Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 14, 2024; 30(38): 4168-4174
Published online Oct 14, 2024. doi: 10.3748/wjg.v30.i38.4168
Is Helicobacter pylori infection protective against esophageal cancer?
Rick Maity, General Medicine, Institute of Post Graduate Medical Education and Research, Kolkata 700020, India
Arkadeep Dhali, Department of Gastroenterology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, United Kingdom
Arkadeep Dhali, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2HQ, United Kingdom
Arkadeep Dhali, Deanery of Clinical Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
Jyotirmoy Biswas, Department of General Medicine, College of Medicine and Sagore Dutta Hospital, Kolkata 700058, India
ORCID number: Rick Maity (0009-0003-5316-2329); Arkadeep Dhali (0000-0002-1794-2569).
Co-first authors: Rick Maity and Arkadeep Dhali.
Author contributions: Maity R conducted literature review and wrote the primary manuscript; Dhali A conceptualized the article; Biswas J conducted literature review and wrote the primary manuscript; All authors agreed with the final version of the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Arkadeep Dhali, MBBS, MPH, PGCert Clin Ed, FRSPH, NIHR Academic Clinical Fellow, Department of Gastroenterology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, United Kingdom. arkadipdhali@gmail.com
Received: August 1, 2024
Revised: September 10, 2024
Accepted: September 14, 2024
Published online: October 14, 2024
Processing time: 58 Days and 16.2 Hours

Abstract

Helicobacter pylori (H. pylori) infection affects a substantial proportion of the global population and causes various gastric disorders, including gastric cancer. Recent studies have found an inverse relationship between H. pylori infection and esophageal cancer (EC), suggesting a protective role against EC. This editorial focuses on the possible mechanisms underlying the role of H. pylori infection in EC and explores the role of gut microbiota in esophageal carcinogenesis and the practicality of H. pylori eradication. EC has two major subtypes: Esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC), which have different etiologies and risk factors. Gut microbiota can contribute to EC via inflammation-induced carcinogenesis, immunomodulation, lactagenesis, and genotoxin production. H. pylori infection is said to be inversely related to EAC, protecting against EAC by inducing atrophic gastritis, altering serum ghrelin levels, and triggering cancer cell apoptosis. Though H. pylori infection has no significant association with ESCC, COX-2-1195 polymorphisms and endogenous nitrosamine production can impact the risk of ESCC in H. pylori-infected individuals. There are concerns regarding a plausible increase in EC after H. pylori eradication treatments. However, H. pylori eradication is not associated with an increased risk of EC, making it safe from an EC perspective.

Key Words: Helicobacter pylori; Helicobacter pylori infection; Esophageal cancer; Esophageal squamous cell carcinoma; Esophageal adenocarcinoma; Barrett’s esophagus; Microbiota; Dysbiosis; Eradication

Core Tip: Helicobacter pylori (H. pylori) infection, while being a risk factor for gastric cancer, may afford protection against esophageal cancer (EC). The two major subtypes of EC, i.e., esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC), have different etiologies and risk factors. Recent studies have unequivocally established the inverse association between H. pylori infection and EAC, however there was no significant association with ESCC. H. pylori infection may protect against EAC by inducing atrophic gastritis, altering serum ghrelin levels, and triggering cancer cell apoptosis. Contrary to prevailing concerns, H. pylori eradication does not increase the risk of EC.



INTRODUCTION

Helicobacter pylori (H. pylori), a Gram-negative anaerobic bacterium that colonizes the stomach, affects nearly 43.9% of adults and 35.1% of children and adolescents globally[1]. Besides causing various gastric disorders such as peptic ulcer, dyspepsia, and gastritis, it is a well-established etiological and risk factor for gastric cancer[2,3]. Eradication of H. pylori infection reduces the risk of gastric cancer in infected individuals[2]. However, startling new evidence has come to light suggesting that H. pylori infection might have a protective role against esophageal cancer (EC)[2,4]. In a recent issue of the World Journal of Gastroenterology, we read with interest an article that investigated the prevalence of H. pylori infection in a retrospective cohort of EC patients from a tertiary-care hospital in Spain[5]. The study findings are consistent with recent systematic reviews suggesting an inverse relationship between H. pylori infection and the development of EC[4,5]. This editorial reviews the current demographics, etiologies, and risk factors associated with EC, as well as the available evidence and possible mechanisms underlying the role of H. pylori infection in EC. It also discusses the role of gut microbiota in esophageal carcinogenesis and feasibility of H. pylori eradication in light of the bacterium’s inverse relationship with EC.

DEMOGRAPHICS, ETIOLOGIES, AND RISK FACTORS OF EC

EC is the seventh most common cancer and sixth-largest cause of cancer-related mortalities in the world[6]. It has various subtypes: Squamous cell carcinoma (SCC), adenocarcinoma (AC), sarcoma, small cell carcinoma, and rare varieties such as lymphomas and melanomas[7]. The two major subtypes, SCC and AC, make up the vast majority of EC cases; SCC accounts for around 85% of cases, whereas AC accounts for 14%[8]. EC is more common in men, with incidence and mortality rates two- to three-times greater than in women[7,8].

The etiologies and risk factors of EC slightly vary across the two main subtypes, although the mechanisms underlying this variation have not yet been fully determined[4]. Smoking is an established risk factor for both esophageal SCC (ESCC) and AC, whereas alcohol consumption is associated only with ESCC[4,9,10]. Obesity, particularly central obesity, can lead to gastroesophageal reflux disease, which causes esophageal AC (EAC) either directly or via a pre-cancerous lesion known as Barrett’s esophagus[9,10]. A low intake of fruits and vegetables is associated with an increased susceptibility to EC, possibly due to the deficiency of vitamins and minerals[4,9]. Table 1 summarizes the risk factors for ESCC and EAC.

Table 1 Risk factors of esophageal squamous cell carcinoma and adenocarcinoma.
Squamous cell carcinoma
Adenocarcinoma
Male genderMale gender
AlcoholTobacco smoking
Tobacco smokingObesity (BMI > 25 kg/m2)
HPV infectionBarrett’s esophagus
Low intake of fruits and vegetablesGERD
Consumption of hot beverages, pickled vegetables, processed and red meatLow intake of fruits and vegetables
Consumption of processed and red meat
Low socioeconomic statusHigh socioeconomic status
Genetic factors: Howel-Evans syndrome, Fanconi anemia, Bloom syndromeGenetic factors: Familial Barrett’s esophagus
ROLE OF GUT MICROBIOTA IN ESOPHAGEAL CARCINOGENESIS

In addition to the above-mentioned risk factors, gut microbiota has been discovered to play a key role in esophageal carcinogenesis[4,11,12]. The term "gut microbiota" primarily refers to the microorganisms (mostly bacteria) that live in the human digestive system, mostly encompassing the esophageal, oral, and intestinal microbiota[12]. The esophageal microbiome can be classified into two subtypes: Type I microbiota (comprising mainly Gram-positive bacteria like Streptococcus) and Type II microbiota (mainly Gram-negative bacteria prevalent in dysbiotic states). The healthy esophageal microbiome is primarily constituted by microorganisms belonging to six phyla (Proteobacteria, Firmicutes, Bacteroidetes, Actinobacteria, Fusobacteria, and TM7), with Streptococcus as the dominant genus[13]. These microbiota achieve symbiosis with the immune system and carry out various physiological functions such as metabolism and immune maturation[12]. Any alterations in the gut microbiota can lead to the development of esophageal diseases, including EC.

Esophageal carcinogenesis can be induced by the gut microbiota via the following mechanisms: (1) Inflammation-induced carcinogenesis: Studies have shown that diet-induced alterations in the gut microbiota lead to increased levels of pro-inflammatory cytokines and immune cells[14]. Dysbalance between the gut microflora and immune system can disrupt the local microenvironment homeostasis and eventually lead to chronic inflammation. Persistent release of pro-inflammatory cytokines can activate toll-like receptors (TLR) and nucleotide-binding oligomeric domain-like receptors, triggering tumorigenesis[12]; (2) Immunomodulation: A taxonomic shift towards Type II microbiota has been observed in patients with gastroesophageal reflux disease (GERD) and BE. Lipopolysaccharide-producing Gram-negative bacteria can cause inducible nitric oxide synthase to be overexpressed, which impairs lower esophageal sphincter relaxation and increases intra-gastric pressure, thus predisposing to GERD, a risk factor for EC. Additionally, lipopolysaccharides can bind to TLR 4, leading to nuclear factor-kappa B activation and expression of cyclooxygenase-2 (COX-2), which blocks apoptosis and induces tumor cell proliferation and angiogenesis[14]. Fusobacterium nucleatum causes aberrant activation of the Wnt/β-catenin pathway, causing an increased production of chemokines that contribute to carcinogenesis and therapeutic resistance in EC[12,14]; (3) Lactagenesis: Studies have found that lactate-producing bacteria, such as Staphylococcus and Lactobacillus, are abundant in patients of GERD, BE, and EAC[14]. According to the Warburg effect, cancer cells are characterized by accelerated glycolysis and excessive lactate formation even under aerobic conditions. Lactate is thought to play a crucial role in all steps of carcinogenesis, i.e., angiogenesis, immune escape, cell migration, metastasis, and self-sufficiency of cancer cells; therefore, the goal of the Warburg effect is now believed to be the augmented production of lactate (also known as lactagenesis)[15]. By converting glucose into lactate, the lactate-producing bacteria support the survival and proliferation of cancer cells[4]. Given that lactate-producing bacteria are significantly increased in EAC, the microbial contribution to lactagenesis and its effect on esophageal cells need to be explored[14]; and (4) Genotoxin production: Certain pathogens are capable of producing compounds called genotoxins, which damage the host DNA, causing cell death, oncogene activation, or downregulation of tumor suppressor genes[16]. A variety of Gram-negative bacteria (such as Escherichia coli, Campylobacter, and H. pylori) can produce cytolethal distending toxin, which damages structural DNA and stimulates carcinogenesis[17]. Colibactin (produced by certain members of Enterobacteriaceae) and nitrosamines are genotoxins that cause DNA damage by alkylation[17,18]. H. pylori produces a toxin named cytotoxin-associated gene A (cagA) that promotes production of reactive oxygen species and causes oxidative DNA damage. While cagA-positive strains of H. pylori have been implicated in gastric cancer, their role in EC remains unknown[17].

H. PYLORI: FRIEND OR FOE IN EC?

Till date, six meta-analyses have investigated the association between H. pylori infection and EC, and they have all indicated a negative correlation. While the meta-analyses have emphatically confirmed the inverse association of H. pylori infection with EAC, no significant association could be found with ESCC[19-24]. But significant regional variances have been observed, with certain regions (such as Asia and the Middle East) showing an inverse association and others displaying a positive association with H. pylori infection, especially with cagA-positive strains; these regional variances may be attributed to dietary cultures and lifestyles that differ from one region to another[19-21]. Thus, H. pylori’s association with ESCC is not clear and needs to be explored by further population-based studies. While analyzing the association between H. pylori infection and the subtypes of EC, it would be prudent to consider the limitations of the meta-analyses, which include heterogeneity among study populations, confounding bias, and varying diagnostic criteria. The relevant information from all the meta-analyses has been summarized in Table 2.

The apparent protective role played by H. pylori in EAC can be explained by the following mechanisms: (1) Development of atrophic gastritis: H. pylori infection can cause atrophic gastritis; this lowers gastric acid secretion, reduces gastroesophageal reflux, and is postulated to protect against GERD, a risk factor for EAC[2,4]. Successful eradication of H. pylori is associated with a higher risk of GERD, especially in Asians[25]; (2) Alteration of plasma ghrelin: H. pylori may influence serum ghrelin levels, which is a key regulator of obesity and is known to stimulate cancer development and progression. H. pylori eradication is postulated to increase serum ghrelin levels, which stimulates adipogenesis and inhibits lipolysis, resulting in obesity[20,26]. By impacting the functioning of the lower esophageal sphincter, obesity predisposes to GERD, which is a risk factor for both BE and EAC[20]; and (3) Induction of cancer cell apoptosis: In vitro, H. pylori has been shown to preferentially trigger apoptosis in Barrett’s-derived EAC cells over normal esophageal cells. By increasing Fas protein expression in tumor cells, H. pylori activates the Fas-caspase pathway, which leads to apoptosis by causing fragmentation of cellular DNA[27]. Table 3 summarizes the above-mentioned protective mechanisms.

In ESCC, the role of H. pylori infection is not entirely clear. The overexpression of COX-2 can influence the inverse association between H. pylori infection and ESCC. COX-2-1195G/A, a single nucleotide polymorphism, can not only modify the transcription of COX-2 but also the risk of developing ESCC. The inverse association between H. pylori infection and ESCC (especially in the lower third of the esophagus) is enhanced in patients carrying the COX-2-1195AA homozygous genotype[28]. On the other hand, H. pylori-induced atrophic gastritis and the ensuing decrease in gastric acidity may favor the proliferation of bacteria that produce nitrosamines, a known genotoxin. Gastric nitrosamines can come in contact with the esophageal mucosa and get converted into carcinogenic compounds by cytochrome P450. Thus, endogenous nitrosamines produced as a secondary effect of H. pylori infection may be implicated in ESCC[18]. Further studies are required to establish the roles of COX-2-1195 polymorphisms, atrophic gastritis, and endogenous nitrosamines in the pathogenesis of ESCC. The mechanisms underlying the protective role of H. pylori infection against EAC have been summarized in Table 3.

Table 2 Summary of meta-analyses regarding the association between Helicobacter pylori infection and esophageal cancer.
Authors
Year of publication
Number of studies
Association of H. pylori infection with ESCC
Association of H. pylori infection with EAC
Gao et al[19]201935No significant association in the general population: OR 0.84 (95%CI: 0.64-1.09)/OR 0.74 (95%CI: 0.54-0.97); Inverse relationship in the Middle Eastern population: OR: 0.34 (95%CI: 0.22-0.52 or 0.26-0.44); Positive association with the North American population: OR: 1.83 (95%CI: 1.17-2.87)Inverse relationship: OR 0.55 (95%CI: 0.43-0.70)/OR 0.23 (95%CI: 0.15-0.36)
Nie et al[20]201428No significant association with the general population: OR 1.16 (95%CI: 0.83-1.60); Inverse association with Asian population: OR 0.74 (95%CI: 0.57–0.97); Positive association with non-Asian population: OR 1.41 (95%CI: 1.02–1.94)Inverse relationship: OR 0.57 (95%CI: 0.44-0.73)
Xie et al[21]201327No significant association in the general population: OR 0.83 (95%CI: 0.63-1.03); Inverse relationship with the East Asian population: OR 0.66 (95%CI: 0.43-0.89)Inverse relationship: OR 0.59 (95%CI: 0.51-0.68)
Islami and Kamangar[22]200819No significant association: OR 1.10 (95%CI: 0.78-1.55)Inverse relationship: OR 0.56 (95%CI: 0.46-0.68)
Zhuo et al[23]2008195No significant association: OR 0.80 (95%CI: 0.45-1.43), Z = 0.75, P > 0.05Inverse relationship: OR 0.58 (95%CI: 0.48-0.70), Z = 5.79, P < 0.01
Rokkas et al[24]200772No significant association: OR 0.85 (95%CI: 0.55-1.33), P = 0.48Inverse relationship: OR 0.52 (95%CI: 0.37-0.73), P < 0.001
Table 3 Mechanisms underlying the protective role of Helicobacter pylori infection against esophageal adenocarcinoma.
Mechanism
Description
Implications
Development of atrophic gastritisThe inflammatory processes in chronic H. pylori infection can cause gastric atrophy by loss of gastric glands and partial replacement by intestinal epithelium. This reduces the number of parietal cells which secrete hydrochloric acid, the main constituent of gastric acidLower gastric acidity reduces the risk of GERD and BE, risk factors for EAC
Alteration of plasma ghrelin levelsH. pylori-induced gastric atrophy leads to reduced gastric ghrelin production, subsequently decreasing plasma ghrelin levels. Contrastingly, eradication of H. pylori increases ghrelin levels, thus leading to obesity. Thus, H. pylori infection is inversely related to obesityGhrelin is a key regulator of obesity and has been implicated in the pathogenesis and differentiation of esophageal cancers. Obesity can predispose individuals to GERD, which is a risk factor for both BE and EAC
Induction of cancer cell apoptosisIn vitro, H. pylori induces apoptosis in Barrett’s-derived EAC cells at a higher rate than in healthy esophageal cells. H. pylori activates the Fas-caspase cascade by increasing Fas protein expression in EAC cells, which leads to apoptosis through the fragmentation of cellular DNAH. pylori infection can induce apoptosis and thus reduce the rate of esophageal cancer progression
FEASIBILITY OF H. PYLORI ERADICATION

Since the dawn of humanity, H. pylori has coexisted with us and was previously commonly prevalent in human stomachs. With the advent of antibiotics and improved sanitation, this bacterium is fast disappearing from human populations, especially in Western nations[22].

H. pylori infection can lead to peptic ulcers, which is the main indication for eradication treatment[29]. Eradication of H. pylori, besides healing chronic active gastritis and peptic ulcer disease, is an effective strategy for preventing gastric cancer[2]. It comprises a regimen of antibiotics and proton pump inhibitors and has been shown to reduce the risk of developing gastric cancer by nearly 50%[2,30].

Keeping in mind the inverse association of H. pylori infection with EAC, eradication treatments should have been associated with an increased incidence of EC following successful eradication. However, recent cohort studies have debunked this hypothesis, proving that H. pylori eradication does not increase the risk of EC[29,31]. A possible explanation is that eradication treatment cannot reverse the chronic gastric atrophy caused by H. pylori infection, which protects against both BE and EAC by causing diminished gastric acid secretion and gastroesophageal reflux[29,31]. The adoption of healthier lifestyles and dietary habits may be another factor contributing to the reduced incidence of EAC post-H. pylori eradication[29]. Thus, H. pylori eradication treatment is safe from an EC perspective, and there is no reason to withhold H. pylori eradication in cases where it is indicated[29,31]. Nevertheless, large multicentric studies with long follow-up periods are required to thoroughly evaluate this topic.

CONCLUSION

H. pylori, a Gram-negative bacterium that causes various gastric disorders, shows an inverse relationship with EC. EC has two major subtypes, i.e., EAC and ESCC, which have slightly different etiologies and risk factors. Additionally, gut microbiota can contribute to carcinogenesis in four ways: Inflammation-induced carcinogenesis, immunomodulation, lactagenesis, and genotoxin production. The inverse association of H. pylori infection with EAC has been unequivocally confirmed, but no significant association has been observed with ESCC. H. pylori infection protects against EAC by inducing atrophic gastritis, influencing serum ghrelin levels, and triggering cancer cell apoptosis. While COX-2-1195 polymorphisms can modify the inverse association between H. pylori infection and ESCC, endogenous nitrosamines produced as a secondary impact of H. pylori-induced atrophic gastritis may increase the risk of ESCC. There are concerns regarding a plausible increase in EC after H. pylori eradication treatments. Fortunately, H. pylori eradication is not associated with an increased risk of EC as determined by recent cohort studies, possibly because H. pylori-induced atrophic gastritis cannot be reversed by eradication treatment. Thus, H. pylori infection affords a degree of protection against the development of EC (especially EAC), and H. pylori eradication treatment is safe from an EC perspective.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: United Kingdom

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Pan YB S-Editor: Li L L-Editor: A P-Editor: Wang WB

References
1.  Chen YC, Malfertheiner P, Yu HT, Kuo CL, Chang YY, Meng FT, Wu YX, Hsiao JL, Chen MJ, Lin KP, Wu CY, Lin JT, O'Morain C, Megraud F, Lee WC, El-Omar EM, Wu MS, Liou JM. Global Prevalence of Helicobacter pylori Infection and Incidence of Gastric Cancer Between 1980 and 2022. Gastroenterology. 2024;166:605-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 19]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
2.  Malfertheiner P, Megraud F, O'Morain CA, Gisbert JP, Kuipers EJ, Axon AT, Bazzoli F, Gasbarrini A, Atherton J, Graham DY, Hunt R, Moayyedi P, Rokkas T, Rugge M, Selgrad M, Suerbaum S, Sugano K, El-Omar EM; European Helicobacter and Microbiota Study Group and Consensus panel. Management of Helicobacter pylori infection-the Maastricht V/Florence Consensus Report. Gut. 2017;66:6-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1710]  [Cited by in F6Publishing: 1817]  [Article Influence: 259.6]  [Reference Citation Analysis (1)]
3.  Sugano K, Tack J, Kuipers EJ, Graham DY, El-Omar EM, Miura S, Haruma K, Asaka M, Uemura N, Malfertheiner P; faculty members of Kyoto Global Consensus Conference. Kyoto global consensus report on Helicobacter pylori gastritis. Gut. 2015;64:1353-1367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 899]  [Cited by in F6Publishing: 1026]  [Article Influence: 114.0]  [Reference Citation Analysis (0)]
4.  Li Y, Wei B, Xue X, Li H, Li J. Microbiome changes in esophageal cancer: implications for pathogenesis and prognosis. Cancer Biol Med. 2023;21:163-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
5.  López-Gómez M, Morales M, Fuerte R, Muñoz M, Delgado-López PD, Gómez-Cerezo JF, Casado E. Prevalence of Helicobacter pylori infection among patients with esophageal carcinoma. World J Gastroenterol. 2024;30:3479-3487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (6)]
6.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50630]  [Cited by in F6Publishing: 51891]  [Article Influence: 17297.0]  [Reference Citation Analysis (122)]
7.  Liu CQ, Ma YL, Qin Q, Wang PH, Luo Y, Xu PF, Cui Y. Epidemiology of esophageal cancer in 2020 and projections to 2030 and 2040. Thorac Cancer. 2023;14:3-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 70]  [Reference Citation Analysis (0)]
8.  Morgan E, Soerjomataram I, Rumgay H, Coleman HG, Thrift AP, Vignat J, Laversanne M, Ferlay J, Arnold M. The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040: New Estimates From GLOBOCAN 2020. Gastroenterology. 2022;163:649-658.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 300]  [Article Influence: 150.0]  [Reference Citation Analysis (0)]
9.  Domper Arnal MJ, Ferrández Arenas Á, Lanas Arbeloa Á. Esophageal cancer: Risk factors, screening and endoscopic treatment in Western and Eastern countries. World J Gastroenterol. 2015;21:7933-7943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 554]  [Cited by in F6Publishing: 659]  [Article Influence: 73.2]  [Reference Citation Analysis (13)]
10.  Castro C, Peleteiro B, Lunet N. Modifiable factors and esophageal cancer: a systematic review of published meta-analyses. J Gastroenterol. 2018;53:37-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
11.  Chen C, Chen L, Lin L, Jin D, Du Y, Lyu J. Research progress on gut microbiota in patients with gastric cancer, esophageal cancer, and small intestine cancer. Appl Microbiol Biotechnol. 2021;105:4415-4425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
12.  Zhou J, Sun S, Luan S, Xiao X, Yang Y, Mao C, Chen L, Zeng X, Zhang Y, Yuan Y. Gut Microbiota for Esophageal Cancer: Role in Carcinogenesis and Clinical Implications. Front Oncol. 2021;11:717242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
13.  Dong L, Yin J, Zhao J, Ma SR, Wang HR, Wang M, Chen W, Wei WQ. Microbial Similarity and Preference for Specific Sites in Healthy Oral Cavity and Esophagus. Front Microbiol. 2018;9:1603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 46]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
14.  Muszyński D, Kudra A, Sobocki BK, Folwarski M, Vitale E, Filetti V, Dudzic W, Kaźmierczak-Siedlecka K, Połom K. Esophageal cancer and bacterial part of gut microbiota - A multidisciplinary point of view. Front Cell Infect Microbiol. 2022;12:1057668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
15.  San-Millán I, Brooks GA. Reexamining cancer metabolism: lactate production for carcinogenesis could be the purpose and explanation of the Warburg Effect. Carcinogenesis. 2017;38:119-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 256]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
16.  Grover K, Gregory S, Gibbs JF, Emenaker NJ. A discussion of the gut microbiome's development, determinants, and dysbiosis in cancers of the esophagus and stomach. J Gastrointest Oncol. 2021;12:S290-S300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
17.  Dan W, Peng L, Yan B, Li Z, Pan F. Human Microbiota in Esophageal Adenocarcinoma: Pathogenesis, Diagnosis, Prognosis and Therapeutic Implications. Front Microbiol. 2021;12:791274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
18.  Ye W, Held M, Lagergren J, Engstrand L, Blot WJ, McLaughlin JK, Nyrén O. Helicobacter pylori infection and gastric atrophy: risk of adenocarcinoma and squamous-cell carcinoma of the esophagus and adenocarcinoma of the gastric cardia. J Natl Cancer Inst. 2004;96:388-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 243]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
19.  Gao H, Li L, Zhang C, Tu J, Geng X, Wang J, Zhou X, Jing J, Pan W. Systematic Review with Meta-analysis: Association of Helicobacter pylori Infection with Esophageal Cancer. Gastroenterol Res Pract. 2019;2019:1953497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
20.  Nie S, Chen T, Yang X, Huai P, Lu M. Association of Helicobacter pylori infection with esophageal adenocarcinoma and squamous cell carcinoma: a meta-analysis. Dis Esophagus. 2014;27:645-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 61]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
21.  Xie FJ, Zhang YP, Zheng QQ, Jin HC, Wang FL, Chen M, Shao L, Zou DH, Yu XM, Mao WM. Helicobacter pylori infection and esophageal cancer risk: an updated meta-analysis. World J Gastroenterol. 2013;19:6098-6107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 108]  [Cited by in F6Publishing: 108]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
22.  Islami F, Kamangar F. Helicobacter pylori and esophageal cancer risk: a meta-analysis. Cancer Prev Res (Phila). 2008;1:329-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 249]  [Article Influence: 16.6]  [Reference Citation Analysis (1)]
23.  Zhuo X, Zhang Y, Wang Y, Zhuo W, Zhu Y, Zhang X. Helicobacter pylori infection and oesophageal cancer risk: association studies via evidence-based meta-analyses. Clin Oncol (R Coll Radiol). 2008;20:757-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 22]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
24.  Rokkas T, Pistiolas D, Sechopoulos P, Robotis I, Margantinis G. Relationship between Helicobacter pylori infection and esophageal neoplasia: a meta-analysis. Clin Gastroenterol Hepatol. 2007;5:1413-1417, 1417.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 137]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
25.  Xie T, Cui X, Zheng H, Chen D, He L, Jiang B. Meta-analysis: eradication of Helicobacter pylori infection is associated with the development of endoscopic gastroesophageal reflux disease. Eur J Gastroenterol Hepatol. 2013;25:1195-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
26.  Pradhan G, Samson SL, Sun Y. Ghrelin: much more than a hunger hormone. Curr Opin Clin Nutr Metab Care. 2013;16:619-624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 192]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
27.  Jones AD, Bacon KD, Jobe BA, Sheppard BC, Deveney CW, Rutten MJ. Helicobacter pylori induces apoptosis in Barrett's-derived esophageal adenocarcinoma cells. J Gastrointest Surg. 2003;7:68-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
28.  Hu HM, Kuo CH, Lee CH, Wu IC, Lee KW, Lee JM, Goan YG, Chou SH, Kao EL, Wu MT, Wu DC. Polymorphism in COX-2 modifies the inverse association between Helicobacter pylori seropositivity and esophageal squamous cell carcinoma risk in Taiwan: a case control study. BMC Gastroenterol. 2009;9:37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
29.  Wiklund AK, Santoni G, Yan J, Radkiewicz C, Xie S, Birgisson H, Ness-Jensen E, von Euler-Chelpin M, Kauppila JH, Lagergren J. Risk of Esophageal Adenocarcinoma After Helicobacter pylori Eradication Treatment in a Population-Based Multinational Cohort Study. Gastroenterology. 2024;167:485-492.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
30.  Li WQ, Zhang JY, Ma JL, Li ZX, Zhang L, Zhang Y, Guo Y, Zhou T, Li JY, Shen L, Liu WD, Han ZX, Blot WJ, Gail MH, Pan KF, You WC. Effects of Helicobacter pylori treatment and vitamin and garlic supplementation on gastric cancer incidence and mortality: follow-up of a randomized intervention trial. BMJ. 2019;366:l5016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 133]  [Article Influence: 26.6]  [Reference Citation Analysis (0)]
31.  Doorakkers E, Lagergren J, Santoni G, Engstrand L, Brusselaers N. Helicobacter pylori eradication treatment and the risk of Barrett's esophagus and esophageal adenocarcinoma. Helicobacter. 2020;25:e12688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]