Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2024

Open Access 01.12.2024 | Review

Unveiling the gastric microbiota: implications for gastric carcinogenesis, immune responses, and clinical prospects

verfasst von: Zhiyi Liu, Dachuan Zhang, Siyu Chen

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2024

Abstract

High-throughput sequencing has ushered in a paradigm shift in gastric microbiota, breaking the stereotype that the stomach is hostile to microorganisms beyond H. pylori. Recent attention directed toward the composition and functionality of this 'community' has shed light on its potential relevance in cancer. The microbial composition in the stomach of health displays host specificity which changes throughout a person's lifespan and is subject to both external and internal factors. Distinctive alterations in gastric microbiome signature are discernible at different stages of gastric precancerous lesions and malignancy. The robust microbes that dominate in gastric malignant tissue are intricately implicated in gastric cancer susceptibility, carcinogenesis, and the modulation of immunosurveillance and immune escape. These revelations offer fresh avenues for utilizing gastric microbiota as predictive biomarkers in clinical settings. Furthermore, inter-individual microbiota variations partially account for differential responses to cancer immunotherapy. In this review, we summarize current literature on the influence of the gastric microbiota on gastric carcinogenesis, anti-tumor immunity and immunotherapy, providing insights into potential clinical applications.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
GC
Gastric cancer
TME
Tumor microenvironment
TGGE
Temperature gradient gel electrophoresis
FISH
Fluorescence in situ hybridization
NGS
Next-generation sequencing
WGCNA
Weighted gene co-expression network analysis
GI
Gastrointestinal
HC
Healthy control
AG
Atrophic gastritis
SG
Superficial gastritis
CAG
Chronic atrophic gastritis
IM
Intestinal metaplasia
GIN
Gastric intraepithelial neoplasia
OTUs
Operational Taxonomic Units
ORs
Odds ratios
WMS
Whole metagenomic shotgun sequencing
EGN
Early gastric neoplasia
NS
Not significant
VacA
Vacuolating cytotoxin A
CagA
Cytotoxin-associated gene A
cagPAI
Cag pathogenicity island
T4SS
Type IV secretion system
SH2
SRC homology 2 domain
PAR1b
Partitioning-defective 1
BRCA1
Breast cancer susceptibility gene 1
DSBs
DNA double-strand breaks
ADAM17
Metalloproteinase domain 17
NF-κB
Nuclear factor-κB
HKα
H,K-ATPase α subunit
IL-1ß
Interleukin-1ß
TNF-α
Tumor Necrosis Factor-α
INS-GAS
Insulin-gastrin
SPF
Specific pathogen-free
ANXA2
Annexin A2
IFN-γ
Interferon-γ
ROS
Reactive oxygen species
DCA
Deoxycholic acid
LPS
Lipopolysaccharide
BRG
Bile reflux gastritis
TDCA
Taurodeoxycholic acid
JAK1
Janus kinase 1
STAT3
Signal transducer and activator of transcription 3
PICRUSt
Phylogenetic investigation of communities by reconstruction of unobserved States
NOC
N-nitroso compounds
MUC
Mucin
LINE1
Long interspersed nuclear elements 1
CRC
Colorectal cancer
HOTTIP
HOXA transcript at the distal tip
TLR
Toll-Like Receptor
MYD88
Myeloid Differentiation Primary Response 88
NOD1
Nucleotide-binding Oligomerization Domain 1
ITS
Internal transcribed spacer
MMP-2
Matrix metalloproteinase-2
LAMP
Lipid-associated membrane protein
NLRP3
Nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein 3
PAMPs
Pathogen-associated molecular patterns
PRRs
Pathogen recognition receptors
ILC2s
Group 2 innate lymphoid cells
IL-7R
Interleukin-7 receptor
TANs
Tumor-activated neutrophils
pDCs
Plasmacytoid dendritic cells
Treg
Regulatory T cells
NK
Natural Killer
TIGIT
T-cell immunoreceptor with Ig and ITIM domains
TRMs
Tissue-resident memory T cells
PD-1/PD-L1
Programmed Cell Death Protein 1/Programmed Death-Ligand 1
shh
Sonic Hedgehog
ICOS
Inducible T-cell CO-Stimulator
Foxp3
Forkhead Box P3
Breg
B regulatory cells
ICIs
Improve immune checkpoint inhibitors
NSCLC
Non-small cell lung cancer
CTLA4
Cytotoxic T-Lymphocyte-Associated Protein 4
ATB
Antibiotics
HP-NAP
Helicobacter pylori neutrophil-activating protein
rMBP-NAP
Hp-NAP recombined with the maltose binding protein
Chi-rNap
HP-NAP-embedded Chitosan nanoparticles
OVs
Oncolytic viruses
CAR
Chimeric antigen receptor

Introduction

Gastric cancer (GC) is a very aggressive cancer and the third main cause of cancer-related mortality worldwide with more than 1 million diagnosed cases and 770,000 deaths globally in 2020 [1]. Approximately 40% of GC cases are identified at an advanced stage annually, with the 5-year survival rates for patients diagnosed in 2000 and 2014 merely 2.7% and 4.7%, respectively [2]. The infectious agent, Helicobacter pylori (H. pylori), is widely recognized as the most influential contributor to the onset of GC. An estimated 50% of people worldwide are infected with H. pylori [3] and approximately 90% of GC on a global scale result from the inflammation and damage caused by H. pylori infection [4]. Despite the well-established involvement of H. pylori in the process of gastric carcinogenesis, only fewer than 3% of H. pylori-infected patients eventually progress to GC, indicating the initial role of H. pylori rather than its exclusive influence on GC development. The scope of other contributing factors extends to genetic polymorphisms, environmental exposures, age, and gender [1, 57]. Recent progress has been made in the field, with the emergence of advanced next-generation sequencing technologies that unveiled the variations in diversity and community structure of gastric microbiota between individuals with good health and those afflicted with GC [8, 9]. These distinctions extend beyond the well-known H. pylori and include a broader range of microbial constituents, enlightening the potential role of these non-H. pylori gastric microbiota in the promotion of malignancy.
The microbiota encompasses a diverse array of bacteria, viruses, bacteriophages, fungi, and protozoa collectively shaping a complex ecosystem. These microbial communities, residing in various body habitats, including the oral cavity, gut, lung, skin and genitourinary tract, have been the subject of an extensive investigation regarding their contributions to maintaining homeostasis and their involvement in disease processes [1014]. In particular, the mounting body of evidence highlights the role of the gut microbiota in driving cancer initiation and progression, orchestrating the intricate landscape of the tumor microenvironment (TME), and providing novel avenues for manipulating the microbiota to modulate anti-tumor immunity [1519].
Owing to the relatively lower biomass of gastric microbiota, which quantifies merely 101 to 103 bacteria per gram of gastric content—significantly fewer than the 104 to 107 bacteria per gram found in the jejunum and ileum, and the 1011 to 1012 bacteria per gram in the colon, the gastric microbiota has received relatively limited attention in prior research [20]. Until recently, studies have conducted in vivo manipulations of gastric microbiota based on sequencing results, thus providing robust evidence to elucidate the causal relationship between perturbations in the gastric microbial ecosystem (referred to as "gastric dysbiosis") and the onset and progression of GC [2123]. In this comprehensive review, we explore the emerging findings regarding the dynamic influence of gastric microbiota on GC and also address H. pylori as the most thoroughly investigated constituent of the microbiota, but are more inclined to discuss the evidence concerning its influence on the coexistence of other bacterial species in the stomach. We emphasize the pivotal role of gastric microbiota in the processes of gastric carcinogenesis, modulation of anti-tumor immune responses, and its potential implications in clinical application.

Microbial composition in the stomach

Due to low biomass of the microbiota and the unique and challenging growth environment in gastric mucosa, methods for isolation and cultivation of the gastric flora, as well as those for strain identification via morphological, biochemical and serotype characteristics, are greatly constrained. Therefore, in early assessment of gastric microbial composition, cultivation-independent methods such as molecular fingerprinting have been widely used [24]. Among these, temperature gradient gel electrophoresis (TGGE) was the most popular technique, which separate microbial DNA into a series of bands for inter-species comparisons based on differential physical–chemical properties of DNA fragments. Apparently, TGGE's resolution to discriminate between microbial taxa is very limited, leading to marked underestimation of gastric microbiome richness [25]. Additionally, other molecular methods such as fluorescence in situ hybridization (FISH), microarrays, and quantitative PCR exhibit high sensitivity in detecting low-abundance species [2628]. However, the requirement for species-specific oligonucleotide probes or primers prevented large-scale and unbiased analysis of the entire microbial community.
In the past decade, the development of next-generation sequencing (NGS) technologies has dramatically improved the precision and coverage of taxa identification, eventually enabling deep profiling of gastric microbiome, and dissecting their interactions with the host. Among the NGS methods, 16S rRNA sequencing has been widely used, because the 16S gene contains both the conserved regions supporting phylogenetic classification at the phylum level, and the rapidly evolving regions suitable for finer taxonomic resolution. However, 16S rRNA gene sequencing can only identify known species, with a resolution limited to the genus level, and losses all the functional information for each species. By contrast, metagenomics relies on whole genome shotgun sequencing, thus enabling higher resolution for species identification and functional analysis. NGS analysis often synergize with other multi-omics methodologies such as metabolomics and proteomics, providing a comprehensive understanding of the structure and functions of gastric microbiota in health and disease [25, 29].
H. pylori initially identified in the 1980s was regarded as a milestone to investigate gastric microenvironment. Subsequent advancements in detection technologies have enabled the meticulous tracking of the microbial ecosystem and unveiled the microbial profile of the stomach (Table 1). This groundbreaking progress challenges the longstanding stereotype that the gastric mucosa is fundamentally sterile attributed to its inhospitable condition [24, 30].
Table 1
Gastric microbial composition in health
Age
Main stomach taxonomic structure in health/control sample
Gastric sample type
Subjects
Country
Method
Ref
Infant (1 hour after birth)
Lactobacillus (vaginally delivery), Ureaplasma (premature birth)
Gastric fluid
29
America
16S rRNA sequencing
[31]
27 + / − 0.5 weeks (gestational age) preterm neonates
Phyla: Firmicutes, Proteobacteria, Tenericute, Actinobacteria in week 1
Firmicutes, Proteobacteria in week 4
Genera: Staphylococcus, Streptococcus, Ureaplasma, Neisseria, Haemophilus, Klesbsiella
Gastric aspirates
12
America
16S rRNA sequencing
[32]
Preterm neonates
Gram-positive:
Staphylococcus, Lactobacillus and Enterococcus,
Gram-negative: Serratia, Klebsiella and Escherichia
Gastric fluid
13
Spain
16S rRNA sequencing
[33]
27.7 + / − 2.8 weeks Preterm neonates
Bacteroides spp., Lactobacillus spp., Escherichia coli, Staphylococcus aureus, Streptococcus pneumoniae, Bifidobacterium spp.(breast-fed neonate)
Gastric aspirates
22
America
16S rRNA-based Denaturing gradient gel electrophoresis
[34]
Adult
Proteobacteria, Firmicutes, Bacteroidetes, Actinobacteria, Fusobacteria
Gastric mucosa
HC:23
America
16S rRNA sequencing
[30]
Adult
Phyla: Proteobacteria, Bacteroidetes, Firmicutes, Actinobacteria, Fusobacteria
Genera: Streptococcus, Prevotella, Neisseria, Haemophilus, Porphyromonas
Gastric mucosa
HC:5
non-NSAID gastritis:5
Hong Kong (China)
16S rRNA sequencing
[35]
Adult
Phyla: Proteobacteria, Bacteroidetes, Firmicutes, Fusobacteria, Actinobacteria
Genera: Neisseria, Prevotella, f;[paraprevotellaceae]-g;[Prevotella], Haemophilus, Fusobacterium, Streptococcus, Veillonella, Capnocytophage, Leptotrichia
Gastric mucosa
HC:7
Japan
16S rRNA sequencing
[36]
Adult
Phyla: Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Fusobacteria
Genera: Streptococcus, Prevotella, Veillonella, Fusobacterium, Gemella, Neisseria, Haemophilus
Gastric mucosa
HC:171
Non-atrophic H. pylori gastritis:33
AG:12
antral chemical gastritis:61
Sweden
16S rRNA sequencing
[37]
Adult
Enterococcus, Pseudomonas, Streptococcus, Staphylococcus, Stomatococcus
Gastric mucosa
HC:5
CG:8
Sweden
Temporal temperature gradient gel electrophoresis
[24]
Adult
Phyla:Firmicutes, Proteobacteria, Bacteroidetes, Fusobacteria, Actinobacteria
Genera: Streptococcus, Prevotella, Pseudomonas, Fusobacterium, Gemella, Neisseria, Veillonell
Gastric mucosa
HC:21
German
16S rRNA sequencing
[38]
Abbreviation: GC Gastric cancer, HC Healthy control, AG Atrophic gastritis, NSAID Non-steroidal anti-Inflammatory drugs

Initial establishment of gastric microbial community

The stomach is not as bacteria-free as expected after birth. Microbes are present in the gastric aspirate from newborn infants, suggesting that the initial establishment of the gastric microbial community may occur at delivery or even earlier. The microbial composition of the neonatal stomach displays variability and potentially bears a connection to the mode of delivery (Fig. 1). Term neonates with vaginal delivery have significantly higher gastric microorganism abundance than those born by cesarean section. Additionally, the identification of the predominant constituent in the neonatal gastric microbial community highlights Lactobacillus Crispatusas the prevailing species within this ecosystem. This bacterium is crucial for maintaining vaginal health and becomes enriched within the vaginal mucosa during pregnancy [31, 39]. Whereas in preterm infants, the initial colonizers in the stomach are instead characterized by a high abundance of Ureaplasma [31, 32]. The detection of Ureaplasm from the maternal placenta and amniotic fluid predicts higher risk of preterm birth and neonatal mortality, and the persistent colonization of Ureaplasm in neonates may be associated with systemic infection and congenital diseases such as bronchopulmonary dysplasia [40]. However, Ureaplasm appeared only in the gastric aspirate of neonates in the first week, but disappeared afterward, probably due to intolerance of the gastric environment and prompt intervention with antibiotics [32]. Subsequent exposure to the environment and breast milk feeding flourish more bacteria species colonization in the stomach. A study involving 13 one-month-old preterm infants collected gastric contents and identified a predominant presence of Staphylococcus, Lactobacillus, and Serratia. These microbial components are recognized as significant constituents of breast milk [33, 41]. In addition, gastric microbial species of these preterm infants with long-term hospitalization were observed to overlap with a large proportion of nosocomial infection-associated bacteria from the hospital environment, such as Staphylococcus aureus, Staphylococcus epidermidis, Enterococcus faecium, Neisseria, Klebsiella pneumoniae, and Escherichia coli [3234]. Environment factors seem to act as an essential selector for inhabitants of the stomach, and even for twins, the initial development of microbial community varies according to different exposure to environmental flora [42].
The initial gastric colonizers can shape the local and systemic immunity in early life. In animal models, it has been demonstrated that the offspring of mothers who were given antibiotics during pregnancy and breastfeeding had a decrease in microbial diversity in gastric contents accompanied by elevated IgG and IgM [43]. Consistently, Trevisi et al. also indicated that high-quality and complex initial microbial residence in the stomach not only favor oxyntic mucosa maturation and development of acid secretion but also mediate gastric immunity by modulating interferon response, antibodies production and immune cells maturation [44].

Gastric microbial composition in Health

As early as 2000, Monstein et al. initially described gastric “indigenous” microbiota other than H. pylori by temporal temperature gradient gel electrophoresis(TGGE). Enterococcus, Pseudomonas, Streptococcus, Staphylococcus, and Stomatococcuswere detected as major colonized members of genera in the stomach [24]. In 2005, a 16S rDNA sequencing of human stomach bacterial profile in America uncovered 128 phylotypes belonging to five predominant phyla which are Proteobacteria, Firmicutes, Bacteroidetes, Actinobacteria and Fusobacteria, accounting for over 90% of the total [30]. Generally, consistent phylum structures were identified in subsequent research in Hong Kong and Japan, whereas the American population showed a higher Firmicutes/Bacteroidetes ratio compared with Asians [35, 36]. High Firmicutes/Bacteroidetes ratio was previously mentioned in both animal study and clinical trial as a biomarker of gut dysbiosis in obesity [45, 46], and mice that were fed high-fiber or high-fat food exhibited overrepresented Bacteroidetes and Firmicutes for each [47, 48]. This inter-ethnic difference in Firmicutes/Bacteroidetes ratios within the gastric microbiota may be influenced by dietary habits. Moreover, a more extensive sequencing was performed in Sweden, an area with low H. pylori prevalence. This study recruited 171 healthy subjects and unveiled the taxonomic composition of their gastric microbial community. Sweden's population showed an unexpectedly low abundance of Proteobacteria [37], which is noteworthy as H. pylori falls within this bacterial clade. Genetic variants and external factors within the Swedish population may contribute to the development of a gastric environment that is less conducive to Proteobacteria colonization, thereby leading to a decreased incidence of H. pylori infections.
The most prevalent gastric mucosa-associated genera frequently described in the scientific literature are Neisseria, Prevotell, Haemophilus, Fusobacterium, Streptococcus, and Veillonella, though the abundance of each varies among subjects [24, 3537]. Neisseria, Haemophilus, Fusobacterium and some species of Streptococcus and Prevotella are typical oral bacteria [8, 36, 49]. The swallowing action allows the oral microbiota to "seed" the lower GI tract. It is noteworthy that these prevailing genera within the stomach are scarcely detected in the lower gastrointestinal (GI) tract, which is primarily characterized by microbial communities dominated by Ruminococcus, Faecalibacterium, and Bacteroides [36, 38]. This large difference highlights the inaccuracy of regarding the lower GI and fecal microbiota as representative of the entire GI microbiota and underscores the importance of studying the GI microbiota by compartmentalization. Nonetheless, the outcomes of sequencing solely capture the configuration of the gastric microbiota at a specific juncture within the digestive process. It remains elusive to ascertain whether these microbial communities represent transient contamination or persistent colonization.

Gastric microbiome, H. pylori and Gastric cancer

GC, a complex and multifactorial disease, is known to develop through a multistep progression involving a series of pathological processes referred to as Correa’s cascade. The sequence of events typically begins with superficial gastritis (SG), followed by chronic atrophic gastritis (CAG), intestinal metaplasia (IM), and dysplasia, ultimately culminating in the occurrence of GC [50]. These sequential pathological changes contribute to the stepwise transformation of normal gastric mucosa into malignant tissue. In addition to H. pylori infection as the main driving factor, characteristic changes in the gastric microbiota concomitant with this process were observed (Table 2).
Table 2
gastric microbial diversity and composition of gastric cancer patients
Subject /Sample Size
Gastric sample type
Country
Method
Gastric microbial diversity alternation in diseases
Gastric microbial composition alternation
Ref
12 GC VS 20 functional
Dyspepsia
Gastric tissue
Malaysia
16S rRNA sequencing
GC: ↑
GC: Lactococcus, Veilonella, Fusobacteriaceae (Fusobacterium), Leptotrichia
[8]
268 GC VS 288 control
Gastric mucosa
Korea
16S rRNA sequencing
GC: ↓
GC: Helicobacter, Propionibacterium acnes, Prevotella copri
Control: Lactococcus lactis
[9]
17 SG VS 10 AG VS 5 GIN VS 15 GC
Gastric mucosa
China
16S rRNA sequencing
NS
GC: Slackia, Selenomonas,
Bergeyella, Capnocytopha, Parvimonas, Eikenella, Prevotella-2, Kroppenstedtia, Lentibacillus, Oceanobacillus
GIN: Romboutsia, Fusicatenibacter, Prevotellaceae-Ga6A1-group, Intestinimonas
[22]
20 HC VS 20 Gastritis VS 40 AG VS 40 IM VS 48 GC
Gastric mucosa
Mongolia
16S rRNA sequencing
HC > IM > GC > gastritis > AG
H. Pylori(-) GC:Lactobacilli and Enterococci 
H. pylori( ±) GC:Carnobacterium, Glutamicibacter, Paeniglutamicibacter, Fusobacterium, Parvimonas 
[23]
17 SG VS 16 IM (low-risk) VS 6 IM (high-risk) VS 4 EGN
Gastric antral biopsies
Singapore
16S rRNA sequencing
NS
EGN: Proteobacteria(genus Proteus), H. pylori ↑ Bacteroidetes(S24-7 group), Bfidobacteria, Lactobacillus ↓
High-risk IM: H. pylori, Actinomyces, Moryella 
Low-risk IM: Phyllobacteriaceae, Enhydrobacter, Moryella ↑ Bifidobacteria, Lactobacillus 
[51]
30 HC VS 21 CG VS 27 IM VS IN VS 29 GC
Gastric mucosa
China
16S rRNA sequencing
HC > CG > IM > GC
IM and GC: Gram-positive and anaerobic bacteria ↑
IN, IM and GC: Nitrospirae ↓
[52]
21 SG VS 23 AG VS 17 IM VS 20 GC from Xi an
56 SG VS 51 AG VS 19 GC from Hohhot
Gastric mucosa
China
16S rRNA sequencing
IM and GC: ↓
GC: Peptostreptococcus stomatis, Streptococcus anginosus, Parvimonas micra, Slackia exigua, Dialister pneumosintes
[53]
61 SG VS 55 IM VS 64 GC
Gastric mucosa and
Gastric fluid
China
16S rRNA sequencing
GC: ↓
GC: Lactobacillus, Veillonella, Gemella
[54]
9 Gastritis VS 7 IM VS 11 GC
Gastric mucosa
China
16S rRNA sequencing
/
GC: Clostridium colicanis, Fusobacterium nucleatum and Lactobacillus speciesH. pylroi-negative patients.: Burkholderia, Enterobacter, and Leclercia
[55]
89 IM VS 89 matched controls
Gastric mucosa
America
Shotgun metagenomic sequencing
NS
IM: Johnsonella ignava, Actinomyces sp. oral taxon 448, Prevotella baroniae, Filifactor alocis, Veillonella sp. oral taxon 780, Leptotrichia goodfellowii
[56]
54 GC VS 81 CG
Gastric mucosa
Portuguese, China and Mexico
16S rRNA sequencing
GC: ↓
GC: phylum level: Firmicutes, Actinobacteria and non-Helicobacter Proteobacteria ↑; Genus level: Citrobacter, Clostridium, Lactobacillus, Achromobacter, Rhodococcus, Phyllobacterium
[57]
230 Normal tissues VS 247 peritumoral tissues VS 229 tumoral tissues
Gastric tissue
China
16S rRNA sequencing
Normal > Tumor > Peritumoral tissue
Tumor: Prevotella melaninogenica, Streptococcus anginosus, Propionibacterium acnesH. pylori, Prevotella copri, Bacteroides uniformis
[58]
10 high GC risk VS 10 low GC risk
Gastric biopsy samples
Colombia
Whole metagenomic shotgun sequencing
NS
high GC risk: Staphylococus, Streptococcus, Bacillus, Aspergillus, and Siphoviridae
[59]
53 tumorous tissue VS 53 adjacent gastric tissue
Gastric mucosa
America
16S rRNA sequencing
Tumor: ↑
worse overall survival in GC: Fusobacterium and Prevotella
[60]
62 cancerous tissues VS 62 neiboring non-cancerous tissues
Gastric mucosa
China
16S rRNA sequencing
Cancerous tissues: ↑
cancerous tissues: Fusobacterium, Streptococcus, Peptostreptococcus
non-cancerous tissues: Lactococcus lactis, Lactobacillus brevis
[61]
25 H. pylori(-) SG VS 34 GC
Gastric mucosa
China
16S rRNA sequencing
NS
CG: Lactobacillus, Streptococcus, Citrobacter, Klebsiella
H. pylori(-) GC: Pseudomonadales, Betaproteobacteria, Gammaproteobacteria H. pylori( +) GC: Campylobacterales
[62]
18 GC tumor samples VS 78 paracancerous samples VS 64 SG samples
Gastric mucosa
China
16S rRNA sequencing
GC: ↓
GC: Dialister spp., Helicobacter spp., Lactobacillus spp., Rhodococcus spp., Rudaea spp., Sediminibacterium spp. ↑
tumor tissue: Bradyrhizobium spp., Mesorhizobium spp.↓
[63]
37 tumor tissues VS 37 adjacent non-tumor tissues
Gastric mucosa
China
16S rRNA sequencing
Tumor tissue: ↑
tumor tissues: Lactobacillus, Streptococcus, Acinetobacter, Prevotella, Sphingomonas, Bacteroides, Fusobacterium, Comamonas, Empedobacter, Faecalibacterium
non-tumor tissues: Helicobacter
[64]
53 GC VS 30 CG
Gastric mucosa
China
16S rRNA sequencing
GC: ↓
GC: Oceanobacter, Methylobacterium, and Syntrophomona 
[65]
16 distal GC (normal and tumor tissue) VS 13 proximal GC (normal and tumor tissue)
Gastric tissue
China
16S rRNA sequencing
Tumor tissue: ↑
tumor: Helicobacter↓, Lactobacillus and Muribaculaceae 
proximal GC: Rikenellaceae_RC9_gut_group, Porphyromonas, Catonella, Proteus, Oribacterium, and Moraxella 
distal GC: Methylobacterium_ Methylorubrum 
[66]
Abbreviation: GC Gastric cancer, SG Superficial gastritis, AG Atrophic gastritis, GIN Gastric intraepithelial neoplasia, NS Not Significant, HC Healthy control, IM Intestinal metaplasia

Gastric microbiome in gastric carcinogenesis

Gastric microbial diversity in gastric carcinogenesis

Recent studies have investigated the variances in gastric microbial diversity across Correa's cascade, ranging from healthy controls (HC) to SG, AG, IM, and ultimately GC [21, 22, 38, 4952]. Among these investigations, two studies did not observe significant differences in gastric microbial diversity among these subgroups [22, 51]. Other larger-scale studies reported significantly reduced microbial richness and diversity in GC when compared with SG or HC groups. Whereas the discrimination of gastric flora diversity between GC and precancerous lesions (IM and dysplasia) did not achieve statistical significance in these studies [53, 54]. Microbial diversity may undergo a substantial decrease between the AG and IM stages [67]. As shown in previous studies, H. pylori eradication significantly decreased the risk of GC whereas its impact is constrained in patients with IM and dysplasia [68, 69]. This indicated that the optimal therapeutic window for antibiotic intervention along Correa's cascade lies before the transition from AG to IM and this timepoint may be correlated with the observed changes in the microbial composition and ecosystem dynamics within the gastric environment.

Gastric microbial composition in gastric carcinogenesis

In parallel, the variations in the composition of the microbiota along the spectrum spanning from healthy gastric mucosa to GC were explored in several studies (Table 2). Through inter-comparison between the gastric microbiome of GC, pre-cancerous stages and normal tissue, these investigations unveiled the distinctive dysbiosis that characterizes gastric carcinogenic progress. A noteworthy observation is that gastric microbial composition is significantly influenced by hypochlorhydria, a main pathological manifestation of atrophy. This alteration in gastric acidity may result in increased vulnerability to colonization by microorganisms from other origins, primarily the oral cavity and intestine [33, 43, 48, 50, 53, 54].
For instance, a prospective cohort study undertaken by Sung et al. recruited 587 H. pylori-positive patients. They reported the presence of a specific assembly of oral bacteria, including Prevotella, Rothia, Peptostreptococcus, Parvimonas and Streptococcus, which were implicated in the occurrence and prolonged course of GA and IM [70]. A similar enrichment of oral microbes was found in a case–control study performed on 89 IM patients, where the abundance of Johnsonella ignava, Peptostreptococcus stomatis, Neisseria elongata, and Neisseria flavescens was found to be significantly higher in IM cases [56]. Moreover, an insightful study was conducted on a well-defined cohort, recruiting 34 cases of SC, 20 cases of AG, 5 cases of gastric intraepithelial neoplasia(GIN), and 15 cases of intestinal-type GC. Remarkably, they revealed a noticeable increase in the abundance of specific oral bacteria, including Slackia, Selenomonas, Bergeyella, and Capnocytophaga, which continuously increased from SG to GC. Concurrently, intestinal bacteria including Romboutsia, Fusicatenibacter, Prevotellaceae-Ga6A1-group and Intestinimonas were observed to be overabundant in GIN patients [22].
The predominance of these “non-indigenous colonizers” persists in GC which may significantly characterize malignant transformation. Coker et al. detected distinct Operational Taxonomic Units (OTUs) that were associated with five oral-derived species, including Peptostreptococcus stomatis, Streptococcus anginosus, Parvimonas micra, Lackia exigua, and Dialister pneumosintes, which can significantly distinguish GC from SG [53]. In another investigation where 54 GC patients and 81 chronic gastritis patients were enrolled, GC-representative genera exhibited a remarkable overabundance of intestinal commensals such as Citrobacter, Clostridium, Lactobacillus, Achromobacter and Rhodococcus [57]. Even by autologous comparison, tumor microhabitats were characterized by the discernible increase of oral pathogen, Prevotella melaninogenica, Streptococcus anginosus and skin pathogen Propionibacterium acnes (P.acnes), setting them apart from both paired paracancerous and normal tissues [58].

Gastric microbiota as predictive markers in gastric cancer

The recognition of microbial features during gastric carcinogenesis instigates exploration into the correlation between gastric microbiota and the multifaceted aspects of GC, such as risk, progression and prognosis. A Korean study, comprising a cohort of 268 GC patients and 288 controls, identified H. pylori, P.acnes and Prevotella copri as significant risk factors for GC, as indicated by odds ratios (ORs) of 1.86, 4.7 and 2.54 respectively [9]. Concurrently, another case–control study employed Whole metagenomic shotgun sequencing (WMS) on the gastric biopsy samples, which were obtained from a cohort of Colombian individuals, including both high-risk (n = 10) and low-risk (n = 10) populations for GC. The results revealed that the typical soil bacteria including Bacillus, Actinomyces and Arthrobacter spp., and Keratinibaculum spp. that was over representative in males, as well as skin commensal Staphylococcus and oral microbe Streptococcus, associated with an increased risk of GC [59].
In addition, a prospective longitudinal study was conducted to investigate the predictive value of gastric microbiota in GC progression. This comprehensive investigation involved 43 participants who underwent initial gastroscopy, followed by regular monitoring through 1–2 yearly gastroscopies over a minimum duration of 5 years. The results showed that patients who progressed from IM to early gastric neoplasia (EGN) during the study period had significantly higher relative abundance of Moryella genus and Vibro genus at baseline, compared to those who did not exhibit such progression [51].
Regarding the prognostic assessment of GC based on the gastric microbiome, multiple retrospective studies delved into distinctions in gastric microbial composition between GC patients with favorable and unfavorable prognoses. Several investigations highlighted the association between heightened levels of Fusobacterium nucleatum (F. nucleatum) in tumor samples and unfavorable prognostic outcomes among GC patients [60, 71, 72]. Notably, in Lauren’s diffuse-type GC, F. nucleatum positivity was found to be associated with markedly diminished overall survival rates. The abundance of F. nucleatum demonstrated a positive correlation with patient age, although it exhibited no significant associations with gender, H. pylori infection status, tumor stage, or tumor location [71]. Survival analysis from an additional cohort study has indicated that colonization of F. nucleatum species correlates with poorer prognosis among late-stage GC patients with H. pylori positivity [72]. These findings were further corroborated by an additional study showing that the presence of F. nucleatum and Prevotella species in tumor specimens has most significant impact on prognosis [60]. Furthermore, emerging evidence has also suggested potential correlations between specific bacterial species and favorable prognoses in GC. Using weighted gene co-expression network analysis (WGCNA), a recent study unveiled that GC patients exhibiting higher microbial diversity experienced poorer outcomes compared to those with lower microbial diversity. However, Tissierella enrichment was frequently observed in TP53 wild-type tumors and was associated with a favorable prognosis in various tumor types including GC, breast cancer and lung adenocarcinoma [73]. Another study revealed that GC patients with favorable prognoses displayed elevated H. pylori abundance alongside a decrease in Halomonas and Shewanella abundance [74]. Moreover, the presence of Asinibacterium in non-tumor adjacent tissues correlated with improved overall survival in GC patients, suggesting a potential influence of the gastric microenvironment on the pathophysiology of GC [60].

Role of the gastric microbiota in gastric carcinogenesis

H. pylori in gastric carcinogenesis through the “hit and run” mechanism

H. pylori, a known Class I carcinogen in the development of GC, exerts a significant predisposing effect on carcinogenesis by employing various virulence factors [75]. Its remarkable feature of urease synthesis enables it to thrive within the acidic environment of the stomach. The primary mechanism underlying H. pylori-induced carcinogenesis involves the expression of two oncogenic effector proteins: vacuolating cytotoxin A (VacA) and cytotoxin-associated gene A(CagA) protein [76] (Fig. 2).
VacA, a potent vacuolating cytotoxin present in all H. pylori strains, is capable of inducing vacuolation in host cells. VacA-forming pores can target mitochondria, causing depolarization of the mitochondrial transmembrane potential and subsequent mitochondrial dysfunction. This event triggers the release of cytochrome c from the mitochondria, which ultimately leads to the initiation of the apoptotic pathway [77]. The cag pathogenicity island (cagPAI) contains genes responsible for the expression of a type IV secretion system (T4SS) and the oncogenic protein CagA [78]. Upon the action of T4SS, H. pylori translocates CagA into the cytosol of host cells, where it serves as a hub protein to disrupt multiple cellular signaling pathways. CagA, when phosphorylated by Src family kinases, can engage with SH2 domain-containing proteins, which are mostly effectors in the mitogenic signaling pathway [79, 80]. Concurrently, unphosphorylated CagA interacts with E-cadherin to disassociate the E-cadherin/β-catenin complex and activate β-catenin-dependent expression of pro-oncogenic genes [81]. In addition to triggering oncogenic signals, CagA also elicits genetic instability, thereby exacerbating the potential for oncogenic transformation. The CagA-mediated sequestration of the polarity-regulating kinase PAR1b, initially known for its disruption of cellular polarity via tight junction perturbation [82], has recently been elucidated to play a role in inhibiting BRCA1 phosphorylation. This inhibition results in the confinement of BRCA1 to the cytoplasm, called BRCAness, where it fails to perform DNA repair in the nucleus, thereby inducing DNA double-strand breaks (DSBs) and genetic instability. The PAR1b kinase inhibited by CagA also activates Hippo signaling, thereby providing DNA-damaged cells with a means to escape apoptosis and facilitating the repair of DSBs via mutagenic repair mechanisms. As the cells with BRCAness expand, the pro-oncogenic role of CagA is eventually replaced by genomic instability which gives rise to cells with cancer predisposition phenotype independent of CagA. Consequently, the neoplastic phenotype no longer relies on the presence of CagA [83].
Moreover, H. pylori seems to assume a pioneering role by establishing a low-acid environment conducive to the colonization of potential pathogenic microorganisms in the stomach. For instance, a longitudinal study showed that patients with successful H. pylori eradication but still with persistent inflammation exhibit an increase in oral bacteria and pathogenic bacterial load [70]. The mechanisms underlying H. pylori-induced hypochlorhydria were partially unveiled. During acute H. pylori infection, T4SS protein CagL plays a dual role: it facilitates the intracellular delivery of CagA by binding to α5β1 integrins and also influences the dissociation of the metalloenzyme ADAM17 from α5β1 integrins. CagL stimulates ADAM17 activation, initiating ADAM17-triggered NF-κB-mediated suppression of H,K-ATPase α subunit (HKα) [84]. Besides, pro-inflammatory cytokines IL-1ß and TNF-α released by immune cells elicited by H. pylori infection are also robust inhibitors on the gastric acid secretion of parietal cells [85].
The “hit and run” theory was initially introduced by Skinner et al. to elucidate how viruses contribute to carcinogenesis by promoting the accumulation of mutations and inducing genomic instability until the virus is no longer necessary for tumor maintenance [86]. Consistently, genetic instability and neutralized PH in the stomach caused by H. pylori infection induces the accumulation of oncogenic insults including driver mutations acquisition in cancer-predisposing cells and pathogenic bacteria colonization in the stomach, which collectively contribute to the establishment and persistence of a neoplastic phenotype. And the actions of H. pylori successively fade in malignancy maintenance [87]. Aforementioned longitudinal study reported that eradicating H. pylori is insufficient to completely reverse the progression of IM [70]. Moreover, a study incorporating 9 gastritis, 7 IM and 11 GC subjects showed a decrease in H. pylori abundance in GC while no significant differences in the abundance of other dominant species between cancer and non-cancer group [55]. Similar conclusions were found in a Portuguese study (54 GC and 81 CG) in which H. pylori load reduced with increased abundance of intestinal commensal in GC versus chronic gastritis [57]. Based on a pathology review conducted by Stewart et al., the prevalence of H. pylori was found to be as high as 90% in cases of active gastritis, but it decreased to a range of 30% to 72% in individuals with atrophic gastritis and to approximately 30% to 35% in those with intestinal metaplasia (IM) and dysplasia. Notably, only 24.6% of GC patients had detectable H. pylori [3]. The “hit and run” model of H. pylori was initially introduced and documented by Hatakeyama. On the one hand, it highlights the action of CagA in cell reprogramming by facilitating genetic and epigenetic alternation, on the other hand, elevated genetic instability evokes host cell protection mechanisms such as cell senescence and apoptosis, impeding the role of CagA in transformed cells, ultimately resulting in the diminishing influence of CagA over time. Likewise, in the context of reduced gastric acid secretion and the absence of H. pylori, invasive microbiota successfully colonizes the stomach, initiating a progressive replacement of H. pylori by other microbial species that eventually come to dominate the gastric environment [87].

The interplay between H. pylori and non-H. pylori microbiota

The presence of H. pylori influences the microecological landscape in the stomach. In turn, gastric microbiota can also modulate the outcome of H. pylori infection. This interplay between H. pylori and the microbiota throughout Correa's cascade is potentially involved in GC development.
H. pylori demonstrates a strong competitive advantage, rendering it the predominant microorganism in the gastric microbiota, constituting 40%–90% of the total gastric microbiota composition [30, 38, 57, 88]. The presence of H. pylori is inversely correlated with the alpha diversity of gastric bacteria [58, 8890]. While the impact of H. pylori on alpha diversity can be reversible after the removal of H. pylori [70, 91]. Evidence from beta diversity analysis suggested that the presence of H. pylori infection leads to significant modifications to microbial composition in the stomach [89, 92]. Individuals infected with H. pylori exhibit a predominant composition of the same phyla as those without H. pylori, but with low levels of Actinobacteria, Bacteroidetes, and the overrepresentation of Proteobacteria, probably attributed to the influence of H. pylori [93]. Co-occurring and co-excluding networks showed the potential interplay between H. pylori and other gastric microbes. In SG, H. pylori manifests a pattern of co-exclusion with Methylobacillus and co-occurrence with Arthrobacter. Within the stomach of subjects with IM, H. pylori demonstrates co-exclusion with various members of the Firmicutes phylum, such as Ruminococcus, Bacillales, SMB53 and Lactobacillus, while concurrently displaying co-occurrence with Prevotella, Moryella and H. ganmani [53]. Among GC patients, a notable negative correlation persists between Lactobacillus and Helicobacter [23].
A study employing germ-free insulin-gastrin (INS-GAS) transgenic mice illuminated the crucial role of gastric microbiota in H. pylori-driven Correa's cascade progression. H. pylori infection in germ-free mice induced milder lesions and delayed progression to gastric intraepithelial neoplasia (GIN), in comparison to H. pylori-infected specific pathogen-free (SPF) INS-GAS mice [94]. Remarkably, it has been documented that gastric colonization involving H. pylori, along with a consortium of commensal microbiota encompassing species such as Clostridium spp., Bacteroides spp., and Lactobacillus murinus, led to the development of GC in INS-GAS mice to a degree on par with that observed in mice harboring a diverse microbiota. Moreover, the colonization of the stomachs of INS-GAS mice with complex microbiota even reduced the level of gastric H. pylori colonization. These findings indicate that the effective colonization with specific commensals seems to exert a greater influence than the overall microbial diversity in the context of H. pylori-associated GC [94, 95].
Several gastric microbiota with potential modulatory effects on the severity and outcomes of H. pylori infection have been identified [96101]. For instance, Staphylococcus epidermidis and Streptococcus salivarius, both urease-positive bacteria, were isolated from individuals in low- and high-cancer-risk regions of Colombia respectively. Co-infection experiments in germ-free mice showed that neither bacterial strain significantly altered H. pylori abundance. However, a more severe pathological lesion was found in H. pylori-S. salivarius dual infection while H. pylori-S. epidermidis co-infection showed alleviation of tissue injury and inhibited gene expression of pro-inflammatory cytokines including IL-22, IL-17A, IL-1β, IFN-γ and TNF-α in contrast to H. pylori mono-infection [96].
Moreover, Weizmannia coagulans (BCF-01), a strain isolated from the healthy gastric mucosa, displayed strong anti-H. pylori activity. This strain could significantly alleviate H. pylori-triggered gastric dysbiosis and reduce inflammation following H. pylori infection. Mechanistically, it enhanced the expression of mucosal barrier proteins and inhibited the TLR4-NFκB-mediated pyroptotic pathway in macrophages [102]. Similarly, Lactobacillus gasseri (Kx110A1), another strain isolated from gastric mucosa, effectively suppressed the synthesis of TNF and IL-6 in macrophages upon H. pylori infection through downregulation of ADAM17 [98]. Also Lactobacillus acidophilus and Lactiplantibacillus plantarum, both indigenous gastric strains, attenuated mucosal inflammation in murine gastritis afflicted with H. pylori [99]. Furthermore, Streptococcus mitis, commonly found in the human stomach, can inhibit the growth of H. pylori and prompt its transformation into a coccoid form when co-cultivated with H. pylori [100, 101]. Variations in the prevalence of these bacteria interacting with H. pylori within the gastric microbial community may underlie the disparate susceptibilities of individuals to H. pylori infection and the development of H. pylori-propelled GC.

The exploration of mechanisms involving non-H. pylori microbiota in gastric carcinogenesis

A recent study by Kwon et al. demonstrated the successful reproduction of premalignant lesions in the stomach by transplanting the gastric microbiota from IM and GC patients to germ-free mice, even in the absence of H. pylori. This discovery underscores the contribution of gastric dysbiosis beyond the involvement of H. pylori to the induction of GC [103]. While a definitive mechanistic elucidation remains insufficient, numerous investigations have indicated the potential implication of specific bacterial species in GC development.
To begin with, Streptococcus anginosus (S. anginosus),a species originating from the oral cavity and known for its marked resilience in acidic conditions (pH 3–5), has shown significant overabundance in the gastric mucosa of GC patients across various human cohorts [53, 58, 104]. Administration of S. anginosus via oral gavage in conventional and germ-free mice induced progressive precancerous lesions, including gastritis, parietal cell atrophy, mucinous metaplasia, and dysplasia. Further, S. anginosus in carcinogen-induced or allograft GC models expedited tumor progression by disrupting gastric barrier function, stimulating cell proliferation and preventing apoptosis. Notably, the influence of S. anginosus on gastric carcinogenesis appeared to be species-autonomous, which relied on the interaction between a bacterial lipoprotein, TMPC, and Annexin A2 (ANXA2) receptor on gastric epithelial cells. This interaction instigated oncogenic MAPK signaling cascades, as evidenced by the phosphorylation of ERK and JNK, as well as activation of downstream oncogenic targets within gastric epithelial cells following S. anginosus infection [105].
Another frequently cited candidate associated with GC, showing significant overabundance in the gastric microbiota of GC patients, is Lactobacillus species [8, 22, 23, 54, 55, 57, 61, 62]. Some hypotheses suggested that Lactobacillus spp. may contribute to GC development by producing lactic acid, which could serve as an energy source for tumor cells, promote reactive oxygen species (ROS) production, and contribute to tumor angiogenesis [67, 106, 107]. Nevertheless, the current body of proofs falls short of providing definitive support for these hypotheses. A converse viewpoint has been proposed by several studies, suggesting that Lactobacillus spp. could play an anti-tumor role in GC. This includes their potential to activate the intrinsic apoptotic pathway in cancer cells [108], prevent H. pylori infection [109, 110], and regulate gastric dysbiosis [111]. Furthermore, recent observations presented an alternative perspective indicating that the overgrowth of Lactobacillus sppmay be a consequence rather than a cause of gastric carcinogenesis [112, 113]. Jin et al. conducted long-term treatment of INS-GAS mice with deoxycholic acid (DCA), a secondary bile acid that increases from chronic gastritis to intestinal metaplasia. DCA treatment accelerated the progression to IM and enriched the population of the Lactobacillus genus [113]. In light of these complexities, further scrutiny is indispensable to elucidate the multifaceted role of Lactobacillus spp. in GC.
Additionally, lipopolysaccharide (LPS)-producing bacteria were found to be present at the high relative abundance in the gastric microenvironment of patients suffering from bile reflux gastritis (BRG) and GC. Among these bacteria, an oral-derived pathogen, Prevotella melaninogenica (P. melaninogenica) was found to be prominently elevated [58, 114, 115]. The abundance of LPS-producing bacteria exhibited a positive correlation with taurodeoxycholic acid (TDCA), a secondary bile acid that can stimulate the growth of gastric epithelial cells by triggering a pro-inflammatory IL-6/JAK1/STAT3 signaling cascade. The gavage instillation of TDCA, LPS, and P. melaninogenica in mice induced gastric inflammation and pre-neoplastic lesions, indicating the potential involvement of LPS-producing microbes in the pathogenesis of GC [114]. Therefore, targeting these microbes through alternative interventions may hold promise as a preventive measure against BRG-associated GC.
Microbial species participating in the metabolism of nitrate and nitrite have been implicated in GC. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) analysis from multiple studies unveiled distinctive microbial functional characteristics in GC that are linked to heightened reduction reactions, converting nitrate to nitrite, a precursor of N-nitroso compounds (NOC) [57, 61, 63]. Indeed, bacteria possessing nitrate reductase enzymes such as Clostridium, Lactobacillus, and Neisseria were found to be significantly overrepresented in GC patients [23, 55, 57, 91]. Conversely, Nitrospira, a bacterium that oxidizes nitrite to nitrate, were found to progressively decrease in individuals with intraepithelial neoplasia (IN), IM and GC. This decrease may indicate the accumulation of nitrite during the progression of neoplastic lesions [52]. The buildup of intragastric nitrite and NOC facilitates malignant transformation by initiating DNA damage through the methylation of purine and guanine with electrophilic species, thereby increasing mutagenicity [116, 117].
A novel investigation unveiled distinctive mucin-microbiome profiles in association with clinical outcomes in GC. The results indicated that tumors displaying an intestinal mucin phenotype with upregulated MUC13 expression are associated with an unfavorable prognosis, whereas tumors characterized by dominant expression of MUC5AC or MUC6 exhibit a more encouraging outcome. The presence of specific oral or intestinal microbes was discovered to be dependent on the mucin phenotype. Notably, oral taxa such as Neisseria, Prevotella, and Veillonella showed a significant association with MUC13 overexpression [118]. In prior studies, MUC13 overexpression was documented in GC [119], and this dysregulated MUC13 signaling was shown to confer protection against apoptosis in colorectal cancer (CRC) cells through the activation of the NF-κB pathway [120], as well as to facilitate the progression of intrahepatic cholangiocarcinoma by activating the EGFR/PI3K/AKT pathways [121]. Nevertheless, its specific involvement in GC has yet to be thoroughly investigated. Whether such interactions between mucin and the microbiome causally contribute to gastric carcinogenesis remains an intriguing area for further exploration.
Microbiota-mediated epigenetic regulation represents a novel mechanism by which gastric bacteria may contribute to carcinogenesis [122]. For instance, tumors harboring F. nucleatum exhibited a trend towards decreased DNA methylation of the long interspersed nuclear elements 1 (LINE1) [71]. Kytococcus sedentarius, Actinomyces oris and Staphylococcus saccharolyticus were implicated in regulating DNA methylation of immune-related genes in gastric adenocarcinoma, which consequently promoted distal metastasis in GC. In an in vitro experiment, the co-culture of GC cells with Staphylococcus saccharolyticus was observed to enhance the proliferation and clonogenicity of GC cells by inducing loss of ZNF215 methylation imprinting [123].
Several microorganisms implicated in other cancers were observed to be also enriched in GC [23, 55, 60, 91]. For instance, F. nucleatum, a periodontal commensal, was reported to be involved in the carcinogenesis of CRC [124]. Two vitro studies provided preliminary insights into the potential role of F. nucleatum in the development of GC. Hsieh et al. revealed that F. nucleatum colonization results in the dysregulation of actin cytoskeletal dynamics, which in turn is likely to alter the motility of GC cells [72]. In another study, it was shown that F. nucleatum is capable of inducing the overexpression of exosomal HOTTIP from GC cells, which subsequently accelerates the progression of GC through the activation of the miR-885-3p/EphB2/PI3K/AKT signaling cascade [125]. However, the impact of F. nucleatum on GC lacks validation from in vivo animal studies, and the molecular mechanisms underlying this effect are not fully understood. In CRC, the carcinogenic effect is primarily mediated by its virulence factors [8, 64, 126, 127]. Fap2 lectin ensures the localization and enrichment of F.nucleatum in cancerous sites [128, 129]. And the interaction of FadA adhesin and E-cadherin of cancer cells over-activate the β-catenin/Wnt signaling pathway [130]. Additionally, LPS of F.nucleatum recognized by the Toll-Like Receptor 4/Myeloid Differentiation Primary Response 88 (TLR4/MYD88) system induces NF-κB signaling pathway activation [131]. Notably, sustained activation and abnormal modulation of the NF-κB signaling pathway play a crucial role in the maintenance of stem-cell-like prosperity of GC cells [131137]. Except for F.nucleatum, H. pylori has been demonstrated to stimulate NF-κB signaling pathway activation via its distinctive effector molecule, ADP-glycerol-β-D-mannoheptulose [138, 139], Peptostreptococcus anaerobius and Bacteroides fragilis which were observed to increase in GC, can activate NF-κB signaling pathway via inducing intracellular synthesis of ROS and IL-17 respectively in CRC [140, 141]. However, it remains unclear whether these mechanisms of the bacteria are replicated in the acidic environment of the stomach.

Non-bacterial component in gastric cancer

Although the majority of investigations into the connection between the microbiome and GC have primarily focused on bacterial components, a limited number of studies adopting a pan-biological perspective have yielded insights into the tumorigenic potential of non-bacterial elements, conferring a more comprehensive depiction of the microbiome ecosystem within the context of cancer [142144].
One published by Zhong et al. in 2021 initially characterized fungal features of GC by applying internal transcribed spacer (ITS) rDNA sequencing on 45 gastric cancerous biopsies and peritumor normal biopsies. Candida and Alternaria were detected as representatives of fungi in cancerous tissue whereas Saitozyma and Thermonyces were observed to decrease in GC. Although the causal role of fungus on gastric carcinogenesis has not been proved, they highlighted the role of Candida albicansas an effective biomarker to distinguish GC tissue from control [143]. Candida albicans is a common fungus in human microbiota that normally colonizes but can cause severe issues in immunocompromised patients [145]. Recently bioinformatics analysis of mycobiome performed by Dohlman et al. demonstrated the unique enrichment of Candida in GC tissues. Concerning endosymbiotic crosstalk, authors conducted trans-kingdom network analysis and identified Candida as “Keystone taxa” in cancer which is co-abundant with a cluster of tumor-specific microbiota including Streptococcus, Clostridium, and Lactobacillus in GC. Moreover, they revealed Candida-corresponding gene expression patterns in GC, where pro-inflammatory cytokines such as IL-8, IL-1β, and IL-6 were mainly upregulated, indicating the involvement of Candida in inflammation in GC. However, it is still elusive whether overrepresented Candida is a cause or secondary to the phenotype of GC [144].
The majority of Mycoplasma implicated in GC are Mycoplasma hyorhinis(M. hyorhinis), a recognized pathogen responsible for various swine diseases [127, 146]. Experimental studies involving co-culturing M. hyorhinis with host cells have shed light on its impact on GC cell lines. Lipoprotein P37, expressed on the membrane of M. hyorhinis, serves as the main virulence factor, significantly enhancing the invasiveness of GC cells. This enhancement is strongly correlated with the activation of the EGFR-dependent NF-κB signaling pathway and the activation of matrix metalloproteinase-2(MMP-2) [147, 148]. Besides, Lipid-associated membrane protein(LAMP) of M.hyorhin, recognized by TLR2, induces the monocyte-derived IL-1ß secretion in an NLRP3 inflammasome-mediated manner to enhance cell migration [149]. Furthermore, M.hyorhin-specific methyltransferases selectively act on CG and GATC sites may lead to pro-carcinogenic epigenetic alternation by silencing tumor-suppressing genes [150].

Microbial involvement in immunosurveillance of gastric cancer

In a state of homeostasis, the persistent mutualistic symbiosis between the host and commensal flora not only serves to safeguard the microbial ecosystem but also plays a crucial role in preserving immunological equilibrium. Nonetheless, when the intricate interplay goes awry due to dysbiosis or disruptions in the host system, opportunistic pathogens can seize the opportunity to initiate pathological processes. In this scenario, pathogen-associated molecular patterns (PAMPs), such as flagellin, LPS, peptidoglycan, and formylated peptides recognized by pathogen recognition receptors (PRRs) of host cells may initiate an aberrant immune response. Similarly, within the realm of cancer, the presence of tumor-enriched pathogens harbors the capacity to disrupt the balance between immune defenses and tumor evasion, ultimately facilitating the progression of the disease [75] (Fig. 3).

Potential role of gastric microbes on the innate immune system in gastric cancer

Innate immunity encompasses a diverse array of myeloid or lymphoid lineages-derived immune cells, cytokines and complement cascades, all working in concert to coordinate immune balance. The innate immune cells do not possess an intrinsic predisposition to either favor or impede tumors. Instead, their phenotype and function exhibit plasticity, influenced by contextual cues within specialized niches and the signaling molecules they encounter. Among these, the microbial pattern acts as a key component that significantly shapes the plasticity, recruitment, activity, and overall functionality of the innate immune system. In this regard, we present pivotal findings that shed light on the remodeling of innate immune cells by the microbiota within the TME of GC, and how these interactions exert profound effects on the course of cancer.
Macrophages are a significant component of the leukocytic infiltrate and exhibit phenotypic plasticity in GC. The M1 phenotype, triggered by IFN-γ and microbes-derived molecules, has anti-tumor activities. Conversely, the M2 phenotype, induced by IL-4, IL-10, and IL-13 cytokines, exerts an immunosuppressive effect, encouraging the immune escape of cancer cells [151]. A recent study revealed that P. acnes, a skin pathogen prevalent in GC, can drive the polarization of macrophages toward the M2 phenotype, facilitating GC cell migration in vitro. Pre-exposure of macrophages to P. acnes induces the expression of TLR4 on their surface. This, in turn, initiates an interaction between TLR4 and P. acnes, activating the downstream PI3K/AKT signaling pathway, which upregulates IL-10 expression, a canonical stimulus for promoting M2 polarization. Furthermore, co-localization analysis of M2 and TLR4 in GC tissue samples observed sustained expression of TLR4 in the majority of M2 macrophages. This observation implies that the overrepresentation of P. acnes within TME may be involved in a positive-feedback loop for M2 polarization, as self-secreted IL-10 further enhances the M2 phenotype [152].
Group 2 innate lymphoid cells (ILC2s), the principal subset of innate lymphoid cells in the stomach, respond to tissue injury and bacterial exposure to expand with the production of type II cytokines (IL-4, IL-5, IL-9, and IL-13) [153]. The mutualistic relationship between ILC2s cells and the microbiota is implicated in gastric homeostasis maintenance. ILC2s-derived IL-5 efficiently stimulates B cells to secrete IgA, which coats gastric bacteria for elimination. Reciprocally commensal bacteria such as Bacteroidales Family S24-7 actively participate in the preservation of the abundance and functionality of ILC2s within the stomach [154]. However, in TME, the microbiota-ILC2 interaction undergoes dysregulation, leading to a pronounced skew favoring a pro-tumor activity. The ILC2s in the peripheral blood of GC patients were found to exhibit an immunosuppressive phenotype [155]. And Li et al. reported that H. pylori infection induces the overexpression of transcription factor GATA-3 in lymphocytes in GC, contributing to the reprogramming of immune responses towards a dominant type 2 immunity, which is predominantly mediated by immunosuppressive ILC2s [156]. Simultaneously, H. pylori infection elicits an upregulation of the surface expression of interleukin-7 receptor (IL-7R) on ILC2s and induces an elevation in the gastric levels of IL-7, which synergistically enhances the recruitment and propagation of ILC2s in the stomach, potentially exacerbating the immunosuppressive nature of the TME [154, 157].
Tumor-activated neutrophils (TANs), a subset of granulocytes, constitute an immune-inhibitory milieu and are also recruited in response to microbial influences in TME [158]. TANs orchestrate a multifaceted role in promoting tumorigenesis by producing pro-angiogenic factors [159], enhancing the metastatic propensity of cancer cells [160] and suppressing anti-tumor immune responses [161]. Their mobilization in GC can be mediated through chemotaxis along the CXCL6/CXCL8-CXCR1 pathway [162]. Studies highlighted that H. pylori-induced upregulation of hepatocyte growth factor and IL-8 (CXCL8) triggers the infiltration of neutrophils in chronic gastritis and GC [163]. Moreover, IL-17, a cytokine abundant within the mucosa, was also demonstrated to engage in crosstalk with cancer cells, coordinating the recruitment of neutrophils [164, 165]. The intracellular inhabitation of F. nucleatum in GC cells was observed to trigger the NF-κB /IL-17 signaling axis, leading to neutrophil recruitment and facilitating immune evasion by tumor cells [166].
In addition, several bacteria, despite their roles remaining partially understood, were identified in association with specific immune cell populations within TME of GC. For instance, an opportunistic pathogen, Stenotrophomonas was found to exhibit a positive correlation with the infiltration frequency of BDCA2 + plasmacytoid dendritic cells (pDCs) in GC tissues [167]. These pDCs, existing in an immature state, are known to positively regulate the activity of regulatory T cells (Treg), thereby promoting the suppression of anti-tumor immunosurveillance [168, 169]. Likewise, Fusobacterium sp. infection showed a positive association with the lymphocyte influx in GC [126]. The direct immunosuppressive impact of F. nucleatum on human Natural Killer (NK) cells were elucidated in CRC, involving its interaction with the inhibitory receptor TIGIT via the virulence protein Fap2 [170]. However, the applicability of this immunosuppressive mechanism in GC requires further investigations to validate.

Potential role of gastric microbes on the adaptive immune system in gastric cancer

The adaptive immune system, composed of T-cell-mediated and B-cell-mediated immunity, plays an important role in the surveillance and defense against neoplastic growth. The intricate interplay between the gastric microbiota and the adaptive immune system emerges as a significant determinant in shaping the anti-tumor immune response, thus offering the potential to modulate the dynamic process of GC development and progression [22, 65, 126].
The plasticity of T cells is dependent on various stimuli and signals encountered during lineage differentiation and activation [171]. Studies illustrated the impact of gastric microbiota on the balances of effector and Tregs as well as the regulation of co-stimulatory and co-inhibitory signal transmission [75, 172, 173]. For instance, the overabundance of Methylobacterium in GC tissue, highly associated with unfavorable prognostic outcomes in GC patients, was demonstrated to advance GC progression by exhausting CD8 + tissue-resident memory T cells (TRMs) in TME [65, 66]. TRMs, distinguished by their elevated expression of immune checkpoint molecules and effector proteins, serve as a reserve force in anti-tumor immunity and was recognized as a favorable prognostic indicator linked to prolonged patient survival in prior studies [174]. Animal experiments unveiled Methylobacterium's capacity to diminish CD8 + TRMs by downregulating TGFß. This is significant since TGFß plays a pivotal role in the induction of CD103 expression, an essential prerequisite for the TRMs generation and enduring residence within the epithelial tissue [65].
Another bacterium that hampers T cell-mediated anti-tumor immunity is H. pylori. Aydın et al. reported an upregulation of immune-checkpoint inhibitor proteins (PD-1/PD-L1) in H. pylori-positive GC samples [175]. Experimental studies unveiled a putative mechanism underlying H. pylori-mediated PD-L1 expression. Using GC organoids, it was discovered that CagA from H. pylori stimulates increased secretion of the signaling molecule Shh, thereby augmenting the Hedgehog signaling pathway. This intricate cascade of events has the potential to modulate PD-L1 expression by regulating the mTOR pathway [176, 177].
Tregs emerge as pivotal orchestrators of the balance of immune tolerance and immune activation in homeostasis. However, within the complex landscape of cancer, a perturbation unfolds, where with the dominance of Treg cells surpasses that of effector T cells, which skews TME towards a state of immunosuppression. This dysregulation, in turn, allows cancer cells to evade vigilant immune surveillance [178]. Foxp3 + Tregs were reported to be over-represented in gastric tumoral and peritumoral samples compared with normal tissue. Selenomonas, a typical gingival bacterium, enriched in GC, exhibited a positive correlation with the elevated level of the Foxp3 + Tregs as shown in Pearson's correlation analysis. This suggests its potential contribution to Tregs accumulation [22, 167]. Moreover, a strong association was observed between H. pylori infection and the presence of ICOS + Tregs, of which higher level was linked to an unfavorable prognosis in late-stage GC [179]. Within the realm of co-stimulatory receptors belonging to the CD28 family, ICOS stands out as a distinct subtype, characterized by its unique ability to enhance the efficiency of producing immunosuppressive cytokines, including IL-10 and TGF-β. The presence of H. pylori in the gastric environment sets off a chain of events, activating TLR9 and instigating the induction of ICOS ligands on pDCs. This process serves as a compelling signal summoning ICOS + Tregs to infiltrate the TME of GC [180].
It was established that B lymphocytes act as anti-tumor immune agents by producing counterpart antibodies [181]. The immunoglobulin pool and repertoire can be influenced by microbial exposure, which may affect B cell recognition and function of tumor cells [182]. A subtype of B cell identified as B regulatory cells (Breg) exhibit a series of immunosuppressive features including synthesis of IL-10, IL-35 and TGF-β, counteracting the activity of effector T cells and NK cells, which are implicated in the immune evasion mechanisms employed by tumor cells [183, 184]. Evidence showed that the density of IL-10-producing B cells was elevated in the presence of H. pylori [185]. However, how Bregs respond to H. pylori and other microbes is still less well-studied.

The impact of microbiota on cancer immunotherapy

Immunotherapy that aims to boost the body's immune system to target and eliminate cancer cells has been well-established in clinics. However, it only works well for a small subset of cancer patients, underscoring substantial challenges in achieving favorable outcomes, as a significant proportion fails to mount an adequate response or develops treatment resistance [186]. Robust evidence highlighted the regulation of the microbiome on immune surveillance, inspiring the investigation into the impact and application of microbes on immunotherapy [75, 175, 187]. Given considerable inter-individual variability in the microbiome, the patient's distinctive microbial profile may serve as a pivotal factor in predicting and optimizing the therapeutic response to immunotherapy.

Gut microbiota and immunotherapy

The lion's share of experimental investigations exploring microbiota's impact on immunotherapy has revolved around gut microbiota, revealing several microbial taxa associated with immunotherapy efficacy [188192]. For instance, Bifidobacterium genus, Bacteroides fragilis, Faecalibacterium spp., Akkermansia muciniphila, Ruminococcaceae and Lachnospiraceae were frequently identified as “favorable” bacteria to improve immune checkpoint inhibitors (ICIs) in multiple solid tumors by potentiating anti-tumor immunity [188, 193198], while the high abundance of Bacteroidales was reported to be associated with poor response and weakened tumor-specific immune response [192, 195]. In GC patients, Peng et al. observed an increase in microbial taxa associated with favorable responses to ICIs, including Prevotealla, Bifidobacterium, Bacteroides, Ruminococcaceae, and Lachnospiraceae, whereas non-responders showed a higher abundance of Megamonas, Butyricimonas, Lachnospiraceae_UCG-001, and Agathobacter [199]. This study exclusively concentrated on gut microbial alternation, even though the modulation of the local immune response in the stomach by the gastric microbiota is anticipated to have a potential influence on the efficacy of ICI.

The impact of H. pylori infection and eradication on Immunotherapy

Recently, the impact of H. pylori in gastric microbiota on immunotherapy is increasingly becoming a topic of great interest. Oster et al. found that the responsiveness of ICIs is hampered by H. pylori infection in both MC38 colon adenocarcinoma and B16-OVA melanoma-bearing mice model as well as non-small cell lung cancer (NSCLC) patients. The tumor size in mice with H. pylori infection was much larger than those without infection after anti-CTLA4 or anti-PD-L1 treatment. Likewise, following treatment with anti-PD-1, the median survival time of NSCLC patients with H. pylori seropositivity was found to be significantly shorter in comparison to their seronegative counterparts. The mechanistic investigation in murine models unveiled that the infection causes the hypoactivation of DCs, ultimately leading to compromised activation and proliferation of tumor-specific CD8 + T cells. Concurrently, a decrease in the abundance of monocyte-derived cells and a noteworthy suppression of genes under the regulation of type I IFN, IFNγ, and IL-6 were detected within the TME of NSCLC patients who exhibited resistance to ICIs therapy and had pre-existing H. pylori infection [200]. The other two retrospective cohort studies confirmed the negative impact of H. pylori infection on the response to ICIs. Both ICI-treated melanoma and GC patients with H. pylori seropositivity experienced lower overall survival rates and worse treatment outcomes than H. pylori-seronegative patients [201, 202].
Notably, despite the adverse impact of H. pylori on immunotherapy, Oster et al. were unable to restore the efficacy of immunotherapy even after the successful eradication of H. pylori. They attributed this outcome to the decrease in immune-boosting bacteria resulting from non-specific antibiotics (ATB) treatment [200]. This finding is consistent with many clinical investigations that demonstrated a correlation between the use of antibiotics and decreased clinical efficacy of ICIs [190, 203205]. Regarding the taxonomic composition of the gut microbiome post-eradication of H. pylori, several studies reported a gut dysbiosis characterized by a reduction in alpha diversity [206, 207] and Bifidobacterium abundance [208, 209]. However, longitudinal studies observed that these effects are transient, with a gradual recuperation of alpha diversity over time [210] and even favorable changes in fecal microbiota composition observed in patients, such as a noteworthy enrichment of Bifidobacterium and a decrease in Bacteroidales, after a period of 6 months post-eradication [91]. Moreover, different time windows of exposure to ATB before ICI initiation may exhibit different therapeutic outcomes. A comprehensive meta-analysis conducted by Lurienne et al. found that ATB exposure within 60 days of ICI administration significantly reduced its effectiveness in NSCLC patients, while exposure more than 90 days prior did not result in worse clinical outcomes [211].
Although the impact of antibiotics in H. pylori eradication on immunotherapy needs to be explored in prospective preclinical and clinical studies that include multiple time points, there seems to be agreement that antibiotics lead to transient ICI-unfavorable perturbations of microbial homeostasis, which should be considered in immunotherapy regimens for cancer patients.

The therapeutic potential of H. pylori in immunotherapy

In addition to the immunosuppressive effects of H. pylori infection within the TME, multiple researchers have delved into the strategic harnessing of its immunostimulatory property to augment the therapeutic efficacy of immunotherapy [212215].
Helicobacter pylori neutrophil-activating protein (HP-NAP) is a virulence factor in H. pylori, known for its ability to activate neutrophils and induce reactive oxygen species via TLR2 activation [216]. HP-NAP activates neutrophils and monocytes, promoting IL-12 and IL-23 production, which polarizes a CD4 + T cell response into an anti-tumor Th1 response characterized by increased IFN-γ and TNF-α [217]. It also aids DC maturation and shifts macrophages towards an anti-tumoral phenotype [218]. These potent immunogenic properties make HP-NAP a promising candidate for the development of novel cancer immunotherapeutic applications.
Given these, Codolo et al. conducted the first investigation of the therapeutic potential of HP-NAP in bladder cancer-bearing mice. Peritumoral administration of HP-NAP resulted in a reduction of tumorigenesis, promotion of tumor necrosis, and augmentation of CD4 + and CD8 + cell populations secreting IFN-γ within both tumor sites and associated lymph nodes [219]. In subsequent studies, the efficacy of Hp-NAP in various formulations was extensively explored for its anticancer properties. Hp-NAP recombined with the maltose binding protein of Escherichia coli(rMBP-NAP) stimulated TLR2-mediated Th-1-dependent anti-tumor immunity in hepatoma, sarcoma [220], and metastatic lung cancer [221]. Recombinant HP-NAP-embedded Chitosan nanoparticles (Chi-rNap) was reported to reduce breast tumor growth by enhancing cytokine production and shifting immune functionality towards tumor-killing [222]. And the genetic manipulation of oncolytic viruses (OVs) to HP-NAP, harnessing the selective oncolysis of OVs with the immunomodulatory properties of HP-NAP, was demonstrated to amplify anti-tumor immune responses in breast cancer and neuroendocrine tumors [212, 213]. Moreover, a recent study introduced chimeric antigen receptor (CAR) T cell-bearing secretory HP-NAP as a solution for CAR-T's limited effectiveness in solid tumors. CAR(NAP) T cells prompted DC maturation, elicited bystander T-cell responses, and activated cytotoxic CD8 + T cells against multiple tumor antigens, not just the CAR's target. Importantly, CAR(NAP) T cells didn't increase non-specific toxicity or hinder CAR T cell therapy, offering promising clinical potential [214]. These remarkable findings highlight the immense potential of H. pylori in revolutionizing immunotherapy, although practical applications in GC are pending.

Conclusion and prospect

Growing numbers of studies in recent years have uncovered the alternation of gastric microbiota during gastric carcinogenesis, particularly changes in diversity and composition. These changes may serve as promising biomarkers for clinical applications of GC. Additionally, the GC-associated microbial community actively helps tilt TME towards a more immunosuppressive state, contributing to the evasion of cancer cells from immune detection and clearance. Notably, H. pylori possesses an intricate immunomodulatory mechanism. On one hand, its immunosuppressive nature poses a detrimental impact on the effectiveness of immunotherapy. On the other hand, the virulence factors of H. pylori exhibit remarkable immune-stimulating properties, holding promise as adjunctive mediators for immunotherapy.
However, the area is still in its infancy and the tasks trying to elucidate the underlying mechanism through which gastric microbiota influence the carcinogenic process and mediate immunomodulation are challenging. For instance, past retrospective investigations presented intriguing and contentious evidence for linking gastric microbes and cancer. This presumably reflects the co-evolutionary dynamics of the host's immune system, its symbiotic microbiota, and tumorigenic processes, and such dynamic changes are typically unable to be captured by cross-sectional comparison. Concurrently, the variability in sequencing methodologies presents a significant hurdle in attaining a consensus regarding the landscape of the gastric microbiota. Thus, it is necessary to allocate increased research efforts to this area, including the establishment of multi-center longitudinal cohort studies and the application of standardized sequencing tools with the capacity to dissect the entire metagenome at the strain level (e.g., emerging third-generation sequencing technologies [223] and single-cell microbial sequencing [224]).
Furthermore, despite the extensive interest in the involvement of non-H. pylori gastric microbiota in GC, there is limited evidence regarding their causality with GC and their roles in the TME [173]. In light of this, there is a compelling need for in-depth research to monitor the co-evolutionary dynamics within the gastric microbiota. These researches are aimed at achieving a more comprehensive understanding of its multifaceted role in the tumorigenesis process. It is noteworthy to underscore that this exploratory venture should transcend the bacterial taxonomic level and encompass the full spectrum of microorganisms.
Exploring the clinical translation related to microbiota is a pivotal area of future research. Nevertheless, significant challenges persist in the clinical application of gastric microbiota. The invasive nature of endoscopy poses a substantial impediment to early GC screening, potentially limiting the clinical adoption of gastric mucosal microbiota testing as a viable biomarker. Moreover, current studies investigating the influence of gastric bacteria on cancer prevention and therapeutic response are primarily focused on H. pylori, with other strains remaining underexplored. Additionally, the applicability of established microbiota-based therapeutic strategies, including prebiotics, symbiotic microbial consortia, microbiota-derived metabolite therapy, microbiota-targeted interventions, and microbiota engineering, to the gastric microbiota ecosystem remains unclear, particularly given the stomach's unique acidic conditions. Interestingly, despite these challenges, emerging technologies and accumulating evidence indicate some limited progress in this field. For example, the detection of GC-related microbial translocation in the blood [225], oral cavity [56], and feces of GC patients has shown promising predictive significance and offers a non-invasive means for early GC screening [226]. Regarding of microbiota-targed treatment, studies have investigated strategies to target H. pylori, such as developing biomaterials that utilize the gastric acid environment as an activation mechanism to selectively eradicate H. pylori from the stomach [227, 228], and genetically modifying probiotics to release H. pylori-guided antimicrobial peptides [229]. These approaches have the potential to eliminate H. pylori, all while avoiding the induction of gastric dysbiosis. However, extensive clinical trials, underpinned by rigorous preclinical experiments, are essential to assess the safety and efficacy of these strategies and enhance our comprehension of GC prevention and treatment through the modulation of gastric microorganisms, extending beyond H. pylori (Fig. 4).
Going forward, the personalized treatment of cancer will increasingly integrate the microbiome with genomic, transcriptomic, and metabolomic data in the future. This multi-level and multi-dimensional approach will enable a more comprehensive and nuanced understanding of the interplay between the gastric microbiome and GC, facilitating the development of targeted and effective treatment strategies.

Acknowledgements

We thank all our research team members for their invaluable contributions, constructive feedback, and insightful suggestions.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Ajani JA, D’Amico TA, Bentrem DJ, Chao J, Cooke D, Corvera C, et al. Gastric cancer, version 2.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2022;20(2):167–92.PubMedCrossRef Ajani JA, D’Amico TA, Bentrem DJ, Chao J, Cooke D, Corvera C, et al. Gastric cancer, version 2.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2022;20(2):167–92.PubMedCrossRef
2.
Zurück zum Zitat Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat Rev Clin Oncol. 2023;20(5):338–49.PubMedCrossRef Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat Rev Clin Oncol. 2023;20(5):338–49.PubMedCrossRef
4.
Zurück zum Zitat Malfertheiner P, Camargo MC, El-Omar E, Liou JM, Peek R, Schulz C, et al. Helicobacter pylori infection. Nat Rev Dis Primer. 2023;9(1):1–24.CrossRef Malfertheiner P, Camargo MC, El-Omar E, Liou JM, Peek R, Schulz C, et al. Helicobacter pylori infection. Nat Rev Dis Primer. 2023;9(1):1–24.CrossRef
5.
Zurück zum Zitat Yeoh KG, Tan P. Mapping the genomic diaspora of gastric cancer. Nat Rev Cancer. 2022;22(2):71–84.PubMedCrossRef Yeoh KG, Tan P. Mapping the genomic diaspora of gastric cancer. Nat Rev Cancer. 2022;22(2):71–84.PubMedCrossRef
6.
Zurück zum Zitat Yin J, Wu X, Li S, Li C, Guo Z. Impact of environmental factors on gastric cancer: a review of the scientific evidence, human prevention and adaptation. J Environ Sci. 2020;89:65–79.CrossRef Yin J, Wu X, Li S, Li C, Guo Z. Impact of environmental factors on gastric cancer: a review of the scientific evidence, human prevention and adaptation. J Environ Sci. 2020;89:65–79.CrossRef
7.
Zurück zum Zitat Asaka M, Kobayashi M, Kudo T, Akino K, Asaka Y, Fujimori K, et al. Gastric cancer deaths by age group in Japan: outlook on preventive measures for elderly adults. Cancer Sci. 2020;111(10):3845–53.PubMedPubMedCentralCrossRef Asaka M, Kobayashi M, Kudo T, Akino K, Asaka Y, Fujimori K, et al. Gastric cancer deaths by age group in Japan: outlook on preventive measures for elderly adults. Cancer Sci. 2020;111(10):3845–53.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Castaño-Rodríguez N, Goh KL, Fock KM, Mitchell HM, Kaakoush NO. Dysbiosis of the microbiome in gastric carcinogenesis. Sci Rep. 2017;7(1):15957.PubMedPubMedCentralCrossRef Castaño-Rodríguez N, Goh KL, Fock KM, Mitchell HM, Kaakoush NO. Dysbiosis of the microbiome in gastric carcinogenesis. Sci Rep. 2017;7(1):15957.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Gunathilake MN, Lee J, Choi IJ, Kim YI, Ahn Y, Park C, et al. Association between the relative abundance of gastric microbiota and the risk of gastric cancer: a case-control study. Sci Rep. 2019;9(1):13589.PubMedPubMedCentralCrossRef Gunathilake MN, Lee J, Choi IJ, Kim YI, Ahn Y, Park C, et al. Association between the relative abundance of gastric microbiota and the risk of gastric cancer: a case-control study. Sci Rep. 2019;9(1):13589.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Chen BY, Lin WZ, Li YL, Bi C, Du LJ, Liu Y, et al. Roles of oral microbiota and oral-gut microbial transmission in hypertension. J Adv Res. 2023;43:147–61.PubMedCrossRef Chen BY, Lin WZ, Li YL, Bi C, Du LJ, Liu Y, et al. Roles of oral microbiota and oral-gut microbial transmission in hypertension. J Adv Res. 2023;43:147–61.PubMedCrossRef
11.
Zurück zum Zitat Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56.PubMedCrossRef Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56.PubMedCrossRef
12.
Zurück zum Zitat O’Dwyer DN, Ashley SL, Gurczynski SJ, Xia M, Wilke C, Falkowski NR, et al. Lung microbiota contribute to pulmonary inflammation and disease progression in pulmonary fibrosis. Am J Respir Crit Care Med. 2019;199(9):1127–38.PubMedPubMedCentralCrossRef O’Dwyer DN, Ashley SL, Gurczynski SJ, Xia M, Wilke C, Falkowski NR, et al. Lung microbiota contribute to pulmonary inflammation and disease progression in pulmonary fibrosis. Am J Respir Crit Care Med. 2019;199(9):1127–38.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Uberoi A, Bartow-McKenney C, Zheng Q, Flowers L, Campbell A, Knight SAB, et al. Commensal microbiota regulates skin barrier function and repair via signaling through the aryl hydrocarbon receptor. Cell Host Microbe. 2021;29(8):1235–1248.e8.PubMedPubMedCentralCrossRef Uberoi A, Bartow-McKenney C, Zheng Q, Flowers L, Campbell A, Knight SAB, et al. Commensal microbiota regulates skin barrier function and repair via signaling through the aryl hydrocarbon receptor. Cell Host Microbe. 2021;29(8):1235–1248.e8.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Adu-Oppong B, Thänert R, Wallace MA, Burnham CAD, Dantas G. Substantial overlap between symptomatic and asymptomatic genitourinary microbiota states. Microbiome. 2022;10(1):6.PubMedPubMedCentralCrossRef Adu-Oppong B, Thänert R, Wallace MA, Burnham CAD, Dantas G. Substantial overlap between symptomatic and asymptomatic genitourinary microbiota states. Microbiome. 2022;10(1):6.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Yachida S, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T, et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med. 2019;25(6):968–76.PubMedCrossRef Yachida S, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T, et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med. 2019;25(6):968–76.PubMedCrossRef
16.
Zurück zum Zitat Li R, Zhou R, Wang H, Li W, Pan M, Yao X, et al. Gut microbiota-stimulated cathepsin K secretion mediates TLR4-dependent M2 macrophage polarization and promotes tumor metastasis in colorectal cancer. Cell Death Differ. 2019;26(11):2447–63.PubMedPubMedCentralCrossRef Li R, Zhou R, Wang H, Li W, Pan M, Yao X, et al. Gut microbiota-stimulated cathepsin K secretion mediates TLR4-dependent M2 macrophage polarization and promotes tumor metastasis in colorectal cancer. Cell Death Differ. 2019;26(11):2447–63.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, Burr AHP, Tometich JT, Bhattacharjee A, et al. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity. 2021;54(12):2812–2824.e4.PubMedPubMedCentralCrossRef Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, Burr AHP, Tometich JT, Bhattacharjee A, et al. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity. 2021;54(12):2812–2824.e4.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Stein-Thoeringer CK, Saini NY, Zamir E, Blumenberg V, Schubert ML, Mor U, et al. A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat Med. 2023;29(4):906–16.PubMedPubMedCentralCrossRef Stein-Thoeringer CK, Saini NY, Zamir E, Blumenberg V, Schubert ML, Mor U, et al. A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat Med. 2023;29(4):906–16.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Andrews MC, Duong CPM, Gopalakrishnan V, Iebba V, Chen WS, Derosa L, et al. Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat Med. 2021;27(8):1432–41.PubMedCrossRef Andrews MC, Duong CPM, Gopalakrishnan V, Iebba V, Chen WS, Derosa L, et al. Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat Med. 2021;27(8):1432–41.PubMedCrossRef
20.
Zurück zum Zitat Sekirov I, Russell SL, Antunes LCM, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90(3):859–904.PubMedCrossRef Sekirov I, Russell SL, Antunes LCM, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90(3):859–904.PubMedCrossRef
21.
Zurück zum Zitat Abate M, Vos E, Gonen M, Janjigian YY, Schattner M, Laszkowska M, et al. A novel microbiome signature in gastric cancer: a two independent cohort retrospective analysis. Ann Surg. 2022;276(4):605–15.PubMedCrossRef Abate M, Vos E, Gonen M, Janjigian YY, Schattner M, Laszkowska M, et al. A novel microbiome signature in gastric cancer: a two independent cohort retrospective analysis. Ann Surg. 2022;276(4):605–15.PubMedCrossRef
22.
Zurück zum Zitat Zhang X, Li C, Cao W, Zhang Z. Alterations of gastric microbiota in gastric cancer and precancerous stages. Front Cell Infect Microbiol. 2021;11:559148.PubMedPubMedCentralCrossRef Zhang X, Li C, Cao W, Zhang Z. Alterations of gastric microbiota in gastric cancer and precancerous stages. Front Cell Infect Microbiol. 2021;11:559148.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Gantuya B, El Serag HB, Matsumoto T, Ajami NJ, Uchida T, Oyuntsetseg K, et al. Gastric mucosal microbiota in a Mongolian population with gastric cancer and precursor conditions. Aliment Pharmacol Ther. 2020;51(8):770–80.PubMedPubMedCentralCrossRef Gantuya B, El Serag HB, Matsumoto T, Ajami NJ, Uchida T, Oyuntsetseg K, et al. Gastric mucosal microbiota in a Mongolian population with gastric cancer and precursor conditions. Aliment Pharmacol Ther. 2020;51(8):770–80.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Monstein HJ, Tiveljung A, Kraft CH, Borch K, Jonasson J. Profiling of bacterial flora in gastric biopsies from patients with Helicobacter pylori-associated gastritis and histologically normal control individuals by temperature gradient gel electrophoresis and 16S rDNA sequence analysis. J Med Microbiol. 2000;49(9):817–22.PubMedCrossRef Monstein HJ, Tiveljung A, Kraft CH, Borch K, Jonasson J. Profiling of bacterial flora in gastric biopsies from patients with Helicobacter pylori-associated gastritis and histologically normal control individuals by temperature gradient gel electrophoresis and 16S rDNA sequence analysis. J Med Microbiol. 2000;49(9):817–22.PubMedCrossRef
25.
Zurück zum Zitat Costa M, Weese JS. Methods and basic concepts for microbiota assessment. Vet J. 2019;249:10–5.PubMedCrossRef Costa M, Weese JS. Methods and basic concepts for microbiota assessment. Vet J. 2019;249:10–5.PubMedCrossRef
26.
Zurück zum Zitat Perkins GA, den Bakker HC, Burton AJ, Erb HN, McDonough SP, McDonough PL, et al. Equine stomachs harbor an abundant and diverse mucosal microbiota. Appl Environ Microbiol. 2012;78(8):2522–32.PubMedPubMedCentralCrossRef Perkins GA, den Bakker HC, Burton AJ, Erb HN, McDonough SP, McDonough PL, et al. Equine stomachs harbor an abundant and diverse mucosal microbiota. Appl Environ Microbiol. 2012;78(8):2522–32.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185(20):3789–3806.e17.PubMedPubMedCentralCrossRef Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185(20):3789–3806.e17.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat von Rosenvinge EC, Song Y, White JR, Maddox C, Blanchard T, Fricke WF. Immune status, antibiotic medication and pH are associated with changes in the stomach fluid microbiota. ISME J. 2013;7(7):1354–66.CrossRef von Rosenvinge EC, Song Y, White JR, Maddox C, Blanchard T, Fricke WF. Immune status, antibiotic medication and pH are associated with changes in the stomach fluid microbiota. ISME J. 2013;7(7):1354–66.CrossRef
29.
Zurück zum Zitat Wensel CR, Pluznick JL, Salzberg SL, Sears CL. Next-generation sequencing: insights to advance clinical investigations of the microbiome. J Clin Invest. 2022;132(7):e154944.PubMedPubMedCentralCrossRef Wensel CR, Pluznick JL, Salzberg SL, Sears CL. Next-generation sequencing: insights to advance clinical investigations of the microbiome. J Clin Invest. 2022;132(7):e154944.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, et al. Molecular analysis of the bacterial microbiota in the human stomach. Proc Natl Acad Sci. 2006;103(3):732–7.PubMedPubMedCentralCrossRef Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, et al. Molecular analysis of the bacterial microbiota in the human stomach. Proc Natl Acad Sci. 2006;103(3):732–7.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Bajorek S, Parker L, Li N, Winglee K, Weaver M, Johnson J, et al. Initial microbial community of the neonatal stomach immediately after birth. Gut Microbes. 2019;10(3):289–97.PubMedCrossRef Bajorek S, Parker L, Li N, Winglee K, Weaver M, Johnson J, et al. Initial microbial community of the neonatal stomach immediately after birth. Gut Microbes. 2019;10(3):289–97.PubMedCrossRef
32.
Zurück zum Zitat Milisavljevic V, Garg M, Vuletic I, Miller JF, Kim L, Cunningham TD, et al. Prospective assessment of the gastroesophageal microbiome in VLBW neonates. BMC Pediatr. 2013;13(1):49.PubMedPubMedCentralCrossRef Milisavljevic V, Garg M, Vuletic I, Miller JF, Kim L, Cunningham TD, et al. Prospective assessment of the gastroesophageal microbiome in VLBW neonates. BMC Pediatr. 2013;13(1):49.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Moles L, Gómez M, Jiménez E, Bustos G, de Andrés J, Melgar A, et al. Bacterial diversity of the gastric content of preterm infants during their first month of life at the hospital. Front Nutr. 2017;4:4.CrossRef Moles L, Gómez M, Jiménez E, Bustos G, de Andrés J, Melgar A, et al. Bacterial diversity of the gastric content of preterm infants during their first month of life at the hospital. Front Nutr. 2017;4:4.CrossRef
34.
Zurück zum Zitat Patel K, Konduru K, Patra AK, Chandel DS, Panigrahi P. Trends and determinants of gastric bacterial colonization of preterm neonates in a NICU setting. Rogers LK, editor. PLoS One. 2015;10(7):e0114664.PubMedPubMedCentralCrossRef Patel K, Konduru K, Patra AK, Chandel DS, Panigrahi P. Trends and determinants of gastric bacterial colonization of preterm neonates in a NICU setting. Rogers LK, editor. PLoS One. 2015;10(7):e0114664.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Li XX, Wong GLH, To KF, Wong VWS, Lai LH, Chow DKL, et al. Bacterial microbiota profiling in gastritis without Helicobacter pylori infection or non-steroidal anti-inflammatory drug use. PLoS ONE. 2009;4(11):e7985.PubMedPubMedCentralCrossRef Li XX, Wong GLH, To KF, Wong VWS, Lai LH, Chow DKL, et al. Bacterial microbiota profiling in gastritis without Helicobacter pylori infection or non-steroidal anti-inflammatory drug use. PLoS ONE. 2009;4(11):e7985.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Kashiwagi S, Naito Y, Inoue R, Takagi T, Nakano T, Inada Y, et al. Mucosa-associated microbiota in the gastrointestinal tract of healthy Japanese subjects. Digestion. 2020;101(2):107–20.PubMedCrossRef Kashiwagi S, Naito Y, Inoue R, Takagi T, Nakano T, Inada Y, et al. Mucosa-associated microbiota in the gastrointestinal tract of healthy Japanese subjects. Digestion. 2020;101(2):107–20.PubMedCrossRef
37.
Zurück zum Zitat Ndegwa N, Ploner A, Andersson AF, Zagai U, Andreasson A, Vieth M, et al. Gastric microbiota in a low–Helicobacter pylori prevalence general population and their associations with gastric lesions. Clin Transl Gastroenterol. 2020;11(7):e00191.PubMedPubMedCentralCrossRef Ndegwa N, Ploner A, Andersson AF, Zagai U, Andreasson A, Vieth M, et al. Gastric microbiota in a low–Helicobacter pylori prevalence general population and their associations with gastric lesions. Clin Transl Gastroenterol. 2020;11(7):e00191.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Vasapolli R, Schütte K, Schulz C, Vital M, Schomburg D, Pieper DH, et al. Analysis of transcriptionally active bacteria throughout the gastrointestinal tract of healthy individuals. Gastroenterology. 2019;157(4):1081–1092.e3.PubMedCrossRef Vasapolli R, Schütte K, Schulz C, Vital M, Schomburg D, Pieper DH, et al. Analysis of transcriptionally active bacteria throughout the gastrointestinal tract of healthy individuals. Gastroenterology. 2019;157(4):1081–1092.e3.PubMedCrossRef
39.
Zurück zum Zitat Chee WJY, Chew SY, Than LTL. Vaginal microbiota and the potential of Lactobacillus derivatives in maintaining vaginal health. Microb Cell Factories. 2020;19(1):203.CrossRef Chee WJY, Chew SY, Than LTL. Vaginal microbiota and the potential of Lactobacillus derivatives in maintaining vaginal health. Microb Cell Factories. 2020;19(1):203.CrossRef
40.
Zurück zum Zitat Sprong KE, Mabenge M, Wright CA, Govender S. Ureaplasma species and preterm birth: current perspectives. Crit Rev Microbiol. 2020;46(2):169–81.PubMedCrossRef Sprong KE, Mabenge M, Wright CA, Govender S. Ureaplasma species and preterm birth: current perspectives. Crit Rev Microbiol. 2020;46(2):169–81.PubMedCrossRef
41.
Zurück zum Zitat Zimmermann P, Curtis N. Breast milk microbiota: a review of the factors that influence composition. J Infect. 2020;81(1):17–47.PubMedCrossRef Zimmermann P, Curtis N. Breast milk microbiota: a review of the factors that influence composition. J Infect. 2020;81(1):17–47.PubMedCrossRef
42.
Zurück zum Zitat Dong Q, Xin Y, Wang L, Meng X, Yu X, Lu L, et al. Characterization of gastric microbiota in twins. Curr Microbiol. 2017;74(2):224–9.PubMedCrossRef Dong Q, Xin Y, Wang L, Meng X, Yu X, Lu L, et al. Characterization of gastric microbiota in twins. Curr Microbiol. 2017;74(2):224–9.PubMedCrossRef
43.
Zurück zum Zitat Nyangahu DD, Lennard KS, Brown BP, Darby MG, Wendoh JM, Havyarimana E, et al. Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity. Microbiome. 2018;6(1):124.PubMedPubMedCentralCrossRef Nyangahu DD, Lennard KS, Brown BP, Darby MG, Wendoh JM, Havyarimana E, et al. Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity. Microbiome. 2018;6(1):124.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Trevisi P, Priori D, Motta V, Luise D, Jansman AJM, Koopmans SJ, et al. The effects of starter microbiota and the early life feeding of medium chain triglycerides on the gastric transcriptome profile of 2- or 3-week-old cesarean delivered piglets. J Anim Sci Biotechnol. 2017;8(1):82.PubMedPubMedCentralCrossRef Trevisi P, Priori D, Motta V, Luise D, Jansman AJM, Koopmans SJ, et al. The effects of starter microbiota and the early life feeding of medium chain triglycerides on the gastric transcriptome profile of 2- or 3-week-old cesarean delivered piglets. J Anim Sci Biotechnol. 2017;8(1):82.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Van Hul M, Cani PD. The gut microbiota in obesity and weight management: microbes as friends or foe? Nat Rev Endocrinol. 2023;19(5):258–71.PubMedCrossRef Van Hul M, Cani PD. The gut microbiota in obesity and weight management: microbes as friends or foe? Nat Rev Endocrinol. 2023;19(5):258–71.PubMedCrossRef
46.
Zurück zum Zitat Zou Y, Ju X, Chen W, Yuan J, Wang Z, Aluko RE, et al. Rice bran attenuated obesity via alleviating dyslipidemia, browning of white adipocytes and modulating gut microbiota in high-fat diet-induced obese mice. Food Funct. 2020;11(3):2406–17.PubMedCrossRef Zou Y, Ju X, Chen W, Yuan J, Wang Z, Aluko RE, et al. Rice bran attenuated obesity via alleviating dyslipidemia, browning of white adipocytes and modulating gut microbiota in high-fat diet-induced obese mice. Food Funct. 2020;11(3):2406–17.PubMedCrossRef
47.
Zurück zum Zitat Dapa T, Ramiro RS, Pedro MF, Gordo I, Xavier KB. Diet leaves a genetic signature in a keystone member of the gut microbiota. Cell Host Microbe. 2022;30(2):183–199.e10.PubMedCrossRef Dapa T, Ramiro RS, Pedro MF, Gordo I, Xavier KB. Diet leaves a genetic signature in a keystone member of the gut microbiota. Cell Host Microbe. 2022;30(2):183–199.e10.PubMedCrossRef
48.
Zurück zum Zitat Xie Y, Pei F, Liu Y, Liu Z, Chen X, Xue D. Fecal fermentation and high-fat diet-induced obesity mouse model confirmed exopolysaccharide from Weissella cibaria PFY06 can ameliorate obesity by regulating the gut microbiota. Carbohydr Polym. 2023;318:121122.PubMedCrossRef Xie Y, Pei F, Liu Y, Liu Z, Chen X, Xue D. Fecal fermentation and high-fat diet-induced obesity mouse model confirmed exopolysaccharide from Weissella cibaria PFY06 can ameliorate obesity by regulating the gut microbiota. Carbohydr Polym. 2023;318:121122.PubMedCrossRef
49.
Zurück zum Zitat Sedghi L, DiMassa V, Harrington A, Lynch SV, Kapila YL. The oral microbiome: role of key organisms and complex networks in oral health and disease. Periodontol 2000. 2021;87(1):107–31.PubMedPubMedCentralCrossRef Sedghi L, DiMassa V, Harrington A, Lynch SV, Kapila YL. The oral microbiome: role of key organisms and complex networks in oral health and disease. Periodontol 2000. 2021;87(1):107–31.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Fann JCY, Chiang TH, Yen AMF, Lee YC, Wu MS, Chen HH. Personalized risk assessment for dynamic transition of gastric neoplasms. J Biomed Sci. 2018;25(1):84.PubMedPubMedCentralCrossRef Fann JCY, Chiang TH, Yen AMF, Lee YC, Wu MS, Chen HH. Personalized risk assessment for dynamic transition of gastric neoplasms. J Biomed Sci. 2018;25(1):84.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Png CW, Lee WJJ, Chua SJ, Zhu F, Gastric Consortium5, Yeoh KG, et al. Mucosal microbiome associates with progression to gastric cancer. Theranostics. 2022;12(1):48–58.PubMedPubMedCentralCrossRef Png CW, Lee WJJ, Chua SJ, Zhu F, Gastric Consortium5, Yeoh KG, et al. Mucosal microbiome associates with progression to gastric cancer. Theranostics. 2022;12(1):48–58.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Wang Z, Gao X, Zeng R, Wu Q, Sun H, Wu W, et al. Changes of the gastric mucosal microbiome associated with histological stages of gastric carcinogenesis. Front Microbiol. 2020;11:997.PubMedPubMedCentralCrossRef Wang Z, Gao X, Zeng R, Wu Q, Sun H, Wu W, et al. Changes of the gastric mucosal microbiome associated with histological stages of gastric carcinogenesis. Front Microbiol. 2020;11:997.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Coker OO, Dai Z, Nie Y, Zhao G, Cao L, Nakatsu G, et al. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67(6):1024–32.PubMedCrossRef Coker OO, Dai Z, Nie Y, Zhao G, Cao L, Nakatsu G, et al. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67(6):1024–32.PubMedCrossRef
54.
Zurück zum Zitat He C, Peng C, Shu X, Wang H, Zhu Z, Ouyang Y, et al. Convergent dysbiosis of gastric mucosa and fluid microbiome during stomach carcinogenesis. Gastric Cancer. 2022;25(5):837–49.PubMedCrossRef He C, Peng C, Shu X, Wang H, Zhu Z, Ouyang Y, et al. Convergent dysbiosis of gastric mucosa and fluid microbiome during stomach carcinogenesis. Gastric Cancer. 2022;25(5):837–49.PubMedCrossRef
55.
Zurück zum Zitat Hsieh YY, Tung SY, Pan HY, Yen CW, Xu HW, Lin YJ, et al. Increased abundance of Clostridium and Fusobacterium in gastric microbiota of patients with gastric cancer in Taiwan. Sci Rep. 2018;8(1):158.PubMedPubMedCentralCrossRef Hsieh YY, Tung SY, Pan HY, Yen CW, Xu HW, Lin YJ, et al. Increased abundance of Clostridium and Fusobacterium in gastric microbiota of patients with gastric cancer in Taiwan. Sci Rep. 2018;8(1):158.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Wu F, Yang L, Hao Y, Zhou B, Hu J, Yang Y, et al. Oral and gastric microbiome in relation to gastric intestinal metaplasia. Int J Cancer. 2022;150(6):928–40.PubMedCrossRef Wu F, Yang L, Hao Y, Zhou B, Hu J, Yang Y, et al. Oral and gastric microbiome in relation to gastric intestinal metaplasia. Int J Cancer. 2022;150(6):928–40.PubMedCrossRef
57.
Zurück zum Zitat Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, Machado JC, et al. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut. 2018;67(2):226–36.PubMedCrossRef Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, Machado JC, et al. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut. 2018;67(2):226–36.PubMedCrossRef
58.
Zurück zum Zitat Liu X, Shao L, Liu X, Ji F, Mei Y, Cheng Y, et al. Alterations of gastric mucosal microbiota across different stomach microhabitats in a cohort of 276 patients with gastric cancer. EBioMedicine. 2019;40:336–48.PubMedCrossRef Liu X, Shao L, Liu X, Ji F, Mei Y, Cheng Y, et al. Alterations of gastric mucosal microbiota across different stomach microhabitats in a cohort of 276 patients with gastric cancer. EBioMedicine. 2019;40:336–48.PubMedCrossRef
59.
Zurück zum Zitat Mannion A, Sheh A, Shen Z, Dzink-Fox J, Piazuelo MB, Wilson KT, et al. Shotgun metagenomics of gastric biopsies reveals compositional and functional microbiome shifts in high- and low-gastric-cancer-risk populations from Colombia, South America. Gut Microbes. 2023;15(1):2186677.PubMedPubMedCentralCrossRef Mannion A, Sheh A, Shen Z, Dzink-Fox J, Piazuelo MB, Wilson KT, et al. Shotgun metagenomics of gastric biopsies reveals compositional and functional microbiome shifts in high- and low-gastric-cancer-risk populations from Colombia, South America. Gut Microbes. 2023;15(1):2186677.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Lehr K, Nikitina D, Vilchez-Vargas R, Steponaitiene R, Thon C, Skieceviciene J, et al. Microbial composition of tumorous and adjacent gastric tissue is associated with prognosis of gastric cancer. Sci Rep. 2023;13(1):4640.PubMedPubMedCentralCrossRef Lehr K, Nikitina D, Vilchez-Vargas R, Steponaitiene R, Thon C, Skieceviciene J, et al. Microbial composition of tumorous and adjacent gastric tissue is associated with prognosis of gastric cancer. Sci Rep. 2023;13(1):4640.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Chen XH, Wang A, Chu AN, Gong YH, Yuan Y. Mucosa-associated microbiota in gastric cancer tissues compared with non-cancer tissues. Front Microbiol. 2019;10:1261.PubMedPubMedCentralCrossRef Chen XH, Wang A, Chu AN, Gong YH, Yuan Y. Mucosa-associated microbiota in gastric cancer tissues compared with non-cancer tissues. Front Microbiol. 2019;10:1261.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Deng Y, Ding X, Song Q, Zhao G, Han L, Ding B, et al. Alterations in mucosa-associated microbiota in the stomach of patients with gastric cancer. Cell Oncol. 2021;44(3):701–14.CrossRef Deng Y, Ding X, Song Q, Zhao G, Han L, Ding B, et al. Alterations in mucosa-associated microbiota in the stomach of patients with gastric cancer. Cell Oncol. 2021;44(3):701–14.CrossRef
63.
Zurück zum Zitat Wu ZF, Zou K, Wu GN, Jin ZJ, Xiang CJ, Xu S, et al. A comparison of tumor-associated and non-tumor-associated gastric microbiota in gastric cancer patients. Dig Dis Sci. 2021;66(5):1673–82.PubMedCrossRef Wu ZF, Zou K, Wu GN, Jin ZJ, Xiang CJ, Xu S, et al. A comparison of tumor-associated and non-tumor-associated gastric microbiota in gastric cancer patients. Dig Dis Sci. 2021;66(5):1673–82.PubMedCrossRef
64.
Zurück zum Zitat Dai D, Yang Y, Yu J, Dang T, Qin W, Teng L, et al. Interactions between gastric microbiota and metabolites in gastric cancer. Cell Death Dis. 2021;12(12):1104.PubMedPubMedCentralCrossRef Dai D, Yang Y, Yu J, Dang T, Qin W, Teng L, et al. Interactions between gastric microbiota and metabolites in gastric cancer. Cell Death Dis. 2021;12(12):1104.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Peng R, Liu S, You W, Huang Y, Hu C, Gao Y, et al. Gastric microbiome alterations are associated with decreased CD8 + tissue-resident memory T cells in the tumor microenvironment of gastric cancer. Cancer Immunol Res. 2022;10(10):1224–40.PubMedCrossRef Peng R, Liu S, You W, Huang Y, Hu C, Gao Y, et al. Gastric microbiome alterations are associated with decreased CD8 + tissue-resident memory T cells in the tumor microenvironment of gastric cancer. Cancer Immunol Res. 2022;10(10):1224–40.PubMedCrossRef
66.
Zurück zum Zitat Yang Y, Dai D, Jin W, Huang Y, Zhang Y, Chen Y, et al. Microbiota and metabolites alterations in proximal and distal gastric cancer patients. J Transl Med. 2022;20(1):439.PubMedPubMedCentralCrossRef Yang Y, Dai D, Jin W, Huang Y, Zhang Y, Chen Y, et al. Microbiota and metabolites alterations in proximal and distal gastric cancer patients. J Transl Med. 2022;20(1):439.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Xi J, Li Y, Zhang H, Bai Z. Dynamic variations of the gastric microbiota: key therapeutic points in the reversal of Correa’s cascade. Int J Cancer. 2023;152(6):1069–84.PubMedCrossRef Xi J, Li Y, Zhang H, Bai Z. Dynamic variations of the gastric microbiota: key therapeutic points in the reversal of Correa’s cascade. Int J Cancer. 2023;152(6):1069–84.PubMedCrossRef
68.
Zurück zum Zitat Yan L, Chen Y, Chen F, Tao T, Hu Z, Wang J, et al. Effect of Helicobacter pylori eradication on gastric cancer prevention: updated report from a randomized controlled trial with 26.5 years of follow-up. Gastroenterology. 2022;163(1):154–162.e3.PubMedCrossRef Yan L, Chen Y, Chen F, Tao T, Hu Z, Wang J, et al. Effect of Helicobacter pylori eradication on gastric cancer prevention: updated report from a randomized controlled trial with 26.5 years of follow-up. Gastroenterology. 2022;163(1):154–162.e3.PubMedCrossRef
69.
Zurück zum Zitat Liou JM, Malfertheiner P, Lee YC, Sheu BS, Sugano K, Cheng HC, et al. Screening and eradication of Helicobacter pylori for gastric cancer prevention: the Taipei global consensus. Gut. 2020;69(12):2093–112.PubMedCrossRef Liou JM, Malfertheiner P, Lee YC, Sheu BS, Sugano K, Cheng HC, et al. Screening and eradication of Helicobacter pylori for gastric cancer prevention: the Taipei global consensus. Gut. 2020;69(12):2093–112.PubMedCrossRef
70.
Zurück zum Zitat Sung JJY, Coker OO, Chu E, Szeto CH, Luk STY, Lau HCH, et al. Gastric microbes associated with gastric inflammation, atrophy and intestinal metaplasia 1 year after Helicobacter pylori eradication. Gut. 2020;69(9):1572–81.PubMedCrossRef Sung JJY, Coker OO, Chu E, Szeto CH, Luk STY, Lau HCH, et al. Gastric microbes associated with gastric inflammation, atrophy and intestinal metaplasia 1 year after Helicobacter pylori eradication. Gut. 2020;69(9):1572–81.PubMedCrossRef
71.
Zurück zum Zitat Boehm ET, Thon C, Kupcinskas J, Steponaitiene R, Skieceviciene J, Canbay A, et al. Fusobacterium nucleatum is associated with worse prognosis in Lauren’s diffuse type gastric cancer patients. Sci Rep. 2020;10(1):16240.PubMedPubMedCentralCrossRef Boehm ET, Thon C, Kupcinskas J, Steponaitiene R, Skieceviciene J, Canbay A, et al. Fusobacterium nucleatum is associated with worse prognosis in Lauren’s diffuse type gastric cancer patients. Sci Rep. 2020;10(1):16240.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Hsieh YY, Tung SY, Pan HY, Chang TS, Wei KL, Chen WM, et al. Fusobacterium nucleatum colonization is associated with decreased survival of helicobacter pylori-positive gastric cancer patients. World J Gastroenterol. 2021;27(42):7311–23.PubMedPubMedCentralCrossRef Hsieh YY, Tung SY, Pan HY, Chang TS, Wei KL, Chen WM, et al. Fusobacterium nucleatum colonization is associated with decreased survival of helicobacter pylori-positive gastric cancer patients. World J Gastroenterol. 2021;27(42):7311–23.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Guan SW, Lin Q, Wu XD, Yu HB. Weighted gene coexpression network analysis and machine learning reveal oncogenome associated microbiome plays an important role in tumor immunity and prognosis in pan-cancer. J Transl Med. 2023;21:537.PubMedPubMedCentralCrossRef Guan SW, Lin Q, Wu XD, Yu HB. Weighted gene coexpression network analysis and machine learning reveal oncogenome associated microbiome plays an important role in tumor immunity and prognosis in pan-cancer. J Transl Med. 2023;21:537.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Yang J, Xu J, Ling Z, Zhou X, Si Y, Liu X, et al. Prognostic effects of the gastric mucosal microbiota in gastric cancer. Cancer Sci. 2023;114(3):1075–85.PubMedCrossRef Yang J, Xu J, Ling Z, Zhou X, Si Y, Liu X, et al. Prognostic effects of the gastric mucosal microbiota in gastric cancer. Cancer Sci. 2023;114(3):1075–85.PubMedCrossRef
75.
Zurück zum Zitat Nasr R, Shamseddine A, Mukherji D, Nassar F, Temraz S. The crosstalk between microbiome and immune response in gastric cancer. Int J Mol Sci. 2020;21(18):6586.PubMedPubMedCentralCrossRef Nasr R, Shamseddine A, Mukherji D, Nassar F, Temraz S. The crosstalk between microbiome and immune response in gastric cancer. Int J Mol Sci. 2020;21(18):6586.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat He J, Hu W, Ouyang Q, Zhang S, He L, Chen W, et al. Helicobacter pylori infection induces stem cell-like properties in Correa cascade of gastric cancer. Cancer Lett. 2022;542:215764.PubMedCrossRef He J, Hu W, Ouyang Q, Zhang S, He L, Chen W, et al. Helicobacter pylori infection induces stem cell-like properties in Correa cascade of gastric cancer. Cancer Lett. 2022;542:215764.PubMedCrossRef
77.
Zurück zum Zitat Chauhan N, Tay ACY, Marshall BJ, Jain U. Helicobacter pylori VacA, a distinct toxin exerts diverse functionalities in numerous cells: an overview. Helicobacter. 2019;24(1):e12544.PubMedCrossRef Chauhan N, Tay ACY, Marshall BJ, Jain U. Helicobacter pylori VacA, a distinct toxin exerts diverse functionalities in numerous cells: an overview. Helicobacter. 2019;24(1):e12544.PubMedCrossRef
78.
Zurück zum Zitat Toh JWT, Wilson RB. Pathways of gastric carcinogenesis, Helicobacter pylori virulence and interactions with antioxidant systems, vitamin C and phytochemicals. Int J Mol Sci. 2020;21(17):6451.PubMedPubMedCentralCrossRef Toh JWT, Wilson RB. Pathways of gastric carcinogenesis, Helicobacter pylori virulence and interactions with antioxidant systems, vitamin C and phytochemicals. Int J Mol Sci. 2020;21(17):6451.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Zhang X, Soutto M, Chen Z, Bhat N, Zhu S, Eissmann MF, et al. Induction of fibroblast growth factor receptor 4 by Helicobacter pylori via signal transducer and activator of transcription 3 with a feedforward activation loop involving steroid receptor coactivator signaling in gastric cancer. Gastroenterology. 2022;163(3):620–636.e9.PubMedCrossRef Zhang X, Soutto M, Chen Z, Bhat N, Zhu S, Eissmann MF, et al. Induction of fibroblast growth factor receptor 4 by Helicobacter pylori via signal transducer and activator of transcription 3 with a feedforward activation loop involving steroid receptor coactivator signaling in gastric cancer. Gastroenterology. 2022;163(3):620–636.e9.PubMedCrossRef
80.
Zurück zum Zitat Wang Z, Shan Y, Wang R, Zhou H, Hu R, Li Y, et al. Structural insights into the binding propensity of human SHIP2 SH2 to oncogenic CagA isoforms from Helicobacter pylori. Int J Mol Sci. 2022;23(19):11299.PubMedPubMedCentralCrossRef Wang Z, Shan Y, Wang R, Zhou H, Hu R, Li Y, et al. Structural insights into the binding propensity of human SHIP2 SH2 to oncogenic CagA isoforms from Helicobacter pylori. Int J Mol Sci. 2022;23(19):11299.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Murata-Kamiya N, Kurashima Y, Teishikata Y, Yamahashi Y, Saito Y, Higashi H, et al. Helicobacter pylori CagA interacts with E-cadherin and deregulates the beta-catenin signal that promotes intestinal transdifferentiation in gastric epithelial cells. Oncogene. 2007;26(32):4617–26.PubMedCrossRef Murata-Kamiya N, Kurashima Y, Teishikata Y, Yamahashi Y, Saito Y, Higashi H, et al. Helicobacter pylori CagA interacts with E-cadherin and deregulates the beta-catenin signal that promotes intestinal transdifferentiation in gastric epithelial cells. Oncogene. 2007;26(32):4617–26.PubMedCrossRef
82.
Zurück zum Zitat Saadat I, Higashi H, Obuse C, Umeda M, Murata-Kamiya N, Saito Y, et al. Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature. 2007;447(7142):330–3.PubMedCrossRef Saadat I, Higashi H, Obuse C, Umeda M, Murata-Kamiya N, Saito Y, et al. Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature. 2007;447(7142):330–3.PubMedCrossRef
83.
Zurück zum Zitat Imai S, Ooki T, Murata-Kamiya N, Komura D, Tahmina K, Wu W, et al. Helicobacter pylori CagA elicits BRCAness to induce genome instability that may underlie bacterial gastric carcinogenesis. Cell Host Microbe. 2021;29(6):941–958.e10.PubMedCrossRef Imai S, Ooki T, Murata-Kamiya N, Komura D, Tahmina K, Wu W, et al. Helicobacter pylori CagA elicits BRCAness to induce genome instability that may underlie bacterial gastric carcinogenesis. Cell Host Microbe. 2021;29(6):941–958.e10.PubMedCrossRef
84.
Zurück zum Zitat Saha A, Backert S, Hammond CE, Gooz M, Smolka AJ. Helicobacter pylori CagL activates ADAM17 to induce repression of the gastric H, K-ATPase α subunit. Gastroenterology. 2010;139(1):239–48.PubMedCrossRef Saha A, Backert S, Hammond CE, Gooz M, Smolka AJ. Helicobacter pylori CagL activates ADAM17 to induce repression of the gastric H, K-ATPase α subunit. Gastroenterology. 2010;139(1):239–48.PubMedCrossRef
85.
Zurück zum Zitat Yao X, Smolka AJ. Gastric parietal cell physiology and Helicobacter pylori–induced disease. Gastroenterology. 2019;156(8):2158–73.PubMedCrossRef Yao X, Smolka AJ. Gastric parietal cell physiology and Helicobacter pylori–induced disease. Gastroenterology. 2019;156(8):2158–73.PubMedCrossRef
86.
Zurück zum Zitat Skinner GR. Transformation of primary hamster embryo fibroblasts by type 2 simplex virus: evidence for a “hit and run” mechanism. Br J Exp Pathol. 1976;57(4):361–76.PubMedPubMedCentral Skinner GR. Transformation of primary hamster embryo fibroblasts by type 2 simplex virus: evidence for a “hit and run” mechanism. Br J Exp Pathol. 1976;57(4):361–76.PubMedPubMedCentral
87.
Zurück zum Zitat Hatakeyama M. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe. 2014;15(3):306–16.PubMedCrossRef Hatakeyama M. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe. 2014;15(3):306–16.PubMedCrossRef
88.
Zurück zum Zitat Gantuya B, El-Serag HB, Matsumoto T, Ajami NJ, Oyuntsetseg K, Azzaya D, et al. Gastric microbiota in Helicobacter pylori-negative and -positive gastritis among high incidence of gastric cancer area. Cancers. 2019;11(4):504.PubMedPubMedCentralCrossRef Gantuya B, El-Serag HB, Matsumoto T, Ajami NJ, Oyuntsetseg K, Azzaya D, et al. Gastric microbiota in Helicobacter pylori-negative and -positive gastritis among high incidence of gastric cancer area. Cancers. 2019;11(4):504.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Miftahussurur M, Waskito LA, El-Serag HB, Ajami NJ, Nusi IA, Syam AF, et al. Gastric microbiota and Helicobacter pylori in Indonesian population. Helicobacter. 2020;25(4):e12695.PubMedCrossRef Miftahussurur M, Waskito LA, El-Serag HB, Ajami NJ, Nusi IA, Syam AF, et al. Gastric microbiota and Helicobacter pylori in Indonesian population. Helicobacter. 2020;25(4):e12695.PubMedCrossRef
90.
Zurück zum Zitat Llorca L, Pérez-Pérez G, Urruzuno P, Martinez MJ, Iizumi T, Gao Z, et al. Characterization of the gastric microbiota in a pediatric population according to Helicobacter pylori status. Pediatr Infect Dis J. 2017;36(2):173–8.PubMedCrossRef Llorca L, Pérez-Pérez G, Urruzuno P, Martinez MJ, Iizumi T, Gao Z, et al. Characterization of the gastric microbiota in a pediatric population according to Helicobacter pylori status. Pediatr Infect Dis J. 2017;36(2):173–8.PubMedCrossRef
91.
Zurück zum Zitat Guo Y, Zhang Y, Gerhard M, Gao JJ, Mejias-Luque R, Zhang L, et al. Effect of Helicobacter pylori on gastrointestinal microbiota: a population-based study in Linqu, a high-risk area of gastric cancer. Gut. 2020;69(9):1598–607.PubMedCrossRef Guo Y, Zhang Y, Gerhard M, Gao JJ, Mejias-Luque R, Zhang L, et al. Effect of Helicobacter pylori on gastrointestinal microbiota: a population-based study in Linqu, a high-risk area of gastric cancer. Gut. 2020;69(9):1598–607.PubMedCrossRef
92.
Zurück zum Zitat Miao R, Wan C, Wang Z. The relationship of gastric microbiota and Helicobacter pylori infection in pediatrics population. Helicobacter. 2020;25(1):e12676.PubMedCrossRef Miao R, Wan C, Wang Z. The relationship of gastric microbiota and Helicobacter pylori infection in pediatrics population. Helicobacter. 2020;25(1):e12676.PubMedCrossRef
93.
Zurück zum Zitat Klymiuk I, Bilgilier C, Stadlmann A, Thannesberger J, Kastner MT, Högenauer C, et al. The human gastric microbiome is predicated upon infection with Helicobacter pylori. Front Microbiol. 2017;8:2508.PubMedPubMedCentralCrossRef Klymiuk I, Bilgilier C, Stadlmann A, Thannesberger J, Kastner MT, Högenauer C, et al. The human gastric microbiome is predicated upon infection with Helicobacter pylori. Front Microbiol. 2017;8:2508.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Lofgren JL, Whary MT, Ge Z, Muthupalani S, Taylor NS, Mobley M, et al. Lack of commensal flora in Helicobacter pylori–Infected INS-GAS mice reduces gastritis and delays intraepithelial neoplasia. Gastroenterology. 2011;140(1):210–220.e4.PubMedCrossRef Lofgren JL, Whary MT, Ge Z, Muthupalani S, Taylor NS, Mobley M, et al. Lack of commensal flora in Helicobacter pylori–Infected INS-GAS mice reduces gastritis and delays intraepithelial neoplasia. Gastroenterology. 2011;140(1):210–220.e4.PubMedCrossRef
95.
Zurück zum Zitat Lertpiriyapong K, Whary MT, Muthupalani S, Lofgren JL, Gamazon ER, Feng Y, et al. 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(1):54–63.PubMedCrossRef Lertpiriyapong K, Whary MT, Muthupalani S, Lofgren JL, Gamazon ER, Feng Y, et al. 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(1):54–63.PubMedCrossRef
96.
Zurück zum Zitat Shen Z, Dzink-Fox J, Feng Y, Muthupalani S, Mannion AJ, Sheh A, et al. Gastric non-Helicobacter pylori urease-positive Staphylococcus epidermidis and Streptococcus salivarius isolated from humans have contrasting effects on H. pylori-associated gastric pathology and host immune responses in a murine model of gastric cancer. Young VB, editor. mSphere. 2022;7(1):e00772–21.PubMedPubMedCentralCrossRef Shen Z, Dzink-Fox J, Feng Y, Muthupalani S, Mannion AJ, Sheh A, et al. Gastric non-Helicobacter pylori urease-positive Staphylococcus epidermidis and Streptococcus salivarius isolated from humans have contrasting effects on H. pylori-associated gastric pathology and host immune responses in a murine model of gastric cancer. Young VB, editor. mSphere. 2022;7(1):e00772–21.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Chen Z, Tang Z, Li W, Deng X, Yu L, Yang J, et al. Weizmannia coagulans BCF-01: a novel gastrogenic probiotic for Helicobacter pylori infection control. Gut Microbes. 2024;16(1):2313770. Chen Z, Tang Z, Li W, Deng X, Yu L, Yang J, et al. Weizmannia coagulans BCF-01: a novel gastrogenic probiotic for Helicobacter pylori infection control. Gut Microbes. 2024;16(1):2313770.
98.
Zurück zum Zitat Gebremariam HG, Qazi KR, Somiah T, Pathak SK, Sjölinder H, Sverremark Ekström E, et al. Lactobacillus gasseri suppresses the production of proinflammatory cytokines in Helicobacter pylori-infected macrophages by inhibiting the expression of ADAM17. Front Immunol. 2019;10:2326.PubMedPubMedCentralCrossRef Gebremariam HG, Qazi KR, Somiah T, Pathak SK, Sjölinder H, Sverremark Ekström E, et al. Lactobacillus gasseri suppresses the production of proinflammatory cytokines in Helicobacter pylori-infected macrophages by inhibiting the expression of ADAM17. Front Immunol. 2019;10:2326.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Shen S, Ren F, Qin H, Bukhari I, Yang J, Gao D, et al. Lactobacillus acidophilus NCFM and Lactiplantibacillus plantarum Lp-115 inhibit Helicobacter pylori colonization and gastric inflammation in a murine model. Front Cell Infect Microbiol. 2023;13:1196084.PubMedPubMedCentralCrossRef Shen S, Ren F, Qin H, Bukhari I, Yang J, Gao D, et al. Lactobacillus acidophilus NCFM and Lactiplantibacillus plantarum Lp-115 inhibit Helicobacter pylori colonization and gastric inflammation in a murine model. Front Cell Infect Microbiol. 2023;13:1196084.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Khosravi Y, Loke MF, Goh KL, Vadivelu J. Proteomics analysis revealed that crosstalk between Helicobacter pylori and Streptococcus mitis may enhance bacterial survival and reduces carcinogenesis. Front Microbiol. 2016;7:7.CrossRef Khosravi Y, Loke MF, Goh KL, Vadivelu J. Proteomics analysis revealed that crosstalk between Helicobacter pylori and Streptococcus mitis may enhance bacterial survival and reduces carcinogenesis. Front Microbiol. 2016;7:7.CrossRef
101.
Zurück zum Zitat Khosravi Y, Dieye Y, Loke MF, Goh KL, Vadivelu J. Streptococcus mitis induces conversion of Helicobacter pylori to coccoid cells during co-culture in vitro. PLoS ONE. 2014;9(11):e112214.PubMedPubMedCentralCrossRef Khosravi Y, Dieye Y, Loke MF, Goh KL, Vadivelu J. Streptococcus mitis induces conversion of Helicobacter pylori to coccoid cells during co-culture in vitro. PLoS ONE. 2014;9(11):e112214.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Chen Z, Tang Z, Li W, Deng X, Yu L, Yang J, et al. Weizmannia coagulans BCF-01: a novel gastrogenic probiotic for Helicobacter pylori infection control. Gut Microbes. 2024;16(1):2313770.PubMedPubMedCentralCrossRef Chen Z, Tang Z, Li W, Deng X, Yu L, Yang J, et al. Weizmannia coagulans BCF-01: a novel gastrogenic probiotic for Helicobacter pylori infection control. Gut Microbes. 2024;16(1):2313770.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Kwon SK, Park JC, Kim KH, Yoon J, Cho Y, Lee B, et al. Human gastric microbiota transplantation recapitulates premalignant lesions in germ-free mice. Gut. 2022;71(7):1266–76.PubMedCrossRef Kwon SK, Park JC, Kim KH, Yoon J, Cho Y, Lee B, et al. Human gastric microbiota transplantation recapitulates premalignant lesions in germ-free mice. Gut. 2022;71(7):1266–76.PubMedCrossRef
104.
Zurück zum Zitat Sasaki M, Kodama Y, Shimoyama Y, Ishikawa T, Kimura S. Aciduricity and acid tolerance mechanisms of Streptococcus anginosus. J Gen Appl Microbiol. 2018;64(4):174–9.PubMedCrossRef Sasaki M, Kodama Y, Shimoyama Y, Ishikawa T, Kimura S. Aciduricity and acid tolerance mechanisms of Streptococcus anginosus. J Gen Appl Microbiol. 2018;64(4):174–9.PubMedCrossRef
105.
Zurück zum Zitat Fu K, Cheung AHK, Wong CC, Liu W, Zhou Y, Wang F, et al. Streptococcus anginosus promotes gastric inflammation, atrophy, and tumorigenesis in mice. Cell. 2024;187(4):882–896.e17.PubMedCrossRef Fu K, Cheung AHK, Wong CC, Liu W, Zhou Y, Wang F, et al. Streptococcus anginosus promotes gastric inflammation, atrophy, and tumorigenesis in mice. Cell. 2024;187(4):882–896.e17.PubMedCrossRef
106.
Zurück zum Zitat Li Y, Huang X, Tong D, Jiang C, Zhu X, Wei Z, et al. Relationships among microbiota, gastric cancer, and immunotherapy. Front Microbiol. 2022;13:987763.PubMedPubMedCentralCrossRef Li Y, Huang X, Tong D, Jiang C, Zhu X, Wei Z, et al. Relationships among microbiota, gastric cancer, and immunotherapy. Front Microbiol. 2022;13:987763.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Vinasco K, Mitchell HM, Kaakoush NO, Castaño-Rodríguez N. Microbial carcinogenesis: lactic acid bacteria in gastric cancer. Biochim Biophys Acta BBA - Rev Cancer. 2019;1872(2):188309.CrossRef Vinasco K, Mitchell HM, Kaakoush NO, Castaño-Rodríguez N. Microbial carcinogenesis: lactic acid bacteria in gastric cancer. Biochim Biophys Acta BBA - Rev Cancer. 2019;1872(2):188309.CrossRef
108.
Zurück zum Zitat Hwang CH, Lee NK, Paik HD. The anti-cancer potential of heat-killed Lactobacillus brevis KU15176 upon AGS Cell lines through intrinsic apoptosis pathway. Int J Mol Sci. 2022;23(8):4073.PubMedPubMedCentralCrossRef Hwang CH, Lee NK, Paik HD. The anti-cancer potential of heat-killed Lactobacillus brevis KU15176 upon AGS Cell lines through intrinsic apoptosis pathway. Int J Mol Sci. 2022;23(8):4073.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Chen MJ, Chen CC, Huang YC, Tseng CC, Hsu JT, Lin YF, et al. The efficacy of Lactobacillus acidophilus and rhamnosus in the reduction of bacterial load of Helicobacter pylori and modification of gut microbiota-a double-blind, placebo-controlled, randomized trial. Helicobacter. 2021;26(6):e12857.PubMedCrossRef Chen MJ, Chen CC, Huang YC, Tseng CC, Hsu JT, Lin YF, et al. The efficacy of Lactobacillus acidophilus and rhamnosus in the reduction of bacterial load of Helicobacter pylori and modification of gut microbiota-a double-blind, placebo-controlled, randomized trial. Helicobacter. 2021;26(6):e12857.PubMedCrossRef
110.
Zurück zum Zitat Zhao Y, Li Z, Zhao L, Wang J, Wang F, Zhang Q, et al. Two novel lactic acid bacteria, limosilactobacillus fermentum MN-LF23 and Lactobacillus gasseri MN-LG80, inhibited Helicobacter pylori infection in C57BL/6 mice. Food Funct. 2022;13(21):11061–9.PubMedCrossRef Zhao Y, Li Z, Zhao L, Wang J, Wang F, Zhang Q, et al. Two novel lactic acid bacteria, limosilactobacillus fermentum MN-LF23 and Lactobacillus gasseri MN-LG80, inhibited Helicobacter pylori infection in C57BL/6 mice. Food Funct. 2022;13(21):11061–9.PubMedCrossRef
111.
Zurück zum Zitat Delgado S, Leite AMO, Ruas-Madiedo P, Mayo B. Probiotic and technological properties of Lactobacillus spp. strains from the human stomach in the search for potential candidates against gastric microbial dysbiosis. Front Microbiol. 2015;5:5.CrossRef Delgado S, Leite AMO, Ruas-Madiedo P, Mayo B. Probiotic and technological properties of Lactobacillus spp. strains from the human stomach in the search for potential candidates against gastric microbial dysbiosis. Front Microbiol. 2015;5:5.CrossRef
112.
Zurück zum Zitat Xu Z, Xiao L, Wang S, Cheng Y, Wu J, Meng Y, et al. Alteration of gastric microbiota and transcriptome in a rat with gastric intestinal metaplasia induced by deoxycholic acid. Front Microbiol. 2023;14:1160821.PubMedPubMedCentralCrossRef Xu Z, Xiao L, Wang S, Cheng Y, Wu J, Meng Y, et al. Alteration of gastric microbiota and transcriptome in a rat with gastric intestinal metaplasia induced by deoxycholic acid. Front Microbiol. 2023;14:1160821.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Jin D, Huang K, Xu M, Hua H, Ye F, Yan J, et al. Deoxycholic acid induces gastric intestinal metaplasia by activating STAT3 signaling and disturbing gastric bile acids metabolism and microbiota. Gut Microbes. 2022;14(1):2120744.PubMedPubMedCentralCrossRef Jin D, Huang K, Xu M, Hua H, Ye F, Yan J, et al. Deoxycholic acid induces gastric intestinal metaplasia by activating STAT3 signaling and disturbing gastric bile acids metabolism and microbiota. Gut Microbes. 2022;14(1):2120744.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Wang S, Kuang J, Zhang H, Chen W, Zheng X, Wang J, et al. Bile acid–microbiome interaction promotes gastric carcinogenesis. Adv Sci. 2022;9(16):2200263.CrossRef Wang S, Kuang J, Zhang H, Chen W, Zheng X, Wang J, et al. Bile acid–microbiome interaction promotes gastric carcinogenesis. Adv Sci. 2022;9(16):2200263.CrossRef
115.
Zurück zum Zitat Gunathilake M, Lee J, Choi IJ, Kim YI, Yoon J, Sul WJ, et al. Alterations in gastric microbial communities are associated with risk of gastric cancer in a Korean population: a case-control study. Cancers. 2020;12(9):2619.PubMedPubMedCentralCrossRef Gunathilake M, Lee J, Choi IJ, Kim YI, Yoon J, Sul WJ, et al. Alterations in gastric microbial communities are associated with risk of gastric cancer in a Korean population: a case-control study. Cancers. 2020;12(9):2619.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Seyyedsalehi MS, Mohebbi E, Tourang F, Sasanfar B, Boffetta P, Zendehdel K. Association of dietary nitrate, nitrite, and N-Nitroso compounds Intake and gastrointestinal cancers: a systematic review and meta-analysis. Toxics. 2023;11(2):190.PubMedPubMedCentralCrossRef Seyyedsalehi MS, Mohebbi E, Tourang F, Sasanfar B, Boffetta P, Zendehdel K. Association of dietary nitrate, nitrite, and N-Nitroso compounds Intake and gastrointestinal cancers: a systematic review and meta-analysis. Toxics. 2023;11(2):190.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Kraus A, McKeague M, Seiwert N, Nagel G, Geisen SM, Ziegler N, et al. Immunological and mass spectrometry-based approaches to determine thresholds of the mutagenic DNA adduct O6-methylguanine in vivo. Arch Toxicol. 2019;93(2):559–72.PubMedCrossRef Kraus A, McKeague M, Seiwert N, Nagel G, Geisen SM, Ziegler N, et al. Immunological and mass spectrometry-based approaches to determine thresholds of the mutagenic DNA adduct O6-methylguanine in vivo. Arch Toxicol. 2019;93(2):559–72.PubMedCrossRef
118.
Zurück zum Zitat Oosterlinck B, Ceuleers H, Arras W, De Man JG, Geboes K, De Schepper H, et al. Mucin-microbiome signatures shape the tumor microenvironment in gastric cancer. Microbiome. 2023;11(1):86.PubMedPubMedCentralCrossRef Oosterlinck B, Ceuleers H, Arras W, De Man JG, Geboes K, De Schepper H, et al. Mucin-microbiome signatures shape the tumor microenvironment in gastric cancer. Microbiome. 2023;11(1):86.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Shimamura T, Ito H, Shibahara J, Watanabe A, Hippo Y, Taniguchi H, et al. Overexpression of MUC13 is associated with intestinal-type gastric cancer. Cancer Sci. 2005;96(5):265–73.PubMedCrossRef Shimamura T, Ito H, Shibahara J, Watanabe A, Hippo Y, Taniguchi H, et al. Overexpression of MUC13 is associated with intestinal-type gastric cancer. Cancer Sci. 2005;96(5):265–73.PubMedCrossRef
120.
Zurück zum Zitat Sheng YH, He Y, Hasnain SZ, Wang R, Tong H, Clarke DT, et al. MUC13 protects colorectal cancer cells from death by activating the NF-κB pathway and is a potential therapeutic target. Oncogene. 2017;36(5):700–13.PubMedCrossRef Sheng YH, He Y, Hasnain SZ, Wang R, Tong H, Clarke DT, et al. MUC13 protects colorectal cancer cells from death by activating the NF-κB pathway and is a potential therapeutic target. Oncogene. 2017;36(5):700–13.PubMedCrossRef
121.
Zurück zum Zitat Tiemin P, Fanzheng M, Peng X, Jihua H, Ruipeng S, Yaliang L, et al. MUC13 promotes intrahepatic cholangiocarcinoma progression via EGFR/PI3K/AKT pathways. J Hepatol. 2020;72(4):761–73.PubMedCrossRef Tiemin P, Fanzheng M, Peng X, Jihua H, Ruipeng S, Yaliang L, et al. MUC13 promotes intrahepatic cholangiocarcinoma progression via EGFR/PI3K/AKT pathways. J Hepatol. 2020;72(4):761–73.PubMedCrossRef
122.
Zurück zum Zitat Cao Y, Xia H, Tan X, Shi C, Ma Y, Meng D, et al. Intratumoural microbiota: a new frontier in cancer development and therapy. Signal Transduct Target Ther. 2024;9:15.PubMedPubMedCentralCrossRef Cao Y, Xia H, Tan X, Shi C, Ma Y, Meng D, et al. Intratumoural microbiota: a new frontier in cancer development and therapy. Signal Transduct Target Ther. 2024;9:15.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Yue K, Sheng D, Xue X, Zhao L, Zhao G, Jin C, et al. Bidirectional mediation effects between intratumoral microbiome and host DNA methylation changes contribute to stomach adenocarcinoma. Microbiol Spectr. 2023;11(4):e0090423.PubMedCrossRef Yue K, Sheng D, Xue X, Zhao L, Zhao G, Jin C, et al. Bidirectional mediation effects between intratumoral microbiome and host DNA methylation changes contribute to stomach adenocarcinoma. Microbiol Spectr. 2023;11(4):e0090423.PubMedCrossRef
124.
125.
Zurück zum Zitat Xin Y, Li X, Zhang M, Shang Z, Luo Z, Wang Y, et al. Fusobacterium nucleatum -induced exosomal HOTTIP promotes gastric cancer progression through the microRNA -885-3p/ EphB2 axis. Cancer Sci. 2023;114(6):2360–74.PubMedPubMedCentralCrossRef Xin Y, Li X, Zhang M, Shang Z, Luo Z, Wang Y, et al. Fusobacterium nucleatum -induced exosomal HOTTIP promotes gastric cancer progression through the microRNA -885-3p/ EphB2 axis. Cancer Sci. 2023;114(6):2360–74.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Nie S, Wang A, Yuan Y. Comparison of clinicopathological parameters, prognosis, micro-ecological environment and metabolic function of gastric cancer with or without Fusobacterium sp. infection. J Cancer. 2021;12(4):1023–32.PubMedPubMedCentralCrossRef Nie S, Wang A, Yuan Y. Comparison of clinicopathological parameters, prognosis, micro-ecological environment and metabolic function of gastric cancer with or without Fusobacterium sp. infection. J Cancer. 2021;12(4):1023–32.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat do Nascimento Araujo C, Amorim AT, Barbosa MS, Alexandre JCPL, Campos GB, Macedo CL, et al. Evaluating the presence of Mycoplasma hyorhinis, Fusobacterium nucleatum, and Helicobacter pylori in biopsies of patients with gastric cancer. Infect Agent Cancer. 2021;16(1):70.PubMedPubMedCentralCrossRef do Nascimento Araujo C, Amorim AT, Barbosa MS, Alexandre JCPL, Campos GB, Macedo CL, et al. Evaluating the presence of Mycoplasma hyorhinis, Fusobacterium nucleatum, and Helicobacter pylori in biopsies of patients with gastric cancer. Infect Agent Cancer. 2021;16(1):70.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Parhi L, Alon-Maimon T, Sol A, Nejman D, Shhadeh A, Fainsod-Levi T, et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat Commun. 2020;11(1):3259.PubMedPubMedCentralCrossRef Parhi L, Alon-Maimon T, Sol A, Nejman D, Shhadeh A, Fainsod-Levi T, et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat Commun. 2020;11(1):3259.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Mereiter S, Polom K, Williams C, Polonia A, Guergova-Kuras M, Karlsson NG, et al. The Thomsen-Friedenreich antigen: a highly sensitive and specific predictor of microsatellite instability in gastric cancer. J Clin Med. 2018;7(9):256.PubMedPubMedCentralCrossRef Mereiter S, Polom K, Williams C, Polonia A, Guergova-Kuras M, Karlsson NG, et al. The Thomsen-Friedenreich antigen: a highly sensitive and specific predictor of microsatellite instability in gastric cancer. J Clin Med. 2018;7(9):256.PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, et al. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 2020;39(1):202.PubMedPubMedCentralCrossRef Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, et al. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 2020;39(1):202.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Yang Y, Weng W, Peng J, Hong L, Yang L, Toiyama Y, et al. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating toll-like receptor 4 signaling to nuclear factor − κB, and up-regulating expression of microRNA-21. Gastroenterology. 2017;152(4):851–866.e24.PubMedCrossRef Yang Y, Weng W, Peng J, Hong L, Yang L, Toiyama Y, et al. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating toll-like receptor 4 signaling to nuclear factor − κB, and up-regulating expression of microRNA-21. Gastroenterology. 2017;152(4):851–866.e24.PubMedCrossRef
132.
Zurück zum Zitat Kang W, Jia Z, Tang D, Zhang Z, Gao H, He K, et al. Fusobacterium nucleatum facilitates apoptosis, ROS generation, and inflammatory cytokine production by activating AKT/MAPK and NF- κ B signaling pathways in human gingival fibroblasts. Oxid Med Cell Longev. 2019;2019:1–22.CrossRef Kang W, Jia Z, Tang D, Zhang Z, Gao H, He K, et al. Fusobacterium nucleatum facilitates apoptosis, ROS generation, and inflammatory cytokine production by activating AKT/MAPK and NF- κ B signaling pathways in human gingival fibroblasts. Oxid Med Cell Longev. 2019;2019:1–22.CrossRef
133.
Zurück zum Zitat Chen Y, Chen Y, Cao P, Su W, Zhan N, Dong W. Fusobacterium nucleatum facilitates ulcerative colitis through activating IL-17F signaling to NF‐κB via the upregulation of CARD3 expression. J Pathol. 2020;250(2):170–82.PubMedCrossRef Chen Y, Chen Y, Cao P, Su W, Zhan N, Dong W. Fusobacterium nucleatum facilitates ulcerative colitis through activating IL-17F signaling to NF‐κB via the upregulation of CARD3 expression. J Pathol. 2020;250(2):170–82.PubMedCrossRef
134.
Zurück zum Zitat Xu C, Fan L, Lin Y, Shen W, Qi Y, Zhang Y, et al. Fusobacterium nucleatum promotes colorectal cancer metastasis through miR-1322/CCL20 axis and M2 polarization. Gut Microbes. 2021;13(1):1980347.PubMedPubMedCentralCrossRef Xu C, Fan L, Lin Y, Shen W, Qi Y, Zhang Y, et al. Fusobacterium nucleatum promotes colorectal cancer metastasis through miR-1322/CCL20 axis and M2 polarization. Gut Microbes. 2021;13(1):1980347.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Kim SY, Park SY, Jang HS, Park YD, Kee SH. Yes-associated protein is required for ZO-1-mediated tight-junction integrity and cell migration in E-cadherin-restored AGS gastric cancer cells. Biomedicines. 2021;9(9):1264.PubMedPubMedCentralCrossRef Kim SY, Park SY, Jang HS, Park YD, Kee SH. Yes-associated protein is required for ZO-1-mediated tight-junction integrity and cell migration in E-cadherin-restored AGS gastric cancer cells. Biomedicines. 2021;9(9):1264.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Peng C, Ouyang Y, Lu N, Li N. The NF-κB signaling pathway, the microbiota, and gastrointestinal tumorigenesis: recent advances. Front Immunol. 2020;11:1387.PubMedPubMedCentralCrossRef Peng C, Ouyang Y, Lu N, Li N. The NF-κB signaling pathway, the microbiota, and gastrointestinal tumorigenesis: recent advances. Front Immunol. 2020;11:1387.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Kim JM, Cho SJ, Oh YK, Jung HY, Kim YJ, Kim N. Nuclear factor-kappa B activation pathway in intestinal epithelial cells is a major regulator of chemokine gene expression and neutrophil migration induced by Bacteroides fragilis enterotoxin. Clin Exp Immunol. 2002;130(1):59–66.PubMedPubMedCentralCrossRef Kim JM, Cho SJ, Oh YK, Jung HY, Kim YJ, Kim N. Nuclear factor-kappa B activation pathway in intestinal epithelial cells is a major regulator of chemokine gene expression and neutrophil migration induced by Bacteroides fragilis enterotoxin. Clin Exp Immunol. 2002;130(1):59–66.PubMedPubMedCentralCrossRef
138.
Zurück zum Zitat Cao L, Zhu S, Lu H, Soutto M, Bhat N, Chen Z, et al. Helicobacter pylori-induced RASAL2 through activation of NF-kB promotes gastric tumorigenesis via β-catenin signaling axis. Gastroenterology. 2022;162(6):1716–1731.e17.PubMedCrossRef Cao L, Zhu S, Lu H, Soutto M, Bhat N, Chen Z, et al. Helicobacter pylori-induced RASAL2 through activation of NF-kB promotes gastric tumorigenesis via β-catenin signaling axis. Gastroenterology. 2022;162(6):1716–1731.e17.PubMedCrossRef
139.
Zurück zum Zitat Maubach G, Vieth M, Boccellato F, Naumann M. Helicobacter pylori-induced NF-κB: trailblazer for gastric pathophysiology. Trends Mol Med. 2022;28(3):210–22.PubMedCrossRef Maubach G, Vieth M, Boccellato F, Naumann M. Helicobacter pylori-induced NF-κB: trailblazer for gastric pathophysiology. Trends Mol Med. 2022;28(3):210–22.PubMedCrossRef
140.
Zurück zum Zitat Chung L, Orberg ET, Geis AL, Chan JL, Fu K, DeStefano Shields CE, et al. Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory cascade via targeting of colonic epithelial cells. Cell Host Microbe. 2018;23(2):203–214.e5.PubMedPubMedCentralCrossRef Chung L, Orberg ET, Geis AL, Chan JL, Fu K, DeStefano Shields CE, et al. Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory cascade via targeting of colonic epithelial cells. Cell Host Microbe. 2018;23(2):203–214.e5.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Long X, Wong CC, Tong L, Chu ESH, Ho Szeto C, Go MYY, et al. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat Microbiol. 2019;4(12):2319–30.PubMedCrossRef Long X, Wong CC, Tong L, Chu ESH, Ho Szeto C, Go MYY, et al. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat Microbiol. 2019;4(12):2319–30.PubMedCrossRef
143.
Zurück zum Zitat Zhong M, Xiong Y, Zhao J, Gao Z, Ma J, Wu Z, et al. Candida albicans disorder is associated with gastric carcinogenesis. Theranostics. 2021;11(10):4945–56.PubMedPubMedCentralCrossRef Zhong M, Xiong Y, Zhao J, Gao Z, Ma J, Wu Z, et al. Candida albicans disorder is associated with gastric carcinogenesis. Theranostics. 2021;11(10):4945–56.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Dohlman AB, Klug J, Mesko M, Gao IH, Lipkin SM, Shen X, et al. A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell. 2022;185(20):3807–3822.e12.PubMedPubMedCentralCrossRef Dohlman AB, Klug J, Mesko M, Gao IH, Lipkin SM, Shen X, et al. A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell. 2022;185(20):3807–3822.e12.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Lohse MB, Gulati M, Johnson AD, Nobile CJ. Development and regulation of single- and multi-species Candida albicans biofilms. Nat Rev Microbiol. 2018;16(1):19–31.PubMedCrossRef Lohse MB, Gulati M, Johnson AD, Nobile CJ. Development and regulation of single- and multi-species Candida albicans biofilms. Nat Rev Microbiol. 2018;16(1):19–31.PubMedCrossRef
146.
Zurück zum Zitat Zhang Y, Gan Y, Wang J, Feng Z, Zhong Z, Bao H, et al. Dysbiosis of gut microbiota and intestinal barrier dysfunction in pigs with pulmonary inflammation induced by Mycoplasma hyorhinis Infection. mSystems. 2022;7(4):e00282–22. Zhang Y, Gan Y, Wang J, Feng Z, Zhong Z, Bao H, et al. Dysbiosis of gut microbiota and intestinal barrier dysfunction in pigs with pulmonary inflammation induced by Mycoplasma hyorhinis Infection. mSystems. 2022;7(4):e00282–22.
147.
Zurück zum Zitat Duan H, Chen L, Qu L, Yang H, Song SW, Han Y, et al. Mycoplasma Hyorhinis infection promotes NF- κB–dependent migration of gastric cancer cells. Cancer Res. 2014;74(20):5782–94.PubMedCrossRef Duan H, Chen L, Qu L, Yang H, Song SW, Han Y, et al. Mycoplasma Hyorhinis infection promotes NF- κB–dependent migration of gastric cancer cells. Cancer Res. 2014;74(20):5782–94.PubMedCrossRef
148.
Zurück zum Zitat Morgan AD, Seely KD, Hagenstein LD, Florey GM, Small JM. Bacterial involvement in progression and metastasis of adenocarcinoma of the stomach. Cancers. 2022;14(19):4886.PubMedPubMedCentralCrossRef Morgan AD, Seely KD, Hagenstein LD, Florey GM, Small JM. Bacterial involvement in progression and metastasis of adenocarcinoma of the stomach. Cancers. 2022;14(19):4886.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Xu Y, Li H, Chen W, Yao X, Xing Y, Wang X, et al. Mycoplasma hyorhinis activates the NLRP3 inflammasome and promotes migration and invasion of gastric cancer cells. PLoS ONE. 2013;8(11):e77955.PubMedPubMedCentralCrossRef Xu Y, Li H, Chen W, Yao X, Xing Y, Wang X, et al. Mycoplasma hyorhinis activates the NLRP3 inflammasome and promotes migration and invasion of gastric cancer cells. PLoS ONE. 2013;8(11):e77955.PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Chernov AV, Reyes L, Xu Z, Gonzalez B, Golovko G, Peterson S, et al. Mycoplasma CG- and GATC-specific DNA methyltransferases selectively and efficiently methylate the host genome and alter the epigenetic landscape in human cells. Epigenetics. 2015;10(4):303–18.PubMedPubMedCentralCrossRef Chernov AV, Reyes L, Xu Z, Gonzalez B, Golovko G, Peterson S, et al. Mycoplasma CG- and GATC-specific DNA methyltransferases selectively and efficiently methylate the host genome and alter the epigenetic landscape in human cells. Epigenetics. 2015;10(4):303–18.PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Li Q, Wu W, Gong D, Shang R, Wang J, Yu H. Propionibacterium acnes overabundance in gastric cancer promote M2 polarization of macrophages via a TLR4/PI3K/Akt signaling. Gastric Cancer. 2021;24(6):1242–53.PubMedCrossRef Li Q, Wu W, Gong D, Shang R, Wang J, Yu H. Propionibacterium acnes overabundance in gastric cancer promote M2 polarization of macrophages via a TLR4/PI3K/Akt signaling. Gastric Cancer. 2021;24(6):1242–53.PubMedCrossRef
153.
Zurück zum Zitat Petersen CP, Meyer AR, De Salvo C, Choi E, Schlegel C, Petersen A, et al. A signalling cascade of IL-33 to IL-13 regulates metaplasia in the mouse stomach. Gut. 2018;67(5):805–17.PubMedCrossRef Petersen CP, Meyer AR, De Salvo C, Choi E, Schlegel C, Petersen A, et al. A signalling cascade of IL-33 to IL-13 regulates metaplasia in the mouse stomach. Gut. 2018;67(5):805–17.PubMedCrossRef
154.
Zurück zum Zitat Satoh-Takayama N, Kato T, Motomura Y, Kageyama T, Taguchi-Atarashi N, Kinoshita-Daitoku R, et al. Bacteria-induced group 2 innate lymphoid cells in the stomach provide immune protection through induction of IgA. Immunity. 2020;52(4):635–649.e4.PubMedCrossRef Satoh-Takayama N, Kato T, Motomura Y, Kageyama T, Taguchi-Atarashi N, Kinoshita-Daitoku R, et al. Bacteria-induced group 2 innate lymphoid cells in the stomach provide immune protection through induction of IgA. Immunity. 2020;52(4):635–649.e4.PubMedCrossRef
155.
Zurück zum Zitat Bie Q, Zhang P, Su Z, Zheng D, Ying X, Wu Y, et al. Polarization of ILC2s in peripheral blood might contribute to immunosuppressive microenvironment in patients with gastric cancer. J Immunol Res. 2014;2014:923135.PubMedPubMedCentralCrossRef Bie Q, Zhang P, Su Z, Zheng D, Ying X, Wu Y, et al. Polarization of ILC2s in peripheral blood might contribute to immunosuppressive microenvironment in patients with gastric cancer. J Immunol Res. 2014;2014:923135.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Li R, Jiang XX, Zhang LF, Liu XM, Hu TZ, Xia XJ, et al. Group 2 innate lymphoid cells are involved in skewed type 2 immunity of gastric diseases induced by Helicobacter pylori infection. Mediators Inflamm. 2017;2017:4927964.PubMedPubMedCentralCrossRef Li R, Jiang XX, Zhang LF, Liu XM, Hu TZ, Xia XJ, et al. Group 2 innate lymphoid cells are involved in skewed type 2 immunity of gastric diseases induced by Helicobacter pylori infection. Mediators Inflamm. 2017;2017:4927964.PubMedPubMedCentralCrossRef
157.
158.
Zurück zum Zitat Hedrick CC, Malanchi I. Neutrophils in cancer: heterogeneous and multifaceted. Nat Rev Immunol. 2022;22(3):173–87.PubMedCrossRef Hedrick CC, Malanchi I. Neutrophils in cancer: heterogeneous and multifaceted. Nat Rev Immunol. 2022;22(3):173–87.PubMedCrossRef
159.
Zurück zum Zitat Li TJ, Jiang YM, Hu YF, Huang L, Yu J, Zhao LY, et al. Interleukin-17–producing neutrophils link inflammatory stimuli to disease progression by promoting angiogenesis in gastric cancer. Clin Cancer Res. 2017;23(6):1575–85.PubMedCrossRef Li TJ, Jiang YM, Hu YF, Huang L, Yu J, Zhao LY, et al. Interleukin-17–producing neutrophils link inflammatory stimuli to disease progression by promoting angiogenesis in gastric cancer. Clin Cancer Res. 2017;23(6):1575–85.PubMedCrossRef
160.
Zurück zum Zitat Li S, Cong X, Gao H, Lan X, Li Z, Wang W, et al. Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. J Exp Clin Cancer Res. 2019;38(1):6.PubMedPubMedCentralCrossRef Li S, Cong X, Gao H, Lan X, Li Z, Wang W, et al. Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. J Exp Clin Cancer Res. 2019;38(1):6.PubMedPubMedCentralCrossRef
161.
Zurück zum Zitat Nie P, Zhang W, Meng Y, Lin M, Guo F, Zhang H, et al. A YAP/TAZ-CD54 axis is required for CXCR2 − CD44 − tumor-specific neutrophils to suppress gastric cancer. Protein Cell. 2022;14(7):515–33.PubMedCentral Nie P, Zhang W, Meng Y, Lin M, Guo F, Zhang H, et al. A YAP/TAZ-CD54 axis is required for CXCR2 − CD44 − tumor-specific neutrophils to suppress gastric cancer. Protein Cell. 2022;14(7):515–33.PubMedCentral
162.
Zurück zum Zitat Shan ZG, Chen J, Liu JS, Zhang JY, Wang TT, Teng YS, et al. Activated neutrophils polarize protumorigenic interleukin-17A-producing T helper subsets through TNF-α-B7-H2-dependent pathway in human gastric cancer. Clin Transl Med. 2021;11(6):e484.PubMedPubMedCentralCrossRef Shan ZG, Chen J, Liu JS, Zhang JY, Wang TT, Teng YS, et al. Activated neutrophils polarize protumorigenic interleukin-17A-producing T helper subsets through TNF-α-B7-H2-dependent pathway in human gastric cancer. Clin Transl Med. 2021;11(6):e484.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Chu TH, Huang ST, Yang SF, Li CJ, Lin HW, Weng BC, et al. Hepatoma-derived growth factor participates in Helicobacter Pylori-induced neutrophils recruitment, gastritis and gastric carcinogenesis. Oncogene. 2019;38(37):6461–77.PubMedCrossRef Chu TH, Huang ST, Yang SF, Li CJ, Lin HW, Weng BC, et al. Hepatoma-derived growth factor participates in Helicobacter Pylori-induced neutrophils recruitment, gastritis and gastric carcinogenesis. Oncogene. 2019;38(37):6461–77.PubMedCrossRef
164.
Zurück zum Zitat Zhang Y, Chandra V, Riquelme Sanchez E, Dutta P, Quesada PR, Rakoski A, et al. Interleukin-17–induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J Exp Med. 2020;217(12):e20190354.PubMedPubMedCentralCrossRef Zhang Y, Chandra V, Riquelme Sanchez E, Dutta P, Quesada PR, Rakoski A, et al. Interleukin-17–induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J Exp Med. 2020;217(12):e20190354.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015;522(7556):345–8.PubMedPubMedCentralCrossRef Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015;522(7556):345–8.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Zhang T, Li Y, Chen J, Liu Y, Huang Z, Xu X, et al. IDDF2023-ABS-0058 Fusobacterium nucleatum intracellular parasitism activates the NF-KB/IL-17 signaling pathway induce neutrophil recruitment and promote gastric cancer progression. Gut. 2023;72(Suppl 1):A70. Zhang T, Li Y, Chen J, Liu Y, Huang Z, Xu X, et al. IDDF2023-ABS-0058 Fusobacterium nucleatum intracellular parasitism activates the NF-KB/IL-17 signaling pathway induce neutrophil recruitment and promote gastric cancer progression. Gut. 2023;72(Suppl 1):A70.
167.
Zurück zum Zitat Ling Z, Shao L, Liu X, Cheng Y, Yan C, Mei Y, et al. Regulatory T cells and plasmacytoid dendritic cells within the tumor microenvironment in gastric cancer are correlated with gastric microbiota dysbiosis: a preliminary study. Front Immunol. 2019;10:533.PubMedPubMedCentralCrossRef Ling Z, Shao L, Liu X, Cheng Y, Yan C, Mei Y, et al. Regulatory T cells and plasmacytoid dendritic cells within the tumor microenvironment in gastric cancer are correlated with gastric microbiota dysbiosis: a preliminary study. Front Immunol. 2019;10:533.PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Liu W, Zhao J, Li Q, Wang Q, Zhou Y, Tong Z. Gastric cancer patients have elevated plasmacytoid and CD1c+ dendritic cells in the peripheral blood. Oncol Lett. 2018;15(4):5087–92.PubMedPubMedCentral Liu W, Zhao J, Li Q, Wang Q, Zhou Y, Tong Z. Gastric cancer patients have elevated plasmacytoid and CD1c+ dendritic cells in the peripheral blood. Oncol Lett. 2018;15(4):5087–92.PubMedPubMedCentral
170.
Zurück zum Zitat Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42(2):344–55.PubMedPubMedCentralCrossRef Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42(2):344–55.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Dutta A, Venkataganesh H, Love PE. New insights into epigenetic regulation of T cell differentiation. Cells. 2021;10(12):533.CrossRef Dutta A, Venkataganesh H, Love PE. New insights into epigenetic regulation of T cell differentiation. Cells. 2021;10(12):533.CrossRef
172.
Zurück zum Zitat Lee K, Hwang H, Nam KT. Immune response and the tumor microenvironment: how they communicate to regulate gastric cancer. Gut Liver. 2014;8(2):131–9.PubMedPubMedCentralCrossRef Lee K, Hwang H, Nam KT. Immune response and the tumor microenvironment: how they communicate to regulate gastric cancer. Gut Liver. 2014;8(2):131–9.PubMedPubMedCentralCrossRef
173.
174.
Zurück zum Zitat Lin R, Zhang H, Yuan Y, He Q, Zhou J, Li S, et al. Fatty acid oxidation controls CD8 + tissue-resident memory T-cell survival in gastric adenocarcinoma. Cancer Immunol Res. 2020;8(4):479–92.PubMedCrossRef Lin R, Zhang H, Yuan Y, He Q, Zhou J, Li S, et al. Fatty acid oxidation controls CD8 + tissue-resident memory T-cell survival in gastric adenocarcinoma. Cancer Immunol Res. 2020;8(4):479–92.PubMedCrossRef
175.
Zurück zum Zitat Aydın EM, Demir TD, Seymen N, Said SS, Oktem-Okullu S, Tiftikci A, et al. The crosstalk between H. pylori virulence factors and the PD1:PD-L1 immune checkpoint inhibitors in progression to gastric cancer. Immunol Lett. 2021;239:1–11.PubMedCrossRef Aydın EM, Demir TD, Seymen N, Said SS, Oktem-Okullu S, Tiftikci A, et al. The crosstalk between H. pylori virulence factors and the PD1:PD-L1 immune checkpoint inhibitors in progression to gastric cancer. Immunol Lett. 2021;239:1–11.PubMedCrossRef
176.
Zurück zum Zitat Holokai L, Chakrabarti J, Broda T, Chang J, Hawkins JA, Sundaram N, et al. Increased programmed death-ligand 1 is an early epithelial cell response to Helicobacter pylori infection. Blanke SR, editor. PLOS Pathog. 2019;15(1):e1007468.PubMedPubMedCentralCrossRef Holokai L, Chakrabarti J, Broda T, Chang J, Hawkins JA, Sundaram N, et al. Increased programmed death-ligand 1 is an early epithelial cell response to Helicobacter pylori infection. Blanke SR, editor. PLOS Pathog. 2019;15(1):e1007468.PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Koh V, Chakrabarti J, Torvund M, Steele N, Hawkins JA, Ito Y, et al. Hedgehog transcriptional effector GLI mediates mTOR-Induced PD-L1 expression in gastric cancer organoids. Cancer Lett. 2021;518:59–71.PubMedPubMedCentralCrossRef Koh V, Chakrabarti J, Torvund M, Steele N, Hawkins JA, Ito Y, et al. Hedgehog transcriptional effector GLI mediates mTOR-Induced PD-L1 expression in gastric cancer organoids. Cancer Lett. 2021;518:59–71.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Li C, Jiang P, Wei S, Xu X, Wang J. Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects. Mol Cancer. 2020;19(1):116.PubMedPubMedCentralCrossRef Li C, Jiang P, Wei S, Xu X, Wang J. Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects. Mol Cancer. 2020;19(1):116.PubMedPubMedCentralCrossRef
179.
Zurück zum Zitat Urakawa S, Yamasaki M, Makino T, Kurokawa Y, Yamamoto K, Goto K, et al. The impact of ICOS + regulatory T cells and Helicobacter pylori infection on the prognosis of patients with gastric and colorectal cancer: potential prognostic benefit of pre-operative eradication therapy. Cancer Immunol Immunother. 2021;70(2):443–52.PubMedCrossRef Urakawa S, Yamasaki M, Makino T, Kurokawa Y, Yamamoto K, Goto K, et al. The impact of ICOS + regulatory T cells and Helicobacter pylori infection on the prognosis of patients with gastric and colorectal cancer: potential prognostic benefit of pre-operative eradication therapy. Cancer Immunol Immunother. 2021;70(2):443–52.PubMedCrossRef
180.
Zurück zum Zitat Nagase H, Takeoka T, Urakawa S, Morimoto-Okazawa A, Kawashima A, Iwahori K, et al. ICOS + Foxp3 + TILs in gastric cancer are prognostic markers and effector regulatory T cells associated with H elicobacter pylori: unique expression of ICOS on TREGS in gastric cancer. Int J Cancer. 2017;140(3):686–95.PubMedCrossRef Nagase H, Takeoka T, Urakawa S, Morimoto-Okazawa A, Kawashima A, Iwahori K, et al. ICOS + Foxp3 + TILs in gastric cancer are prognostic markers and effector regulatory T cells associated with H elicobacter pylori: unique expression of ICOS on TREGS in gastric cancer. Int J Cancer. 2017;140(3):686–95.PubMedCrossRef
181.
Zurück zum Zitat Engelhard V, Conejo-Garcia JR, Ahmed R, Nelson BH, Willard-Gallo K, Bruno TC, et al. B cells and cancer. Cancer Cell. 2021;39(10):1293–6.PubMedCrossRef Engelhard V, Conejo-Garcia JR, Ahmed R, Nelson BH, Willard-Gallo K, Bruno TC, et al. B cells and cancer. Cancer Cell. 2021;39(10):1293–6.PubMedCrossRef
182.
Zurück zum Zitat Li H, Limenitakis JP, Greiff V, Yilmaz B, Schären O, Urbaniak C, et al. Mucosal or systemic microbiota exposures shape the B cell repertoire. Nature. 2020;584(7820):274–8.PubMedCrossRef Li H, Limenitakis JP, Greiff V, Yilmaz B, Schären O, Urbaniak C, et al. Mucosal or systemic microbiota exposures shape the B cell repertoire. Nature. 2020;584(7820):274–8.PubMedCrossRef
183.
Zurück zum Zitat Catalán D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillón JC, et al. Immunosuppressive mechanisms of regulatory B cells. Front Immunol. 2021;12:611795.PubMedPubMedCentralCrossRef Catalán D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillón JC, et al. Immunosuppressive mechanisms of regulatory B cells. Front Immunol. 2021;12:611795.PubMedPubMedCentralCrossRef
184.
Zurück zum Zitat Horii M, Matsushita T. Regulatory B cells and T cell regulation in cancer. J Mol Biol. 2021;433(1):166685.PubMedCrossRef Horii M, Matsushita T. Regulatory B cells and T cell regulation in cancer. J Mol Biol. 2021;433(1):166685.PubMedCrossRef
185.
Zurück zum Zitat Nahid-Samiei M, Rahimian G, Shafigh M, Taheri F, Karami-Hurestani M, Sanaei MJ, et al. Enhanced frequency of CD19 + IL-10 + B cells in human gastric mucosa infected by Helicobacter pylori. Am J Med Sci. 2020;359(6):347–53.PubMedCrossRef Nahid-Samiei M, Rahimian G, Shafigh M, Taheri F, Karami-Hurestani M, Sanaei MJ, et al. Enhanced frequency of CD19 + IL-10 + B cells in human gastric mucosa infected by Helicobacter pylori. Am J Med Sci. 2020;359(6):347–53.PubMedCrossRef
187.
188.
Zurück zum Zitat Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science. 2015;350(6264):1084–9.PubMedPubMedCentralCrossRef Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science. 2015;350(6264):1084–9.PubMedPubMedCentralCrossRef
189.
Zurück zum Zitat Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–84.PubMedPubMedCentralCrossRef Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–84.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.PubMedCrossRef Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.PubMedCrossRef
191.
Zurück zum Zitat Si W, Liang H, Bugno J, Xu Q, Ding X, Yang K, et al. Lactobacillus rhamnosus GG induces cGAS/STING- dependent type I interferon and improves response to immune checkpoint blockade. Gut. 2022;71(3):521–33.PubMedCrossRef Si W, Liang H, Bugno J, Xu Q, Ding X, Yang K, et al. Lactobacillus rhamnosus GG induces cGAS/STING- dependent type I interferon and improves response to immune checkpoint blockade. Gut. 2022;71(3):521–33.PubMedCrossRef
192.
Zurück zum Zitat Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.PubMedCrossRef Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.PubMedCrossRef
193.
Zurück zum Zitat Lee SH, Cho SY, Yoon Y, Park C, Sohn J, Jeong JJ, et al. Bifidobacterium bifidum strains synergize with immune checkpoint inhibitors to reduce tumour burden in mice. Nat Microbiol. 2021;6(3):277–88.PubMedCrossRef Lee SH, Cho SY, Yoon Y, Park C, Sohn J, Jeong JJ, et al. Bifidobacterium bifidum strains synergize with immune checkpoint inhibitors to reduce tumour burden in mice. Nat Microbiol. 2021;6(3):277–88.PubMedCrossRef
194.
Zurück zum Zitat Newsome RC, Gharaibeh RZ, Pierce CM, da Silva WV, Paul S, Hogue SR, et al. Interaction of bacterial genera associated with therapeutic response to immune checkpoint PD-1 blockade in a United States cohort. Genome Med. 2022;14(1):35.PubMedPubMedCentralCrossRef Newsome RC, Gharaibeh RZ, Pierce CM, da Silva WV, Paul S, Hogue SR, et al. Interaction of bacterial genera associated with therapeutic response to immune checkpoint PD-1 blockade in a United States cohort. Genome Med. 2022;14(1):35.PubMedPubMedCentralCrossRef
195.
Zurück zum Zitat Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol off J Eur Soc Med Oncol. 2019;30(12):2012.CrossRef Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol off J Eur Soc Med Oncol. 2019;30(12):2012.CrossRef
196.
Zurück zum Zitat Hakozaki T, Richard C, Elkrief A, Hosomi Y, Benlaïfaoui M, Mimpen I, et al. The gut microbiome associates with immune checkpoint inhibition outcomes in patients with advanced non-small cell lung cancer. Cancer Immunol Res. 2020;8(10):1243–50.PubMedCrossRef Hakozaki T, Richard C, Elkrief A, Hosomi Y, Benlaïfaoui M, Mimpen I, et al. The gut microbiome associates with immune checkpoint inhibition outcomes in patients with advanced non-small cell lung cancer. Cancer Immunol Res. 2020;8(10):1243–50.PubMedCrossRef
197.
Zurück zum Zitat Mao J, Wang D, Long J, Yang X, Lin J, Song Y, et al. Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers. J Immunother Cancer. 2021;9(12):e003334.PubMedPubMedCentralCrossRef Mao J, Wang D, Long J, Yang X, Lin J, Song Y, et al. Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers. J Immunother Cancer. 2021;9(12):e003334.PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat McCulloch JA, Davar D, Rodrigues RR, Badger JH, Fang JR, Cole AM, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med. 2022;28(3):545–56.PubMedPubMedCentralCrossRef McCulloch JA, Davar D, Rodrigues RR, Badger JH, Fang JR, Cole AM, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med. 2022;28(3):545–56.PubMedPubMedCentralCrossRef
199.
Zurück zum Zitat Peng Z, Cheng S, Kou Y, Wang Z, Jin R, Hu H, et al. The gut microbiome is associated with clinical response to Anti–PD-1/PD-L1 immunotherapy in gastrointestinal cancer. Cancer Immunol Res. 2020;8(10):1251–61.PubMedCrossRef Peng Z, Cheng S, Kou Y, Wang Z, Jin R, Hu H, et al. The gut microbiome is associated with clinical response to Anti–PD-1/PD-L1 immunotherapy in gastrointestinal cancer. Cancer Immunol Res. 2020;8(10):1251–61.PubMedCrossRef
200.
Zurück zum Zitat Oster P, Vaillant L, Riva E, McMillan B, Begka C, Truntzer C, et al. Helicobacter pylori infection has a detrimental impact on the efficacy of cancer immunotherapies. Gut. 2022;71(3):457–66.PubMedCrossRef Oster P, Vaillant L, Riva E, McMillan B, Begka C, Truntzer C, et al. Helicobacter pylori infection has a detrimental impact on the efficacy of cancer immunotherapies. Gut. 2022;71(3):457–66.PubMedCrossRef
201.
Zurück zum Zitat Che H, Xiong Q, Ma J, Chen S, Wu H, Xu H, et al. Association of Helicobacter pylori infection with survival outcomes in advanced gastric cancer patients treated with immune checkpoint inhibitors. BMC Cancer. 2022;22(1):904.PubMedPubMedCentralCrossRef Che H, Xiong Q, Ma J, Chen S, Wu H, Xu H, et al. Association of Helicobacter pylori infection with survival outcomes in advanced gastric cancer patients treated with immune checkpoint inhibitors. BMC Cancer. 2022;22(1):904.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Tonneau M, Nolin-Lapalme A, Kazandjian S, Auclin E, Panasci J, Benlaifaoui M, et al. Helicobacter pylori serology is associated with worse overall survival in patients with melanoma treated with immune checkpoint inhibitors. Oncoimmunology. 2022;11(1):2096535.PubMedPubMedCentralCrossRef Tonneau M, Nolin-Lapalme A, Kazandjian S, Auclin E, Panasci J, Benlaifaoui M, et al. Helicobacter pylori serology is associated with worse overall survival in patients with melanoma treated with immune checkpoint inhibitors. Oncoimmunology. 2022;11(1):2096535.PubMedPubMedCentralCrossRef
203.
Zurück zum Zitat Derosa L, Routy B, Fidelle M, Iebba V, Alla L, Pasolli E, et al. Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. Eur Urol. 2020;78(2):195–206.PubMedCrossRef Derosa L, Routy B, Fidelle M, Iebba V, Alla L, Pasolli E, et al. Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. Eur Urol. 2020;78(2):195–206.PubMedCrossRef
204.
Zurück zum Zitat Pinato DJ, Howlett S, Ottaviani D, Urus H, Patel A, Mineo T, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 2019;5(12):1774–8.PubMedPubMedCentralCrossRef Pinato DJ, Howlett S, Ottaviani D, Urus H, Patel A, Mineo T, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 2019;5(12):1774–8.PubMedPubMedCentralCrossRef
205.
Zurück zum Zitat Ahmed J, Kumar A, Parikh K, Anwar A, Knoll BM, Puccio C, et al. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. Oncoimmunology. 2018;7(11):e1507670.PubMedPubMedCentralCrossRef Ahmed J, Kumar A, Parikh K, Anwar A, Knoll BM, Puccio C, et al. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. Oncoimmunology. 2018;7(11):e1507670.PubMedPubMedCentralCrossRef
206.
Zurück zum Zitat Hsu PI, Pan CY, Kao JY, Tsay FW, Peng NJ, Kao SS, et al. Helicobacter pylori eradication with bismuth quadruple therapy leads to dysbiosis of gut microbiota with an increased relative abundance of proteobacteria and decreased relative abundances of bacteroidetes and actinobacteria. Helicobacter. 2018;23(4):e12498.PubMedCrossRef Hsu PI, Pan CY, Kao JY, Tsay FW, Peng NJ, Kao SS, et al. Helicobacter pylori eradication with bismuth quadruple therapy leads to dysbiosis of gut microbiota with an increased relative abundance of proteobacteria and decreased relative abundances of bacteroidetes and actinobacteria. Helicobacter. 2018;23(4):e12498.PubMedCrossRef
207.
Zurück zum Zitat Schulz C, Schütte K, Koch N, Vilchez-Vargas R, Wos-Oxley ML, Oxley APA, et al. The active bacterial assemblages of the upper GI tract in individuals with and without Helicobacter infection. Gut. 2018;67(2):216–25.PubMedCrossRef Schulz C, Schütte K, Koch N, Vilchez-Vargas R, Wos-Oxley ML, Oxley APA, et al. The active bacterial assemblages of the upper GI tract in individuals with and without Helicobacter infection. Gut. 2018;67(2):216–25.PubMedCrossRef
208.
Zurück zum Zitat Olekhnovich EI, Manolov AI, Samoilov AE, Prianichnikov NA, Malakhova MV, Tyakht AV, et al. Shifts in the human gut microbiota structure caused by quadruple Helicobacter pylori eradication therapy. Front Microbiol. 2019;10:1902.PubMedPubMedCentralCrossRef Olekhnovich EI, Manolov AI, Samoilov AE, Prianichnikov NA, Malakhova MV, Tyakht AV, et al. Shifts in the human gut microbiota structure caused by quadruple Helicobacter pylori eradication therapy. Front Microbiol. 2019;10:1902.PubMedPubMedCentralCrossRef
209.
Zurück zum Zitat Kakiuchi T, Mizoe A, Yamamoto K, Imamura I, Hashiguchi K, Kawakubo H, et al. Effect of probiotics during vonoprazan-containing triple therapy on gut microbiota in Helicobacter pylori infection: a randomized controlled trial. Helicobacter. 2020;25(3):e12690.PubMedCrossRef Kakiuchi T, Mizoe A, Yamamoto K, Imamura I, Hashiguchi K, Kawakubo H, et al. Effect of probiotics during vonoprazan-containing triple therapy on gut microbiota in Helicobacter pylori infection: a randomized controlled trial. Helicobacter. 2020;25(3):e12690.PubMedCrossRef
210.
Zurück zum Zitat Liou JM, Chen CC, Chang CM, Fang YJ, Bair MJ, Chen PY, et al. Long-term changes of gut microbiota, antibiotic resistance, and metabolic parameters after Helicobacter pylori eradication: a multicentre, open-label, randomised trial. Lancet Infect Dis. 2019;19(10):1109–20.PubMedCrossRef Liou JM, Chen CC, Chang CM, Fang YJ, Bair MJ, Chen PY, et al. Long-term changes of gut microbiota, antibiotic resistance, and metabolic parameters after Helicobacter pylori eradication: a multicentre, open-label, randomised trial. Lancet Infect Dis. 2019;19(10):1109–20.PubMedCrossRef
211.
Zurück zum Zitat Lurienne L, Cervesi J, Duhalde L, de Gunzburg J, Andremont A, Zalcman G, et al. NSCLC immunotherapy efficacy and antibiotic use: a systematic review and meta-analysis. J Thorac Oncol. 2020;15(7):1147–59.PubMedCrossRef Lurienne L, Cervesi J, Duhalde L, de Gunzburg J, Andremont A, Zalcman G, et al. NSCLC immunotherapy efficacy and antibiotic use: a systematic review and meta-analysis. J Thorac Oncol. 2020;15(7):1147–59.PubMedCrossRef
212.
Zurück zum Zitat Iankov ID, Allen C, Federspiel MJ, Myers RM, Peng KW, Ingle JN, et al. Expression of immunomodulatory neutrophil-activating protein of Helicobacter pylori enhances the antitumor activity of oncolytic measles virus. Mol Ther. 2012;20(6):1139–47.PubMedPubMedCentralCrossRef Iankov ID, Allen C, Federspiel MJ, Myers RM, Peng KW, Ingle JN, et al. Expression of immunomodulatory neutrophil-activating protein of Helicobacter pylori enhances the antitumor activity of oncolytic measles virus. Mol Ther. 2012;20(6):1139–47.PubMedPubMedCentralCrossRef
213.
Zurück zum Zitat Ma J, Jin C, Čančer M, Wang H, Ramachandran M, Yu D. Concurrent expression of HP-NAP enhances antitumor efficacy of oncolytic vaccinia virus but not for Semliki Forest virus. Mol Ther - Oncolytics. 2021;21:356–66.PubMedPubMedCentralCrossRef Ma J, Jin C, Čančer M, Wang H, Ramachandran M, Yu D. Concurrent expression of HP-NAP enhances antitumor efficacy of oncolytic vaccinia virus but not for Semliki Forest virus. Mol Ther - Oncolytics. 2021;21:356–66.PubMedPubMedCentralCrossRef
214.
Zurück zum Zitat Jin C, Ma J, Ramachandran M, Yu D, Essand M. CAR T cells expressing a bacterial virulence factor trigger potent bystander antitumour responses in solid cancers. Nat Biomed Eng. 2022;6(7):830–41.PubMedPubMedCentralCrossRef Jin C, Ma J, Ramachandran M, Yu D, Essand M. CAR T cells expressing a bacterial virulence factor trigger potent bystander antitumour responses in solid cancers. Nat Biomed Eng. 2022;6(7):830–41.PubMedPubMedCentralCrossRef
215.
Zurück zum Zitat Xue LJ, Mao XB, Liu XB, Gao H, Chen YN, Dai TT, et al. Activation of CD3 + T cells by Helicobacter pylori DNA vaccines in potential immunotherapy of gastric carcinoma. Cancer Biol Ther. 2019;20(6):866–76.PubMedPubMedCentralCrossRef Xue LJ, Mao XB, Liu XB, Gao H, Chen YN, Dai TT, et al. Activation of CD3 + T cells by Helicobacter pylori DNA vaccines in potential immunotherapy of gastric carcinoma. Cancer Biol Ther. 2019;20(6):866–76.PubMedPubMedCentralCrossRef
216.
Zurück zum Zitat Codolo G, Coletta S, D’Elios MM, de Bernard M. HP-NAP of Helicobacter pylori: the power of the Immunomodulation. Front Immunol. 2022;13:944139.PubMedPubMedCentralCrossRef Codolo G, Coletta S, D’Elios MM, de Bernard M. HP-NAP of Helicobacter pylori: the power of the Immunomodulation. Front Immunol. 2022;13:944139.PubMedPubMedCentralCrossRef
217.
Zurück zum Zitat Ramachandran M, Jin C, Yu D, Eriksson F, Essand M. Correction: Vector-encoded Helicobacter pylori neutrophil-activating protein promotes maturation of dendritic cells with Th1 polarization and improved migration. J Immunol Baltim Md 1950. 2015;194(11):5568. Ramachandran M, Jin C, Yu D, Eriksson F, Essand M. Correction: Vector-encoded Helicobacter pylori neutrophil-activating protein promotes maturation of dendritic cells with Th1 polarization and improved migration. J Immunol Baltim Md 1950. 2015;194(11):5568.
218.
Zurück zum Zitat Codolo G, Facchinello N, Papa N, Bertocco A, Coletta S, Benna C, et al. Macrophage-mediated melanoma reduction after HP-NAP treatment in a zebrafish xenograft model. Int J Mol Sci. 2022;23(3):1644.PubMedPubMedCentralCrossRef Codolo G, Facchinello N, Papa N, Bertocco A, Coletta S, Benna C, et al. Macrophage-mediated melanoma reduction after HP-NAP treatment in a zebrafish xenograft model. Int J Mol Sci. 2022;23(3):1644.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Codolo G, Fassan M, Munari F, Volpe A, Bassi P, Rugge M, et al. HP-NAP inhibits the growth of bladder cancer in mice by activating a cytotoxic Th1 response. Cancer Immunol Immunother. 2012;61(1):31–40.PubMedCrossRef Codolo G, Fassan M, Munari F, Volpe A, Bassi P, Rugge M, et al. HP-NAP inhibits the growth of bladder cancer in mice by activating a cytotoxic Th1 response. Cancer Immunol Immunother. 2012;61(1):31–40.PubMedCrossRef
220.
Zurück zum Zitat Wang T, Liu X, Ji Z, Men Y, Du M, Ding C, et al. Antitumor and immunomodulatory effects of recombinant fusion protein rMBP-NAP through TLR-2 dependent mechanism in tumor bearing mice. Int Immunopharmacol. 2015;29(2):876–83.PubMedCrossRef Wang T, Liu X, Ji Z, Men Y, Du M, Ding C, et al. Antitumor and immunomodulatory effects of recombinant fusion protein rMBP-NAP through TLR-2 dependent mechanism in tumor bearing mice. Int Immunopharmacol. 2015;29(2):876–83.PubMedCrossRef
221.
Zurück zum Zitat Wang T, Du M, Ji Z, Ding C, Wang C, Men Y, et al. Recombinant protein rMBP-NAP restricts tumor progression by triggering antitumor immunity in mouse metastatic lung cancer. Can J Physiol Pharmacol. 2018;96(2):113–9.PubMedCrossRef Wang T, Du M, Ji Z, Ding C, Wang C, Men Y, et al. Recombinant protein rMBP-NAP restricts tumor progression by triggering antitumor immunity in mouse metastatic lung cancer. Can J Physiol Pharmacol. 2018;96(2):113–9.PubMedCrossRef
222.
Zurück zum Zitat Mohabati Mobarez A, Soleimani N, Esmaeili SA, Farhangi B. Nanoparticle-based immunotherapy of breast cancer using recombinant Helicobacter pylori proteins. Eur J Pharm Biopharm. 2020;155:69–76.PubMedCrossRef Mohabati Mobarez A, Soleimani N, Esmaeili SA, Farhangi B. Nanoparticle-based immunotherapy of breast cancer using recombinant Helicobacter pylori proteins. Eur J Pharm Biopharm. 2020;155:69–76.PubMedCrossRef
223.
Zurück zum Zitat Li Y, Jin Y, Zhang J, Pan H, Wu L, Liu D, et al. Recovery of human gut microbiota genomes with third-generation sequencing. Cell Death Dis. 2021;12(6):569.PubMedPubMedCentralCrossRef Li Y, Jin Y, Zhang J, Pan H, Wu L, Liu D, et al. Recovery of human gut microbiota genomes with third-generation sequencing. Cell Death Dis. 2021;12(6):569.PubMedPubMedCentralCrossRef
224.
Zurück zum Zitat Zheng W, Zhao S, Yin Y, Zhang H, Needham DM, Evans ED, et al. High-throughput, single-microbe genomics with strain resolution, applied to a human gut microbiome. Science. 2022;376(6597):eabm1483.PubMedCrossRef Zheng W, Zhao S, Yin Y, Zhang H, Needham DM, Evans ED, et al. High-throughput, single-microbe genomics with strain resolution, applied to a human gut microbiome. Science. 2022;376(6597):eabm1483.PubMedCrossRef
225.
Zurück zum Zitat Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, Wandro S, et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature. 2020;579(7800):567–74.PubMedPubMedCentralCrossRef Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, Wandro S, et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature. 2020;579(7800):567–74.PubMedPubMedCentralCrossRef
226.
Zurück zum Zitat Zhou CB, Pan SY, Jin P, Deng JW, Xue JH, Ma XY, et al. Fecal signatures of Streptococcus anginosus and Streptococcus constellatus for noninvasive screening and early warning of gastric cancer. Gastroenterology. 2022;162(7):1933–1947.e18.PubMedCrossRef Zhou CB, Pan SY, Jin P, Deng JW, Xue JH, Ma XY, et al. Fecal signatures of Streptococcus anginosus and Streptococcus constellatus for noninvasive screening and early warning of gastric cancer. Gastroenterology. 2022;162(7):1933–1947.e18.PubMedCrossRef
227.
Zurück zum Zitat Zhang W, Zhou Y, Fan Y, Cao R, Xu Y, Weng Z, et al. Metal–organic-framework-based hydrogen-release platform for multieffective Helicobacter pylori targeting therapy and intestinal flora protective capabilities. Adv Mater. 2022;34(2):2105738.CrossRef Zhang W, Zhou Y, Fan Y, Cao R, Xu Y, Weng Z, et al. Metal–organic-framework-based hydrogen-release platform for multieffective Helicobacter pylori targeting therapy and intestinal flora protective capabilities. Adv Mater. 2022;34(2):2105738.CrossRef
228.
Zurück zum Zitat Yu J, Guo Z, Yan J, Bu C, Peng C, Li C, et al. Gastric acid-responsive ROS nanogenerators for effective treatment of Helicobacter pylori infection without disrupting homeostasis of intestinal flora. Adv Sci Weinh Baden-Wurtt Ger. 2023;10(20):e2206957. Yu J, Guo Z, Yan J, Bu C, Peng C, Li C, et al. Gastric acid-responsive ROS nanogenerators for effective treatment of Helicobacter pylori infection without disrupting homeostasis of intestinal flora. Adv Sci Weinh Baden-Wurtt Ger. 2023;10(20):e2206957.
229.
Zurück zum Zitat Choudhury A, Ortiz PS, Young M, Mahmud MT, Stoffel RT, Greathouse KL, et al. Control of Helicobacter pylori with engineered probiotics secreting selective guided antimicrobial peptides. Microbiol Spectr. 2023;11(5):e02014–23. Choudhury A, Ortiz PS, Young M, Mahmud MT, Stoffel RT, Greathouse KL, et al. Control of Helicobacter pylori with engineered probiotics secreting selective guided antimicrobial peptides. Microbiol Spectr. 2023;11(5):e02014–23.
Metadaten
Titel
Unveiling the gastric microbiota: implications for gastric carcinogenesis, immune responses, and clinical prospects
verfasst von
Zhiyi Liu
Dachuan Zhang
Siyu Chen
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2024
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-024-03034-7

Weitere Artikel der Ausgabe 1/2024

Journal of Experimental & Clinical Cancer Research 1/2024 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.