Skip to main content

Unravelling the role of intratumoral bacteria in digestive system cancers: current insights and future perspectives

Abstract

Recently, research on the human microbiome, especially concerning the bacteria within the digestive system, has substantially advanced. This exploration has unveiled a complex interplay between microbiota and health, particularly in the context of disease. Evidence suggests that the gut microbiome plays vital roles in digestion, immunity and the synthesis of vitamins and neurotransmitters, highlighting its significance in maintaining overall health. Conversely, disruptions in these microbial communities, termed dysbiosis, have been linked to the pathogenesis of various diseases, including digestive system cancers. These bacteria can influence cancer progression through mechanisms such as DNA damage, modulation of the tumour microenvironment, and effects on the host’s immune response. Changes in the composition and function within the tumours can also impact inflammation, immune response and cancer therapy effectiveness. These findings offer promising avenues for the clinical application of intratumoral bacteria for digestive system cancer treatment, including the potential use of microbial markers for early cancer detection, prognostication and the development of microbiome-targeted therapies to enhance treatment outcomes. This review aims to provide a comprehensive overview of the pivotal roles played by gut microbiome bacteria in the development of digestive system cancers. Additionally, we delve into the specific contributions of intratumoral bacteria to digestive system cancer development, elucidating potential mechanisms and clinical implications. Ultimately, this review underscores the intricate interplay between intratumoral bacteria and digestive system cancers, underscoring the pivotal role of microbiome research in transforming diagnostic, prognostic and therapeutic paradigms for digestive system cancers.

Introduction

Digestive system cancers pose a significant global health challenge due to their high incidence and mortality rates [1,2,3,4,5]. Epidemiological analyses demonstrated that digestive system cancers, encompassing malignancies of the oesophagus, stomach, liver, pancreas, colon and rectum, significantly contribute to the overall health burden worldwide [6,7,8]. Recent advancements in endoscopic techniques and minimally invasive surgery have greatly improved the outcomes and quality of life for individuals diagnosed with early-stage digestive system cancers [9,10,11,12,13]. Simultaneously, the advent of personalised medicine, facilitated by molecular profiling and targeted therapies, has revolutionised the landscape of digestive system cancer management [14,15,16]. Notably, immunotherapeutic interventions, particularly effective for treating mismatch repair-deficient colorectal cancer and hepatocellular carcinoma, offer promising prospects for enhancing patient outcomes [17,18,19,20]. Despite these advances in detection and treatment modalities, managing digestive system cancers continues to encounter challenges [21, 22]. Early detection remains a formidable obstacle, particularly for pancreatic and oesophageal cancers, which often manifest symptoms at advanced stages [23,24,25]. Variability in survival rates across different digestive system cancers reflects the inherent biological diversity, tumour heterogeneity and varied treatment responses, highlighting the complexity of these malignancies [26,27,28,29]. As research progresses, the potential of intratumoral bacteria in digestive system cancer development is increasingly recognised, paving the way for innovative approaches to digestive system cancer therapy (Table 1) [30,31,32].

Table 1 The profiling and functions of intratumoral bacteria in digestive system cancers

The human microbiome constitutes a complex ecosystem of microorganisms inhabiting the body, comprising trillions of bacteria, viruses, fungi and other microbes [33,34,35,36]. The seminal work by the Human Microbiome Project (HMP) has elucidated the extensive diversity and functionality of the microbiome, emphasising its significance in maintaining health and contributing to disease pathogenesis [37, 38]. Particularly dense in the gastrointestinal tract, the microbiome plays a pivotal role in numerous physiological processes, including digestion, immune function, and even brain health and behaviour [39,40,41]. Its impact extends beyond infectious diseases, encompassing a spectrum of human disorders, from metabolic disorders, neurodegenerative diseases, allergies and cardiovascular conditions to various cancers [42,43,44,45,46]. Subsequent research has deepened our understanding of the microbiome’s role in modulating the immune system, with evidence suggesting that dysbiosis, an imbalance in microbial communities, can influence autoimmune disease development and infectious disease response [47,48,49,50].

Moreover, recent advancements in sequencing technologies and microbiome research have unveiled the complex interplay between intratumoral bacteria and cancer, prompting significant interest in their potential roles in oncogenesis and malignancy progression [51,52,53,54,55]. Emerging evidence indicates that intratumoral bacteria, situated within the tumour microenvironment, contribute to the modulation of carcinogenic processes through diverse mechanisms, including carcinogen production, host inflammation modulation and tumour microenvironment alteration [56,57,58]. The complex and dynamic nature of the bacterial ecosystem within the digestive system underscores the pivotal role of intratumoral bacteria in digestive system cancers (Fig. 1) [59]. These bacteria exhibit dual functions in digestive system cancers, serving as promoters of oncogenesis while also acting as protective agents against tumorigenesis, depending on their interactions with host cells and the local microenvironment (Fig. 2) [60,61,62,63]. For instance, the association between Helicobacter pylori (H. pylori) infection and gastric cancer (GC) exemplifies the significant impact of certain intratumoral bacteria on the onset of digestive system malignancies [64]. Conversely, certain bacterial populations within colorectal cancer (CRC) may beneficially modulate immune responses by producing short-chain fatty acids from dietary fibres, suggesting the potential of dietary interventions in CRC management [65,66,67,68]. Importantly. the influence of intratumoral bacteria extends beyond cancer initiation and progression to significantly affect patients’ responses to treatment and overall prognoses [69, 70]. Intratumoral bacteria play a role in modulating the tumour microenvironment, influencing drug metabolism, and interacting with the host immune system, which can either enhance or impair the efficacy of existing therapies. For instance, specific bacteria within CRC have been associated with improved immunotherapy efficacy, potentially due to the influence on the immune microenvironment, favouring T cell activation and infiltration [71]. This observation highlights the potential for microbiome profiling to predict response to immunotherapy and guide treatment decisions. Additionally, certain bacteria have been found to metabolise chemotherapeutic drugs, such as gemcitabine, reducing the drug’s availability and efficacy in pancreatic cancer treatment [72, 73]. This emphasises the importance of considering intratumoral microbiota in designing and administering chemotherapeutic regimens. As research progresses, manipulating intratumoral bacteria through antibiotics, probiotics, or dietary interventions is emerging as a novel strategy to enhance treatment efficacy in multiple digestive system cancers [71, 74,75,76,77]. Furthermore, the composition of intratumoral microbiomes correlates with patient prognosis, with specific microbial signatures associated with either improved or worsened survival outcomes [78, 79]. For example, reduced diversity of intratumoral bacteria in CRC has been associated with a better prognosis, possibly due to the beneficial effects of microbial diversity on immune system regulation [79]. Collectively, these findings underscore the profound impacts of intratumoral bacteria in digestive system cancers, highlighting the implications for integrating microbiome research into digestive system cancer treatment (Table 2) [80,81,82,83].

Fig. 1
figure 1

The pivotal roles of intratumoral bacteria in the pathogenesis of gastrointestinal cancers. Diverse gastrointestinal cancers exhibit significant alterations in the diversity and abundance of bacterial communities, with strong links to cancer progression. The enrichment and colonisation of specific bacterial populations within tumour tissues lead to significant reshaping of the tumour microenvironment. Through various direct and indirect mechanisms, these bacteria contribute to cancer development by promoting chronic inflammation, inducing DNA damage and enhancing cellular proliferation and invasion. Moreover, the bacteria associated with digestive system cancers exert a dual role in cancer dynamics, capable of both driving and impeding tumour progression. Their influence extends across multiple malignant processes, including immune system evasion, persistent inflammation stimulation and genetic instability promotion within host cells

Fig. 2
figure 2

Major characteristic alterations and molecular mechanisms of intratumoral bacteria in digestive system cancers. In digestive system cancers, significant changes in the diversity and abundance of bacterial communities underscore an abnormal bacterial landscape, highlighting their significant roles in cancer development. These aberrant bacterial communities influence cancer progression through a myriad of mechanisms, engaging in both direct and indirect interactions. Directly, bacteria interact with cancer cells or modulate the tumour microenvironment, whereas indirectly, they exert effects through the secretion of virulence factors or the generation of metabolic byproducts. Such interactions significantly affect cancer-associated molecular pathways and cellular processes, thereby playing a critical role in the regulation of tumour progression. Therefore, these altered bacteria are implicated in various malignant processes. They contribute to the proliferation and invasion of tumour cells, facilitate the epithelial-mesenchymal transition (EMT) and regulate inflammation, apoptosis, immune evasion and bile acid metabolism. Furthermore, they influence oxidative stress and DNA damage, underscoring their comprehensive impact on the pathogenesis of digestive system cancers

Table 2 The clinical significance of intratumoral bacteria in digestive system cancers

This review provides a comprehensive analysis of intratumoral bacteria in digestive system cancers, with a specific focus on liver cancer (LC), CRC, GC, pancreatic cancer (PC) and oesophageal cancer (EC). Our objective is to consolidate the diversity of bacteria found within digestive system cancers, elucidate their proposed mechanisms, and discuss implications for digestive system cancer therapy. By exploring the intersection of microbiology and digestive system cancers, this review illuminates the potential of intratumoral bacteria as biomarkers for diagnosis and prognosis, as well as potential targets for innovative therapeutic strategies in digestive system cancers (Fig. 3).

Fig. 3
figure 3

Clinical potential of intratumoral bacteria in digestive system cancers The unique bacterial signatures correlated with various stages and types of digestive system cancers offer novel avenues for utilising certain bacterial species as non-invasive biomarkers, with promise potential as early detection and accurate diagnostic markers of digestive system cancers. In terms of prognosis, the composition of bacterial communities within tumours is intricately linked to a range of clinicopathological features and can profoundly influence survival outcomes. Therapeutically, innovative approaches targeting these bacterial populations and their metabolic outputs have shown promising results. The strategic application of antibiotics, probiotics and faecal microbiota transplantation has been found to significantly inhibit tumour growth. Furthermore, these microbial-based therapies enhance the sensitivity of tumours to conventional chemotherapies and immunotherapies

Roles and mechanisms of intratumoral bacteria in digestive system cancers

Roles and mechanisms of bacteria in LC

The complex relationship between gut microbiota and the development of LC is increasingly evident, influenced by both local and systemic factors. The liver, which receives nutrient-rich blood from the intestine via the portal vein, is a key site for interactions with the gut microbiome and microbe-associated molecular patterns (MAMPs). These MAMPs typically consist of microbial metabolites and byproducts [84]. Extensive research has highlighted the critical role of intestinal permeability and dysbiosis in facilitating the progression of chronic liver diseases to hepatocellular carcinoma (HCC), highlighting bidirectional regulation and interconnection between these factors [85, 86].

Recent studies have identified distinct variations in microbial diversity between patients with liver cirrhosis and those with HCC. Notably, the progression from liver cirrhosis induced by chronic hepatitis B virus (HBV) infection to early-stage HCC is marked by an increase in fecal microbial diversity. This includes a rise in the Phylum Actinobacteria and lipopolysaccharide-producing genera, coupled with a decrease in butyrate-producing genera. A random forest model utilising microbiota with differential expression, validated across 486 faecal samples from diverse regions, achieved an 80.64% area under the curve for early HCC diagnosis, indicating the potential of microbial profiles in non-invasive diagnostics [61]. Additionally, faecal microbial richness is significantly higher in patients with HBV-related HCC compared to healthy controls and patients with non-HBV non-hepatitis C virus (NBNC)-related HCC. Faecal samples from patients with NBNC-related HCC exhibit a higher abundance of Escherichia coli and Shigella and lower concentrations of faecal Bacteroides, Ruminococcus and Lachnoclostridium [87]. Compared to NASH-induced liver cirrhosis without HCC, patients with HCC showed elevated levels of Bacteroides and Ruminococcaceae and decreased levels of Akkermansia [62]. The variable features of specific microbial profiles in patients with HCC offer a promising non-invasive approach based on microbial-based diagnostic tests. Moreover, elevated levels of lipopolysaccharides (LPS), a cell wall component of gram-negative bacteria, have been detected in various HCC cell lines, mouse models and blood samples, confirming the phenomenon of intestinal leakage across different stages of chronic liver disease progression to HCC [88]. Increased intestinal permeability induced by agents like dextran sodium sulfate elevates portal LPS levels and promotes tumour formation in a choline-deficient high-fat diet-induced non-alcoholic steatohepatitis (NASH) mouse model [89]. Furthermore, enhanced intestinal bacterial translocation contributes to chronic liver inflammation through the interaction of MAMPs with pattern recognition receptors (PRRs) on host liver-resident cells (including hepatocytes, human hepatic stellate cells and Kupffer cells), thereby facilitating liver cancer development [88]. Increased LPS levels stimulate both human and mouse hepatic stellate cells to upregulate epiregulin mRNA and protein in a nuclear factor kappa B (NF-κB)-dependent manner, exerting a potent mitogenic effect on hepatocytes [90]. Additionally, activation of the LPS-TLR4 axis results in reduced cleavage of the apoptotic marker caspase-3 mediated by NF-κB, thereby preventing hepatocyte apoptosis [90]. Furthermore, sustained activation of the LPS-TLR4 axis promotes NF-κB activation and weakens reactive oxygen species-induced toxicity, amplifying inflammation-mediated hepatocyte proliferation and tumorigenic response [88]. Additionally, LPS-induced constitutive overexpression of TLR4 directly activates NF-κB signalling in HCC SMMC-772 cells, leading to the activation of the main transcription factors involved in epithelial-mesenchymal transition (EMT) such as Snail [78]. Consequently, Snail induction in SMMC-7721 cells enhances EMT processes and invasion in HCC SMMC-7721 cells. Notably, TLR4 is expressed in the majority of clinical tissue specimens from 106 patients with HCC, and its high expression carries worse prognostic implications, correlating with unfavourable cancer-free or overall survival. TLR4 overexpression is closely associated with poor clinicopathologic characteristics, including cirrhosis, tumour size, margin, vascular invasion, UICC T stage, portal vein thrombosis and tumour thrombus.

Furthermore, dysbiosis has been implicated in mediating liver cancer development through bacterial metabolites. Studies in obesity-associated HCC mouse models have revealed alterations in gut microbiota and increased levels of deoxycholic acid (DCA) levels, promoting the transformation of hepatic stellate cells into a senescence-associated secretory phenotype (SASP). This phenotype releases inflammatory and tumour-promoting factors such as IL-6, CXCL1 and CXCL9. Interestingly, vancomycin treatment, which targets Gram-positive bacteria, attenuated signs of SASP and slowed HCC progression, suggesting that the increase in obesity-related Gram-positive bacteria may facilitate HCC development via the enterohepatic circulation of gut bacterial metabolites. Importantly, the phenomenon of cellular senescence and SASP has also been observed in the HSCs of NASH-related patients with HCC. Moreover, gut bacteria-controlled bile acid metabolism has been implicated in liver antitumor immunosurveillance. The role of Gram-positive bacteria in primary-to-secondary bile acid transformation reduces chenodeoxycholic acid (CDCA) concentration and increases glycolithocholate (GLCA), thereby decreasing CXCL16 expression on liver sinusoidal endothelial cells. This imbalance suppresses CXCR6 + Natural Killer T (NKT) cell accumulation and antitumor activity, thus promoting liver tumour growth [75]. Similar regulatory effects of bile acids CXCL16 expression have been confirmed in human liver sinusoidal endothelial cells and tissue samples. Additionally, dietary soluble fibres and their fermentation products, short-chain fatty acids (SCFA), exhibit complex effects during icteric HCC development. Toll-like receptor 5 deficient (T5KO) mouse models, prone to icteric HCC, demonstrate gut dysbiosis characterised by an increase in fibre-fermenting bacteria and Proteobacteria. A high-fat diet supplemented with inulin in T5KO mouse models leads to gut dysbiosis, early onset of cholestasis, hepatocyte death, neutrophilic inflammation and eventual formation of icteric HCC. Targeted interventions aimed at reducing fibre-fermenting bacteria, inhibiting fermentation, lowering soluble fiber intake, or preventing bile acid reabsorption have significantly reduced HCC incidence. These findings underscore the gut-microbiota-liver axis as a critical target for HCC treatment strategies [76].

Targeting the gut-microbiota-liver axis, with interventions directed at LPS and its receptor TLR4, has shown promise in mitigating HCC development across preclinical studies [91]. Continuous gut decontamination with antibiotics such as ampicillin, neomycin, metronidazole and vancomycin effectively reduces tumour number and size in HCC mouse models. Furthermore, the modulation of the gut microbiome influences responses to chemotherapy and immunomodulatory therapy [84, 92]. Interventions including faecal microbiota transplantation (FMT), TLR antagonists and the use of bile acids and their receptors to protect the intestinal barrier have demonstrated therapeutic potential in various experimental mouse models [85]. However, the translation of these preclinical findings from mice and rats into clinical trials remains a gap, underscoring the therapeutic potential of targeting the gut-microbiota-liver axis in future HCC treatment strategies.

Roles and mechanisms of bacteria in CRC

Increasing large-scale metagenomic studies in human CRC have underscored species-specific microbial compositional and ecological alterations, highlighting the pivotal role of the intestinal microbiota in the tumorigenesis of CRC [68, 93,94,95].

A strong association between gut microbial dysbiosis and CRC pathogenesis implicates the involvement of the gut microbiome in CRC initiation and progression through diverse complex mechanisms [96]. For instance, Fusobacteria, commonly found in the human oral cavity, has been detected in higher abundance in CRC tissues compared to healthy controls, suggesting its role in promoting tumour growth and CRC progression [97,98,99,100]. Recent research indicates that the host factor D-galactose-β (1–3)-N-acetyl-D-galactosamine (Gal-GalNAc) is overexpressed in CRC, facilitating Fusobacterium enrichment in CRC by binding with the galactose-binding lectin, Fap2, on Fusobacterium nucleatum’s surface. This interaction promotes CRC metastasis, emphasising the critical role of host-microbe interactions in CRC progression [101]. Additionally, Fusobacterium nucleatum activates β-catenin signalling by binding its FadA adhesin to E-cadherin on CRC cells, promoting invasion, inflammatory responses and CRC cell proliferation, thereby contributing to CRC oncogenesis [102]. Mechanistically, Fusobacterium nucleatum stimulates the NFκB pathway through the activation of the TLR4/MYD88 cascade in HCT116 cells, leading to the upregulation of downstream target miR21 expression. Subsequently, Fusobacterium nucleatum manipulates miR21-mediated RAS p21 protein activator 1 (RASA1) downregulation to activate the MAPK pathway, enhancing invasiveness and proliferation of CRC cells. Importantly, higher levels of Fusobacterium nucleatum and miR21 are associated with advanced CRC phenotypes, including late T stage, elevated Ki-67 expression and lymphatic invasion, leading to reduced overall survival rates in patients with CRC [79]. These findings suggest a close association between Fusobacterium nucleatum abundance and adverse clinical outcomes in CRC, with implications for prognosis prediction and clinical management.

Furthermore, Fusobacterium nucleatum develops evasion mechanisms by inhibiting the infiltration and cytotoxic activity of Natural Killer (NK) cells within CRC tumours. The interaction between the Fap2 protein of Fusobacterium nucleatum and the inhibitory receptor T cell immunoglobulin and ITIM domain (TIGIT) on human NK cells hampers the activity of NK cells, protecting human colon tumours from immune cell attack [103]. Similarly, studies have demonstrated the causative roles of Bacteroides fragilis in CRC [104, 105]. Enteropathogenic Escherichia coli (EPEC) infection induces the translocation of the EPEC-secreted effector protein EspF to the host cell membrane and mitochondria, depleting DNA mismatch repair (MMR) proteins in host HT29 and SW480 cells and increasing the spontaneous mutation frequency, contributing to CRC development [106]. Additionally, Enterotoxigenic Bacteroides fragilis (ETBF) secrets Bacteroides fragilis toxin (BFT) to activate colonic epithelial cells via STAT3/NF-κB/IL-17R signalling, leading to Th17 cell response and CXCR2-expressing polymorphonuclear immature myeloid cell recruitment and consequently inducing a pro-carcinogenic inflammatory response and myeloid-cell-dependent distal colon tumorigenesis [71]. These findings demonstrate that bacteria can disrupt the tumour’s immune microenvironment, facilitating CRC formation.

Roles and mechanisms of bacteria in GC

GC presents a significant global health challenge, with H. pylori infection identified as the most unequivocal risk factor, implicated in nearly all GC cases and instigating pathogenic mechanisms crucial to disease progression [107,108,109,110,111,112,113,114]. These mechanisms include initiating a chronic inflammatory response, manipulation of the host’s innate immune system, inducing DNA damage and dysregulating autophagy pathways [115,116,117,118,119]. H. pylori colonises the gastric mucosa of over half of the global population, yet only a subset develops GC, suggesting a complex interplay of bacterial, host and environmental factors. For example, H. pylori infection initiates a persistent inflammatory response in the gastric mucosa, characterised by the recruitment of inflammatory cells and the release of pro-inflammatory cytokines such as IL-1β, IL-8 and TNF-α. This inflammatory milieu fosters a conducive environment for neoplastic transformation, seen as a precursor to atrophic gastritis, a well-established GC risk factor [120,121,122]. Moreover, H. pylori secret virulence factors, notably cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA), that manipulate host cell proliferation and apoptosis [123,124,125]. CagA, upon injection into host cells, undergoes phosphorylation and interacts with multiple signal transduction pathways, leading to dysregulated cell signalling, proliferation and increased mutations [126,127,128,129]. H. pylori infection is also associated with increased DNA damage within gastric epithelial cells due to the production of reactive oxygen and nitrogen species inducing oxidative stress, resulting in genomic instability in key genes involved in gastric carcinogenesis [130,131,132,133]. Beyond H. pylori, advancements in polymerase chain reaction (PCR) technology and metagenomics have revealed a complex gastric microbiota potentially contributing to GC development [134,135,136,137]. Studies also explore H. pylori’s role in modulating the gastric microbiome, suggesting H. pylori-induced dysbiosis may further contribute to carcinogenesis [138]. Consistently, reduced gastric microbial diversity is observed from atrophic gastritis (AG) to intestinal metaplasia (IM), and finally to GC, suggesting a dysbiotic shift favouring oncogenic processes [139,140,141,142]. Conversely, some findings show increased microbial evenness and diversity in patients with GC compared to those with other gastritis forms, indicating a complex relationship between microbial diversity and cancer development [143, 144]. Recent research focusing on microbial profiling from healthy gastric mucosa to GC in individuals has identified a significant decrease in microbial diversity and richness within peritumoral and tumoral microhabitats [145]. Moreover, the combination of specific dysregulated bacterial clusters has been identified with a strong discriminatory capability for distinguishing between gastritis and GC, highlighting their potential as biomarkers for early detection. Additionally, variations in gastric microbiota among patients with GC have prognostic significance, with certain bacteria like Fusobacterium and Prevotella significantly associated with poorer overall survival rates, suggesting their potential as prognostic targets [146]. Investigation into the gastric microbiome’s role in GC underscores the need for a comprehensive understanding of cancer development, considering not only pathogenic bacteria like H. pylori but also the broader microbial ecosystem. This approach holds promise for innovative diagnostic, prognostic and therapeutic strategies, potentially revolutionising GC management through targeted modulation of the gastric microbiota [136, 147,148,149].

Roles and mechanisms of bacteria in PC

PC, notorious for its dismal prognosis and rapid progression, has recently been associated with a distinct intratumoral microbiome [150,151,152,153]. Studies indicate that bacteria residing within the PC tumour microenvironment significantly contribute to cancer development, immune evasion and treatment resistance [154,155,156]. Pushalkar et al. pioneered the discovery of a unique bacterial population within pancreatic tumours, challenging the previous notion of the pancreas as a sterile environment [77]. Their research revealed a substantially richer microbiome in cancerous pancreas compared to normal pancreatic tissue in both mice and humans. This microbiome plays a crucial role in shaping the tumour’s immune landscape to foster immune tolerance. The enriched microbial ecosystem activates TLR on macrophages, leading to an increase in myeloid-derived suppressor cells (MDSCs), suppression of Th1 polarisation of CD4 + T cells and activation of CD8 + T cells. These changes diminish the effectiveness of immune checkpoint therapies by inhibiting PD-1 expression. Depleting the tumour-associated microbiome with oral antibiotics has shown promise in reversing this microbiome-induced immune tolerance, reducing tumour burden by approximately 50% and enhancing the efficacy of PD-1-based immunotherapies [77]. These findings underscore the complex interplay between the tumour microbiome and the host immune system in PC, highlighting the potential of microbiome-targeted therapies as adjuncts to immunotherapy. Previous research also highlights the intricate impact of intratumoral bacteria on chemotherapy efficacy. Bacterial species within PC, particularly Gammaproteobacteria, metabolise gemcitabine, a standard PC chemotherapy drug, into its inactive form by expressing a specific form of the bacterial enzyme cytidine deaminase (CDDL), contributing to chemoresistance [72]. Additionally, recent studies elucidate a complex interplay between microbiome-derived metabolites and the innate immune response within the tumour microenvironment, ultimately influencing chemotherapy efficacy in PC. A significant correlation was observed between the levels of the microbiota-derived tryptophan metabolite indole-3-acetic acid (3-IAA) and treatment outcomes in two independent PC cohorts, with higher concentrations of 3-IAA enriched in patients who responded to chemotherapy [157]. Neutrophil-derived myeloperoxidase plays a pivotal role in this context by oxidising 3-IAA. Combined with chemotherapy, this oxidation process induces the downregulation of reactive oxygen species (ROS)-degrading enzymes, specifically glutathione peroxidase 3 (GPx3) and GPx7, impairing cancer cell metabolic adaptability and hindering their proliferative capacity. While some bacteria contribute to immune evasion and therapy resistance, others can enhance immune surveillance and suppression of tumour growth [154]. Beneficial microbiota also exists within PC tissues. 16 S rRNA gene sequencing analysis was conducted using two independent cohorts of patients with PC who survived for more than five years (long-term survivors, LTS) and those who did not survive for five years. The results revealed a higher microbial diversity in LTS tissues. A univariate Cox proportional hazards model further demonstrated that patients with higher microbial diversity within PC exhibited significantly prolonged overall survival. A combination of microbial genera consisting of Pseudoxanthomonas, Saccharopolyspora and Streptomyces within the tumour microbiome has been identified as a strong predictor of favourable long-term survival outcomes in patients with PC, thereby offering new avenues for PC therapeutic interventions. Human-to-mouse FMT experiments also reveal that modulating these three bacterial genera within PC tumours enhances CD8 + T cell recruitment and immune response, inhibiting tumour growth. This mechanism suggests a direct link between specific components of the tumour microbiome and the activation of effective antitumor immunity [63]. Additionally, tumour-microbiome crosstalk is increasingly recognised as pivotal in regulating PC tumorigenesis. Single-cell analysis of host-microbiome interactions (SAHMI), drawing on data from two large, independent scRNA-seq cohorts of pancreatic ductal adenocarcinoma (PDA), has highlighted the significant role of somatic-cell-associated bacteria in association with tumour cells, influencing cell motility and immune signalling pathways [158]. Furthermore, the presence of cell-associated bacteria within PC across multiple independent datasets emphasises their potential as valuable prognostic markers. High intratumoral levels of Fusobacterium nucleatum in PC have also been linked to shorter survival in patients. Fusobacterium nucleatum infection induces both normal pancreatic epithelial cells and PC cells, driven by Fap2, to secrete elevated levels of cytokines such as granulocyte-macrophage colony-stimulating factor (GM-CSF), chemokine (C-X-C motif) ligand 1 (CXCL1), Interleukin-8 (IL-8) and macrophage inflammatory protein-3α (MIP-3α). These cytokines, through host autocrine and paracrine signalling, enhance phenotypes in PC BxPC3 and Panc1 cells associated with tumour progression, including cell proliferation, migration and invasion, thus contributing to the aggressive nature of PC [159].

The findings underscore highlights the potential of microbiome-targeted therapies as complementary to conventional cancer treatments. By selectively promoting the growth of beneficial bacteria or reducing harmful microbial populations, it may be possible to improve patient outcomes in PC.

Roles and mechanisms of bacteria in EC

Recent investigations into the oesophageal microbiome have revealed significant alterations in patients with EC, shedding light on the potential influence of the microbiome on the development of this malignancy [160,161,162]. Comparisons between healthy individuals, patients with Barrett’s oesophagus (BE) and those diagnosed with EC have shown clear distinct microbial compositions, highlighting the dynamic changes accompanying disease progression. Particularly noteworthy is the observed decrease in microbial diversity among patients with EC, characterised by a shift from the abundance of Veillonella and Streptococcus to the predominance of Lactobacillus [163]. This shift is believed to significantly impact the tumour microenvironment, potentially contributing to tumorigenesis. Further analysis of microbial diversity in EC tissues, conducted using 16 S rDNA sequencing, has shown a significant decrease compared to nontumor tissues, accompanied by an increase in Fusobacterium abundance and a corresponding decrease in Streptococcus [164]. Innovative research utilising microbial prediction models has identified the combination of Streptococcus and Neisseria as effective predictors of EC progression and its precancerous lesions, demonstrating high diagnostic performance, with an area under the curve (AUC) value of 0.738, and potential for early detection and progression monitoring [165]. Subsequent studies involving 325 resected EC specimens have revealed significantly higher levels of Fusobacterium nucleatum DNA in EC tissues compared to normal oesophageal mucosa. The presence of Fusobacterium nucleatum DNA strongly correlated with advanced tumour stages and shorter cancer-specific survival, indicating its potential as a prognostic biomarker in EC [166]. Additionally, a significant correlation has been observed between Fusobacterium nucleatum and the chemokine CCL20, suggesting a potential mechanism through which Fusobacterium nucleatum may promote tumour aggressiveness [166]. Porphyromonas gingivalis has also been identified as more prevalent in EC tissues, with its abundance varying across different stages of EC [167, 168]. Its presence is often associated with poor tumour differentiation, advanced stages, metastasis and reduced survival outcomes, further emphasising its prognostic significance. Emerging evidence supports the role of the oesophageal microbiome in modulating responses to immune checkpoint inhibitors in EC, with different microbial compositions correlating with varying responses to radiotherapy and chemotherapy [161, 169, 170]. These insights underscore the intricate relationship between intratumoral bacteria and the host immune system in EC, highlighting the potential of microbiome-targeted therapies to complement traditional cancer treatments and improve therapeutic efficacy and patient outcomes [171, 172].

Conclusion

The intricate interactions of bacteria within digestive system cancers present a rapidly expanding area of research at the intersection of microbiology and oncology. Intratumoral bacteria contribute to digestive system cancer pathogenesis through direct and indirect interactions, influencing cancer cell proliferation, genetic and epigenetic alterations, and the tumour microenvironment conducive to tumour growth and metastasis. This review provides a comprehensive summary of the current understanding of how intratumoral bacteria shape the pathogenesis, diagnosis and treatment of digestive system cancers. By examining recent studies, we elucidate the distinct microbial profiles associated with various digestive system cancers and their roles in inflammation promotion, immune response modulation and tumour microenvironment alteration. We further explore the diagnostic and prognostic potential of microbial signatures in digestive system tumours, indicating their value in predicting disease progression and treatment outcomes. Specific bacterial signatures within tumours have been linked to disease stage, treatment responses and patient survival, suggesting potential applications in precision medicine. Nevertheless, capitalizing on the therapeutic possibilities of targeting these microbial communities poses significant challenges, necessitating a delicate balance between disrupting pathogenic interactions and preserving beneficial host-microbiome relationships.

This review underscores several key limitations in the current research, primarily the associative rather than causative understanding of intratumoral bacteria’s roles in digestive system cancers, and the microbiome’s dynamic responses to disease progression and treatment. Future studies should focus on elucidating the mechanistic foundations of how intratumoral bacteria influence the pathogenesis and progression of digestive system cancers. Advanced models enabling real-time study of tumour-microbiome interactions in situ will be essential. Moreover, exploring the therapeutic potential of modulating the tumour microbiome through targeted antibiotics, probiotic supplementation or FMT holds promise for improving cancer treatment outcomes.

In summary, investigating intratumoral bacteria in digestive system cancers offers new insights into tumour biology, with significant implications for diagnosis, prognostication and therapy. As the field progresses, employing multidisciplinary approaches that integrate microbiology, oncology and immunology will be crucial in fully harnessing the potential of microbiome-targeted therapies in cancer management.

Data availability

Not applicable.

Abbreviations

MDSCs:

myeloid-derived suppressor cells

HMP:

Human Microbiome Project

H:

pylori Helicobacter pylori

GC:

gastric cancer

CRC:

colorectal cancer

LC:

liver cancer

HCC:

hepatocellular carcinoma

PC:

pancreatic cancer

EC:

oesophageal cancer

MAMPs:

microbe-associated molecular patterns

HBV:

hepatitis B virus

NBNC:

non-HBV non-hepatitis C virus

LPS:

lipopolysaccharides

PRRs:

pattern recognition receptors

NF-κB:

nuclear factor kappa B

EMT:

epithelial-mesenchymal transition

DCA:

deoxycholic acid

SASP:

senescence-associated secretory phenotype

CDCA:

chenodeoxycholic acid

GLCA:

glycolithocholate

NKT:

Natural Killer T

SCFA:

short-chain fatty acids

T5KO:

Toll-like receptor 5 deficient

FMT:

faecal microbiota transplantation

Gal-GalNAc:

D-galactose-β (1–3)-N-acetyl-D-galactosamine

RASA1:

RAS p21 protein activator 1

TIGIT T:

cell immunoglobulin and ITIM domain

EPEC:

Enteropathogenic Escherichia coli

MMR:

mismatch repair

ETBF:

Enterotoxigenic Bacteroides fragilis

References

  1. Huang J, Lucero-Prisno DE 3rd, Zhang L, Xu W, Wong SH, Ng SC, et al. Updated epidemiology of gastrointestinal cancers in East Asia. Nat Rev Gastroenterol Hepatol. 2023;20:271–87. https://doi.org/10.1038/s41575-022-00726-3.

    Article  PubMed  Google Scholar 

  2. Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA et al. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology. 2020;159:335 – 49.e15. https://doi.org/10.1053/j.gastro.2020.02.068.

  3. Wang S, Zheng R, Li J, Zeng H, Li L, Chen R, et al. Global, regional, and national lifetime risks of developing and dying from gastrointestinal cancers in 185 countries: a population-based systematic analysis of GLOBOCAN. Lancet Gastroenterol Hepatol. 2024;9:229–37. https://doi.org/10.1016/s2468-1253(23)00366-7.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Yu Z, Bai X, Zhou R, Ruan G, Guo M, Han W, et al. Differences in the incidence and mortality of digestive cancer between Global Cancer Observatory 2020 and global burden of Disease 2019. Int J Cancer. 2024;154:615–25. https://doi.org/10.1002/ijc.34740.

    Article  CAS  PubMed  Google Scholar 

  5. Ma C, Congly SE, Chyou DE, Ross-Driscoll K, Forbes N, Tsang ES et al. Factors Associated With Geographic Disparities in Gastrointestinal Cancer Mortality in the United States. Gastroenterology. 2022;163:437 – 48.e1. https://doi.org/10.1053/j.gastro.2022.04.019.

  6. Murphy CC, Tavakkoli A, Wani S, Singal AG. Pandemic-related changes in incidence and mortality rates of gastrointestinal cancers during 2020. Am J Gastroenterol. 2024;119:382–7. https://doi.org/10.14309/ajg.0000000000002526.

    Article  PubMed  Google Scholar 

  7. Lu L, Mullins CS, Schafmayer C, Zeißig S, Linnebacher M. A global assessment of recent trends in gastrointestinal cancer and lifestyle-associated risk factors. Cancer Commun (Lond). 2021;41:1137–51. https://doi.org/10.1002/cac2.12220.

    Article  PubMed  Google Scholar 

  8. 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. https://doi.org/10.3389/fimmu.2020.01387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lagergren J, Lagergren P. Recent developments in esophageal adenocarcinoma. CA Cancer J Clin. 2013;63:232–48. https://doi.org/10.3322/caac.21185.

    Article  PubMed  Google Scholar 

  10. 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;8:1251–61. https://doi.org/10.1158/2326-6066.Cir-19-1014.

    Article  PubMed  Google Scholar 

  11. Temel JS, Greer JA, El-Jawahri A, Pirl WF, Park ER, Jackson VA, et al. Effects of Early Integrated Palliative Care in patients with lung and GI Cancer: a Randomized Clinical Trial. J Clin Oncol. 2017;35:834–41. https://doi.org/10.1200/jco.2016.70.5046.

    Article  PubMed  Google Scholar 

  12. Xing S, Zhu Y, You Y, Wang S, Wang H, Ning M, et al. Cell-free RNA for the liquid biopsy of gastrointestinal cancer. Wiley Interdiscip Rev RNA. 2023;14:e1791. https://doi.org/10.1002/wrna.1791.

    Article  CAS  PubMed  Google Scholar 

  13. Kurumi H, Yamaguchi N, Isomoto H. Appropriate interval of surveillance endoscopy for early detection of gastric cancer. Dig Endosc. 2023;35:603–5. https://doi.org/10.1111/den.14571.

    Article  PubMed  Google Scholar 

  14. Long J, Lin J, Wang A, Wu L, Zheng Y, Yang X, et al. PD-1/PD-L blockade in gastrointestinal cancers: lessons learned and the road toward precision immunotherapy. J Hematol Oncol. 2017;10:146. https://doi.org/10.1186/s13045-017-0511-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cencioni C, Trestini I, Piro G, Bria E, Tortora G, Carbone C, et al. Gastrointestinal Cancer Patient Nutritional Management: from specific needs to Novel Epigenetic Dietary approaches. Nutrients. 2022;14. https://doi.org/10.3390/nu14081542.

  16. Zhou J, Chen L, Chen L, Zhang Y, Yuan Y. Emerging role of nanoparticles in the diagnostic imaging of gastrointestinal cancer. Semin Cancer Biol. 2022;86:580–94. https://doi.org/10.1016/j.semcancer.2022.04.009.

    Article  CAS  PubMed  Google Scholar 

  17. Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet. 2018;391:1301–14. https://doi.org/10.1016/s0140-6736(18)30010-2.

    Article  PubMed  Google Scholar 

  18. Abolarinwa BA, Ibrahim RB, Huang YH. Conceptual development of immunotherapeutic approaches to gastrointestinal Cancer. Int J Mol Sci. 2019;20. https://doi.org/10.3390/ijms20184624.

  19. Koustas E, Trifylli EM, Sarantis P, Papadopoulos N, Karapedi E, Aloizos G, et al. Immunotherapy as a therapeutic strategy for gastrointestinal Cancer-current treatment options and future perspectives. Int J Mol Sci. 2022;23. https://doi.org/10.3390/ijms23126664.

  20. Zhang Y, Xu J, Zhang N, Chen M, Wang H, Zhu D. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett. 2019;458:123–35. https://doi.org/10.1016/j.canlet.2019.05.017.

    Article  CAS  PubMed  Google Scholar 

  21. Anwanwan D, Singh SK, Singh S, Saikam V, Singh R. Challenges in liver cancer and possible treatment approaches. Biochim Biophys Acta Rev Cancer. 2020;1873:188314. https://doi.org/10.1016/j.bbcan.2019.188314.

    Article  CAS  PubMed  Google Scholar 

  22. Halama N, Haberkorn U. The unmet needs of the diagnosis, staging, and treatment of gastrointestinal tumors. Semin Nucl Med. 2020;50:389–98. https://doi.org/10.1053/j.semnuclmed.2020.06.003.

    Article  PubMed  Google Scholar 

  23. Guo X, Peng Y, Song Q, Wei J, Wang X, Ru Y, et al. A Liquid Biopsy Signature for the early detection of gastric Cancer in patients. Gastroenterology. 2023;165:402–e1313. https://doi.org/10.1053/j.gastro.2023.02.044.

    Article  CAS  PubMed  Google Scholar 

  24. Kandimalla R, Xu J, Link A, Matsuyama T, Yamamura K, Parker MI, et al. EpiPanGI Dx: a cell-free DNA methylation fingerprint for the early detection of gastrointestinal cancers. Clin Cancer Res. 2021;27:6135–44. https://doi.org/10.1158/1078-0432.Ccr-21-1982.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hoare M, Fitzgerald RC. What is the current status of multicancer early detection tests and upper gastrointestinal cancer? Lancet Gastroenterol Hepatol. 2023;8:1065–6. https://doi.org/10.1016/s2468-1253(23)00249-2.

    Article  PubMed  Google Scholar 

  26. Zhao Y, Li ZX, Zhu YJ, Fu J, Zhao XF, Zhang YN, et al. Single-cell transcriptome analysis uncovers Intratumoral Heterogeneity and underlying mechanisms for Drug Resistance in Hepatobiliary Tumor Organoids. Adv Sci (Weinh). 2021;8:e2003897. https://doi.org/10.1002/advs.202003897.

    Article  CAS  PubMed  Google Scholar 

  27. Adam RS, Blomberg I, Ten Hoorn S, Bijlsma MF, Vermeulen L. The recurring features of molecular subtypes in distinct gastrointestinal malignancies-A systematic review. Crit Rev Oncol Hematol. 2021;164:103428. https://doi.org/10.1016/j.critrevonc.2021.103428.

    Article  PubMed  Google Scholar 

  28. Hakuno SK, Michiels E, Kuhlemaijer EB, Rooman I, Hawinkels L, Slingerland M. Multicellular modelling of difficult-to-treat gastrointestinal cancers: current possibilities and challenges. Int J Mol Sci. 2022;23. https://doi.org/10.3390/ijms23063147.

  29. Parikh AR, Leshchiner I, Elagina L, Goyal L, Levovitz C, Siravegna G, et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat Med. 2019;25:1415–21. https://doi.org/10.1038/s41591-019-0561-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Garrett WS. Cancer and the microbiota. Science. 2015;348:80–6. https://doi.org/10.1126/science.aaa4972.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Martinelli S, Lamminpää I, Dübüş EN, Sarıkaya D, Niccolai E. Synergistic strategies for gastrointestinal Cancer Care: unveiling the benefits of Immunonutrition and Microbiota Modulation. Nutrients. 2023;15. https://doi.org/10.3390/nu15204408.

  32. Saus E, Iraola-Guzmán S, Willis JR, Brunet-Vega A, Gabaldón T. Microbiome and colorectal cancer: roles in carcinogenesis and clinical potential. Mol Aspects Med. 2019;69:93–106. https://doi.org/10.1016/j.mam.2019.05.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, et al. Microbiota in health and diseases. Signal Transduct Target Ther. 2022;7:135. https://doi.org/10.1038/s41392-022-00974-4.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Dominguez-Bello MG, Godoy-Vitorino F, Knight R, Blaser MJ. Role of the microbiome in human development. Gut. 2019;68:1108–14. https://doi.org/10.1136/gutjnl-2018-317503.

    Article  CAS  PubMed  Google Scholar 

  35. VanEvery H, Franzosa EA, Nguyen LH, Huttenhower C. Microbiome epidemiology and association studies in human health. Nat Rev Genet. 2023;24:109–24. https://doi.org/10.1038/s41576-022-00529-x.

    Article  CAS  PubMed  Google Scholar 

  36. Coburn B, Guttman DS. The human microbiome. CMAJ. 2015;187:825. https://doi.org/10.1503/cmaj.141072.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449:804–10. https://doi.org/10.1038/nature06244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. The Integrative Human Microbiome Project. Nature. 2019;569:641–8. https://doi.org/10.1038/s41586-019-1238-8.

    Article  CAS  Google Scholar 

  39. Cai J, Sun L, Gonzalez FJ. Gut microbiota-derived bile acids in intestinal immunity, inflammation, and tumorigenesis. Cell Host Microbe. 2022;30:289–300. https://doi.org/10.1016/j.chom.2022.02.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Yang G, Wei J, Liu P, Zhang Q, Tian Y, Hou G, et al. Role of the gut microbiota in type 2 diabetes and related diseases. Metabolism. 2021;117:154712. https://doi.org/10.1016/j.metabol.2021.154712.

    Article  CAS  PubMed  Google Scholar 

  41. Bäumler AJ, Sperandio V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature. 2016;535:85–93. https://doi.org/10.1038/nature18849.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Adelman MW, Woodworth MH, Langelier C, Busch LM, Kempker JA, Kraft CS, et al. The gut microbiome’s role in the development, maintenance, and outcomes of sepsis. Crit Care. 2020;24:278. https://doi.org/10.1186/s13054-020-02989-1.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Clemente JC, Manasson J, Scher JU. The role of the gut microbiome in systemic inflammatory disease. BMJ. 2018;360:j5145. https://doi.org/10.1136/bmj.j5145.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Mou Y, Du Y, Zhou L, Yue J, Hu X, Liu Y, et al. Gut microbiota interact with the Brain through systemic chronic inflammation: implications on Neuroinflammation, Neurodegeneration, and aging. Front Immunol. 2022;13:796288. https://doi.org/10.3389/fimmu.2022.796288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. 2017;2:135–43. https://doi.org/10.1016/s2468-1253(16)30119-4.

    Article  PubMed  Google Scholar 

  46. Boulangé CL, Neves AL, Chilloux J, Nicholson JK, Dumas ME. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016;8:42. https://doi.org/10.1186/s13073-016-0303-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121–41. https://doi.org/10.1016/j.cell.2014.03.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Shim JA, Ryu JH, Jo Y, Hong C. The role of gut microbiota in T cell immunity and immune mediated disorders. Int J Biol Sci. 2023;19:1178–91. https://doi.org/10.7150/ijbs.79430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Koboziev I, Reinoso Webb C, Furr KL, Grisham MB. Role of the enteric microbiota in intestinal homeostasis and inflammation. Free Radic Biol Med. 2014;68:122–33. https://doi.org/10.1016/j.freeradbiomed.2013.11.008.

    Article  CAS  PubMed  Google Scholar 

  50. Annunziata G, Sureda A, Orhan IE, Battino M, Arnone A, Jiménez-García M, et al. The neuroprotective effects of polyphenols, their role in innate immunity and the interplay with the microbiota. Neurosci Biobehav Rev. 2021;128:437–53. https://doi.org/10.1016/j.neubiorev.2021.07.004.

    Article  CAS  PubMed  Google Scholar 

  51. Cullin N, Azevedo Antunes C, Straussman R, Stein-Thoeringer CK, Elinav E. Microbiome and cancer. Cancer Cell. 2021;39:1317–41. https://doi.org/10.1016/j.ccell.2021.08.006.

    Article  CAS  PubMed  Google Scholar 

  52. El Tekle G, Garrett WS. Bacteria in cancer initiation, promotion and progression. Nat Rev Cancer. 2023;23:600–18. https://doi.org/10.1038/s41568-023-00594-2.

    Article  CAS  PubMed  Google Scholar 

  53. Rayan M, Sayed TS, Hussein OJ, Therachiyil L, Maayah ZH, Maccalli C, et al. Unlocking the secrets: exploring the influence of the aryl hydrocarbon receptor and microbiome on cancer development. Cell Mol Biol Lett. 2024;29:33. https://doi.org/10.1186/s11658-024-00538-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang Z, Sun W, Hua R, Wang Y, Li Y, Zhang H. Promising dawn in tumor microenvironment therapy: engineering oral bacteria. Int J Oral Sci. 2024;16:24. https://doi.org/10.1038/s41368-024-00282-3.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 2020;368:973–80. https://doi.org/10.1126/science.aay9189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Dohlman AB, Arguijo Mendoza D, Ding S, Gao M, Dressman H, Iliev ID, et al. The cancer microbiome atlas: a pan-cancer comparative analysis to distinguish tissue-resident microbiota from contaminants. Cell Host Microbe. 2021;29:281–e985. https://doi.org/10.1016/j.chom.2020.12.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yang L, Li A, Wang Y, Zhang Y. Intratumoral microbiota: roles in cancer initiation, development and therapeutic efficacy. Signal Transduct Target Ther. 2023;8:35. https://doi.org/10.1038/s41392-022-01304-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Xue C, Chu Q, Zheng Q, Yuan X, Su Y, Bao Z, et al. Current understanding of the intratumoral microbiome in various tumors. Cell Rep Med. 2023;4:100884. https://doi.org/10.1016/j.xcrm.2022.100884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human gut microbiota and gastrointestinal Cancer. Genomics Proteom Bioinf. 2018;16:33–49. https://doi.org/10.1016/j.gpb.2017.06.002.

    Article  CAS  Google Scholar 

  60. Achiwa K, Ishigami M, Ishizu Y, Kuzuya T, Honda T, Hayashi K, et al. DSS colitis promotes tumorigenesis and fibrogenesis in a choline-deficient high-fat diet-induced NASH mouse model. Biochem Biophys Res Commun. 2016;470:15–21. https://doi.org/10.1016/j.bbrc.2015.12.012.

    Article  CAS  PubMed  Google Scholar 

  61. Ren Z, Li A, Jiang J, Zhou L, Yu Z, Lu H, et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut. 2019;68:1014–23. https://doi.org/10.1136/gutjnl-2017-315084.

    Article  CAS  PubMed  Google Scholar 

  62. Ponziani FR, Bhoori S, Castelli C, Putignani L, Rivoltini L, Del Chierico F, et al. Hepatocellular Carcinoma is Associated with Gut Microbiota Profile and inflammation in nonalcoholic fatty liver disease. Hepatology. 2019;69:107–20. https://doi.org/10.1002/hep.30036.

    Article  CAS  PubMed  Google Scholar 

  63. Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer outcomes. Cell. 2019;178:795–e80612. https://doi.org/10.1016/j.cell.2019.07.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Polk DB, Peek RM. Jr. Helicobacter pylori: gastric cancer and beyond. Nat Rev Cancer. 2010;10:403–14. https://doi.org/10.1038/nrc2857.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014;12:661–72. https://doi.org/10.1038/nrmicro3344.

    Article  CAS  PubMed  Google Scholar 

  66. Tsoi H, Chu ESH, Zhang X, Sheng J, Nakatsu G, Ng SC, et al. Peptostreptococcus anaerobius induces intracellular cholesterol biosynthesis in Colon cells to Induce Proliferation and causes dysplasia in mice. Gastroenterology. 2017;152:1419–33. https://doi.org/10.1053/j.gastro.2017.01.009.

    Article  PubMed  Google Scholar 

  67. Zou S, Yang C, Zhang J, Zhong D, Meng M, Zhang L, et al. Multi-omic profiling reveals associations between the gut microbiome, host genome and transcriptome in patients with colorectal cancer. J Transl Med. 2024;22:175. https://doi.org/10.1186/s12967-024-04984-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Clay SL, Fonseca-Pereira D, Garrett WS. Colorectal cancer: the facts in the case of the microbiota. J Clin Invest. 2022;132. https://doi.org/10.1172/jci155101.

  69. Fan JX, Niu MT, Qin YT, Sun YX, Zhang XZ. Progress of engineered bacteria for tumor therapy. Adv Drug Deliv Rev. 2022;185:114296. https://doi.org/10.1016/j.addr.2022.114296.

    Article  CAS  PubMed  Google Scholar 

  70. Duong MT, Qin Y, You SH, Min JJ. Bacteria-cancer interactions: bacteria-based cancer therapy. Exp Mol Med. 2019;51:1–15. https://doi.org/10.1038/s12276-019-0297-0.

    Article  CAS  PubMed  Google Scholar 

  71. Chung L, Thiele Orberg E, 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;23:203–e145. https://doi.org/10.1016/j.chom.2018.01.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357:1156–60. https://doi.org/10.1126/science.aah5043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Wong-Rolle A, Wei HK, Zhao C, Jin C. Unexpected guests in the tumor microenvironment: microbiome in cancer. Protein Cell. 2021;12:426–35. https://doi.org/10.1007/s13238-020-00813-8.

    Article  PubMed  Google Scholar 

  74. Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499:97–101. https://doi.org/10.1038/nature12347.

    Article  CAS  PubMed  Google Scholar 

  75. Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018;360. https://doi.org/10.1126/science.aan5931.

  76. Singh V, Yeoh BS, Chassaing B, Xiao X, Saha P, Aguilera Olvera R et al. Dysregulated Microbial Fermentation of Soluble Fiber Induces Cholestatic Liver Cancer. Cell. 2018;175:679 – 94.e22. https://doi.org/10.1016/j.cell.2018.09.004.

  77. Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, et al. The pancreatic Cancer Microbiome promotes oncogenesis by induction of Innate and Adaptive Immune suppression. Cancer Discov. 2018;8:403–16. https://doi.org/10.1158/2159-8290.Cd-17-1134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Jing YY, Han ZP, Sun K, Zhang SS, Hou J, Liu Y, et al. Toll-like receptor 4 signaling promotes epithelial-mesenchymal transition in human hepatocellular carcinoma induced by lipopolysaccharide. BMC Med. 2012;10:98. https://doi.org/10.1186/1741-7015-10-98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. 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;152:851–e6624. https://doi.org/10.1053/j.gastro.2016.11.018.

    Article  CAS  PubMed  Google Scholar 

  80. Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, Knight R. The microbiome and human cancer. Science. 2021;371. https://doi.org/10.1126/science.abc4552.

  81. Hussain A, Patwekar U, Mongad DS, Shouche YS. Strategizing the human microbiome for small molecules: approaches and perspectives. Drug Discov Today. 2023;28:103459. https://doi.org/10.1016/j.drudis.2022.103459.

    Article  CAS  PubMed  Google Scholar 

  82. Sharma AK. Special issue: mining human microbiome bringing newer paradigms to anticancer therapeutics. Semin Cancer Biol. 2021;70:1–2. https://doi.org/10.1016/j.semcancer.2020.08.008.

    Article  PubMed  Google Scholar 

  83. Kustrimovic N, Bombelli R, Baci D, Mortara L. Microbiome and prostate Cancer: a Novel Target for Prevention and Treatment. Int J Mol Sci. 2023;24. https://doi.org/10.3390/ijms24021511.

  84. Schwabe RF, Greten TF. Gut microbiome in HCC - mechanisms, diagnosis and therapy. J Hepatol. 2020;72:230–8. https://doi.org/10.1016/j.jhep.2019.08.016.

    Article  CAS  PubMed  Google Scholar 

  85. Yu LX, Schwabe RF. The gut microbiome and liver cancer: mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol. 2017;14:527–39. https://doi.org/10.1038/nrgastro.2017.72.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology. 2014;146:1513–24. https://doi.org/10.1053/j.gastro.2014.01.020.

    Article  CAS  PubMed  Google Scholar 

  87. Liu Q, Li F, Zhuang Y, Xu J, Wang J, Mao X, et al. Alteration in gut microbiota associated with hepatitis B and non-hepatitis virus related hepatocellular carcinoma. Gut Pathog. 2019;11:1. https://doi.org/10.1186/s13099-018-0281-6.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Yu LX, Yan HX, Liu Q, Yang W, Wu HP, Dong W, et al. Endotoxin accumulation prevents carcinogen-induced apoptosis and promotes liver tumorigenesis in rodents. Hepatology. 2010;52:1322–33. https://doi.org/10.1002/hep.23845.

    Article  CAS  PubMed  Google Scholar 

  89. Zhang HL, Yu LX, Yang W, Tang L, Lin Y, Wu H, et al. Profound impact of gut homeostasis on chemically-induced pro-tumorigenic inflammation and hepatocarcinogenesis in rats. J Hepatol. 2012;57:803–12. https://doi.org/10.1016/j.jhep.2012.06.011.

    Article  PubMed  Google Scholar 

  90. Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, Mederacke I, et al. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell. 2012;21:504–16. https://doi.org/10.1016/j.ccr.2012.02.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Gori S, Inno A, Belluomini L, Bocus P, Bisoffi Z, Russo A, et al. Gut microbiota and cancer: how gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit Rev Oncol Hematol. 2019;143:139–47. https://doi.org/10.1016/j.critrevonc.2019.09.003.

    Article  PubMed  Google Scholar 

  92. Lv LX, Fang DQ, Shi D, Chen DY, Yan R, Zhu YX, et al. Alterations and correlations of the gut microbiome, metabolism and immunity in patients with primary biliary cirrhosis. Environ Microbiol. 2016;18:2272–86. https://doi.org/10.1111/1462-2920.13401.

    Article  CAS  PubMed  Google Scholar 

  93. Tilg H, Adolph TE, Gerner RR, Moschen AR. The intestinal microbiota in Colorectal Cancer. Cancer Cell. 2018;33:954–64. https://doi.org/10.1016/j.ccell.2018.03.004.

    Article  CAS  PubMed  Google Scholar 

  94. Cheng Y, Ling Z, Li L. The intestinal microbiota and colorectal Cancer. Front Immunol. 2020;11:615056. https://doi.org/10.3389/fimmu.2020.615056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Wong SH, Yu J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16:690–704. https://doi.org/10.1038/s41575-019-0209-8.

    Article  CAS  PubMed  Google Scholar 

  96. Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, et al. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 2019;68:654–62. https://doi.org/10.1136/gutjnl-2018-317178.

    Article  CAS  PubMed  Google Scholar 

  97. Zepeda-Rivera M, Minot SS, Bouzek H, Wu H, Blanco-Míguez A, Manghi P, et al. A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche. Nature. 2024. https://doi.org/10.1038/s41586-024-07182-w.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22:292–8. https://doi.org/10.1101/gr.126573.111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299–306. https://doi.org/10.1101/gr.126516.111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Park CH, Han DS, Oh YH, Lee AR, Lee YR, Eun CS. Role of Fusobacteria in the serrated pathway of colorectal carcinogenesis. Sci Rep. 2016;6:25271. https://doi.org/10.1038/srep25271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Abed J, Emgård JE, Zamir G, Faroja M, Almogy G, Grenov A, et al. Fap2 mediates Fusobacterium nucleatum Colorectal Adenocarcinoma Enrichment by binding to tumor-expressed Gal-GalNAc. Cell Host Microbe. 2016;20:215–25. https://doi.org/10.1016/j.chom.2016.07.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14:195–206. https://doi.org/10.1016/j.chom.2013.07.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. 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;42:344–55. https://doi.org/10.1016/j.immuni.2015.01.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Martin HM, Campbell BJ, Hart CA, Mpofu C, Nayar M, Singh R, et al. Enhanced Escherichia coli adherence and invasion in Crohn’s disease and colon cancer. Gastroenterology. 2004;127:80–93. https://doi.org/10.1053/j.gastro.2004.03.054.

    Article  CAS  PubMed  Google Scholar 

  105. Swidsinski A, Khilkin M, Kerjaschki D, Schreiber S, Ortner M, Weber J, et al. Association between intraepithelial Escherichia coli and colorectal cancer. Gastroenterology. 1998;115:281–6. https://doi.org/10.1016/s0016-5085(98)70194-5.

    Article  CAS  PubMed  Google Scholar 

  106. Maddocks OD, Scanlon KM, Donnenberg MS. An Escherichia coli effector protein promotes host mutation via depletion of DNA mismatch repair proteins. mBio. 2013;4:e00152–13. https://doi.org/10.1128/mBio.00152-13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Chen CC, Liou JM, Lee YC, Hong TC, El-Omar EM, Wu MS. The interplay between Helicobacter pylori and gastrointestinal microbiota. Gut Microbes. 2021;13:1–22. https://doi.org/10.1080/19490976.2021.1909459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Yang J, Zhou X, Liu X, Ling Z, Ji F. Role of the gastric microbiome in gastric Cancer: from carcinogenesis to treatment. Front Microbiol. 2021;12:641322. https://doi.org/10.3389/fmicb.2021.641322.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Nabavi-Rad A, Yadegar A, Sadeghi A, Aghdaei HA, Zali MR, Klionsky DJ, et al. The interaction between autophagy, Helicobacter pylori, and gut microbiota in gastric carcinogenesis. Trends Microbiol. 2023;31:1024–43. https://doi.org/10.1016/j.tim.2023.04.001.

    Article  CAS  PubMed  Google Scholar 

  110. Amieva M, Peek RM. Jr. Pathobiology of Helicobacter pylori-Induced Gastric Cancer. Gastroenterology. 2016;150:64–78. https://doi.org/10.1053/j.gastro.2015.09.004.

    Article  CAS  PubMed  Google Scholar 

  111. Sexton RE, Al Hallak MN, Diab M, Azmi AS. Gastric cancer: a comprehensive review of current and future treatment strategies. Cancer Metastasis Rev. 2020;39:1179–203. https://doi.org/10.1007/s10555-020-09925-3.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Waldum H, Fossmark R, Gastritis. Gastric polyps and gastric Cancer. Int J Mol Sci. 2021;22. https://doi.org/10.3390/ijms22126548.

  113. Lee YC, Chiang TH, Chou CK, Tu YK, Liao WC, Wu MS, et al. Association between Helicobacter pylori Eradication and Gastric Cancer incidence: a systematic review and Meta-analysis. Gastroenterology. 2016;150:1113–24.e5.

    Article  PubMed  Google Scholar 

  114. Mentis AA, Boziki M, Grigoriadis N, Papavassiliou AG. Helicobacter pylori infection and gastric cancer biology: tempering a double-edged sword. Cell Mol Life Sci. 2019;76:2477–86. https://doi.org/10.1007/s00018-019-03044-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Peek RM Jr., Fiske C, Wilson KT. Role of innate immunity in Helicobacter pylori-induced gastric malignancy. Physiol Rev. 2010;90:831–58. https://doi.org/10.1152/physrev.00039.2009.

    Article  CAS  PubMed  Google Scholar 

  116. Zhang L, Sung JJ, Yu J, Ng SC, Wong SH, Cho CH, et al. Xenophagy in Helicobacter pylori- and Epstein-Barr virus-induced gastric cancer. J Pathol. 2014;233:103–12. https://doi.org/10.1002/path.4351.

    Article  CAS  PubMed  Google Scholar 

  117. Wessler S, Krisch LM, Elmer DP, Aberger F. From inflammation to gastric cancer - the importance of Hedgehog/GLI signaling in Helicobacter pylori-induced chronic inflammatory and neoplastic diseases. Cell Commun Signal. 2017;15:15. https://doi.org/10.1186/s12964-017-0171-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Israel DA, Peek RM. Surreptitious manipulation of the human host by Helicobacter pylori. Gut Microbes. 2010;1:119–27. https://doi.org/10.4161/gmic.1.2.11991.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Mommersteeg MC, Simovic I, Yu B, van Nieuwenburg SAV, Bruno IMJ, Doukas M, et al. Autophagy mediates ER stress and inflammation in Helicobacter pylori-related gastric cancer. Gut Microbes. 2022;14:2015238. https://doi.org/10.1080/19490976.2021.2015238.

    Article  CAS  PubMed  Google Scholar 

  120. Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014;345:196–202. https://doi.org/10.1016/j.canlet.2013.08.016.

    Article  CAS  PubMed  Google Scholar 

  121. de Brito BB, da Silva FAF, Soares AS, Pereira VA, Santos MLC, Sampaio MM, et al. Pathogenesis and clinical management of Helicobacter pylori gastric infection. World J Gastroenterol. 2019;25:5578–89. https://doi.org/10.3748/wjg.v25.i37.5578.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Lamb A, Chen LF. Role of the Helicobacter pylori-induced inflammatory response in the development of gastric cancer. J Cell Biochem. 2013;114:491–7. https://doi.org/10.1002/jcb.24389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Wen Y, Huang H, Tang T, Yang H, Wang X, Huang X, et al. AI-2 represses CagA expression and bacterial adhesion, attenuating the Helicobacter pylori-induced inflammatory response of gastric epithelial cells. Helicobacter. 2021;26:e12778. https://doi.org/10.1111/hel.12778.

    Article  CAS  PubMed  Google Scholar 

  124. De Falco M, Lucariello A, Iaquinto S, Esposito V, Guerra G, De Luca A. Molecular mechanisms of Helicobacter pylori Pathogenesis. J Cell Physiol. 2015;230:1702–7. https://doi.org/10.1002/jcp.24933.

    Article  CAS  PubMed  Google Scholar 

  125. Shimizu T, Marusawa H, Watanabe N, Chiba T. Molecular Pathogenesis of Helicobacter pylori-related gastric Cancer. Gastroenterol Clin North Am. 2015;44:625–38. https://doi.org/10.1016/j.gtc.2015.05.011.

    Article  PubMed  Google Scholar 

  126. Tohidpour A. CagA-mediated pathogenesis of Helicobacter pylori. Microb Pathog. 2016;93:44–55. https://doi.org/10.1016/j.micpath.2016.01.005.

    Article  CAS  PubMed  Google Scholar 

  127. Cover TL, Lacy DB, Ohi MD. The Helicobacter pylori Cag type IV Secretion System. Trends Microbiol. 2020;28:682–95. https://doi.org/10.1016/j.tim.2020.02.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Takahashi-Kanemitsu A, Knight CT, Hatakeyama M. Molecular anatomy and pathogenic actions of Helicobacter pylori CagA that underpin gastric carcinogenesis. Cell Mol Immunol. 2020;17:50–63. https://doi.org/10.1038/s41423-019-0339-5.

    Article  CAS  PubMed  Google Scholar 

  129. Hatakeyama M. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe. 2014;15:306–16. https://doi.org/10.1016/j.chom.2014.02.008.

    Article  CAS  PubMed  Google Scholar 

  130. 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;29:941–e5810. https://doi.org/10.1016/j.chom.2021.04.006.

    Article  CAS  PubMed  Google Scholar 

  131. Hardbower DM, Peek RM Jr., Wilson KT. At the Bench: Helicobacter pylori, dysregulated host responses, DNA damage, and gastric cancer. J Leukoc Biol. 2014;96:201–12. https://doi.org/10.1189/jlb.4BT0214-099R.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Fu L, Xie C. A lucid review of Helicobacter pylori-induced DNA damage in gastric cancer. Helicobacter. 2019;24:e12631. https://doi.org/10.1111/hel.12631.

    Article  PubMed  Google Scholar 

  133. Murata-Kamiya N, Hatakeyama M. Helicobacter pylori-induced DNA double-stranded break in the development of gastric cancer. Cancer Sci. 2022;113:1909–18. https://doi.org/10.1111/cas.15357.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Engstrand L, Graham DY. Microbiome and Gastric Cancer. Dig Dis Sci. 2020;65:865–73. https://doi.org/10.1007/s10620-020-06101-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Li J, Perez Perez GI. Is there a role for the Non-helicobacter pylori Bacteria in the risk of developing gastric Cancer? Int J Mol Sci. 2018;19. https://doi.org/10.3390/ijms19051353.

  136. Stewart OA, Wu F, Chen Y. The role of gastric microbiota in gastric cancer. Gut Microbes. 2020;11:1220–30. https://doi.org/10.1080/19490976.2020.1762520.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Mathebela P, Damane BP, Mulaudzi TV, Mkhize-Khwitshana ZL, Gaudji GR, Dlamini Z. Influence of the Microbiome Metagenomics and Epigenomics on gastric Cancer. Int J Mol Sci. 2022;23. https://doi.org/10.3390/ijms232213750.

  138. Ralser A, Dietl A, Jarosch S, Engelsberger V, Wanisch A, Janssen KP, et al. Helicobacter pylori promotes colorectal carcinogenesis by deregulating intestinal immunity and inducing a mucus-degrading microbiota signature. Gut. 2023;72:1258–70. https://doi.org/10.1136/gutjnl-2022-328075.

    Article  CAS  PubMed  Google Scholar 

  139. Coker OO, Dai Z, Nie Y, Zhao G, Cao L, Nakatsu G, et al. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67:1024–32. https://doi.org/10.1136/gutjnl-2017-314281.

    Article  CAS  PubMed  Google Scholar 

  140. Aviles-Jimenez F, Vazquez-Jimenez F, Medrano-Guzman R, Mantilla A, Torres J. Stomach microbiota composition varies between patients with non-atrophic gastritis and patients with intestinal type of gastric cancer. Sci Rep. 2014;4:4202. https://doi.org/10.1038/srep04202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. 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;67:226–36. https://doi.org/10.1136/gutjnl-2017-314205.

    Article  CAS  PubMed  Google Scholar 

  142. Hu YL, Pang W, Huang Y, Zhang Y, Zhang CJ. The gastric microbiome is perturbed in Advanced gastric adenocarcinoma identified through Shotgun Metagenomics. Front Cell Infect Microbiol. 2018;8:433. https://doi.org/10.3389/fcimb.2018.00433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Castaño-Rodríguez N, Goh KL, Fock KM, Mitchell HM, Kaakoush NO. Dysbiosis of the microbiome in gastric carcinogenesis. Sci Rep. 2017;7:15957. https://doi.org/10.1038/s41598-017-16289-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Eun CS, Kim BK, Han DS, Kim SY, Kim KM, Choi BY, et al. Differences in gastric mucosal microbiota profiling in patients with chronic gastritis, intestinal metaplasia, and gastric cancer using pyrosequencing methods. Helicobacter. 2014;19:407–16. https://doi.org/10.1111/hel.12145.

    Article  CAS  PubMed  Google Scholar 

  145. 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. https://doi.org/10.1016/j.ebiom.2018.12.034.

    Article  PubMed  Google Scholar 

  146. 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;13:4640. https://doi.org/10.1038/s41598-023-31740-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Zhao LY, Mei JX, Yu G, Lei L, Zhang WH, Liu K, et al. Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications. Signal Transduct Target Ther. 2023;8:201. https://doi.org/10.1038/s41392-023-01406-7.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Doocey CM, Finn K, Murphy C, Guinane CM. The impact of the human microbiome in tumorigenesis, cancer progression, and biotherapeutic development. BMC Microbiol. 2022;22:53. https://doi.org/10.1186/s12866-022-02465-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Yu X, Ou J, Wang L, Li Z, Ren Y, Xie L, et al. Gut microbiota modulate CD8(+) T cell immunity in gastric cancer through Butyrate/GPR109A/HOPX. Gut Microbes. 2024;16:2307542. https://doi.org/10.1080/19490976.2024.2307542.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. McAllister F, Khan MAW, Helmink B, Wargo JA. The Tumor Microbiome in Pancreatic Cancer: Bacteria and Beyond. Cancer Cell. 2019;36:577–9. https://doi.org/10.1016/j.ccell.2019.11.004.

    Article  CAS  PubMed  Google Scholar 

  151. Wei MY, Shi S, Liang C, Meng QC, Hua J, Zhang YY, et al. The microbiota and microbiome in pancreatic cancer: more influential than expected. Mol Cancer. 2019;18:97. https://doi.org/10.1186/s12943-019-1008-0.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Nagata N, Nishijima S, Kojima Y, Hisada Y, Imbe K, Miyoshi-Akiyama T, et al. Metagenomic identification of Microbial signatures Predicting Pancreatic Cancer from a multinational study. Gastroenterology. 2022;163:222–38. https://doi.org/10.1053/j.gastro.2022.03.054.

    Article  CAS  PubMed  Google Scholar 

  153. Lu SY, Hua J, Xu J, Wei MY, Liang C, Meng QC, et al. Microorganisms in chemotherapy for pancreatic cancer: an overview of current research and future directions. Int J Biol Sci. 2021;17:2666–82. https://doi.org/10.7150/ijbs.59117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Vitiello GA, Cohen DJ, Miller G. Harnessing the Microbiome for Pancreatic Cancer Immunotherapy. Trends Cancer. 2019;5:670–6. https://doi.org/10.1016/j.trecan.2019.10.005.

    Article  CAS  PubMed  Google Scholar 

  155. Sobocki BK, Kaźmierczak-Siedlecka K, Folwarski M, Hawryłkowicz V, Makarewicz W, Stachowska E. Pancreatic Cancer and gut microbiome-related aspects: a Comprehensive Review and Dietary recommendations. Nutrients. 2021;13. https://doi.org/10.3390/nu13124425.

  156. Thomas H. Pancreatic cancer: intra-tumour bacteria promote gemcitabine resistance in pancreatic adenocarcinoma. Nat Rev Gastroenterol Hepatol. 2017;14:632. https://doi.org/10.1038/nrgastro.2017.142.

    Article  PubMed  Google Scholar 

  157. Tintelnot J, Xu Y, Lesker TR, Schönlein M, Konczalla L, Giannou AD, et al. Microbiota-derived 3-IAA influences chemotherapy efficacy in pancreatic cancer. Nature. 2023;615:168–74. https://doi.org/10.1038/s41586-023-05728-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Ghaddar B, Biswas A, Harris C, Omary MB, Carpizo DR, Blaser MJ, et al. Tumor microbiome links cellular programs and immunity in pancreatic cancer. Cancer Cell. 2022;40:1240–53.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Udayasuryan B, Ahmad RN, Nguyen TTD, Umaña A, Monét Roberts L, Sobol P, et al. Fusobacterium nucleatum induces proliferation and migration in pancreatic cancer cells through host autocrine and paracrine signaling. Sci Signal. 2022;15:eabn4948. https://doi.org/10.1126/scisignal.abn4948.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Deshpande NP, Riordan SM, Castaño-Rodríguez N, Wilkins MR, Kaakoush NO. Signatures within the esophageal microbiome are associated with host genetics, age, and disease. Microbiome. 2018;6:227. https://doi.org/10.1186/s40168-018-0611-4.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Pandey A, Lieu CH, Kim SS. The local Microbiome in Esophageal Cancer and Treatment Response: a review of Emerging Data and future directions. Cancers (Basel). 2023;15. https://doi.org/10.3390/cancers15143562.

  162. Kumar B, Lam S, Adam M, Gilroy R, Pallen MJ. The oesophageal microbiome and cancer: hope or hype? Trends Microbiol. 2022;30:322–9. https://doi.org/10.1016/j.tim.2021.08.007.

    Article  CAS  PubMed  Google Scholar 

  163. Elliott DRF, Walker AW, O’Donovan M, Parkhill J, Fitzgerald RC. A non-endoscopic device to sample the oesophageal microbiota: a case-control study. Lancet Gastroenterol Hepatol. 2017;2:32–42. https://doi.org/10.1016/s2468-1253(16)30086-3.

    Article  PubMed  Google Scholar 

  164. Shao D, Vogtmann E, Liu A, Qin J, Chen W, Abnet CC, et al. Microbial characterization of esophageal squamous cell carcinoma and gastric cardia adenocarcinoma from a high-risk region of China. Cancer. 2019;125:3993–4002. https://doi.org/10.1002/cncr.32403.

    Article  CAS  PubMed  Google Scholar 

  165. Li M, Shao D, Zhou J, Gu J, Qin J, Chen W, et al. Signatures within esophageal microbiota with progression of esophageal squamous cell carcinoma. Chin J Cancer Res. 2020;32:755–67. https://doi.org/10.21147/j.issn.1000-9604.2020.06.09.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Yamamura K, Baba Y, Nakagawa S, Mima K, Miyake K, Nakamura K, et al. Human Microbiome Fusobacterium Nucleatum in Esophageal Cancer tissue is Associated with Prognosis. Clin Cancer Res. 2016;22:5574–81. https://doi.org/10.1158/1078-0432.Ccr-16-1786.

    Article  CAS  PubMed  Google Scholar 

  167. Peters BA, Wu J, Pei Z, Yang L, Purdue MP, Freedman ND, et al. Oral Microbiome Composition reflects prospective risk for esophageal cancers. Cancer Res. 2017;77:6777–87. https://doi.org/10.1158/0008-5472.Can-17-1296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Gao S, Li S, Ma Z, Liang S, Shan T, Zhang M, et al. Presence of Porphyromonas gingivalis in esophagus and its association with the clinicopathological characteristics and survival in patients with esophageal cancer. Infect Agent Cancer. 2016;11:3. https://doi.org/10.1186/s13027-016-0049-x.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Li Y, Wei B, Xue X, Li H, Li J. Microbiome changes in esophageal cancer: implications for pathogenesis and prognosis. Cancer Biol Med. 2023;21:163–74. https://doi.org/10.20892/j.issn.2095-3941.2023.0177.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Moreira C, Figueiredo C, Ferreira RM. The role of the Microbiota in Esophageal Cancer. Cancers (Basel). 2023;15. https://doi.org/10.3390/cancers15092576.

  171. Liang Y, Li Q, Liu Y, Guo Y, Li Q. Awareness of intratumoral bacteria and their potential application in cancer treatment. Discov Oncol. 2023;14:57. https://doi.org/10.1007/s12672-023-00670-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Lu YQ, Qiao H, Tan XR, Liu N. Broadening oncological boundaries: the intratumoral microbiota. Trends Microbiol. 2024. https://doi.org/10.1016/j.tim.2024.01.007.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Author information

Authors and Affiliations

Authors

Contributions

All the authors contributed to the manuscript and reviewed and approved it.

as presented here. Huihui Gao and Weiqin Lu contributed to the concept of the review.

and writing the manuscript. Aihemaitijiang Aihaiti, Paziliya Abudukeranmu, designed the figure. Yajun Liu aided in revising the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Huihui Gao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors declare no competing interests related to this review.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Rights and permissions

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, W., Aihaiti, A., Abudukeranmu, P. et al. Unravelling the role of intratumoral bacteria in digestive system cancers: current insights and future perspectives. J Transl Med 22, 545 (2024). https://doi.org/10.1186/s12967-024-05320-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05320-6

Keywords