Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Sep 15, 2021; 13(9): 1099-1108
Published online Sep 15, 2021. doi: 10.4251/wjgo.v13.i9.1099
Overgrowth of Lactobacillus in gastric cancer
Zhi-Peng Li, Lin-Lin Lu, Li-Li Wang, Lin Xu, Quan-Jiang Dong, Department of Gastroenterology and Central Laboratories, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
Ju-Xin Liu, Clinical Laboratories, Qingdao Municipal Hospital, Qingdao 266071, Shandong Province, China
Zong-Hao Guo, Department of Clinical Medicine, Jining Medical University, Jining 272000, Shandong Province, China
ORCID number: Zhi-Peng Li (0000-0002-9800-5293); Ju-Xin Liu (0000-0002-9035-4237); Lin-Lin Lu (0000-0003-4884-6032); Li-Li Wang (0000-0002-3607-0786); Lin Xu (0000-0003-3098-8251); Zong-Hao Guo (0000-0002-3893-8622); Quan-Jiang Dong (0000-0002-5226-7853).
Author contributions: Li ZP and Guo ZH collected the literature; Li ZP and Lu LL analyzed the data and wrote the manuscript; Liu JX, Wang LL, and Xu L reviewed the literature and wrote the discussion part of the manuscript; Dong QJ designed the research and supervised the manuscript; all authors reviewed the manuscript and approved the final version of the manuscript.
Supported by The National Natural Science Foundation of China, No. 31870777.
Conflict-of-interest statement: The authors declare no conflict of interests 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: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Quan-Jiang Dong, MD, PhD, Chief Doctor, Professor, Department of Gastroenterology and Central Laboratories, Qingdao Municipal Hospital, Qingdao University, No. 5 Donghai Middle Road, Qingdao 266071, Shandong Province, China. allyking114@126.com
Received: February 22, 2021
Peer-review started: February 22, 2021
First decision: April 19, 2021
Revised: April 30, 2021
Accepted: July 23, 2021
Article in press: July 23, 2021
Published online: September 15, 2021

Abstract

Dysbiosis of the gastric microbiome is involved in the development of gastric cancer (GC). A number of studies have demonstrated an increase in the relative abundance of Lactobacillus in GC. In this review, we present data that support the overgrowth of Lactobacillus in GC from studies on molecular and bacterial culture of the gastric microbiome, discuss the heterogenic effects of Lactobacillus on the health of human stomach, and explore the potential roles of the overgrowth of Lactobacillus in gastric carcinogenesis. Further studies are required to examine the association between Lactobacillus and GC at strain and species levels, which would facilitate to elucidate its role in the carcinogenic process.

Key Words: Lactobacillus, Gastric cancer, Gastritis, Microbiome, Next generation sequencing

Core Tip: Many strains of Lactobacillus have been used as probiotics in the clinical setting. However, recent molecular analyses of the gastric microbiome demonstrate a close association between an increased abundance of Lactobacillus and gastric cancer. In this paper, we review the current understanding of heterogenic effects of Lactobacillus on the health of the human stomach and discuss potential roles of the overgrowth of Lactobacillus in the gastric carcinogenesis.



INTRODUCTION

Gastric cancer (GC) is a major health burden worldwide, which is the fifth most common cancer worldwide[1]. Host genetic variations, environmental factors, and microbial infection participate in the development of GC[2-6]. Helicobacter pylori (H. pylori), which is highly prevalent in the human population, is classified as a class I carcinogen[7]. The pathogen has been suggested to act as a trigger of the carcinogenic cascade of the stomach.

The human stomach harbors a number of bacteria. These bacteria may participate in the development of GC through production of carcinogenic compounds[8]. Recent molecular analysis reveals bacteria of the gastric microbiome are mainly from the phyla Proteobacteria, Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria[9]. In GC, the microbiome was altered substantially[10,11]. Data of studies on gnotobiotic mice demonstrate that the presence of artificial microbiota in the stomach promotes the development of H. pylori induced cancer[12]. Thus, dysbiosis of the gastric microbiome is likely involved in the development of GC[13].

A prominent change of the gastric microbiome is the altered relative abundance of certain bacteria in GC[14]. Results from a number of studies show an increase in the abundance of Lactobacillus in GC. Considering the wide use of Lactobacillus in the clinical setting as a probiotic organism, it is surprising that the bacterium is enriched in GC. It is of great importance to clarify whether the enrichment of Lactobacillus is involved in the gastric carcinogenesis[15]. In this paper, we review the literature published recently to verify the finding that Lactobacillus is enriched in GC, discuss possible mechanisms for the overgrowth of Lactobacillus, and explore potential roles of Lactobacillus in the carcinogenesis of the stomach.

INCREASED ABUNDANCE OF LACTOBACILLUS IN GC

In GC, the gastric microbiome shows a substantial alteration[16]. Bacteria with varied abundance are potentially associated with cancer development. A close association has been suggested between Lactobacillus and GC[10]. To verify this notion, we review the literature involving studies designed to identify compositional changes of gastric mucosa associated microbiota in GC. The key words “gastric cancer” and “microbiota” or “microbiome” were used to search the PubMed database from January 2014 to January 2021. A total of 20 articles were included for further analyses, after exclusion of the articles published in non-English languages, letters, case reports, reviews, or conference reports. Studies were also excluded when the microbiome examined was not from the stomach (i.e., oral or fecal microbiome).

According to experimental design, these studies were divided into three categories. Category I consists of three case-control studies comparing GC with healthy controls (normal stomach) (Table 1)[17-19], category II includes nine case-control studies comparing GC with non-cancerous stomach (Table 2)[10,16,20-26], and category III is composed of eight studies observing the compositional changes of the gastric microbiome in multiple stages of the carcinogenic process of the stomach, including normal stomach, intestinal metaplasia, intraepithelial neoplasia, and GC (Table 3)[11,14,27-32].

Table 1 Relative abundance of Lactobacillus from category I studies.
Ref.
HC (n)
GC (n)
Lactobacillus
Fold change
LDA score
Gunathilake et al[17] 88268-
Gunathilake et al[18]288268+47.63.2
Cavadas et al[19] 16483-
Table 2 Changes in relative abundance of Lactobacillus from category II studies.
Ref.
Control group (n)
GC group (n)
Lactobacillus
Fold change
LDA score
Jo et al[20]29 (NC)34-
Sohn et al[21]5 (NC)7+194.6/1.9
Wang et al[16]6 (CG)6+
Ferreira et al[22]81 (CG)54+188.04.4
Hu et al[23]5 (CG)6-
Wu et al[24]32 (SG)18+2.5> 2.4
Wang et al[10]60 (CG)60-> 3.6
Castaño-Rodríguez et al[25]20 (FD)12+17.73.6
Spiegelhauer et al[26]22 (FD)12+148.1
Table 3 Variations in relative abundance of Lactobacillus from category III studies.
Ref.
HCSGAGIMINGCLactobacillusFold change (GC vs SG/CG)LDA score
Aviles-Jimenez et al[27]555+
Eun et al[28]101011-
Li et al[29]8997-
Coker et al[30]21231720+
Hsieh et al[11]9711+81.0/91.1
Wang et al[14]3021272529+22.04.1
Dang et al[31]171330-
Gantuya et al[32]2020404048+17.94.4

Studies from category I have demonstrated that Lactobacillus was rarely present in the gastric microbiome of healthy controls (Table 1)[17-19]. The abundance of Lactobacillus was dramatically increased in GC[18]. However, results of the other two studies do not support this finding. Analyses of category II studies reveal that 7/9 studies show an enrichment of Lactobacillus in GC in comparison with chronic gastritis, functional dyspepsia, or non-cancer conditions. The fold change in the abundance of Lactobacillus varies from 1.9 to 194.6.

Results from the majority of (5/8) category III studies demonstrated that the relative abundance of Lactobacillus is significantly increased in GC compared to that in precancerous lesions, chronic gastritis, or normal mucosa (Table 3). There was a trend that the relative abundance of Lactobacillus was enhanced gradually from the normal mucosa to cancer of the stomach, suggesting a close association between the bacterium and the carcinogenic process[14,32]. Two studies from category III have employed the random forest model, a machine learning algorithm, to identify bacteria members of gastric microbiome capable of discriminating cancer from non-cancer diseases. The results demonstrated that a set of bacteria including Lactobacillus has high capacity to distinguish GC from precancerous diseases[14].

Compositional changes of the gastric microbiome are a distinctive feature in GC. However, bacteria with altered abundance in GC varied greatly among studies. Few bacterial members in the gastric microbiome, except for Lactobacillus, have been universally identified to have a varied abundance in GC. An increase in the relative abundance of Lactobacillus suggests the overgrowth of Lactobacillus in GC.

OVERGROWTH OF LACTOBACILLUS IN GASTRIC DISEASES

The human stomach is a hostile niche for the inhibition of bacteria. The gastric fluid has a median pH value of 1.4 which is able to kill most ingested bacteria[13]. Rapid peristalsis and epithelial regeneration further prohibit bacteria colonization of the gastric mucosa[33]. In healthy individuals, the stomach was once considered to be microbiologically sterile. Data from recent molecular analyses, however, demonstrated that gastric fluid and the luminal surface of the stomach harbor a low microbial population. The total bacterial count is lower than 1 × 104 per millimeter in gastric fluid, which is much lower than that in the intestine[13]. The species richness, which is the number of bacteria species in the stomach, is pretty low in contrast to the intestinal microflora[34].

A reduction in gastric acid may lead to bacterial overgrowth in the stomach. In pernicious anemia caused by autoimmune gastritis, loss of parietal cell results in decreased acid secretion from the stomach[35]. Distal gastrectomy also has lowered gastric acid output[35]. In these conditions, bacterial overgrowth occurs in the stomach[35]. As the intragastric pH rises, increased number of bacteria survive and proliferate to the point of being cultivated from the stomach[36]. In addition to the pH value, the total number of bacterial count in the stomach is also dependent on the time that bacteria are exposed to an environment at a pH lower than 4.0. Bacterial growth is inhibited when the pH is < 4.0 for a few hours a day[37-39]. Bacteria colonizing in the stomach are mainly from ingestion, the migration of commensal bacteria in the oropharyngeal cavity, or otherwise of the bowel through enterogastric reflux[35]. Bacteria moving into the stomach suffer from acid stress. Those finally colonizing the human stomach must possess properties of acid resistance or tolerance. There is, however, a substantial variation in the ability of different microorganisms to survive in the acidic environment. Colonization of the human stomach by H. pylori is dependent on its urease activity that produces an alkaline buffer zone of ammonia around cells. Yersinia enterocolitica also possesses a urease gene cluster, making it possible to grow in the stomach. Lactic acid bacteria are usually capable of proliferating in the weak acidic environment. Many lactic acid bacteria possess the urease activity. Some enteric pathogens have the ability to adapt themselves and consequently stay alive in different ways in this acidic condition. Recent studies demonstrate that prolonged treatment with proton pump inhibitors results in elevated pH of the gastric lumen, which would cause bacterial overgrowth[40]. Particularly, Streptococcus, one of the lactic acid bacteria, is associated with the use of proton pump inhibitors[41].

A prominent pathophysiological change in the stomach is a gradual drop in the gastric secretion during the carcinogenic process of the stomach. It has been found that patients with GC had reduced gastric acid secretion caused by oxyntic atrophic gastritis[42]. There is varying degrees of impaired acid secretion being reported in GC[43]. The pH value of gastric fluid may increase to 3.5-7.0. The acid hypo-secretion in intestinal-type GC has been further confirmed by the determination of serum biomarkers[44-47]. Reduction in the acid output is likely to be the major cause of the enrichment of Lactobacillus in GC. Lactobacillus, a lactic acid bacterium, is capable of tolerating acid and proliferating under weak acid conditions[48]. An elevation of the pH value in the stomach during proton pump inhibitor treatments increases the relative abundance of Lactobacillus[49]. With the increase in pH during the carcinogenic process, Lactobacillus may thus grow over other bacteria and become enriched in the cancerous stomach.

HETEROGENEOUS EFFECT OF LACTOBACILLUS ON HOST

The genus of Lactobacillus is a common member of gut microbiota. In many vertebrates (for example, mice, rats, and chickens) and insects, the genus is the most abundant bacteria in the gastric microbiota[50-53]. In humans, however, the frequency of Lactobacillus detected in the stomach is about 25.5%[54]. Prolonged symbiosis of the bacterium with animals promotes the mutualism and commensalism. In other words, Lactobacillus provides benefits for its host. Studies on the human stomach demonstrate that Lactobacillus possesses antimicrobial activities, alleviates mucosal inflammation, modulates mucosal immunity, and even has anti-cancer effects.

Benefits of Lactobacillus for its host

It has been found that the co-existence rate of Lactobacillus spp. and the gastric pathogen H. pylori is pretty low in the gastric mucosa of symptomatic patients with gastrointestinal discomfort[54,55]. Two studies involving 197 and 427 patients with gastrointestinal discomfort, respectively, revealed that the co-existence rate of these two bacteria is only 8%[55] and 6.1%[54], respectively. In contrast, H. pylori has been detected in approximately half of patients in both studies. These data indicate the pathogen exclusion effect of Lactobacillus. Clinical trials with supplement of strains of Lactobacillus may increase the eradication rate of H. pylori. In addition, Lactobacillus could reduce the incidence of adverse events caused by anti-H. pylori therapies, alleviating disease-related clinical symptoms[56,57]. Thus, Lactobacillus benefits its host through anti-H. pylori effects. Further studies reveal that Lactobacillus can create competitive conditions, inhibiting pathogen adherence to mucus and epithelial layer of the stomach[58]. L. plantarum strains and L. rhamnosus strains increase the expression of MUC2 and MUC3 genes by epithelial cells, inhibiting the binding of pathogens[59]. Many Lactobacillus species produce bacteriocin which is capable of killing pathogens[60].

Lactobacillus alleviates mucosal inflammation of the stomach[61]. The microbe modulates signaling pathways, which results in a reduction in the expression of IL-8[62]. Some probiotic strains of Lactobacillus and their metabolic products stimulate dendritic cells, leading to activation of anti-inflammatory pathways, and inhibition the inflammatory function of Th1 and Th17 lymphocytes[63]. The bacterium may change cytokine profiles, which increases the production of secretory IgA by B cells[62]. These findings suggest that Lactobacillus possesses an anti-inflammatory effect.

Lactobacillus shows anti-cancer activities. L. plantarum up-regulates the expression of PTEN, Bax, and TLR4 and down-regulates the AKT genes, and then promote the apoptosis of AGS cells[64]. In in vitro studies, L. gasseri causes a significant decrease in the expression of oncogenes including Bcl-2, β-catenin, integrin α5, and integrin β1. Overall, current evidence supports the notion that certain strains of Lactobacillus provide benefits for the human stomach through their properties of pathogen exclusion, maintaining the gastric barrier function, anti-inflammation, and anti-cancer effects.

Carcinogenic potentials of Lactobacillus

It is widely recognized that Lactobacillus has probiotic effects, which is capable of benefiting the human health at an adequate quantity. Recently, however, data supporting the bacterium occasionally shows carcinogenic potentials. In an INS-GAS transgenic mouse model, colonization with artificial microbiota (L. murinus ASF361, Clostridium ASF356, and Bacteroides ASF519) could promote the development of gastric intraepithelial neoplasia[12]. This is associated with a strong upregulation of pro-inflammatory genes and oncogenes[12]. These findings suggest a potential role of Lactobacillus in gastric carcinogenesis. Lactobacillus produces lactate, which may serve as a fuel for the tumor cells, accelerating their growth[15]. In a need of rapid growth, tumor cells rely primarily on anerobic glycolysis rather than oxidative phosphorylation, which provides more lactic acid[65,66]. The lactate concentration in glycolytic tumors is approximately 10 times higher than the basal lactate production in an average human[67-70]. It has been assumed that an increased production of lactic acid by Lactobacillus promotes the growth of tumor cells. Furthermore, Lactobacillus has been shown to reduce nitrate to nitrite, which leads to the formation of large amounts of N-nitroso compounds[71,72]. These compounds promote mutagenesis, angiogenesis, and protooncogene expression by epithelial cells, thus leading to the occurrence of GC. Lactobacillus, like other lactic acid bacteria, is a very potent inducer of reactive oxygen species in cultured cells and in vivo[73]. These reactive oxygen species induce intensively DNA damages[73]. It has been shown that lactic acid bacteria enhance the expression of NANOG, a known multipotency marker, turning human adult fibroblasts into multipotent cells[74]. This supports a direct cancer promoting activity of Lactobacillus.

RELATIONSHIP BETWEEN LACTOBACILLUS OVERGROWTH AND GASTRIC CARCINOGENESIS

Molecular analyses of the gastric microbiome have demonstrated overgrowth of Lactobacillus in GC. Recently, overgrowth of the bacterium has been found in cervical cancer[75]. L. iners was more abundant in cervical cancer and precancerous lesions. Similarly, some species of Lactobacillus have been found to have an increased abundance in GC[21]. These data suggest a close association between Lactobacillus and cancer. Considering that foods and drugs containing strains of Lactobacillus are widely used in daily life and clinical settings, it is of immense importance to clarify whether overgrowth of Lactobacillus participates in the development of GC.

The genus Lactobacillus consists of more than 100 species. The biological behaviors vary substantially among different species. For instances, certain Lactobacillus strains possess urease activity which facilitates the inhibition of acidic environments of the stomach. In the normal human stomach, in contrast to that of mice, Lactobacillus was barely isolated[76,77]. To clarify the role of Lactobacillus in gastric carcinogenesis, a key issue remains to be resolved is whether the increased abundance of Lactobacillus detected by molecular analyses results from the overgrowth of a particular species of Lactobacillus. Results of preliminary studies, however, found that a diverse species of Lactobacillus have been isolated from the stomach of patients with GC[78-81]. These species include L. paracasei, L. fermentum, L. rhamosus, L. salivarius, L. delbreuckii, and L. acidophilus. These findings suggest that a great number of Lactobacillus species are capable of surviving and proliferating in the cancerous stomach. Although Lactobacillus was more frequently isolated from GC compared with non-cancer conditions, non-particular species has been found to be associated with GC[78-81]. Therefore, it appears that the overgrowth of Lactobacillus in GC is not caused by a particular species. Biological roles of Lactobacillus vary greatly among strains, no matter which species it is. Further studies are required to elucidate whether virulent strains, which have increased carcinogenic potentials mentioned above, are enriched in GC. Such studies would be beneficial for clarifying the role of Lactobacillus in GC.

Interactions between bacteria play a vital role in maintaining homeostasis of the microbiome. The co-occurrence network analyses demonstrate that Lactobacillus interacts with other bacterial members of the gastric microbiome[10]. Thus, overgrowth of Lactobacillus may alter the structure of the gastric microbiome through interactions with other bacteria, possibly leading to dysbiosis. This results in changes in the community behaviors, which enhance the carcinogenic potentials of the gastric microbiome. Thus, it is likely that Lactobacillus itself does not play any significant role in the carcinogenesis. Instead, it indirectly promotes carcinogenesis through altering the gastric microbial community. Bacterial overgrowth is commonly found in GC[13]. Overgrowth of Lactobacillus, thus, may merely reflect an altered state of the gastric microbiome that has many overgrown bacterial members. That is, the overgrowth of Lactobacillus plays little, if any, roles in the development of GC.

CONCLUSION

In summary, there is strong evidence supporting Lactobacillus overgrowth in GC. It may promote the development of GC directly through metabolic products, or indirectly through altering the microbial community. However, the possibility that it just acts as a marker for bacterial overgrowth cannot be excluded. To clarify the role of Lactobacillus in the development of GC, further studies at strain and species levels are indicated.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Microbiology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B, B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Hudler P, Kosuga T, Mansilla-Vivar R, Suzuki H S-Editor: Zhang H L-Editor: Wang TQ P-Editor: Li JH

References
1.  Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Prz Gastroenterol. 2019;14:26-38.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Mommersteeg MC, Yu J, Peppelenbosch MP, Fuhler GM. Genetic host factors in Helicobacter pylori-induced carcinogenesis: Emerging new paradigms. Biochim Biophys Acta Rev Cancer. 2018;1869:42-52.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev. 2014;23:700-713.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Wiseman M. The second World Cancer Research Fund/American Institute for Cancer Research expert report. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Proc Nutr Soc. 2008;67:253-256.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Moss SF. The Clinical Evidence Linking Helicobacter pylori to Gastric Cancer. Cell Mol Gastroenterol Hepatol. 2017;3:183-191.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Xu W, Liu Z, Bao Q, Qian Z. Viruses, Other Pathogenic Microorganisms and Esophageal Cancer. Gastrointest Tumors. 2015;2:2-13.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Biological agents. Volume 100 B. A review of human carcinogens. IARC Monogr Eval Carcinog Risks Hum. 2012;100:1-441.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Correa P. Human gastric carcinogenesis: a multistep and multifactorial process--First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res. 1992;52:6735-6740.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Stewart OA, Wu F, Chen Y. The role of gastric microbiota in gastric cancer. Gut Microbes. 2020;11:1220-1230.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Wang L, Xin Y, Zhou J, Tian Z, Liu C, Yu X, Meng X, Jiang W, Zhao S, Dong Q. Gastric Mucosa-Associated Microbial Signatures of Early Gastric Cancer. Front Microbiol. 2020;11:1548.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Hsieh YY, Tung SY, Pan HY, Yen CW, Xu HW, Lin YJ, Deng YF, Hsu WT, Wu CS, Li C. Increased Abundance of Clostridium and Fusobacterium in Gastric Microbiota of Patients with Gastric Cancer in Taiwan. Sci Rep. 2018;8:158.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Lertpiriyapong K, Whary MT, Muthupalani S, Lofgren JL, Gamazon ER, Feng Y, Ge Z, Wang TC, Fox JG. Gastric colonisation with a restricted commensal microbiota replicates the promotion of neoplastic lesions by diverse intestinal microbiota in the Helicobacter pylori INS-GAS mouse model of gastric carcinogenesis. Gut. 2014;63:54-63.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Wang LL, Yu XJ, Zhan SH, Jia SJ, Tian ZB, Dong QJ. Participation of microbiota in the development of gastric cancer. World J Gastroenterol. 2014;20:4948-4952.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Wang Z, Gao X, Zeng R, Wu Q, Sun H, Wu W, Zhang X, Sun G, Yan B, Wu L, Ren R, Guo M, Peng L, Yang Y. Changes of the Gastric Mucosal Microbiome Associated With Histological Stages of Gastric Carcinogenesis. Front Microbiol. 2020;11:997.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Vinasco K, Mitchell HM, Kaakoush NO, Castaño-Rodríguez N. Microbial carcinogenesis: Lactic acid bacteria in gastric cancer. Biochim Biophys Acta Rev Cancer. 2019;1872:188309.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Wang L, Zhou J, Xin Y, Geng C, Tian Z, Yu X, Dong Q. Bacterial overgrowth and diversification of microbiota in gastric cancer. Eur J Gastroenterol Hepatol. 2016;28:261-266.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Gunathilake MN, Lee J, Choi IJ, Kim YI, Ahn Y, Park C, Kim J. Association between the relative abundance of gastric microbiota and the risk of gastric cancer: a case-control study. Sci Rep. 2019;9:13589.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Gunathilake M, Lee J, Choi IJ, Kim YI, Yoon J, Sul WJ, Kim JF, Kim J. Alterations in Gastric Microbial Communities Are Associated with Risk of Gastric Cancer in a Korean Population: A Case-Control Study. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Cavadas B, Camacho R, Ferreira JC, Ferreira RM, Figueiredo C, Brazma A, Fonseca NA, Pereira L. Gastric Microbiome Diversities in Gastric Cancer Patients from Europe and Asia Mimic the Human Population Structure and Are Partly Driven by Microbiome Quantitative Trait Loci. Microorganisms. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Jo HJ, Kim J, Kim N, Park JH, Nam RH, Seok YJ, Kim YR, Kim JS, Kim JM, Lee DH, Jung HC. Analysis of Gastric Microbiota by Pyrosequencing: Minor Role of Bacteria Other Than Helicobacter pylori in the Gastric Carcinogenesis. Helicobacter. 2016;21:364-374.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Sohn SH, Kim N, Jo HJ, Kim J, Park JH, Nam RH, Seok YJ, Kim YR, Lee DH. Analysis of Gastric Body Microbiota by Pyrosequencing: Possible Role of Bacteria Other Than Helicobacter pylori in the Gastric Carcinogenesis. J Cancer Prev. 2017;22:115-125.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, Machado JC, Figueiredo C. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut. 2018;67:226-236.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Hu YL, Pang W, Huang Y, Zhang Y, Zhang CJ. The Gastric Microbiome Is Perturbed in Advanced Gastric Adenocarcinoma Identified Through Shotgun Metagenomics. Front Cell Infect Microbiol. 2018;8:433.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Wu ZF, Zou K, Wu GN, Jin ZJ, Xiang CJ, Xu S, Wang YH, Wu XY, Chen C, Xu Z, Li WS, Yao XQ, Zhang JF, Liu FK. A Comparison of Tumor-Associated and Non-Tumor-Associated Gastric Microbiota in Gastric Cancer Patients. Dig Dis Sci. 2021;66:1673-1682.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Castaño-Rodríguez N, Goh KL, Fock KM, Mitchell HM, Kaakoush NO. Dysbiosis of the microbiome in gastric carcinogenesis. Sci Rep. 2017;7:15957.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Spiegelhauer MR, Kupcinskas J, Johannesen TB, Urba M, Skieceviciene J, Jonaitis L, Frandsen TH, Kupcinskas L, Fuursted K, Andersen LP. Transient and Persistent Gastric Microbiome: Adherence of Bacteria in Gastric Cancer and Dyspeptic Patient Biopsies after Washing. J Clin Med. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Aviles-Jimenez F, Vazquez-Jimenez F, Medrano-Guzman R, Mantilla A, Torres J. Stomach microbiota composition varies between patients with non-atrophic gastritis and patients with intestinal type of gastric cancer. Sci Rep. 2014;4:4202.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Eun CS, Kim BK, Han DS, Kim SY, Kim KM, Choi BY, Song KS, Kim YS, Kim JF. Differences in gastric mucosal microbiota profiling in patients with chronic gastritis, intestinal metaplasia, and gastric cancer using pyrosequencing methods. Helicobacter. 2014;19:407-416.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Li TH, Qin Y, Sham PC, Lau KS, Chu KM, Leung WK. Alterations in Gastric Microbiota After H. pylori Eradication and in Different Histological Stages of Gastric Carcinogenesis. Sci Rep. 2017;7:44935.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Coker OO, Dai Z, Nie Y, Zhao G, Cao L, Nakatsu G, Wu WK, Wong SH, Chen Z, Sung JJY, Yu J. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67:1024-1032.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Dang YN, Dong Y, Mu YZ, Yan J, Lu M, Zhu YL, Zhang GX. Identification of gastric microbiota biomarker for gastric cancer. Chin Med J (Engl). 2020;133:2765-2767.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Gantuya B, El Serag HB, Matsumoto T, Ajami NJ, Uchida T, Oyuntsetseg K, Bolor D, Yamaoka Y. Gastric mucosal microbiota in a Mongolian population with gastric cancer and precursor conditions. Aliment Pharmacol Ther. 2020;51:770-780.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Nardone G, Compare D. The human gastric microbiota: Is it time to rethink the pathogenesis of stomach diseases? United European Gastroenterol J. 2015;3:255-260.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Vuik F, Dicksved J, Lam SY, Fuhler GM, van der Laan L, van de Winkel A, Konstantinov SR, Spaander M, Peppelenbosch MP, Engstrand L, Kuipers EJ. Composition of the mucosa-associated microbiota along the entire gastrointestinal tract of human individuals. United European Gastroenterol J. 2019;7:897-907.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Del Piano M, Anderloni A, Balzarini M, Ballarè M, Carmagnola S, Montino F, Orsello M, Pagliarulo M, Tari R, Soattini L, Sforza F, Mogna L, Mogna G. The innovative potential of Lactobacillus rhamnosus LR06, Lactobacillus pentosus LPS01, Lactobacillus plantarum LP01, and Lactobacillus delbrueckii Subsp. delbrueckii LDD01 to restore the "gastric barrier effect" in patients chronically treated with PPI: a pilot study. J Clin Gastroenterol. 2012;46 Suppl:S18-S26.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Gray JD, Shiner M. Influence of gastric pH on gastric and jejunal flora. Gut. 1967;8:574-581.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Wandall JH. Effects of omeprazole on neutrophil chemotaxis, super oxide production, degranulation, and translocation of cytochrome b-245. Gut. 1992;33:617-621.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Meuwissen SG, Craanen ME, Kuipers EJ. Gastric mucosal morphological consequences of acid suppression: a balanced view. Best Pract Res Clin Gastroenterol. 2001;15:497-510.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Mowat C, Williams C, Gillen D, Hossack M, Gilmour D, Carswell A, Wirz A, Preston T, McColl KE. Omeprazole, Helicobacter pylori status, and alterations in the intragastric milieu facilitating bacterial N-nitrosation. Gastroenterology. 2000;119:339-347.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Arroyo Vázquez JA, Henning C, Park PO, Bergström M. Bacterial colonization of the stomach and duodenum in a Swedish population with and without proton pump inhibitor treatment. JGH Open. 2020;4:405-409.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Paroni Sterbini F, Palladini A, Masucci L, Cannistraci CV, Pastorino R, Ianiro G, Bugli F, Martini C, Ricciardi W, Gasbarrini A, Sanguinetti M, Cammarota G, Posteraro B. Effects of Proton Pump Inhibitors on the Gastric Mucosa-Associated Microbiota in Dyspeptic Patients. Appl Environ Microbiol. 2016;82:6633-6644.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Grossman MI, Kirsner JB, Gillespie IE. Basal and histalog-stimulated gastric secretion in control subjects and in patients with peptic ulcer or gastric cancer. Gastroenterology. 1963;45:14-26.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Horii T, Koike T, Abe Y, Kikuchi R, Unakami H, Iijima K, Imatani A, Ohara S, Shimosegawa T. Two distinct types of cancer of different origin may be mixed in gastroesophageal junction adenocarcinomas in Japan: evidence from direct evaluation of gastric acid secretion. Scand J Gastroenterol. 2011;46:710-719.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Ohata H, Kitauchi S, Yoshimura N, Mugitani K, Iwane M, Nakamura H, Yoshikawa A, Yanaoka K, Arii K, Tamai H, Shimizu Y, Takeshita T, Mohara O, Ichinose M. Progression of chronic atrophic gastritis associated with Helicobacter pylori infection increases risk of gastric cancer. Int J Cancer. 2004;109:138-143.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Varis K, Sipponen P, Laxén F, Samloff IM, Huttunen JK, Taylor PR, Heinonen OP, Albanes D, Sande N, Virtamo J, Härkönen M. Implications of serum pepsinogen I in early endoscopic diagnosis of gastric cancer and dysplasia. Helsinki Gastritis Study Group. Scand J Gastroenterol. 2000;35:950-956.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Miki K, Fujishiro M, Kodashima S, Yahagi N. Long-term results of gastric cancer screening using the serum pepsinogen test method among an asymptomatic middle-aged Japanese population. Dig Endosc. 2009;21:78-81.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Leung WK, Wu MS, Kakugawa Y, Kim JJ, Yeoh KG, Goh KL, Wu KC, Wu DC, Sollano J, Kachintorn U, Gotoda T, Lin JT, You WC, Ng EK, Sung JJ;  Asia Pacific Working Group on Gastric Cancer. Screening for gastric cancer in Asia: current evidence and practice. Lancet Oncol. 2008;9:279-287.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Azcarate-Peril MA, Altermann E, Hoover-Fitzula RL, Cano RJ, Klaenhammer TR. Identification and inactivation of genetic loci involved with Lactobacillus acidophilus acid tolerance. Appl Environ Microbiol. 2004;70:5315-5322.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Shin CM, Kim N, Kim YS, Nam RH, Park JH, Lee DH, Seok YJ, Kim YR, Kim JH, Kim JM, Kim JS, Jung HC. Impact of Long-Term Proton Pump Inhibitor Therapy on Gut Microbiota in F344 Rats: Pilot Study. Gut Liver. 2016;10:896-901.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Abbas Hilmi HT, Surakka A, Apajalahti J, Saris PE. Identification of the most abundant lactobacillus species in the crop of 1- and 5-week-old broiler chickens. Appl Environ Microbiol. 2007;73:7867-7873.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Brooks SP, McAllister M, Sandoz M, Kalmokoff ML. Culture-independent phylogenetic analysis of the faecal flora of the rat. Can J Microbiol. 2003;49:589-601.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Leser TD, Amenuvor JZ, Jensen TK, Lindecrona RH, Boye M, Møller K. Culture-independent analysis of gut bacteria: the pig gastrointestinal tract microbiota revisited. Appl Environ Microbiol. 2002;68:673-690.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Salzman NH, de Jong H, Paterson Y, Harmsen HJM, Welling GW, Bos NA. Analysis of 16S libraries of mouse gastrointestinal microflora reveals a large new group of mouse intestinal bacteria. Microbiology (Reading). 2002;148:3651-3660.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  García A, Sáez K, Delgado C, González CL. Low co-existence rates of Lactobacillus spp. and Helicobacter pylori detected in gastric biopsies from patients with gastrointestinal symptoms. Rev Esp Enferm Dig. 2012;104:473-478.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  García CA, Henríquez AP, Retamal RC, Pineda CS, Delgado Sen C, González CC. Propiedades probióticas de Lactobacillus spp aislados de biopsias gástricas de pacientes con y sin infección por Helicobacter pylori [Probiotic properties of Lactobacillus spp isolated from gastric biopsies of Helicobacter pylori infected and non-infected individuals]. Rev Med Chil. 2009;137:369-376.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Lü M, Yu S, Deng J, Yan Q, Yang C, Xia G, Zhou X. Efficacy of Probiotic Supplementation Therapy for Helicobacter pylori Eradication: A Meta-Analysis of Randomized Controlled Trials. PLoS One. 2016;11:e0163743.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Fang HR, Zhang GQ, Cheng JY, Li ZY. Efficacy of Lactobacillus-supplemented triple therapy for Helicobacter pylori infection in children: a meta-analysis of randomized controlled trials. Eur J Pediatr. 2019;178:7-16.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Eslami M, Yousefi B, Kokhaei P, Jazayeri Moghadas A, Sadighi Moghadam B, Arabkari V, Niazi Z. Are probiotics useful for therapy of Helicobacter pylori diseases? Comp Immunol Microbiol Infect Dis. 2019;64:99-108.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Mack DR, Ahrne S, Hyde L, Wei S, Hollingsworth MA. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut. 2003;52:827-833.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Behrens HM, Six A, Walker D, Kleanthous C. The therapeutic potential of bacteriocins as protein antibiotics. Emerg Top Life Sci. 2017;1:65-74.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Blum S, Haller D, Pfeifer A, Schiffrin EJ. Probiotics and immune response. Clin Rev Allergy Immunol. 2002;22:287-309.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Kabir AM, Aiba Y, Takagi A, Kamiya S, Miwa T, Koga Y. Prevention of Helicobacter pylori infection by lactobacilli in a gnotobiotic murine model. Gut. 1997;41:49-55.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Cassatella MA, Guasparri I, Ceska M, Bazzoni F, Rossi F. Interferon-gamma inhibits interleukin-8 production by human polymorphonuclear leucocytes. Immunology. 1993;78:177-184.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Maleki-Kakelar H, Dehghani J, Barzegari A, Barar J, Shirmohamadi M, Sadeghi J, Omidi Y. Lactobacillus plantarum induces apoptosis in gastric cancer cells via modulation of signaling pathways in Helicobacter pylori. Bioimpacts. 2020;10:65-72.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell. 2012;21:297-308.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Warburg O. On the origin of cancer cells. Science. 1956;123:309-314.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Fall PJ, Szerlip HM. Lactic acidosis: from sour milk to septic shock. J Intensive Care Med. 2005;20:255-271.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfør K, Rofstad EK, Mueller-Klieser W. High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Res. 2000;60:916-921.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Walenta S, Salameh A, Lyng H, Evensen JF, Mitze M, Rofstad EK, Mueller-Klieser W. Correlation of high lactate levels in head and neck tumors with incidence of metastasis. Am J Pathol. 1997;150:409-415.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Brizel DM, Schroeder T, Scher RL, Walenta S, Clough RW, Dewhirst MW, Mueller-Klieser W. Elevated tumor lactate concentrations predict for an increased risk of metastases in head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2001;51:349-353.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Forsythe SJ, Cole JA. Nitrite accumulation during anaerobic nitrate reduction by binary suspensions of bacteria isolated from the achlorhydric stomach. J Gen Microbiol. 1987;133:1845-1849.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Calmels S, Béréziat JC, Ohshima H, Bartsch H. Bacterial formation of N-nitroso compounds from administered precursors in the rat stomach after omeprazole-induced achlorhydria. Carcinogenesis. 1991;12:435-439.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Jones RM, Mercante JW, Neish AS. Reactive oxygen production induced by the gut microbiota: pharmacotherapeutic implications. Curr Med Chem. 2012;19:1519-1529.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Ohta K, Kawano R, Ito N. Lactic acid bacteria convert human fibroblasts to multipotent cells. PLoS One. 2012;7:e51866.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Yang X, Da M, Zhang W, Qi Q, Zhang C, Han S. Role of Lactobacillus in cervical cancer. Cancer Manag Res. 2018;10:1219-1229.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Walter J. Ecological role of Lactobacilli in the gastrointestinal tract: implications for fundamental and biomedical research. Appl Environ Microbiol. 2008;74:4985-4996.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Molin G, Jeppsson B, Johansson ML, Ahrné S, Nobaek S, Ståhl M, Bengmark S. Numerical taxonomy of Lactobacillus spp. associated with healthy and diseased mucosa of the human intestines. J Appl Bacteriol. 1993;74:314-323.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Roberts PJ, Dickinson RJ, Whitehead A, Laughton CR, Foweraker JE. The culture of Lactobacilli species in gastric carcinoma. J Clin Pathol. 2002;55:477.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Sanders DS, Hussein KA. Filamentous organisms in gastric brushings and gastric cancer diagnosis. Diagn Cytopathol. 1991;7:11-13.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Wilson CA, Young JA, Sanders DS. Filamentous organisms in benign and malignant gastric cytology brushings. Cytopathology. 1996;7:268-273.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Sjöstedt S, Kager L, Heimdahl A, Nord CE. Microbial colonization of tumors in relation to the upper gastrointestinal tract in patients with gastric carcinoma. Ann Surg. 1988;207:341-346.  [PubMed]  [DOI]  [Cited in This Article: ]