ, Yang JC, Chow LP. Combination of OipA, BabA, and SabA as candidate biomarkers for predicting Helicobacter pylori-related gastric cancer. Sci Rep 2016; 6:36442. [PMID: 27819260 PMCID: PMC5098209 DOI: 10.1038/srep36442] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori (H. pylori ) infection is a major cause of chronic gastritis and is highly related to duodenal ulcer (DU) and gastric cancer (GC). To identify H. pylori-related GC biomarkers with high seropositivity in GC patients, differences in levels of protein expression between H. pylori from GC and DU patients were analyzed by isobaric tag for relative and absolute quantitation (iTRAQ). In total, 99 proteins showed increased expression (>1.5-fold) in GC patients compared to DU patients, and 40 of these proteins were categorized by KEGG pathway. The four human disease-related adhesin identified, AlpA, OipA, BabA, and SabA, were potential GC-related antigens, with a higher seropositivity in GC patients (n = 76) than in non-GC patients (n = 100). Discrimination between GC and non-GC patients was improved using multiple antigens, with an odds ratio of 9.16 (95% CI, 2.99-28.07; p < 0.0001) for three antigens recognized. The optimized combination of OipA, BabA, and SabA gave a 77.3% correct prediction rate. A GC-related protein microarray was further developed using these antigens. The combination of OipA, BabA, and SabA showed significant improvement in the diagnostic accuracy and the protein microarray containing above antigens should provide a rapid and convenient diagnosis of H. pylori-associated GC.
Collapse
Affiliation(s)
- Yu-Lin Su
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsiang-Ling Huang
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bo-Shih Huang
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Chung Chen
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Chien-Sheng Chen
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Hong-Long Wang
- Department of Statistics, National Taipei University, New Taipei City, Taiwan
| | - Pin-Hsin Lin
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Shu Chieh
- First Core Laboratory, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jiunn-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jyh-Chin Yang
- Department of Internal Medicine, Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Lu-Ping Chow
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
31
|
Cai H, Ye F, Michel A, Murphy G, Sasazuki S, Taylor PR, Qiao YL, Park SK, Yoo KY, Jee SH, Cho ER, Kim J, Chen SC, Abnet CC, Tsugane S, Cai Q, Shu XO, Zheng W, Pawlita M, Epplein M. Helicobacter pylori blood biomarker for gastric cancer risk in East Asia. Int J Epidemiol 2016; 45:774-81. [PMID: 27170766 DOI: 10.1093/ije/dyw078] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Incidence and mortality rates for gastric cancer, the fifth most commonly diagnosed and third most deadly cancer worldwide, are highest in East Asia. We sought to identify gastric cancer risk biomarkers among eight prospective studies from China, Japan and Korea. METHODS This pooled nested case-control study included 1608 incident non-cardia gastric cancer cases and 1958 matched controls. Pre-diagnostic antibody levels to 15 Helicobacter pylori proteins were assessed using multiplex serology. Conditional logistic regression models were used to calculate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Sero-positivity to 10 H. pylori antigens (Omp, CagA, VacA, HcpC, HP 0305, GroEL, NapA, HyuA, Cad, HpaA) was associated with a 1.29- to 3.26-fold increase in odds of gastric cancer. Omp and HP 0305 consistently remained associated with gastric cancer risk after mutually adjusting for all other markers. Sero-positivity to both Omp and HP 0305 was associated with an over 4-fold increase in gastric cancer incidence (OR, 4.09; 95% CI 3.26-5.13). When limited to only those who are CagA+ H. pylori+, Omp/HP 0305 sero-positivity remained strongly associated with an over 3-fold increase in the odds of gastric cancer (OR, 3.34; 95% CI 2.27-4.91). The results were highly consistent among the cohorts. CONCLUSIONS We have confirmed new H. pylori biomarkers that are strongly associated with gastric cancer risk, even among those infected with the known H. pylori virulence factor CagA. These results may help to design cost-efficient prevention strategies to reduce gastric cancer incidence in East Asia.
Collapse
Affiliation(s)
- Hui Cai
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center
| | - Fei Ye
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Gwen Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Shizuka Sasazuki
- Epidemiology and Prevention Group, National Cancer Center, Tokyo, Japan
| | - Philip R Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - You-Lin Qiao
- Department of Cancer Epidemiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sue K Park
- Cancer Research Institute, Department of Biomedical Sciences and Department of Preventive Medicine
| | - Keun-Young Yoo
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Yonsei University, Seoul, Korea
| | - Eo Rin Cho
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Yonsei University, Seoul, Korea
| | - Jeongseon Kim
- Division of Cancer Epidemiology and Prevention, Research Institute, National Cancer Center, Goyang, Korea
| | - Sheau-Chiann Chen
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, National Cancer Center, Tokyo, Japan
| | - Qiuyin Cai
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center
| | - Xiao-Ou Shu
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center
| | - Wei Zheng
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Meira Epplein
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center
| |
Collapse
|
32
|
Werner S, Chen H, Butt J, Michel A, Knebel P, Holleczek B, Zörnig I, Eichmüller SB, Jäger D, Pawlita M, Waterboer T, Brenner H. Evaluation of the diagnostic value of 64 simultaneously measured autoantibodies for early detection of gastric cancer. Sci Rep 2016; 6:25467. [PMID: 27140836 PMCID: PMC4853774 DOI: 10.1038/srep25467] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/18/2016] [Indexed: 02/08/2023] Open
Abstract
Autoantibodies against tumor-associated antigens (TAAs) have been suggested as biomarkers for early detection of gastric cancer. However, studies that systematically assess the diagnostic performance of a large number of autoantibodies are rare. Here, we used bead-based multiplex serology to simultaneously measure autoantibody responses against 64 candidate TAAs in serum samples from 329 gastric cancer patients, 321 healthy controls and 124 participants with other diseases of the upper digestive tract. At 98% specificity, sensitivities for the 64 tested autoantibodies ranged from 0–12% in the training set and a combination of autoantibodies against five TAAs (MAGEA4 + CTAG1 + TP53 + ERBB2_C + SDCCAG8) was able to detect 32% of the gastric cancer patients at a specificity of 87% in the validation set. Sensitivities for early and late stage gastric cancers were similar, while chronic atrophic gastritis, a precursor lesion of gastric cancer, was not detectable. However, the 5-marker combination also detected 26% of the esophageal cancer patients. In conclusion, the tested autoantibodies and combinations alone did not reach sufficient sensitivity for gastric cancer screening. Nevertheless, some autoantibodies, such as anti-MAGEA4, anti-CTAG1 or anti-TP53 and their combinations could possibly contribute to the development of cancer early detection tests (not necessarily restricted to gastric cancer) when being combined with other markers.
Collapse
Affiliation(s)
- Simone Werner
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Hongda Chen
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Julia Butt
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP &T cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
33
|
Chen XZ, Schöttker B, Castro FA, Chen H, Zhang Y, Holleczek B, Brenner H. Association of helicobacter pylori infection and chronic atrophic gastritis with risk of colonic, pancreatic and gastric cancer: A ten-year follow-up of the ESTHER cohort study. Oncotarget 2016; 7:17182-93. [PMID: 26958813 PMCID: PMC4941379 DOI: 10.18632/oncotarget.7946] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 02/09/2016] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES To assess the association of H. pylori and chronic atrophic gastritis (AG) with colonic, pancreatic and gastric cancer in a population-based prospective cohort. METHODS Serum antibodies against H. pylori in general and specific to cytotoxin-associated gene A (CagA), as well as serum pepsinogen I and II were analyzed in 9,506 men and women, aged 50-75 years in a cohort study from Saarland, Germany. Incident cases of colonic, pancreatic and gastric cancer were ascertained by record linkage with data from the Saarland Cancer Registry. RESULTS During an average follow-up of 10.6 years, 108 colonic, 46 pancreatic and 27 gastric incident cancers were recorded. There was no association between H. pylori infection and colonic cancer (HR = 1.07; 95% CI 0.73-1.56) or pancreatic cancer (HR = 1.32; 0.73-2.39), regardless of either CagA seropositivity or AG status. In contrast, CagA+ infection was associated with a strongly increased risk of gastric cancer, especially non-cardia gastric cancer, and this association was particularly pronounced in the presence of AG. Compared to people without AG and without CagA+ infection, people with both risk factors had a significantly increased risk of non-cardia gastric cancer (HR = 32.4; 7.6-137.6). CONCLUSIONS This large cohort study did not observe an association of H. pylori infection or AG with colonic or pancreatic cancer, but underlines that the vast majority of non-cardia gastric cancers arise from AG and infection with CagA+ H. pylori strains.
Collapse
Affiliation(s)
- Xin-Zu Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felipe Andres Castro
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hongda Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yan Zhang
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernd Holleczek
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Saarland Cancer Registry, Saarbrücken, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
34
|
Seo JH, Lim CW, Park JS, Yeom JS, Lim JY, Jun JS, Woo HO, Youn HS, Baik SC, Lee WK, Cho MJ, Rhee KH. Correlations between the CagA Antigen and Serum Levels of Anti-Helicobacter pylori IgG and IgA in Children. J Korean Med Sci 2016; 31:417-22. [PMID: 26955243 PMCID: PMC4779867 DOI: 10.3346/jkms.2016.31.3.417] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/10/2015] [Indexed: 11/20/2022] Open
Abstract
We tested correlations between anti-Helicobacter pylori IgG and IgA levels and the urease test, anti-CagA protein antibody, degree of gastritis, and age. In total, 509 children (0-15 years) were enrolled. Subjects were stratified as 0-4 years (n = 132), 5-9 years (n = 274), and 10-15 years (n = 103) and subjected to the urease test, histopathology, ELISA, and western blot using whole-cell lysates of H. pylori strain 51. The positivity rate in the urease test (P = 0.003), the degree of chronic gastritis (P = 0.021), and H. pylori infiltration (P < 0.001) increased with age. The median titer for anti-H. pylori IgG was 732.5 IU/mL at 0-4 years, 689.0 IU/mL at 5-9 years, and 966.0 IU/mL at 10-15 years (P < 0.001); the median titer for anti-H. pylori IgA was 61.0 IU/mL at 0-4 years, 63.5 IU/mL at 5-9 years, and 75.0 IU/mL at 10-15 years (P < 0.001). The CagA-positivity rate was 26.5% at 0-4 years, 36.5% at 5-9 years, and 46.6% at 10-15 years for IgG (P = 0.036), and 11.3% at 0-4 years, 18.6% at 5-9 years, and 23.3% at 10-15 years for IgA (P < 0.001). Anti-H. pylori IgG and IgA titers increased with the urease test grade, chronic gastritis degree, active gastritis, and H. pylori infiltration. Presence of CagA-positivity is well correlated with a high urease test grade and high anti-H. pylori IgG/IgA levels.
Collapse
Affiliation(s)
- Ji-Hyun Seo
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Chun Woo Lim
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Ji Sook Park
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Jung Sook Yeom
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Jae-Young Lim
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Jin-Su Jun
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Hyang-Ok Woo
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Hee-Shang Youn
- Department of Pediatrics, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Seung-Chul Baik
- Department of Microbiology, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Woo-Kon Lee
- Department of Microbiology, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Myung-Je Cho
- Department of Microbiology, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| | - Kwang-Ho Rhee
- Department of Microbiology, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju, Korea
| |
Collapse
|
35
|
Camargo MC, Beltran M, Conde-Glez C, Harris PR, Michel A, Waterboer T, Flórez AC, Torres J, Ferreccio C, Sampson JN, Pawlita M, Rabkin CS. Serological response to Helicobacter pylori infection among Latin American populations with contrasting risks of gastric cancer. Int J Cancer 2015; 137:3000-5. [PMID: 26178251 PMCID: PMC4817269 DOI: 10.1002/ijc.29678] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/26/2015] [Indexed: 12/20/2022]
Abstract
Gastric cancer is a rare outcome of chronic Helicobacter pylori infection. Serologic profiles may reveal bacterial, environmental and/or host factors associated with cancer risk. We therefore compared specific anti-H. pylori antibodies among populations with at least twofold differences in gastric cancer mortality from Mexico, Colombia and Chile. Our study included 1,776 adults (mean age 42 years) from three nationally representative surveys, equally divided between residents of high- and low-risk areas. Antibodies to 15 immunogenic H. pylori antigens were measured by fluorescent bead-based multiplex assays; results were summarized to identify overall H. pylori seropositivity. We used logistic regression to model associations between antibody seroreactivity and regional cancer risk (high vs. low), adjusting for country, age and sex. Both risk areas had similar H. pylori seroprevalence. Residents in high- and low-risk areas were seroreactive to a similar number of antigens (means 8.2 vs. 7.9, respectively; adjusted odds ratio, OR: 1.02, p = 0.05). Seroreactivities to Catalase and the known virulence proteins CagA and VacA were each significantly (p < 0.05) associated with residence in high-risk areas, but ORs were moderate (1.26, 1.42 and 1.41, respectively) and their discriminatory power was low (area under the curve < 0.6). The association of Catalase was independent from effects of either CagA or VacA. Sensitivity analyses for antibody associations restricted to H. pylori-seropositive individuals generally replicated significant associations. Our findings suggest that humoral responses to H. pylori are insufficient to distinguish high and low gastric cancer risk in Latin America. Factors determining population variation of gastric cancer burden remain to be identified.
Collapse
Affiliation(s)
- M. Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Rockville, Maryland, USA
| | - Mauricio Beltran
- Dirección de Redes en Salud Pública, Instituto
Nacional de Salud, Bogotá, Colombia
| | - Carlos Conde-Glez
- Centro de Investigación en Salud Poblacional, Instituto
Nacional de Salud Pública, Cuernavaca, Morelos, México
| | - Paul R. Harris
- Departamento de Gastroenterología y Nutrición
Pediátrica, Pontificia Universidad Católica de Chile, Santiago,
Chile
| | - Angelika Michel
- Division of Genome Modifications and Carcinogenesis, Infection and
Cancer Program, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Genome Modifications and Carcinogenesis, Infection and
Cancer Program, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Astrid Carolina Flórez
- Laboratorio de Parasitología, Dirección de Redes en
Salud Pública, Laboratorio Nacional de Referencia, Instituto Nacional de
Salud, Bogotá, Colombia
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas, UMAE
Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, México
City, México
| | - Catterina Ferreccio
- Crónicas Advanced Center for Chronic Diseases, Departamento
de Salud Pública, Pontificia Universidad Católica de Chile,
Santiago, Chile
| | - Joshua N. Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Rockville, Maryland, USA
| | - Michael Pawlita
- Division of Genome Modifications and Carcinogenesis, Infection and
Cancer Program, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Charles S. Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Rockville, Maryland, USA
| |
Collapse
|
36
|
Shakeri R, Malekzadeh R, Nasrollahzadeh D, Pawlita M, Pawilta M, Murphy G, Islami F, Sotoudeh M, Michel A, Etemadi A, Waterboer T, Poustchi H, Brennan P, Boffetta P, Dawsey SM, Kamangar F, Abnet CC. Multiplex H. pylori Serology and Risk of Gastric Cardia and Noncardia Adenocarcinomas. Cancer Res 2015; 75:4876-83. [PMID: 26383162 PMCID: PMC4792189 DOI: 10.1158/0008-5472.can-15-0556] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 08/09/2015] [Indexed: 02/06/2023]
Abstract
The reported associations with gastric adenocarcinoma and seropositivity to different Helicobacter pylori antigens using multiplex serology have not been consistent across studies. We aimed to investigate the association between 15 different multiplex serology antigens and the risk of gastric cardia (GCA) and gastric noncardia (GNCA) adenocarcinomas in northeastern Iran, a population with high rates of gastric adenocarcinoma. We included 272 cases of gastric adenocarcinoma (142 GCA, 103 GNCA, and 27 unspecified) and 524 controls who were individually matched to cases for age, sex, and place of residence in a population-based case-control study. Seropositivity to H. pylori was assessed using both multiplex serology and H. pylori IgG ELISA. Ninety-five percent of controls were seropositive to H. pylori. Of the 15 antibodies in the multiplex assay, 11 showed no significant association with gastric adenocarcinomas. CagA and VacA were associated with a significantly increased risk of all gastric adenocarcinoma and GNCA in multivariate models. Surprisingly, GroEL and NapA were significantly associated with a reduced risk of these tumors. Only CagA antigen was associated with significantly elevated risk of GCA. We found no associations between H. pylori seropositivity overall either by whole-cell ELISA test or multiplex serology, likely due to the high prevalence of seropositivity. Individual antigen testing showed that CagA positivity was associated with increased risk of both noncardia and cardia adenocarcinoma, which is similar to some other Asian populations, whereas two antigens were associated with lower risk of gastric cancer. This latter result was unexpected and should be retested in other populations.
Collapse
Affiliation(s)
- Ramin Shakeri
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Dariush Nasrollahzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran. Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | | | | | - Gwen Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Farhad Islami
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran. American Cancer Society, Atlanta
| | - Masoud Sotoudeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Arash Etemadi
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran. Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | | | - Hossein Poustchi
- Digestive Diseases Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | - Paolo Boffetta
- Institute for Translational Epidemiology and Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Farin Kamangar
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran. Department of Public Health Analysis, School of Community Health and Policy, Morgan State University, Baltimore, Maryland
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
37
|
Shiota S, Yamaoka Y. Biomarkers for Helicobacter pylori infection and gastroduodenal diseases. Biomark Med 2015; 8:1127-37. [PMID: 25402582 DOI: 10.2217/bmm.14.72] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori infection is a major cause of gastric cancer. Although identifying H. pylori infected subjects is the first approach for delineating the high-risk population for gastric cancer, the presence of H. pylori antibodies is not sufficient for gastric cancer screening. Among H. pylori infected subjects, only a minority of infected individuals develop gastric cancer. Serologic markers of H. pylori infection can serve as potential predictors for the development of gastric cancer. Serum or urinary H. pylori antibodies, cytotoxin-associated gene A antibodies, pepsinogen and microRNAs were reported to be associated with precancerous lesions or gastric cancer. In this review, we summarized the utilities and limitations of each strategy.
Collapse
Affiliation(s)
- Seiji Shiota
- Department of Environmental & Preventive Medicine, Oita University Faculty of Medicine, Yufu City, Oita, Japan
| | | |
Collapse
|
38
|
Kalali B, Formichella L, Gerhard M. Diagnosis of Helicobacter pylori: Changes towards the Future. Diseases 2015; 3:122-135. [PMID: 28943614 PMCID: PMC5548244 DOI: 10.3390/diseases3030122] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/18/2015] [Accepted: 06/19/2015] [Indexed: 12/19/2022] Open
Abstract
Since the first evidence demonstrating the dramatically high incidence of H. pylori infection and the subsequent medical challenges it incurs, health management of H. pylori infection has been a high priority for health authorities worldwide. Despite a decreasing rate of infection in western countries, prevalence of H. pylori infection in developing and in some industrial countries is still very high. Whereas treatment and vaccination against H. pylori is a contemporary issue in medical communities, selective treatment and prior high-throughput screening of the subject population is a major concern of health organizations. So far, diagnostic tests are either elaborative and require relatively advanced medical care infrastructure or they do not fulfill the criteria recommended by the Maastricht IV/Florence consensus report. In this review, in light of recent scientific studies, we highlight current and possible future approaches for the diagnosis of H. pylori. We point out that novel non-invasive tests may not only cover the requirements of gold standard methods in H. pylori detection but also offer the potential for risk stratification of infection in a high throughput manner.
Collapse
Affiliation(s)
- Behnam Kalali
- Institute of Medical Microbiology, Immunology and Hygiene, TU-Munich, Troger Str.30, 81675 Munich, Germany.
- ImevaX GmbH, Grillparzer Str.18, 81675 Munich, Germany.
| | - Luca Formichella
- Institute of Medical Microbiology, Immunology and Hygiene, TU-Munich, Troger Str.30, 81675 Munich, Germany.
| | - Markus Gerhard
- Institute of Medical Microbiology, Immunology and Hygiene, TU-Munich, Troger Str.30, 81675 Munich, Germany.
- ImevaX GmbH, Grillparzer Str.18, 81675 Munich, Germany.
- German Center for Infection Research (DZIF), Partner site Munich, Troger Str.30, 81675 Munich, Germany.
| |
Collapse
|
39
|
Eichelberger L, Murphy G, Etemadi A, Abnet CC, Islami F, Shakeri R, Malekzadeh R, Dawsey SM. Risk of gastric cancer by water source: evidence from the Golestan case-control study. PLoS One 2015; 10:e0128491. [PMID: 26023788 PMCID: PMC4449025 DOI: 10.1371/journal.pone.0128491] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 04/27/2015] [Indexed: 02/07/2023] Open
Abstract
Background Gastric cancer (GC) is the world’s fifth most common cancer, and the third leading cause of cancer-related death. Over 70% of incident cases and deaths occur in developing countries. We explored whether disparities in access to improved drinking water sources were associated with GC risk in the Golestan Gastric Cancer Case Control Study. Methods and Findings 306 cases and 605 controls were matched on age, gender, and place of residence. We conducted unconditional logistic regression to calculate odds ratios (ORs) and 95% confidence intervals (CI), adjusted for age, gender, ethnicity, marital status, education, head of household education, place of birth and residence, homeownership, home size, wealth score, vegetable consumption, and H. pylori seropositivity. Fully-adjusted ORs were 0.23 (95% CI: 0.05–1.04) for chlorinated well water, 4.58 (95% CI: 2.07–10.16) for unchlorinated well water, 4.26 (95% CI: 1.81–10.04) for surface water, 1.11 (95% CI: 0.61–2.03) for water from cisterns, and 1.79 (95% CI: 1.20–2.69) for all unpiped sources, compared to in-home piped water. Comparing unchlorinated water to chlorinated water, we found over a two-fold increased GC risk (OR 2.37, 95% CI: 1.56–3.61). Conclusions Unpiped and unchlorinated drinking water sources, particularly wells and surface water, were significantly associated with the risk of GC.
Collapse
Affiliation(s)
- Laura Eichelberger
- Department of Anthropology, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, United States of America; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., Bethesda, MD, 20892, United States of America
| | - Gwen Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., Bethesda, MD, 20892, United States of America
| | - Arash Etemadi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., Bethesda, MD, 20892, United States of America; Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., Bethesda, MD, 20892, United States of America
| | - Farhad Islami
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Surveillance and Health Services Research, American Cancer Society, 250 Williams St., Atlanta, GA, 30303, United States of America
| | - Ramin Shakeri
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., Bethesda, MD, 20892, United States of America
| |
Collapse
|
40
|
Murphy G, Freedman ND, Michel A, Fan JH, Taylor PR, Pawlita M, Qiao YL, Zhang H, Yu K, Abnet CC, Dawsey SM. Prospective study of Helicobacter pylori antigens and gastric noncardia cancer risk in the nutrition intervention trial cohort. Int J Cancer 2015; 137:1938-46. [PMID: 25845708 DOI: 10.1002/ijc.29543] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/12/2015] [Indexed: 12/26/2022]
Abstract
Helicobacter pylori (H. pylori) infection is the strongest known risk factor for gastric noncardia adenocarcinoma (GNCA). We used multiplex serology to determine whether seropositivity to 15 H. pylori proteins is associated with the subsequent development of noncardia gastric cancer in Linxian, China. We included 448 GNCA cases and 1242 controls from two time points within the Linxian General Population Nutrition Intervention Trial, Linxian. H. pylori multiplex seropositivity was defined as positivity to ≥4 of the 15 included antigens. Odds ratios (ORs) and 95% confidence intervals (CIs) were adjusted for major GNCA risk factors. In addition, we undertook a meta-analysis combining H. pylori multiplex serology data from both time points. H. pylori multiplex seropositivity was associated with a significant increase in risk of GNCA at one time point (1985; OR: 3.44, 95% CI: 1.91, 6.19) and this association remained significant following adjustment for H. pylori or CagA ELISA seropositivity (OR: 2.92, 95% CI: 1.56, 5.47). Combining data from both time points in a meta-analysis H. pylori multiplex seropositivity was associated with an increased risk of GNCA, as were six individual antigens: GroEL, HP0305, CagA, VacA, HcpC and Omp. CagM was inversely associated with risk of GNCA. We identified six individual antigens that confer an increase in risk of GNCA within this population of high H. pylori seroprevalence, as well as a single antigen that may be inversely associated with GNCA risk. We further determined that the H. pylori multiplex assay provides additional information to the conventional ELISA methods on risk of GNCA.
Collapse
Affiliation(s)
- Gwen Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Angelika Michel
- Division of Genome Modifications and Carcinogenesis, Infection and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jin-Hu Fan
- Department of Epidemiology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Philip R Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michael Pawlita
- Division of Genome Modifications and Carcinogenesis, Infection and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - You-Lin Qiao
- Department of Epidemiology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Han Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
41
|
Abstract
Epstein-Barr virus (EBV) infection is found in a subset of gastric cancers. Previous reviews have exclusively focused on EBV-encoded small RNA (EBER) positivity in gastric cancer tissues, but a comprehensive evaluation of other type of studies is lacking.We searched the PubMed database up to September, 2014, and performed a systematic review.We considered studies comparing EBV nucleic acids positivity in gastric cancer tissue with positivity in either adjacent non-tumor tissue of cancer patients or non-tumor mucosa from healthy individuals, patients with benign gastric diseases, or deceased individuals. We also considered studies comparing EBV antibodies in serum from cancer patients and healthy controls.Selection of potentially eligible studies and data extraction were performed by 2 independent reviewers. Due to the heterogeneity of studies, we did not perform formal meta-analysis.Forty-seven studies (8069 cases and 1840 controls) were identified. EBER positivity determined by in situ hybridization (ISH) was significantly higher in cancer tissues (range 5.0%-17.9%) than in adjacent mucosa from the same patients or biopsies from all control groups (almost 0%). High EBV nuclear antigen-1 (EBNA-1) positivity by PCR was found in gastric cancer tissues, but most were not validated by ISH or adjusted for inflammatory severity and lymphocyte infiltration. Only 4 studies tested for EBV antibodies, with large variation in the seropositivities of different antibodies in both cases and controls, and did not find an association between EBV seropositivity and gastric cancer.In summary, tissue-based ISH methods strongly suggest an association between EBV infection and gastric cancer, but PCR method alone is invalid to confirm such association. Very limited evidence from serological studies and the lack of novel antibodies warrant further investigations to identify potential risk factors of EBV for gastric cancer.
Collapse
Affiliation(s)
- Xin-Zu Chen
- From the Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany (X-ZC, HC, FAC, HB); Department of Gastrointestinal Surgery (X-ZC, J-KH); Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China (X-ZC, J-KH); and German Cancer Consortium (DKTK), Heidelberg, Germany (HB)
| | | | | | | | | |
Collapse
|
42
|
Murphy G, Michel A, Taylor PR, Albanes D, Weinstein SJ, Virtamo J, Parisi D, Snyder K, Butt J, McGlynn KA, Koshiol J, Pawlita M, Lai GY, Abnet CC, Dawsey SM, Freedman ND. Association of seropositivity to Helicobacter species and biliary tract cancer in the ATBC study. Hepatology 2014; 60:1963-71. [PMID: 24797247 PMCID: PMC4216769 DOI: 10.1002/hep.27193] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 04/29/2014] [Indexed: 12/15/2022]
Abstract
UNLABELLED Helicobacter have been detected in human bile and hepatobiliary tissue. Despite evidence that Helicobacter species promote gallstone formation and hepatobiliary tumors in laboratory studies, it remains unclear whether Helicobacter species contribute to these cancers in humans. We used a multiplex panel to assess whether seropositivity to 15 Helicobacter pylori proteins was associated with subsequent incidence of hepatobiliary cancers in the Finnish Alpha-Tocopherol, Beta-Carotene Cancer Prevention (ATBC) Study. We included 64 biliary cancers, 122 liver cancers, and 224 age-matched controls which occurred over the course of 22 years. Helicobacter pylori seropositivity was defined as those positive to ≥ 4 antigens. Odds ratios (OR) and 95% confidence intervals were adjusted for major hepatobiliary cancer risk factors. Among the controls, 88% were seropositive to H. pylori at baseline. Among those who subsequently developed hepatobiliary cancer, the prevalence of seropositivity was higher: 100% for gallbladder cancer, 97% of extrahepatic bile duct cancer, 91% of ampula of Vater cancer, 96% of intrahepatic bile duct cancer, and 94% of hepatocellular carcinoma. Although the OR for gallbladder cancer could not be calculated, the OR for the other sites were 7.01 (95% confidence interval [CI]: 0.79-62.33), 2.21 (0.19-25.52), 10.67 (0.76-150.08), and 1.20 (0.42-3.45), respectively, with an OR of 5.47 (95% CI: 1.17-25.65) observed for the biliary tract cancers combined. ORs above 1 were observed for many of the investigated antigens, although most of these associations were not statistically significant. CONCLUSION Seropositivity to H. pylori proteins was associated with an increased risk of biliary tract cancers in ATBC. Further studies are needed to confirm our findings and to determine how H. pylori might influence the risk of biliary tract cancer.
Collapse
Affiliation(s)
- Gwen Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Angelika Michel
- Division of Genome Modifications and Carcinogenesis, Research Program Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philip R. Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie J. Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | | | - Kirk Snyder
- Information Management Services, Inc., Rockville, MD, USA
| | - Julia Butt
- Division of Genome Modifications and Carcinogenesis, Research Program Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katherine A. McGlynn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jill Koshiol
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Pawlita
- Division of Genome Modifications and Carcinogenesis, Research Program Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gabriel Y. Lai
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christian C. Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sanford M. Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Neal D. Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Michel A, Pawlita M, Boeing H, Gissmann L, Waterboer T. Helicobacter pylori antibody patterns in Germany: a cross-sectional population study. Gut Pathog 2014; 6:10. [PMID: 24782915 PMCID: PMC4004453 DOI: 10.1186/1757-4749-6-10] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/17/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Helicobacter pylori infection that is usually acquired in childhood and lasts for lifetime is mostly asymptomatic but associated with severe gastrointestinal disease including cancer. During chronic infection, the gastric mucosa is histologically changing. This forces H. pylori to permanent adaptation in its gastric habitat by expression of different proteins which might be reflected in distinctive antibody patterns. METHODS To characterize dynamics of the immune response to H. pylori we analysed 1797 sera of a cross-sectional study representative for the German population (age range 1-82 years) with multiplex serology, a fluorescent bead-based antibody binding assay that allows simultaneous and quantitative detection of antibodies. Fifteen recombinant, affinity-purified H. pylori proteins (UreA, GroEL, Catalase, NapA, CagA, CagM, Cagδ, HP0231, VacA, HpaA, Cad, HyuA, Omp, HcpC and HP0305) were used as antigens. RESULTS H. pylori seroprevalence (positivity for at least three antigens) was 48% and increased with age from 12% in children <15 years to 69% in females and 90% in males >65 years. Prevalences were highest (>83%) for Omp, VacA and GroEL. For 11 proteins, seroprevalence was higher in males than females (P < 0.05) from age 55 onwards. For all antigens, the median prevalence increase per age decade was stronger in males (8.4%, range 3.8-12.9%) than females (6.1%, range 3.4-10.8%). However, among seropositives the median number of antigens recognized increased from children <15 years to individuals >65 years stronger in females (P = 0.02). Antibody reactivities to GroEL, HyuA, CagM, Catalase, NapA and UreA also increased stronger in females (average 1.7-fold/decade, SD 0.5) than in males (1.5-fold/decade, SD 0.4). CONCLUSION H. pylori antibody response accumulates qualitatively and quantitatively with age. This may reflect a lifelong stimulation of the immune response by chronically active infection.
Collapse
Affiliation(s)
- Angelika Michel
- Infections and Cancer Epidemiology (F020), Infection and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Michael Pawlita
- Department of Genome Modifications and Carcinogenesis, Infection and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition (DIFE) Potsdam-Rehbrücke, Arthur Scheunert Strasse 114-116, Nuthetal 14558, Germany
| | - Lutz Gissmann
- Department of Genome Modifications and Carcinogenesis, Infection and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Tim Waterboer
- Infections and Cancer Epidemiology (F020), Infection and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| |
Collapse
|
44
|
Oxidative DNA damage as a potential early biomarker of Helicobacter pylori associated carcinogenesis. Pathol Oncol Res 2014; 20:839-46. [PMID: 24664859 DOI: 10.1007/s12253-014-9762-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 03/06/2014] [Indexed: 02/06/2023]
Abstract
Helicobacter pylori infection is an established risk factor for gastritis, gastric ulcer, peptic ulcer and gastric cancer. CagA +ve H. pylori has been associated with oxidative DNA damage of gastric mucosa but their combined role in the development of gastric cancer is still unknown. Here we compare the combined expression of cagA and 8-hydroxy-2'-deoxyguanosine (8-OHdG) in normal, gastritis and gastric cancer tissues. Two hundred gastric biopsies from patients with dyspeptic symptoms, 70 gastric cancer tissue samples and 30 gastric biopsies from non-dyspeptic individuals (controls) were included in this study and 8-OHdG was detected by immunohistochemistry (IHC). Histological features and the presence of H. pylori infection were demonstrated by Hematoxylin and Eosin (HE), Giemsa and alcian blue-periodic acid-Schiff ± diastase (AB-PAS ± D) staining. DNA was extracted from tissues and polymerase chain reaction (PCR) performed to determine the presence of ureaseA and cagA genes of H. pylori. The results showed the presence of H. pylori in 106 (53 %) gastric biopsies out of 200 dyspeptic patients, including 70 (66 %) cases of cagA + ve H. pylori. The presence of cagA gene and high expression of 8-OHdG was highly correlated with severe gastric inflammation and gastric cancer particularly, in cases with infiltration of chronic inflammatory cells (36.8 % cagA + ve, 18 %), neutrophilic activity (47.2 %, 25.5 %), intestinal metaplasia (77.7 %, 35.7 %) and intestinal type gastric cancer (95 %, 95.4 %) (p ≤ 0.01). In conclusion, H. Pylori cagA gene expression and the detection of 8-OHdG adducts in gastric epithelium can serve as potential early biomarkers of H. Pylori-associated gastric carcinogenesis.
Collapse
|
45
|
Serum Helicobacter pylori NapA antibody as a potential biomarker for gastric cancer. Sci Rep 2014; 4:4143. [PMID: 24553293 PMCID: PMC3929916 DOI: 10.1038/srep04143] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/04/2014] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori (H. pylori) infection is strongly associated with gastric cancer. However, only a minority of infected individuals ever develop gastric cancer. This risk stratification may be in part due to differences among strains. The relationship between neutrophil-activating protein (NapA) and gastric cancer is unclear. The purpose of this study is to evaluate the significance of NapA as a biomarker in gastric cancer. We used enzyme linked immunosorbent assay (ELISA) to determine the status of H. pylori infection. Indirect ELISA method was used for detection of NapA antibody titer in the serum of H. pylori infected individuals. Unconditional logistic regressions were adopted to analyze the variables and determine the association of NapA and gastric cancer. The results of study indicated serum H. pylori NapA antibody level were associated with a reduced risk for development of gastric cancer. It may be used in conjugation with other indicators for gastric cancer detection.
Collapse
|
46
|
Tian W, Jia Y, Yuan K, Huang L, Nadolny C, Dong X, Ren X, Liu J. Serum antibody against Helicobacter pylori FlaA and risk of gastric cancer. Helicobacter 2014; 19:9-16. [PMID: 24118166 DOI: 10.1111/hel.12095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection is a major risk factor for gastric cancer (GC); however, only a minority of infected individuals develops GC. We aim to assess the association between serostatus of antibody against H. pylori flagellin A (FlaA) and risk of GC and to evaluate the value of serum FlaA antibody as a novel screening biomarker for GC risk. METHODS A hospital-based case-control study including 232 cases and 264 controls was conducted. Logistic regression was adopted to analyze the association between the serostatus of FlaA antibody and risk of GC. Serum FlaA antibody was measured by an enzyme-linked immunosorbent assay (ELISA). Receiver operating characteristic (ROC) curve was used to evaluate the screening efficacy and to identify a cutoff point of serum FlaA antibody level. RESULTS Helicobacter pylori infection was associated with an increased risk of GC (p = .007). A positive association between serum FlaA antibody and GC risk was observed in overall subjects and H. pylori-positive subjects (OR [95% CI]: 6.8 [4.3-10.7] and 6.9 [3.6-13.4], respectively; p < .001). The seropositivity of FlaA antibody was strongly related to GC risk in a dose-dependent manner (p for trend < .001). The optimal cutoff value (OD) was 0.1403, providing a sensitivity of 74.1% and a specificity of 64.4%. The area under the ROC curve (AUC) was 0.74 in overall subjects and 0.73 in H. pylori-positive subjects, respectively. CONCLUSIONS FlaA was an independent risk factor for H. pylori-related GC. Serum FlaA antibody may serve as a novel noninvasive biomarker for early detection of GC.
Collapse
Affiliation(s)
- Wenjing Tian
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang SS, Nieters A. Unraveling the interactions between environmental factors and genetic polymorphisms in non-Hodgkin lymphoma risk. Expert Rev Anticancer Ther 2014; 10:403-13. [DOI: 10.1586/era.09.194] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
48
|
Kim SH, Woo H, Park M, Rhee KJ, Moon C, Lee D, Seo WD, Kim JB. Cyanidin 3-O-glucoside reduces Helicobacter pylori VacA-induced cell death of gastric KATO III cells through inhibition of the SecA pathway. Int J Med Sci 2014; 11:742-7. [PMID: 24904230 PMCID: PMC4045794 DOI: 10.7150/ijms.7167] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 04/21/2014] [Indexed: 02/06/2023] Open
Abstract
Two key virulence factors of Helicobacter pylori are the secreted virulent proteins of vacuolating toxin A (VacA) and cytotoxin associated protein A (CagA) which lead to damages of gastric epithelial cells. We previously identified that the cyanidin 3-O-glucoside (C3G) inhibits the secretion of both VacA and CagA. In the current report, we show that C3G inhibits VacA secretion in a dose-dependent manner by inhibiting secretion system subunit protein A (SecA) synthesis. As SecA is involved in translocation of bacterial proteins, we predicted that inhibition of the SecA pathway by C3G should decrease H. pylori-induced cell death. To test this hypothesis, the human gastric cell line KATO III cells were co-cultured with H. pylori 60190 (VacA(+)/CagA(+)) and C3G. We found that C3G treatment caused a decrease in activation of the pro-apoptotic proteins caspase-3/-8 in H. pylori-infected cells leading to a decrease in cell death. Our data suggest that consumption of foods containing anthocyanin may be beneficial in reducing cell damage due to H. pylori infection.
Collapse
Affiliation(s)
- Sa-Hyun Kim
- 1. Department of Clinical Laboratory Science, Semyung University, Jaecheon 390-711, Republic of Korea
| | - Hyunjun Woo
- 2. Department of Biomedical Laboratory Science, College of Health Science, Yonsei University, Wonju 220-710, Republic of Korea
| | - Min Park
- 2. Department of Biomedical Laboratory Science, College of Health Science, Yonsei University, Wonju 220-710, Republic of Korea
| | - Ki-Jong Rhee
- 2. Department of Biomedical Laboratory Science, College of Health Science, Yonsei University, Wonju 220-710, Republic of Korea
| | - Cheol Moon
- 1. Department of Clinical Laboratory Science, Semyung University, Jaecheon 390-711, Republic of Korea
| | - Dongsup Lee
- 3. Department of Clinical Laboratory Science, Hyegeon College, Hongseong 350-702, Republic of Korea
| | - Woo Duck Seo
- 4. Department of Functional Crops, National Institute of Crop Science, Rural Development Administration, Miryang 627-803, Republic of Korea
| | - Jong Bae Kim
- 2. Department of Biomedical Laboratory Science, College of Health Science, Yonsei University, Wonju 220-710, Republic of Korea
| |
Collapse
|
49
|
Song H, Michel A, Nyrén O, Ekström AM, Pawlita M, Ye W. A CagA-independent cluster of antigens related to the risk of noncardia gastric cancer: associations between Helicobacter pylori antibodies and gastric adenocarcinoma explored by multiplex serology. Int J Cancer 2013; 134:2942-50. [PMID: 24259284 DOI: 10.1002/ijc.28621] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/28/2013] [Accepted: 11/07/2013] [Indexed: 12/23/2022]
Abstract
Because of the differences in bacterial epitopes and host characteristics, infections with Helicobacter pylori (H. pylori) induce different immune responses. We explored the possibility that certain antibody response patterns are more closely linked to gastric adenocarcinoma (GAC) than others. In a Swedish population-based case-control study, serum samples were obtained from 268 cases and 222 controls, aged 40-79 years and frequency-matched according to age and sex. We measured antibodies against 17 H. pylori proteins using multiplex serology. Associations were estimated with multivariably adjusted logistic regression models, using odds ratio (OR) with 95% confidence interval (CI) as measures of relative risk. Associations were essentially confined to non-cardia GAC but did not differ significantly between intestinal and diffuse subtypes. Point estimates for all antibodies were above unity, 15 significant with top three being CagA (OR = 9.2), GroEL (6.6), HyuA (3.6). ORs were substantially attenuated in individuals with chronic atrophic gastritis. Principal component analysis identified two significant factors: a CagA-dominant factor (antibodies against CagA, VacA and Omp as prominent markers), and a non-CagA factor (antibodies against NapA and Catalase as prominent markers). Both factors showed dose-dependent associations with non-cardia GAC risk (CagA-dominant factor, highest vs. lowest quartiles, OR = 16.2 [95% CI 4.8-54.9]; non-CagA factor OR = 5.3 [95% CI 2.1-13.3]). Overall, our results confirm that serum antibodies against different H. pylori proteins are associated with the presence of non-cardia GAC. Although strongest association is detected by antibodies against CagA and covarying proteins, a pattern of antibodies unrelated to CagA is also significantly linked to the risk of non-cardia GAC.
Collapse
Affiliation(s)
- Huan Song
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
50
|
Helicobacter pylori FliD protein is a highly sensitive and specific marker for serologic diagnosis of H. pylori infection. Int J Med Microbiol 2013; 303:618-23. [DOI: 10.1016/j.ijmm.2013.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 08/07/2013] [Accepted: 08/18/2013] [Indexed: 02/06/2023] Open
|