Skip to main content

Intratumoral microorganisms in tumors of the digestive system

Abstract

Tumors of the digestive system pose a significant threat to human health and longevity. These tumors are associated with high morbidity and mortality rates, leading to a heavy economic burden on healthcare systems. Several intratumoral microorganisms are present in digestive system tumors, and their sources and abundance display significant heterogeneity depending on the specific tumor subtype. These microbes have a complex and precise function in the neoplasm. They can facilitate tumor growth through various mechanisms, such as inducing DNA damage, influencing the antitumor immune response, and promoting the degradation of chemotherapy drugs. Therefore, these microorganisms can be targeted to inhibit tumor progression for improving overall patient prognosis. This review focuses on the current research progress on microorganisms present in the digestive system tumors and how they influence the initiation, progression, and prognosis of tumors. Furthermore, the primary sources and constituents of tumor microbiome are delineated. Finally, we summarize the application potential of intratumoral microbes in the diagnosis, treatment, and prognosis prediction of digestive system tumors.

Video Abstract

Introduction

In 2019, the World Health Organization redefined the criteria for classifying digestive system tumors and included esophageal cancer (EC), gastric cancer (GC), colorectal cancer (CRC), pancreatic cancer (PC), and hepatocellular carcinoma (HCC) [1], among others in this classification. Tumor types are based on their molecular phenotype in addition to histopathological characteristics, reflecting the latest advancements in the understanding of digestive system tumors. Although considerable improvements have been achieved in traditional treatment modalities for neoplasms of the digestive system, patient prognosis remains poor. This happens because of inadequate early screening, limitations of current treatments, and the increased frequency of metastasis and recurrence of digestive system tumors [2, 3]. Consequently, identifying novel diagnostic biomarkers and treatment modalities, mitigating the emergence of antitumor drug resistance, and improving the outcomes and quality of life of patients is crucial for healthcare professionals.

The surface barriers of human body is populated by complex groups of bacteria, fungi, protozoa, and viruses [4]. The intestinal mucosa that lines the inner surface of various organs contains trillions of microbial species, and bacteria are the most dominant group among them [5, 6]. In 2015, Garrett et al. suggested that bacteria can promote tumorigenesis by disrupting cell growth homeostasis and regulating immune responses and drug metabolism [7]. The gastrointestinal tract acts as a repository of microorganisms in the human body and exerts a significant influence on the pathogenesis of various malignancies. Therefore, the gut microbiota can influence the development of tumors by acting as regulators of immune system stimulation [8]. In a word, gut microbes may trigger inflammation associated with cancer and impact the efficacy of various cancer therapies.

Several authors reported the presence of microbes in tumors in the 19th century [9]. However, the origin of the microbes was unclear because the lack of molecular detection techniques was not available to ascertain whether the detected microbes originated from within the tumor or from external contamination. Moreover, the low microbial density within tumors hindered research on their significance for over a century. The progress in the detection technology and an increased understanding of tumor microenvironments have validated the presence of intratumoral microorganisms in the past few years. The gastrointestinal system —the largest reservoir of microorganisms in the human body—is a comprehensive physiological network that interfaces with the external environment through the oral cavity and digestive tract. Notably, intratumoral microbes are most frequently detected in digestive system neoplasms [10,11,12,13].

In this review, we have detailed the fundamental attributes of microbiota present in tumors of the digestive system and the recent advancements in this field. Furthermore, we have innovatively elucidated the functions of typical microorganisms in gastrointestinal neoplasms and the underlying mechanisms by which they facilitate tumor initiation or inhibition of tumor progression. Finally, we focused on the prospective use of these microorganisms in the diagnosis, therapy, and predicting the outcomes of gastrointestinal malignancies. We express our genuine anticipation that our analysis will provide the baseline to explore novel diagnostic and therapeutic approaches for malignancies of the gastrointestinal tract.

Detection methods for intratumoral microbes and their general characteristics

The continual advancements in technology have enabled us to trace the origin of tumor-associated bacteria, determine the specific composition of intratumoral microbiota, and gauge the abundance of microorganisms in different types of digestive system tumors (Fig. 1).

Fig. 1
figure 1

Intratumoral microbes: Detection methods and general characteristics. a. Several methods are used to detect microorganisms in tumors, and next-generation sequencing technology is the primary detection method.b. Microbes can enter and colonize tumor tissue by crossing damaged mucosal barriers, migrating from adjacent normal tissues, or through the circulatory system.c. Fusobacterium, Bacteroides fragilis, Escherichia coli, Helicobacter pylori, and Malassezia are the common microbes in digestive system tumors.d. Microbes show substantial heterogeneity in various types of tumors and during different stages of the same tumor. Moreover, microbial populations in tumors differ from those in neighboring healthy tissues

Detection methods for microbes in digestive system tumors

Next-generation sequencing has emerged as a promising technique to obtain a comprehensive microbial profile of the sample without the requirement for culturing [14]. Deep sequencing techniques involving universal marker gene amplicons are frequently utilized for investigating microbiomes, and 16 S rRNA sequencing and shotgun metagenomic sequencing are the most commonly used methods [15, 16]. 16 S rRNA sequencing is an efficient technique for characterizing bacterial diversity but with less detailed functional information. Whole metagenomic shotgun sequencing has several advantages over 16 S rRNA sequencing, including identification of both non-bacterial and bacterial taxa, strain/species level resolution, and functional annotation of the microbiota [17, 18]. In addition to sequencing techniques, intratumoral microorganisms can also be detected using fluorescent labeling of microbial antigens, genetic material, or metabolites [14, 19]. Overall, the use of advanced detection methods has enabled the rapid identification of microorganisms present within tumor tissues.

Sources of microbiota in digestive system tumors

Bacteria can infiltrate gastrointestinal neoplasms by passing through disrupted mucosal barriers, migrating from adjacent normal tissue, or blood circulation [20]. Tjalsma et al. [21]. (2012) proposed a unique bacterial driver–passenger model of CRC pathogenesis. They suggested that specific intratumoral bacteria (drivers) can facilitate the infiltration of other gut microorganisms (passengers) into the tumor microenvironment. Interestingly, fluorescently labeled Enterococcus faecalis and GFP-labeled Escherichia coli (E. coli) were detected within the pancreatic microenvironment in mice [22]. The use of advanced detection methods has highlighted the existence of bacteria in otherwise normally sterile organs, including the pancreas [23]. Notably, the composition of bacterial detected in PC tissues is extremely similar to that observed in healthy adjacent pancreatic tissues [24, 25]. Abed et al. reported that Fusobacterium nucleatum (F. nucleatum), a major constituent of the human oral microbiome, uses a hematogenous pathway to infiltrate colon adenocarcinomas [26].

Microbial diversity in digestive system cancers

The utilization of next-generation sequencing technology enables the identification of all microorganisms, without the requirement of culturing, while enabling the identification of bacterial species based on their genetic profiles [16]. Bacterial colonization within tumors of the digestive system is a well-documented phenomenon, and these bacteria originate from diverse sources and show significant species-level variability [11, 27, 28]. Several common bacteria have been identified in digestive system tumors. Helicobacter pylori (H. pylori), a recognized risk factor for chronic gastritis and stomach cancer [29], is present not only in the cases of gastric malignancies but also in those of HCC and PC [23, 30]. The bacterial genus Fusobacterium, frequently detected in CRC [31], has also been associated with an unfavorable prognosis in PC [32]. Bacteroides fragilis (B. fragilis) colonizes the tumor microenvironment of colitis-associated CRC and can cause DNA damage [33]. The presence of E. coli in CRC can disrupt the gut vascular barrier and promote the development of a conducive microenvironment for liver metastases [34]. The diversity of bacteria present in digestive system tumors is extensive, thereby necessitating comprehensive investigations involving large sample sizes.

Microbial heterogeneity within digestive system tumors

The composition of bacterial species in each tumor is considerably variable. The microbiome in the cohorts of patients with upper gastrointestinal tract EC and GC predominantly comprised Bacteroidetes and Firmicutes at the phylum level. In contrast, only Bacteroidetes were dominant in the samples collected from the patients with lower gastrointestinal tract cancers [27]. Additionally, bacterial diversity was significantly different between cancerous and adjacent non-cancerous tissues [27, 35]. Yuan and colleagues reported that Porphyromonas gingivalis was predominant and frequently present in the samples of EC and esophageal dysplasia tissues [36]. However, the bacterium was infrequently present in matched noncancerous segments. Similarly, Fusobacterium species were highly abundant in human colonic adenomas compared to the adjacent normal tissues [37].

Further, the composition of bacteria can vary during different phases of tumor progression. As the GC advances, H. pylori gradually diminishes in the tumor tissues and eventually disappears [38, 39]. B. fragilis toxin (BFT) facilitates inflammation, and advanced-stage CRC tissues show high BFT positivity compared to early-stage counterparts [33]. Nevertheless, it is unclear whether this happens due to an increased number of bacterial colonies in late-stage CRC or an increased secretion of toxins. Overall, the variations in microbial diversity among distinct tumors, cancerous versus non-cancerous tissues, and different stages of tumor progression can be exploited to develop new diagnoses and treatment strategies for digestive system tumors.

Furthermore, the relative abundance and source of bacteria within digestive tumors also exhibits substantial heterogeneity. In the microenvironment of HCC, there was a significant alteration in the abundance of microorganisms, including a reduction in the prevalence of Pseudomonadaceae and an increase in the prevalence of Agrobacterium and Rhizobiaceae [40]. Specially, a positive and linear correlation was observed between the presence of Pseudomonadaceae and the prognosis of patients with HCC. Bifidobacteria, as intestinal commensal bacteria, can infiltrate CRC tissues through disrupted intestinal intestinal barrier, leading to elevated abundance of Bifidobacteria [21, 41, 42]. Additionally, the secretion of lactic acid and acetic acid by Bifidobacteria enhances the growth and immune evasion of CRC, thereby negatively impacting prognosis [43]. Further investigations are required to identify variations in the abundance and source of microorganisms and their influence on the prognosis of distinct subtypes of digestive tumors.

Intratumoral microbiome in different digestive system tumors

The role of microorganisms in the development, detection,  prognostication, and therapy of cancer has been a matter of debate [44,45,46]. Next-generation sequencing technology has provided an unparalleled opportunity to investigate the genomes of tumor cells and hosts, as well as the diverse microorganisms that inhabit living organisms [47]. The following sections summarize the published studies on microorganisms identified in tumors of the digestive system using novel sequencing techniques (Table 1; Fig. 2).

Table 1 Intratumoral microbiome in different digestive system tumors
Fig. 2
figure 2

Research studies on intratumoral microbiomes in different digestive system tumors. The number of samples included in different studies on bacteria in tumors of the digestive system is listed in a chronological order using a time axis

Esophageal cancer

Esophageal cancer ranks sixth in terms of global cancer mortality and has two main subtypes, namely esophageal adenocarcinoma (EADC) and esophageal squamous cell carcinoma (ESCC) [90]. ESCC is the most prevalent histological subtype of EC worldwide [91, 92], and EADC is usually associated with Barrett’s esophagus [93]. Recently, several researchers have reported the changes in the microbiome inside tumors during the progression of EC and their influence on the cancer treatment.

Yamamura et al. (2016) quantified the presence of F. nucleatum DNA in 325 excised specimens of EC using quantitative polymerase chain reaction (qPCR). They observed a significant association between the presence of F. nucleatum and decreased survival rates [48]. Additionally, the prevalence of F. nucleatum was strongly correlated with the growth of ESCC tissues, indicating its potential significance in the progression of ESCC [52]. Furthermore, F. nucleatum modulated the expression of endogenous LC3 and ATG7 proteins and facilitated the formation of autophagosomes, which resulted in the development of chemoresistance against 5-fluorouracil, cisplatin, and docetaxel [51]. Notably, high F. nucleatum load was also associated with negative side effects during neoadjuvant chemotherapy [50]. The bacterial abundance varies during EC progression, and these alterations are associated with the clinical characteristics and prognosis of EC [28, 49]. Snider et al. observed variations in the microbial community associated with Barrett’s esophagus in patients with high-grade dysplasia and EADC [12]. These shifts were characterized by an increase in specific bacterial groups potentially associated with pathogenicity, such as Proteobacteria. Overall, these findings indicate that alterations in tumor microbiota are significantly associated with clinical prognoses and chemotherapy-related side effects in patients.

Gastric cancer

Gastric cancer ranks fifth among the most prevalent types of cancer and is the fourth leading cause of cancer-related deaths worldwide [2, 94]. Chronic infection with H. pylori is considered the primary underlying factor of noncardia GC, and approximately all instances of this type of cancer are linked to this bacterium [95, 96]. However, the development of carcinoma of the cardia is not correlated with H. pylori infection and shows an inverse correlation in certain populations [97, 98].

Wang et al. (2015) evaluated a Chinese cohort and indicated that the primary impact of H. pylori on the microbial community is the increased bacterial load in the stomach rather than a change in the proportional abundances of non- H. pylori bacterial groups [53]. However, two years later, Liu et al. determined that the proportion of H. pylori in GC tissues obtained from patient groups in China and Mexico was lower than that in the adjacent nontumor tissues obtained from these patients. Moreover, the relative abundance of non-H. pylori bacteria was also altered in the tumor tissues, suggesting a potential alteration of the microbial composition in GC samples [39]. Subsequently, Ferreira et al. analyzed the gastric microbial community using 16 S rRNA sequencing and real-time qPCR [54]. The authors revealed the dysbiotic microbiota associated with GC, indicated by reduced microbial diversity, decreased abundance of H. pylori, and enrichment of other intestinal commensal bacteria. Furthermore, Liu et al. analyzed gastric microbiota by targeting the 16 S rRNA gene in a cohort of 276 patients with GC who had not received preoperative chemotherapy [55]. The authors proposed that the composition and diversity of the gastric microbiota were determined by specific stomach microhabitats associated with GC rather than being influenced by the stages or types of GC. Mannion et al. (2023) used the shotgun metagenomic approach to determine the microbial functional and composition variations of the microbiome within high-risk and low-risk cohorts of GC [56]. They claimed that it was possible to identify specific intratumoral microorganisms associated with GC by taking advantage of next-generation sequencing methods. Overall, these findings offer novel insights for the prophylaxis, therapy, and diagnosis of GC based on the associated microbiome.

Colorectal cancer

Colorectal cancer is the third leading cause of cancer-associated morbidity, and despite a gradual reduction in mortality since 1978, it remains the second most prevalent cause of cancer-related fatalities in both males and females [2, 3]. Several authors have suggested a close association between CRC and the gut microbiota [99,100,101].

Several researchers have used novel identification methodologies such as qPCR, 16 S rRNA sequencing, and metagenomic analysis and reported the prevalence of Fusobacterium in individuals diagnosed with CRC [57,58,59]. Additionally, the concentration of F. nucleatum DNA in CRC tissue is negatively correlated with patient survival, ultimately resulting in shorter survival times [60, 64, 72]. In the research, this negative correlation was attributed to the ability of bacteria to promote nerve invasion, vascular tumor thrombus formation, and location of tumor [72]. Hamada et al. indicated that F. nucleatum modulated immune response within the tumor microenvironment [70]. The bacterium inhibited adaptive antitumor immune responses in MSI-High CRC, whereas showed pro-inflammatory effects in MSI-Low CRC. Moreover, increased F. nucleatum DNA within cancerous tissues was correlated with increased macrophage permeation and CDKN2A hypermethylation in MSI-High CRCs [66]. F. nucleatum impaired T-cell-mediated immune responses against colorectal tumors, thereby promoting the tumor growth. Therefore, F. nucleatum DNA can be considered a prognostic biomarker for CRC [64]. Thus, the administration of antibiotics targeting F. nucleatum may potentially affect the development and advancement of CRC [67, 74]. Interestingly, the load and abundance of microorganisms in the tumor may also vary depending on various factors, including the stage of cancer and location of tumor [61, 65, 73]. In addition to F. nucleatum, other bacteria may also influence the development, progression, and clinical manifestations of CRC [62, 63, 68, 77]. Coker et al. performed metabolomic and metagenomic analysis on stool samples from 386 subjects and demonstrated that bacterial metabolites, such as l-alanine, glycine, and l-proline, can promote the development of CRC and serve as markers for the non-invasive diagnosis of colorectal neoplasia [76].

The intratumoral enteric viruses, a critical constituent and regulator of the gut microbiota, influence the composition and abundance of intestinal microorganisms. Therefore, these viruses can influence the incidence, development, and outcomes of CRC [69, 99]. Furthermore, modifications in the composition of the intestinal fungal community were associated with CRC. The concentrations of Malassezia spp. increased, whereas those of Saccharomyces and Pneumocystis markedly declined in CRC tissue samples [71, 75].

Pancreatic cancer

Pancreatic cancer is a malignant neoplasm with high mortality rates, where the incidence and mortality rates are almost similar [2]. The disease has a highly unfavorable prognosis and is the third most common cause of cancer-associated deaths in the United States [102, 103]. The presence of intratumoral microorganisms not only facilitates the onset and progression of PC but also influences treatment responsiveness and prognostication [11, 22].

Several authors have reported the colonization of the pancreas by multiple microorganisms both in healthy and diseased states [24, 80, 81]. However, the mechanisms by which microorganisms can infiltrate the supposedly aseptic pancreas are debatable. Nalluri et al. identified a considerable rise in the prevalence of Enterobacteriaceae in PC tissues among patients who underwent biliary stent implantation, suggesting that the biliary tract could potentially serve as a channel for microbial entry into the pancreas [25]. Kohi et al. found that the microbes in the duodenal fluid of adenocarcinoma patients were different from those in the duodenal fluid of healthy controls [87]. Moreover, microbial composition in PC tissues was similar to that in the duodenum, suggesting that bacteria may migrate from the intestine to the pancreas. Geller et al. also supported this hypothesis [80]. However, further studies are required to explore the mechanisms by which microorganisms gain access to the pancreas and determine whether these entry routes influence the progression and outcome of PC.

Several studies have reported that microbiota composition will significantly varies in response to a variety of influences [11, 82, 85,86,87]. Furthermore, several authors have determined that oral microbial communities may be implicated in the origin and progression of PC [79, 84]. The association of H. pylori with pancreatic ductal adenocarcinoma (PDAC) is controversial, although H. pylori is considered a risk factor for GC and can be found in cancer tissues. Nilsson et al. reported a detection rate of H. pylori DNA in 75% (30/40) of PC samples [23]. Conversely, Jesnowski et al. [78]. reported no evidence of H. pylori DNA in chronic pancreatitis or other PDAC tissue samples. In addition to bacteria, fungi have also been identified in PC tissue [104]. The findings reported by Aykut and colleagues provided confirmatory evidence for the enrichment of Malassezia in the fungal communities associated with PDAC [83]. Moreover, the study reveals that pathogenic fungi were enabled to activate mannose-bound lectins, which can promote tumorigenesis in the pancreatic environment.

Hepatocellular carcinoma

Liver cancer ranks as the sixth most prevalent and the fourth most lethal neoplasm globally; its incidence is rapidly increasing worldwide [105,106,107]. HCC constitutes approximately 90% of the cases of liver cancer. Hepatitis B virus infection is a major risk factor for HCC, contributing to approximately 50% of reported cases [108].

The intestinal/hepatic axis establishes communication between the intestinal microbiota and the liver through the portal vein system. Microorganisms migrate from the intestine to the liver through the portal vein and may influence the pathological condition of the liver [109, 110]. Although the effect of gut microbiota on liver diseases has been extensively studied, the intratumoral microbiota of HCC remains unexplored [111,112,113]. In 2004, Huang et al. identified the 16 S rDNA of H. pylori in 8 out of 20 primary liver cancer samples; however, it was unclear whether these H. pylori strains were present within the tumor cells at that time [30]. In 2020, Sookonian et al. verified the presence of intrahepatic bacteria DNA in hepatocytes. They conducted high-throughput sequencing analysis of the 16 S rRNA gene in liver tissues obtained from 97 obese patients with NAFLD and 19 non-obese patients with NAFLD (control group) [114]. Their results indicated that the bacterial DNA profiles of the liver of morbidly obese patients with NAFLD differed significantly from those of non-obese patients with NAFLD. Nevertheless, experimental evidence for the presence of microorganisms in liver cancer cells is yet to be established. Interestingly, Huang et al. observed a notable increase in the levels of microbial α- and β-diversities of both the peritumor area and HCC tissue compared to the normal controls [88]. He et al. have discovered that the microbial diversity within HCC tissue is significantly higher than its surrounding tissues, as determined by a thorough analysis [115]. Moreover, they investigated the underlying mechanism by which microbial communities affect lipid metabolism and their impact on HCC progression. Recently, Sun et al. reported significant heterogeneity in the intratumoral microbiome of HCC [89]. They used microbial profile-based clustering and developed a hepatotype, which may serve as an independent biomarker for the prediction of HCC prognosis after surgery.

Effect of intratumoral bacteria on digestive system tumors

Microorganisms are involved in the initiation, progression, and inhibition of tumors in the digestive tract [116,117,118]. Owing to their distinct physiological characteristics, different microbes manifest differential effects on tumor survival, proliferation, and invasion [119]. The microbial communities of digestive system tumors show considerable diversity. Herein, we elaborate on the effects of specific bacteria on digestive system tumors and the respective mechanisms underlying these effects (Table 2; Fig. 3).

Table 2 Regulatory effect of typical intratumoral bacteria on digestive system tumors
Fig. 3
figure 3

Effect of intratumoral bacteria on digestive system tumors. Bacteria can modulate diverse signaling pathways or initiate DNA damage leading to carcinogenesis in cells. Chronic inflammation constitutes a notable risk factor for tumor development. Bacteria may influence inflammatory cells and can directly induce inflammation, or they may regulate immune cells and inhibit inflammation

Bacteroides fragilis

B. fragilis is a gram-negative anaerobic bacterium, primarily inhabiting the intestinal tract [151]. B. fragilis is an opportunistic pathogen comprising two distinct groups, namely non-toxigenic Bacteroides fragilis (NTBF) and enterotoxigenic Bacteroides fragilis (ETBF) [152]. NTBF can cause limited intestinal damage [153], whereas ETBF causes intestinal mucosal injury and inflammation by secreting BFT, a 20 kDa zinc-dependent metalloproteinase toxin [33, 153, 154]. Notably, intestinal tissue inflammation and damage can induce carcinogenesis, particularly CRC [152, 155].

The exposure of the mucosal tissue to BFT has been strongly implicated in the pathogenesis of CRC. Boleij et al. indicated that BFT exposure may constitute a significant risk factor for the development of CRC [33]. The authors also suggested that the incidence of BFT detection is significantly higher in patients with advanced-stage CRC compared with those with early-stage disease. Subsequently, several authors have investigated the precise mechanisms by which BFT promotes the development of CRC. The co-colonization of B. fragilis and E. coli induced interleukin (IL)-17 secretion, leading to colonic epithelial DNA damage [68]. Consequently, tumor onset was accelerated and mortality rates increased in patients with CRC. Goodwin et al. reported that BFT upregulated the expression of spermine oxidase (SMO), and reactive oxygen species (ROS), formed as a byproduct of spermine oxidation, lead to DNA damage [120]. They also observed that SMO mediated by BFT can increase the generation of SMO-dependent ROS and γ-H2A.x (a DNA damage marker). The accumulation of DNA damage promoted the uncontrolled growth of epithelial cells, eventually progressing into CRC [156]. However, Snezhkina et al. performed qPCR analysis and revealed that SMO and BFT were not significantly correlated with each other. In addition, BFT can induce proteolytic degradation of E-cadherin in the E-cadherin/β-catenin complex, consequently facilitating the nuclear translocation of β-catenin and resulting in the malignant proliferation of cells [121, 122]. The nuclear factor (NF-κB) pathway is a prototypical pro-inflammatory signaling mechanism [157]. Bacterial effector protein BFT can exert a negative effect on IκB and consequently trigger the activation of the NF-κB pathway, resulting in the induction of tumorigenesis [123,124,125].

In contrast, some investigators have reported that B. fragilis can exert antitumor effects. The bacterium can induce the upregulation of human β-defensin-2 gene expression by activating the MAPK pathway. Notably, the insufficient expression of this gene in the intestine increase susceptibility to ETBF-associated diseases, including colitis, irritable bowel disease, and colon tumorigenesis [126]. Furthermore, B. fragilis exerts a negative effect on the NLRP3-mediated pathways of inflammatory signaling by stimulating the secretion of butyrate [127]. This compound inhibits macrophage activation and secretion of pro-inflammatory mediators such as IL-18 and IL-1β, thereby reducing the levels of intestinal inflammation and restricting the development of CRC. B. fragilis can also impair tumor formation and invasion by activating CD4 + T cells, thereby inducing the production of anti-inflammatory molecules such as IL-10 [128, 129]. Taken together, these pathways can be targeted for the development of novel therapeutic regimens.

Escherichia coli

E. coli is a facultatively anaerobic gram-negative bacterium commonly present in the normal intestinal microbiota [158]. Most E. coli strains are commensals and rarely cause diseases in their host. Nevertheless, specific strains can produce toxins having genotoxic properties, which can regulate cell differentiation, apoptosis, and proliferation [159,160,161]. The possible role of these toxins in either stimulating tumor development or inhibiting tumor progression is an active area of cancer research.

E. coli possesses the pathogenic polyketide synthase (pks) genomic island, which encodes a cluster of enzymes responsible for synthesizing colibactin [130]. This peptide alkylates DNA at the adenine residues [131, 132], leading to double-stranded breaks in the host DNA and cell-cycle arrest [130]. Consequently, the uncontrolled proliferation of cells leads to malignancy [133]. Arthur et al. indicated that the removal of the pks genotoxic island from E. coli NC101 reduced tumor occurrence and invasiveness in mice, whereas no changes were observed in intestinal inflammation [162]. Liu et al. suggested that the pathogen engages with the intestinal epithelial cells through its type III secretion system and translocates the UshA protein [163]. UshA is a potent genotoxin that can degrade the intestinal epithelial cell DNA, ultimately leading to carcinogenesis [135]. Furthermore, E. coli can induce double-stranded breaks in the host DNA through copper-mediated oxidative cleavage and has been shown to promote DNA damage by inducing IL-17, ultimately resulting in tumorigenesis [68, 134]. Colibactin interacts with exchangeable copper in the gut to create a complex that coordinates with oxygen in epithelial cells to produce activated colibactin, which attacks and cleaves DNA [134]. In addition, the onset of CRC facilitates the colonization of E. coli in the hepatic tissues by impairing the integrity of the intestinal vascular barricade through virulence regulator VirF, consequently leading to liver metastases of CRC [34].

In contrast, E. coli also has anti-inflammatory and anticancer properties. Nakkarach et al. reported that E. coli secretes short-chain fatty acids (SCFA), which can inhibit the production of pro-inflammatory mediators, such as IL-1β, IL-6, IL-8, and TNF-α [136]. This downregulation may result in the inhibition of cancers linked to inflammation. However, the authors mainly examined the effect of bacteria on breast cancer, and further studies are required to evaluate their effect on CRC cells. E. coli elicits pro-apoptotic effects on CRC cells by upregulating PTEN and AKT1 [137]. Dalmasto et al. proposed an alternative perspective on the contribution of pks + E. coli in the pathogenesis of CRC [164]. The researchers infected xenografts with pks + E. coli at a multiplicity of infection of 100 and did not detect any obvious pro-proliferative effect. Moreover, they observed a reduction in tumor growth. Therefore, further investigations on the role of E. coli in tumor development hold significant potential in CRC diagnostics and therapeutics.

Fusobacterium nucleatum

F. nucleatum is an anaerobic, gram-negative, and opportunistic bacterium that colonizes both the gastrointestinal and oral tracts [165]. F. nucleatum can attach to and infiltrate endothelial and epithelial cells through its virulence factors, including adhesin A (FadA), Fusobacterium autotransporter protein 2 (Fap2), and fusobacterial outer membrane protein A (FomA) [26, 140, 142, 166]. Several authors have indicated a possible connection between F. nucleatum and the onset of carcinogenesis.

F. nucleatum can promote the initiation, proliferation, and progression of tumors by eliciting an inflammatory response and suppressing the anticancer immune response. F. nucleatum regulates microRNA-21 and cytochrome P450 monooxygenases, mainly CYP2J2, as well as its mediated product 12,13-EpOME through the TLR4 signaling pathway to facilitate the epithelial–mesenchymal transition and tumor invasion [138, 139]. Notably, patients with high microRNA-21 expression tend to have shorter survival times compared to those with low micro-RNA-21 expressions [139]. Additionally, F. nucleatum exerts its modulatory effect on the tumor immune microenvironment and promotes tumor progression by inducing NF-κB-mediated inflammation, which, in turn, facilitates the recruitment of myeloid-derived suppressor cells to the tumor microenvironment [37]. This recruitment is accompanied by inhibition of T-cell proliferation and induction of T-cell apoptosis, further underscoring the role of F. nucleatum in promoting tumor growth [141].

Immune checkpoints play a critical role in regulating the immune response and facilitating T-cell dysfunction in autoimmunity and inflammation [167, 168]. Nonetheless, these inhibitory pathways can be co-opted by neoplastic cells to facilitate tumor immune evasion [169, 170]. Gur et al. reported that the Fap2 outer surface protein of F. nucleatum curtailed antitumor immunity by binding to and inducing activation of inhibitory receptors, i.e., T-cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) and carcinoembryonic antigen-related cell adhesion molecules 1 (CEACAM1), which are expressed by T and natural killer cells [142, 143]. Interestingly, F. nucleatum triggers distinct immunological reactions in CRC cases with different MSI statuses [70]. The activation of STING signaling by F. nucleatum resulted in increased PD-L1 expression and accumulation of interferon-gamma and subsequently CD8 + tumor-infiltrating lymphocytes, leading to tumor inhibition [171]. Such effects, when combined with PD-L1 blockade treatment, enhanced tumor sensitivity and response to the immune checkpoint blockade, leading to a marked improvement in the overall survival rates of patients [171]. Overall, investigating the precise mechanism underlying the role of F. nucleatum in the development, invasion, and suppression of tumors will lead to the development of new therapeutic interventions for gastrointestinal cancers.

Other microorganisms

Apart from the bacteria mentioned above, other microorganisms also exert a distinct influence on the metabolism or microenvironment of tumors. H. pylori is a gram-negative bacterium that can selectively colonize gastric epithelium and is associated with the development of digestive system tumors [172]. The presence of H. pylori in gastrointestinal tumors may induce inflammation, thereby facilitating the proliferation of malignant cells through the activation of β-catenin or NF-kB pathways [144,145,146]. Moreover, H. pylori may contribute to the progression of GC through the upregulation of NAT10 expression, which subsequently stabilizes MDM2 mRNA [147]. Furthermore, Salmonella typhi can enhance the proliferation of tumors in the digestive system by activating the β-catenin pathway [148,149,150].

Potential applications of intratumoral bacteria in digestive system tumors

Diagnostic value of intratumoral bacteria

Intratumoral microorganisms have great potential as independent diagnostic markers because of their richness and heterogeneity in different tumors [79, 173,174,175], as well as their varying composition at different stages of the same tumor [12, 39]. For example, altered microbiome composition in Barrett’s esophagus, characterized by a decrease in the abundance of Firmicutes and an increase in the abundance of Proteobacteria, has been associated with high-grade dysplasia and esophageal adenocarcinoma [12]. Therefore, microbiota alterations in Barrett’s esophagus may be tracked for diagnosing esophageal cancer. In addition, an increase in the abundance of Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans has been linked to an increased probability of developing PC [79]. Therefore, multiple diagnostic approaches are recommended to detect any increase in the concentrations of these specific bacteria in pancreatic tissue, thereby minimizing the risk of missing the diagnosis of PC. Furthermore, in addition to the oral microbiota, the presence of fungal genera, such as Malassezia, can also be considered for detecting PC [83, 176]. Overall, identifying the microbial composition and alterations in pathological tissues can be used as an adjunct approach to the diagnosis of cancer.

Therapeutic effects of intratumoral bacteria

Despite rapid advancements in cancer diagnosis and treatment, the global burden of cancer-related deaths is rapidly increasing [2]. According to data published by WHO in 2019, cancer remained a leading cause of death before the age of 70 in most countries worldwide. Microorganisms have functional significance in the initiation and progression of gastrointestinal tumors; therefore, it is imperative to examine their potential use as therapeutic tools in cancer management (Fig. 4).

Fig. 4
figure 4

Therapeutic effect of intratumoral bacteria. Bacteria can promote or inhibit the effects of immunotherapy on the host by regulating immune cells. Bacteria possess oncolytic properties. They may serve as vectors or can be genetically modified to facilitate targeted therapeutics. Antibiotics and bacteriophages can modify the composition of intratumoral bacteria, offering potential therapeutic options for the prevention and treatment of cancer

Intratumoral microorganisms influence anticancer treatment

Currently, the modalities for cancer management include surgical intervention to excise neoplastic tissues, radiotherapy to inhibit the proliferation of tumor cells, and chemotherapy to kill malignant cells by inducing cytotoxic mechanisms or using direct chemical agents [177]. Immunotherapy has emerged as a novel approach, which boosts the immune response of the host for recognizing and eliminating tumor cells, resulting in promising therapeutic outcomes [178]. Intratumoral microorganisms can modulate the immune response within the tumor microenvironment, and consequently, these microbes can influence the effectiveness of immunotherapies for digestive system tumors [173].

Nalluri et al. evaluated intratumoral microorganisms in PC and revealed that individuals who underwent Whipple surgery or preoperative biliary stent implantation showed increased bacterial colonization in the pancreas [25]. However, the authors were not able to determine the correlation between these bacteria and the prognosis of PC. Furthermore, the influence of intestinal bacteria and fungi on the response to radiotherapy has been observed in murine models of breast cancer and melanoma [179]. Nevertheless, it remains uncertain whether these gut microorganisms are present in the digestive system tumor tissues or they exert a significant effect on the response of tumors to radiotherapy. Chemotherapy resistance occurs in neoplastic tissues partly due to the metabolic activities of intratumoral microorganisms [180]. Geller et al. suggested that intratumoral bacteria in PC can produce enzymes that metabolize the chemotherapeutic drug gemcitabine, rendering cancer cells resistant to the drug [181]. The aforementioned discovery has been thoroughly substantiated through a multitude of experimental trials [182, 183].

Introtumoral microorganisms play a positive or negative role in anti-tumor immune responses by mediating diverse immune cells and PD-1/PD-L1 axis [184, 185]. Numerous literatures have reported the facilitating role of intratumoral microorganisms in the anti-tumor immune responses. For example, Lachnospiraceae family bacteria residing in normal tissues of patients with CRC can degrade lysoglycerophospholipids which injury CD8+ T cell activity, thereby controlling the development of CRC by improving immune tumor immune surveillance [186]. E. coli is a common bacterium isolated from solid tumors including CRC. E. coli TOP10 induces the activation of CD8+ and CD4+, which are the effector cells to inhibit tumor occurrence and progression [187]. Shi et al. observed that systemic administration and local delivery of Bifidobacterium promote innate immune responses in mice with CRC [188]. Additionally, depletion of microorganisms in PDAC upregulates the expression of PD-1, which leads to positive effects on immunotherapy targeting PD-1 [22]. However, intratumoral microorganisms can impede the efficacy of anti-tumor immunotherapy. Commensal bacteria enhance the generation of immunosuppressive Treg cells and stimulate the growth of cancerous cells via metabolites such as butyrate [189, 190]. Zhang et al. discovered a correlation between intratumoral bacteria and the proportion of PD-L1 epithelial cells. Moreover, their findings illustrated that elevated levels of intratumoral Lactobacilli contributed to the formation of an immunosuppressive tumor microenvironment, which predicted the poor prognosis of ESCC [191].

Intratumoral microorganisms show therapeutic effects on cancer

Goto et al. identified the presence of intratumoral lytic bacteria that can exert anticancer effects [192]. Bacteria have high targeting properties and minimal toxicity, making them viable delivery vectors [177]. Listeria species, co-incubated with cytotoxic and labeled antibodies, can specifically target and lyse PC cells, thereby inhibiting cancer metastasis [193]. Interestingly, the use of Salmonella strains expressing Fas ligands has demonstrated notable efficacy in inducing antineoplastic responses against colon cancer [194].

Genetic engineering techniques have enabled us to modify bacterial genomes to enhance their safety, antitumor activity, and carrier function to fulfill the unique requirements of complex pathological environments. These genetically engineered bacteria have been used for treating several tumors. The attenuated strain of Salmonella typhimurium VNP20009 has shown significant anticancer properties in several animal models of cancer [195]. The bacteria negatively affected the growth of PC by inducing severe necrosis and apoptosis in a dose-dependent manner [196]. The engineered microorganisms, serving as vectors, can penetrate necrotic and hypoxic regions within tumor tissue, which is beyond the reach of normal bacteria [197]. In addition, engineered bacteria expressing immunomodulatory factors can to potentiate the immune response mediated by immune cells and cytokines, thereby inhibiting the progression and invasion of malignant tumors. Zheng et al. reported that engineered bacteria secreting FlaB can stimulate the infiltration and differentiation of immune cells via Toll-like receptors, suppressing the growth and metastasis of colon cancer [198].

Targeting intratumoral microorganisms for cancer treatment

The majority of intratumoral microorganisms inhibit the antitumor response and instead promote tumor proliferation [22, 47]. Therefore, eliminating intratumoral microorganisms may be a potential adjunct anticancer treatment. Bullman et al. administered metronidazole to mice hosting colon cancer xenografts and found a decrease in Fusobacterium load and subsequent inhibition of cancer cell proliferation and overall tumor growth [67]. The administration of a nonsteroidal anti-inflammatory drug, namely aspirin, has been reported to show direct antibacterial activity against F. nucleatum and decrease the prevalence of F. nucleatum in CRC [74]. Moreover, the treatment distinctly inhibited the promotion of intestinal tumorigenesis by F. nucleatum. Apart from bacteria, the protective effect of fungi on the progression of PDAC has been demonstrated in mouse models, indicating that targeting the mycobiome could be a promising avenue for cancer treatment [38].

However, antibiotics are not selective for intratumoral bacteria, thereby limiting their potential antitumor effects. In contrast, bacteriophages are highly specific for their target bacteria and can precisely lyse intracellular microbes [199]. Scientists injected bacteriophages specifically targeting F. nucleatum into the mice and found that bacteriophages were able to penetrate tumor tissue and infect their target bacterium [200]. Zheng et al. used phage-modified nanoparticles for intravenous or oral administration in mice suffering from CRC [201]. The authors found an enhanced chemotherapy effect and a reduction in the F. nucleatum load. Taken together, bacteriophages precisely target specific intratumoral bacteria and eliminate them, thereby providing a meticulous approach to cancer treatment.

Apart from eliminating intratumoral microorganisms through the aforementioned pathways, manipulating the composition of the microbiota accomplished by fecal microbiota transplantation is also an effective strategy to inhibit cancer growth [47]. Fecal microbiota transplantation bolsters the body’s immune response against tumors and effectively inhibits the progression of patients with CRC [202]. Consequently, therapeutic interventions focused on intratumoral microorganisms exhibit significant potential in enhancing the overall prognosis of individuals diagnosed with digestive system tumors.

Prognostic potential of intratumoral bacteria

A statistically significant association exists between intratumoral microorganisms and survival and mortality rates in patients with cancer [191, 203]. Therefore, the presence of intratumoral microorganisms can be used as a prognostic tool for predicting the outcome in cancer patients. The mucosal microbiome exhibits dynamic association with CRC and thus can be explored for developing microbiota-based prognostic approaches for CRC [61]. The prevalence of F. nucleatum simultaneously increases with tumor progression [60]. Additionally, a higher concentration of F. nucleatum DNA in CRC tissue is linked to decreased survival rates [64]. F. nucleatum has been historically linked to unfavorable prognostic outcomes and is significantly associated with negative prognosis in colorectal carcinomas, as well as esophageal and pancreatic malignancies [32, 48, 50]. However, Oh et al. challenged this assumption by showing that the influence of F. nucleatum on the prognosis of CRC depends on other determining factors [204]. They indicated that the positive prognostic impact of F. nucleatum was solely detected in sub-categories of non-sigmoid carcinoma patients with high levels of non-MSI. Therefore, they concluded that the position of the tumor and the combined status of MSI may play a crucial role in influencing the diverse prognostic impact of F. nucleatum in patients with CRC undergoing adjuvant chemotherapy. In addition, several other microorganisms are closely associated with the prognosis of gastrointestinal malignancies. H. pylori releases CagA through its type IV secretion system, thereby resulting in increased susceptibility and unfavorable clinical outcomes in patients diagnosed with gastric or colorectal cancers [205, 206]. Streptococcus and Prevotella are more abundant in patients with ESCC having lymph node metastasis [49]. The presence of these microorganisms may be an independent predictive indicator for the prognosis of patients with ESCC. Overall, the detection of microorganisms in digestive system tumors can predict the prognosis of patients, thereby guiding timely modifications in treatment strategies for achieving favorable outcomes in affected patients.

Intratumoral microorganisms as tumor biomarkers exhibit significant potential in advancing the development of more efficacious therapeutic approaches and prognosis prediction models. Hermida et al. reported that RNA-seq and whole-genome sequencing provided by The Cancer Genome Atlas documented not only extensive information on thousands of cancer cases but also genetic information from intratumoral microorganisms [207]. Furthermore, they suggested that combining information about tumor gene expression and differences between the microbes in the tumor and those in non-tumor tissue can predict cancer prognosis and drug response. Additionally, Sun et al. indicated hepototype distinguished by differences in intratumoral microorganisms is demonstrated to be an independent prognostic factors for patients with postoperative HCC [89]. In their study, high level of Akkermansia and Methylobacterium is associated with favorable prognosis and can be used to construct clinical predictive models.

Conclusion

Currently, intratumoral microorganisms are being actively investigated in several types of tumors and some remarkable results have been obtained in this context. Their diversity and functions have been described in detail using next-generation sequencing technology. Here, we reviewed the fundamental features, sources, types, and heterogeneity of microorganisms present in digestive system tumors. In addition, we summarized the role of some typical intratumoral microorganisms present in specific tumors. Finally, we elaborated on the potential use of tumor-associated microorganisms for developing novel diagnosis and treatment strategies for digestive system tumors. This information will be valuable for further investigating the role of microorganisms in neoplastic growth and developing microbial therapy for neoplasms.

Availability of data and materials

No original datasets were generated for this review. All data supporting the information given here can be found in the references cited within the paper.

References

  1. Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, et al. The 2019 WHO classification of tumours of the digestive system. Histopathology. 2020;76(2):182–8.

    Article  PubMed  Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. Cancer J Clin. 2021;71(3):209–49.

    Article  Google Scholar 

  3. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. Cancer J Clin. 2022;72(1):7–33.

    Article  Google Scholar 

  4. Matson V, Chervin CS, Gajewski TF. Cancer and the Microbiome-Influence of the commensal microbiota on Cancer, Immune responses, and Immunotherapy. Gastroenterology. 2021;160(2):600–13.

    Article  CAS  PubMed  Google Scholar 

  5. Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human gut microbiota and gastrointestinal Cancer. Genom Proteom Bioinform. 2018;16(1):33–49.

    Article  CAS  Google Scholar 

  6. Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, et al. Metagenomic analysis of the human distal gut microbiome. Science. 2006;312(5778):1355–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Garrett WS. Cancer and the microbiota. Science. 2015;348(6230):80–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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.

    Article  CAS  PubMed  Google Scholar 

  9. Gagliani N, Hu B, Huber S, Elinav E, Flavell RA. The Fire within: microbes inflame tumors. Cell. 2014;157(4):776–83.

    Article  CAS  PubMed  Google Scholar 

  10. Okuda S, Shimada Y, Tajima Y, Yuza K, Hirose Y, Ichikawa H, et al. Profiling of host genetic alterations and intra-tumor microbiomes in Colorectal cancer. Comput Struct Biotechnol J. 2021;19:3330–8.

    Article  CAS  PubMed  Google Scholar 

  11. 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(4):795–806e712.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Snider EJ, Compres G, Freedberg DE, Khiabanian H, Nobel YR, Stump S, et al. Alterations to the Esophageal Microbiome Associated with Progression from Barrett’s Esophagus to Esophageal Adenocarcinoma. Cancer Epidemiol Biomarkers Prev. 2019;28(10):1687–93.

    Article  PubMed  Google Scholar 

  13. Xuan SY, Li N, Qiang X, Zhou RR, Shi YX, Jiang WJ. Helicobacter Infection in hepatocellular carcinoma tissue. World J Gastroenterol. 2006;12(15):2335–40.

    Article  CAS  PubMed  Google Scholar 

  14. 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(1):100884.

    Article  CAS  PubMed  Google Scholar 

  15. Liu YX, Qin Y, Chen T, Lu M, Qian X, Guo X, et al. A practical guide to amplicon and metagenomic analysis of microbiome data. Protein Cell. 2021;12(5):315–30.

    Article  PubMed  Google Scholar 

  16. Calgaro M, Romualdi C, Waldron L, Risso D, Vitulo N. Assessment of statistical methods from single cell, bulk RNA-seq, and metagenomics applied to microbiome data. Genome Biol. 2020;21(1):191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bharti R, Grimm DG. Current challenges and best-practice protocols for microbiome analysis. Brief Bioinform. 2021;22(1):178–93.

    Article  CAS  PubMed  Google Scholar 

  18. Wensel CR, Pluznick JL, Salzberg SL, Sears CL. Next-generation sequencing: insights to advance clinical investigations of the microbiome. J Clin Investig. 2022;132(7):e154944.

  19. Desmarais SM, De Pedro MA, Cava F, Huang KC. Peptidoglycan at its peaks: how chromatographic analyses can reveal bacterial cell wall structure and assembly. Mol Microbiol. 2013;89(1):1–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Xie Y, Xie F, Zhou X, Zhang L, Yang B, Huang J, et al. Microbiota in Tumors: From Understanding to Application. Advanced science.  (Weinheim, Baden-Wurttemberg, Germany). 2022;9(21):e2200470.

    PubMed  Google Scholar 

  21. Tjalsma H, Boleij A, Marchesi JR, Dutilh BE. A bacterial driver-passenger model for Colorectal cancer: beyond the usual suspects. Nat Rev Microbiol. 2012;10(8):575–82.

    Article  CAS  PubMed  Google Scholar 

  22. 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(4):403–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nilsson HO, Stenram U, Ihse I, Wadstrom T. Helicobacter species ribosomal DNA in the pancreas, stomach and duodenum of Pancreatic cancer patients. World J Gastroenterol. 2006;12(19):3038–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Thomas RM, Gharaibeh RZ, Gauthier J, Beveridge M, Pope JL, Guijarro MV, et al. Intestinal microbiota enhances pancreatic carcinogenesis in preclinical models. Carcinogenesis. 2018;39(8):1068–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Nalluri H, Jensen E, Staley C. Role of biliary stent and neoadjuvant chemotherapy in the pancreatic Tumor microbiome. BMC Microbiol. 2021;21(1):280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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(2):215–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang J, Wang Y, Li Z, Gao X, Huang D. Global Analysis of Microbiota Signatures in four major types of gastrointestinal Cancer. Front Oncol. 2021;11:685641.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Wang Y, Guo H, Gao X, Wang J. The Intratumor Microbiota signatures Associate with Subtype, Tumor Stage, and Survival Status of Esophageal Carcinoma. Front Oncol. 2021;11:754788.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Senchukova MA, Tomchuk O, Shurygina EI. Helicobacter pylori in gastric cancer: features of Infection and their correlations with long-term results of treatment. World J Gastroenterol. 2021;27(37):6290–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Huang Y, Fan XG, Wang ZM, Zhou JH, Tian XF, Li N. Identification of helicobacter species in human liver samples from patients with primary hepatocellular carcinoma. J Clin Pathol. 2004;57(12):1273–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yu J, Chen Y, Fu X, Zhou X, Peng Y, Shi L, et al. Invasive Fusobacterium nucleatum may play a role in the carcinogenesis of proximal colon Cancer through the serrated neoplasia pathway. Int J Cancer. 2016;139(6):1318–26.

    Article  CAS  PubMed  Google Scholar 

  32. Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, et al. Association of Fusobacterium species in Pancreatic cancer tissues with molecular features and prognosis. Oncotarget. 2015;6(9):7209–20.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Boleij A, Hechenbleikner EM, Goodwin AC, Badani R, Stein EM, Lazarev MG, et al. The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of Colorectal cancer patients. Clin Infect Diseases: Official Publication Infect Dis Soc Am. 2015;60(2):208–15.

    Article  CAS  Google Scholar 

  34. Bertocchi A, Carloni S, Ravenda PS, Bertalot G, Spadoni I, Lo Cascio A, et al. Gut vascular barrier impairment leads to intestinal bacteria dissemination and Colorectal cancer Metastasis to liver. Cancer Cell. 2021;39(5):708–724e711.

    Article  CAS  PubMed  Google Scholar 

  35. Rodriguez RM, Hernandez BY, Menor M, Deng Y, Khadka VS. The landscape of bacterial presence in Tumor and adjacent normal tissue across 9 major cancer types using TCGA exome sequencing. Comput Struct Biotechnol J. 2020;18:631–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yuan X, Liu Y, Kong J, Gu B, Qi Y, Wang X, et al. Different frequencies of Porphyromonas gingivalis Infection in cancers of the upper digestive tract. Cancer Lett. 2017;404:1–7.

    Article  CAS  PubMed  Google Scholar 

  37. Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14(2):207–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. An Y, Zhang W, Liu T, Wang B, Cao H. The intratumoural microbiota in cancer: new insights from inside. Biochimica et biophysica acta. Reviews on cancer. 2021;1876(2):188626.

    CAS  PubMed  Google Scholar 

  39. Yu G, Torres J, Hu N, Medrano-Guzman R, Herrera-Goepfert R, Humphrys MS, et al. Molecular characterization of the human stomach microbiota in gastric Cancer patients. Front Cell Infect Microbiol. 2017;7:302.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Qu D, Wang Y, Xia Q, Chang J, Jiang X, Zhang H. Intratumoral Microbiome of Human Primary Liver Cancer. Hepatol Commun. 2022;6(7):1741–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. O’Callaghan A, van Sinderen D. Bifidobacteria and their role as members of the human gut microbiota. Front Microbiol. 2016;7:925.

    PubMed  PubMed Central  Google Scholar 

  42. Wang IK, Lai HC, Yu CJ, Liang CC, Chang CT, Kuo HL, et al. Real-time PCR analysis of the intestinal microbiotas in peritoneal dialysis patients. Appl Environ Microbiol. 2012;78(4):1107–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wang M, Yu F, Li P. Intratumor Microbiota in cancer pathogenesis and immunity: from mechanisms of action to therapeutic opportunities. Front Immunol. 2023;14:1269054.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Starnes CO. Coley’s toxins in perspective. Nature. 1992;357(6373):11–2.

    Article  CAS  PubMed  Google Scholar 

  45. Livingston-Wheeler therapy. Cancer J Clin. 1990;40(2):103–8.

    Article  Google Scholar 

  46. White MK, Pagano JS, Khalili K. Viruses and human cancers: a long road of discovery of molecular paradigms. Clin Microbiol Rev. 2014;27(3):463–81.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, Knight R. The microbiome and human cancer. Science. 2021;371(6536):eabc4552.

  48. 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 Research: Official J Am Association Cancer Res. 2016;22(22):5574–81.

    Article  CAS  Google Scholar 

  49. Liu Y, Lin Z, Lin Y, Chen Y, Peng XE, He F, et al. Streptococcus and Prevotella are associated with the prognosis of oesophageal squamous cell carcinoma. J Med Microbiol. 2018;67(8):1058–68.

    Article  CAS  PubMed  Google Scholar 

  50. Yamamura K, Izumi D, Kandimalla R, Sonohara F, Baba Y, Yoshida N, et al. Intratumoral Fusobacterium Nucleatum levels predict therapeutic response to Neoadjuvant Chemotherapy in Esophageal squamous cell carcinoma. Clin cancer Research: Official J Am Association Cancer Res. 2019;25(20):6170–9.

    Article  CAS  Google Scholar 

  51. Liu Y, Baba Y, Ishimoto T, Tsutsuki H, Zhang T, Nomoto D, et al. Fusobacterium nucleatum confers chemoresistance by modulating autophagy in oesophageal squamous cell carcinoma. Br J Cancer. 2021;124(5):963–74.

    Article  CAS  PubMed  Google Scholar 

  52. Li Z, Shi C, Zheng J, Guo Y, Fan T, Zhao H, et al. Fusobacterium nucleatum predicts a high risk of Metastasis for esophageal squamous cell carcinoma. BMC Microbiol. 2021;21(1):301.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Wang L, Zhou J, Xin Y, Geng C, Tian Z, Yu X, et al. Bacterial overgrowth and diversification of microbiota in gastric cancer. Eur J Gastroenterol Hepatol. 2016;28(3):261–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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.

    Article  CAS  PubMed  Google Scholar 

  55. 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.

    Article  PubMed  Google Scholar 

  56. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  57. 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(2):299–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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(2):292–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Warren RL, Freeman DJ, Pleasance S, Watson P, Moore RA, Cochrane K, et al. Co-occurrence of anaerobic bacteria in colorectal carcinomas. Microbiome. 2013;1(1):16.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Flanagan L, Schmid J, Ebert M, Soucek P, Kunicka T, Liska V, et al. Fusobacterium nucleatum associates with stages of colorectal neoplasia development, Colorectal cancer and Disease outcome. Eur J Clin Microbiol Infect Dis. 2014;33(8):1381–90.

    Article  CAS  Google Scholar 

  61. Gao Z, Guo B, Gao R, Zhu Q, Qin H. Microbiota disbiosis is associated with Colorectal cancer. Front Microbiol. 2015;6:20.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Viljoen KS, Dakshinamurthy A, Goldberg P, Blackburn JM. Quantitative profiling of colorectal cancer-associated bacteria reveals associations between fusobacterium spp., enterotoxigenic Bacteroides fragilis (ETBF) and clinicopathological features of Colorectal cancer. PLoS ONE. 2015;10(3):e0119462.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Burns MB, Lynch J, Starr TK, Knights D, Blekhman R. Virulence genes are a signature of the microbiome in the colorectal Tumor microenvironment. Genome Med. 2015;7(1):55.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, Nowak JA, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016;65(12):1973–80.

    Article  CAS  PubMed  Google Scholar 

  65. Flemer B, Lynch DB, Brown JM, Jeffery IB, Ryan FJ, Claesson MJ, et al. Tumour-associated and non-tumour-associated microbiota in Colorectal cancer. Gut. 2017;66(4):633–43.

    Article  CAS  PubMed  Google Scholar 

  66. Park HE, Kim JH, Cho NY, Lee HS, Kang GH. Intratumoral Fusobacterium nucleatum abundance correlates with macrophage infiltration and CDKN2A methylation in microsatellite-unstable colorectal carcinoma. Virchows Archiv: An International Journal of Pathology. 2017;471(3):329–36.

    Article  CAS  PubMed  Google Scholar 

  67. Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, et al. Analysis of Fusobacterium persistence and antibiotic response in Colorectal cancer. Science. 2017;358(6369):1443–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Dejea CM, Fathi P, Craig JM, Boleij A, Taddese R, Geis AL, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359(6375):592–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Nakatsu G, Zhou H, Wu WKK, Wong SH, Coker OO, Dai Z, et al. Alterations in enteric virome are Associated with Colorectal Cancer and Survival outcomes. Gastroenterology. 2018;155(2):529–541e525.

    Article  PubMed  Google Scholar 

  70. Hamada T, Zhang X, Mima K, Bullman S, Sukawa Y, Nowak JA, et al. Fusobacterium nucleatum in Colorectal Cancer relates to Immune Response differentially by Tumor microsatellite instability status. Cancer Immunol Res. 2018;6(11):1327–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. 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(4):654–62.

    Article  CAS  PubMed  Google Scholar 

  72. Chen Y, Lu Y, Ke Y, Li Y. Prognostic impact of the Fusobacterium nucleatum status in colorectal cancers. Medicine. 2019;98(39):e17221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Liu W, Zhang X, Xu H, Li S, Lau HC, Chen Q, et al. Microbial Community Heterogeneity within Colorectal Neoplasia and its correlation with colorectal carcinogenesis. Gastroenterology. 2021;160(7):2395–408.

    Article  CAS  PubMed  Google Scholar 

  74. Brennan CA, Nakatsu G, Gallini Comeau CA, Drew DA, Glickman JN, Schoen RE et al. Aspirin modulation of the Colorectal Cancer-Associated Microbe Fusobacterium nucleatum. mBio. 2021;12(2):e00547.

  75. Liu NN, Jiao N, Tan JC, Wang Z, Wu D, Wang AJ, et al. Multi-kingdom microbiota analyses identify bacterial-fungal interactions and biomarkers of Colorectal cancer across cohorts. Nat Microbiol. 2022;7(2):238–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Coker OO, Liu C, Wu WKK, Wong SH, Jia W, Sung JJY, et al. Altered gut metabolites and microbiota interactions are implicated in colorectal carcinogenesis and can be non-invasive diagnostic biomarkers. Microbiome. 2022;10(1):35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Lo CH, Wu DC, Jao SW, Wu CC, Lin CY, Chuang CH, et al. Enrichment of Prevotella intermedia in human Colorectal cancer and its additive effects with Fusobacterium nucleatum on the malignant transformation of colorectal adenomas. J Biomed Sci. 2022;29(1):88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Jesnowski R, Isaksson B, Möhrcke C, Bertsch C, Bulajic M, Schneider-Brachert W, et al. Helicobacter pylori in autoimmune pancreatitis and pancreatic carcinoma. Pancreatology. : Official journal of the International Association of Pancreatology (IAP) [et al]. 2010;10(4):462–6.

    Article  PubMed  Google Scholar 

  79. Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, et al. Human oral microbiome and prospective risk for Pancreatic cancer: a population-based nested case-control study. Gut. 2018;67(1):120–7.

    Article  CAS  PubMed  Google Scholar 

  80. 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(6356):1156–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Zhu Y, He C, Li X, Cai Y, Hu J, Liao Y, et al. Gut microbiota dysbiosis worsens the severity of acute Pancreatitis in patients and mice. J Gastroenterol. 2019;54(4):347–58.

    Article  CAS  PubMed  Google Scholar 

  82. Goel N, Nadler A, Reddy S, Hoffman JP, Pitt HA. Biliary microbiome in Pancreatic cancer: alterations with neoadjuvant therapy. HPB: The Official Journal of the International Hepato Pancreato Biliary Association. 2019;21(12):1753–60.

    Article  PubMed  Google Scholar 

  83. Aykut B, Pushalkar S, Chen R, Li Q, Abengozar R, Kim JI, et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature. 2019;574(7777):264–7.

    Article  CAS  PubMed  Google Scholar 

  84. Vogtmann E, Han Y, Caporaso JG, Bokulich N, Mohamadkhani A, Moayyedkazemi A, et al. Oral microbial community composition is associated with Pancreatic cancer: a case-control study in Iran. Cancer Med. 2020;9(2):797–806.

    Article  CAS  PubMed  Google Scholar 

  85. Jeong JY, Kim TB, Kim J, Choi HW, Kim EJ, Yoo HJ et al. Diversity in the Extracellular vesicle-derived Microbiome of tissues according to Tumor Progression in Pancreatic Cancer. Cancers. 2020;12(9):2346.

  86. Langheinrich M, Wirtz S, Kneis B, Gittler MM, Tyc O, Schierwagen R et al. Microbiome patterns in matched bile, duodenal, pancreatic Tumor tissue, drainage, and Stool Samples: Association with Preoperative Stenting and postoperative pancreatic Fistula Development. J Clin Med. 2020;9(9):2785.

  87. Kohi S, Macgregor-Das A, Dbouk M, Yoshida T, Chuidian M, Abe T, et al. Alterations in the duodenal fluid microbiome of patients with Pancreatic Cancer. Clinical gastroenterology and hepatology: the official clinical practice. J Am Gastroenterological Association. 2022;20(2):e196–e227.

    CAS  Google Scholar 

  88. Huang JH, Wang J, Chai XQ, Li ZC, Jiang YH, Li J, et al. The Intratumoral Bacterial Metataxonomic signature of Hepatocellular Carcinoma. Microbiol Spectr. 2022;10(5):e0098322.

    Article  PubMed  Google Scholar 

  89. Sun L, Ke X, Guan A, Jin B, Qu J, Wang Y, et al. Intratumoural microbiome can predict the prognosis of hepatocellular carcinoma after Surgery. Clin Translational Med. 2023;13(7):e1331.

    Article  Google Scholar 

  90. Abnet CC, Arnold M, Wei WQ. Epidemiology of esophageal squamous cell carcinoma. Gastroenterology. 2018;154(2):360–73.

    Article  PubMed  Google Scholar 

  91. Rustgi AK, El-Serag HB. Esophageal carcinoma. N Engl J Med. 2014;371(26):2499–509.

    Article  PubMed  Google Scholar 

  92. Arnold M, Soerjomataram I, Ferlay J, Forman D. Global incidence of oesophageal cancer by histological subtype in 2012. Gut. 2015;64(3):381–7.

    Article  PubMed  Google Scholar 

  93. Beydoun AS, Stabenau KA, Altman KW., and Johnston N. Cancer Risk in Barrett’s Esophagus: A Clinical Review. Int J Mol Sci. 2023;24(7):6018.

  94. Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet (London England). 2020;396(10251):635–48.

    Article  CAS  PubMed  Google Scholar 

  95. Infection with Helicobacter pylori. IARC monographs on the evaluation of carcinogenic risks to humans. 1994;61:177–240.

  96. Plummer M, Franceschi S, Vignat J, Forman D, de Martel C. Global burden of gastric cancer attributable to Helicobacter pylori. Int J Cancer. 2015;136(2):487–90.

    Article  CAS  PubMed  Google Scholar 

  97. Koshiol J, Wei WQ, Kreimer AR, Ren JS, Gravitt P, Chen W et al. The gastric cardia is not a target for human papillomavirus-induced carcinogenesis. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2010;19(4):1137–9.

  98. Kamangar F, Dawsey SM, Blaser MJ, Perez-Perez GI, Pietinen P, Newschaffer CJ, et al. Opposing risks of gastric cardia and noncardia gastric adenocarcinomas associated with Helicobacter pylori seropositivity. J Natl Cancer Inst. 2006;98(20):1445–52.

    Article  PubMed  Google Scholar 

  99. Wang Z, Dan W, Zhang N, Fang J, Yang Y. Colorectal cancer and gut microbiota studies in China. Gut Microbes. 2023;15(1):2236364.

    Article  PubMed  Google Scholar 

  100. Tilg H, Adolph TE, Gerner RR, Moschen AR. The intestinal microbiota in Colorectal Cancer. Cancer Cell. 2018;33(6):954–64.

    Article  CAS  PubMed  Google Scholar 

  101. Cheng Y, Ling Z, Li L. The intestinal microbiota and Colorectal Cancer. Front Immunol. 2020;11:615056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Halbrook CJ, Lyssiotis CA, Pasca di Magliano M, Maitra A. Pancreatic cancer: advances and challenges. Cell. 2023;186(8):1729–54.

    Article  CAS  PubMed  Google Scholar 

  103. Siegel RL, Miller KD, Jemal A, Cancer statistics, 2019. CA: a cancer journal for clinicians. 2019;69(1):7–34.

  104. Kaźmierczak-Siedlecka K, Dvořák A, Folwarski M, Daca A, Przewłócka K. and Makarewicz W. Fungal gut microbiota dysbiosis and its role in colorectal, oral, and pancreatic carcinogenesis. Cancers. 2020;12(5):1326.

  105. Kanwal F, Singal AG. Surveillance for Hepatocellular Carcinoma: current best practice and future direction. Gastroenterology. 2019;157(1):54–64.

    Article  PubMed  Google Scholar 

  106. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al. Hepatocellular carcinoma. Nature reviews. Disease Primers. 2016;2:16018.

    Article  PubMed  Google Scholar 

  107. Villanueva A. Hepatocellular Carcinoma. N Engl J Med. 2019;380(15):1450–62.

    Article  CAS  PubMed  Google Scholar 

  108. Akinyemiju T, Abera S, Ahmed M, Alam N, Alemayohu MA, Allen C, et al. The Burden of Primary Liver Cancer and underlying etiologies from 1990 to 2015 at the Global, Regional, and National Level: results from the global burden of Disease Study 2015. JAMA Oncol. 2017;3(12):1683–91.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Albillos A, de Gottardi A, Rescigno M. The gut-liver axis in Liver Disease: pathophysiological basis for therapy. J Hepatol. 2020;72(3):558–77.

    Article  CAS  PubMed  Google Scholar 

  110. Tilg H, Adolph TE, Trauner M. Gut-liver axis: pathophysiological concepts and clinical implications. Cell Metabol. 2022;34(11):1700–18.

    Article  CAS  Google Scholar 

  111. Rao Y, Kuang Z, Li C, Guo S, Xu Y, Zhao D, et al. Gut Akkermansia muciniphila ameliorates metabolic dysfunction-associated fatty Liver Disease by regulating the metabolism of L-aspartate via gut-liver axis. Gut Microbes. 2021;13(1):1–19.

    Article  PubMed  Google Scholar 

  112. Chen Q, Zhao L, Mei L, Zhao X, Han P, Liu J, et al. Vitamin C and vitamin D(3) alleviate metabolic-associated fatty Liver Disease by regulating the gut microbiota and bile acid metabolism via the gut-liver axis. Front Pharmacol. 2023;14:1163694.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Shek D, Chen D, Read SA, Ahlenstiel G. Examining the gut-liver axis in Liver cancer using organoid models. Cancer Lett. 2021;510:48–58.

    Article  CAS  PubMed  Google Scholar 

  114. Sookoian S, Salatino A, Castaño GO, Landa MS, Fijalkowky C, Garaycoechea M, et al. Intrahepatic bacterial metataxonomic signature in non-alcoholic fatty Liver Disease. Gut. 2020;69(8):1483–91.

    Article  CAS  PubMed  Google Scholar 

  115. He Y, Zhang Q, Yu X, Zhang S, Guo W. Overview of microbial profiles in human hepatocellular carcinoma and adjacent nontumor tissues. J Translational Med. 2023;21(1):68.

    Article  CAS  Google Scholar 

  116. Abreu MT, Peek RM Jr. Gastrointestinal malignancy and the microbiome. Gastroenterology. 2014;146(6):1534–1546e1533.

    Article  CAS  PubMed  Google Scholar 

  117. LaCourse KD, Johnston CD, Bullman S. The relationship between gastrointestinal cancers and the microbiota. The lancet. Gastroenterol Hepatol. 2021;6(6):498–509.

    Google Scholar 

  118. Wroblewski LE, Peek RM Jr., Coburn LA. The role of the Microbiome in Gastrointestinal Cancer. Gastroenterol Clin N Am. 2016;45(3):543–56.

    Article  Google Scholar 

  119. Lam SY, Yu J, Wong SH, Peppelenbosch MP, Fuhler GM. The gastrointestinal microbiota and its role in oncogenesis. Best practice & research. Clin Gastroenterol. 2017;31(6):607–18.

    CAS  Google Scholar 

  120. Goodwin AC, Destefano Shields CE, Wu S, Huso DL, Wu X, Murray-Stewart TR, et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc Natl Acad.  Sci U S A. 2011;108(37):15354–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Wu S, Rhee KJ, Zhang M, Franco A, Sears CL. Bacteroides fragilis toxin stimulates intestinal epithelial cell shedding and gamma-secretase-dependent E-cadherin cleavage. J Cell Sci. 2007;120(Pt 11):1944–52.

    Article  CAS  PubMed  Google Scholar 

  122. Wu S, Morin PJ, Maouyo D, Sears CL. Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation. Gastroenterology. 2003;124(2):392–400.

    Article  CAS  PubMed  Google Scholar 

  123. Kim JM, Jung HY, Lee JY, Youn J, Lee CH, Kim KH. Mitogen-activated protein kinase and activator protein-1 dependent signals are essential for Bacteroides fragilis enterotoxin-induced enteritis. Eur J Immunol. 2005;35(9):2648–57.

    Article  CAS  PubMed  Google Scholar 

  124. 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.

    Article  CAS  PubMed  Google Scholar 

  125. Wu S, Powell J, Mathioudakis N, Kane S, Fernandez E, Sears CL. Bacteroides fragilis enterotoxin induces intestinal epithelial cell secretion of interleukin-8 through mitogen-activated protein kinases and a tyrosine kinase-regulated nuclear factor-kappab pathway. Infect Immun. 2004;72(10):5832–9.

    Article  CAS  PubMed  Google Scholar 

  126. Yoon YM, Lee JY, Yoo D, Sim YS, Kim YJ, Oh YK, et al. Bacteroides fragilis enterotoxin induces human beta-defensin-2 expression in intestinal epithelial cells via a mitogen-activated protein kinase/I kappaB kinase/NF-kappaB-dependent pathway. Infect Immun. 2010;78(5):2024–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Shao X, Sun S, Zhou Y, Wang H, Yu Y, Hu T, et al. Bacteroides fragilis restricts colitis-associated cancer via negative regulation of the NLRP3 axis. Cancer Lett. 2021;523:170–81.

    Article  CAS  PubMed  Google Scholar 

  128. Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, et al. The toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science. 2011;332(6032):974–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122(1):107–18.

    Article  CAS  PubMed  Google Scholar 

  130. Nougayrède JP, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, Gottschalk G, et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 2006;313(5788):848–51.

    Article  PubMed  Google Scholar 

  131. Wilson MR, Jiang Y, Villalta PW, Stornetta A, Boudreau PD, Carrá A et al. The human gut bacterial genotoxin colibactin alkylates DNA. Science. 2019;363(6428):eaar7785.

  132. Xue M, Kim CS, Healy AR, Wernke KM, Wang Z, Frischling MC et al. Structure elucidation of colibactin and its DNA cross-links. Science. 2019;365(6457):eaax2685.

  133. Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A, van Hoeck A, Wood HM, Nomburg J, et al. Mutational signature in Colorectal cancer caused by genotoxic pks(+) E. coli. Nature. 2020;580(7802):269–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Li ZR, Li J, Cai W, Lai JY, H, McKinnie SMK, Zhang WP, et al. Macrocyclic colibactin induces DNA double-strand breaks via copper-mediated oxidative cleavage. Nat Chem. 2019;11(10):880–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Liu Y, Fu K, Wier EM, Lei Y, Hodgson A, Xu D, et al. Bacterial genotoxin accelerates transient infection-driven murine Colon tumorigenesis. Cancer Discov. 2022;12(1):236–49.

    Article  PubMed  Google Scholar 

  136. Nakkarach A, Foo HL, Song AA, Mutalib NEA, Nitisinprasert S, Withayagiat U. Anti-cancer and anti-inflammatory effects elicited by short chain fatty acids produced by Escherichia coli isolated from healthy human gut microbiota. Microb Cell Fact. 2021;20(1):36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Alizadeh S, Esmaeili A, Omidi Y. Anti-cancer properties of Escherichia coli Nissle 1917 against HT-29 colon Cancer cells through regulation of Bax/Bcl-xL and AKT/PTEN signaling pathways. Iran J Basic Med Sci. 2020;23(7):886–93.

  138. 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):851–866e824.

    Article  CAS  PubMed  Google Scholar 

  139. Kong C, Yan X, Zhu Y, Zhu H, Luo Y, Liu P, et al. Fusobacterium Nucleatum promotes the development of Colorectal Cancer by activating a cytochrome P450/Epoxyoctadecenoic Acid Axis via TLR4/Keap1/NRF2 signaling. Cancer Res. 2021;81(17):4485–98.

    Article  CAS  PubMed  Google Scholar 

  140. 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(2):195–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Mima K, Sukawa Y, Nishihara R, Qian ZR, Yamauchi M, Inamura K, et al. Fusobacterium nucleatum and T cells in Colorectal Carcinoma. JAMA Oncol. 2015;1(5):653–61.

    Article  PubMed  PubMed Central  Google Scholar 

  142. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Gur C, Maalouf N, Shhadeh A, Berhani O, Singer BB, Bachrach G, et al. Fusobacterium nucleatum supresses anti-tumor immunity by activating CEACAM1. Oncoimmunology. 2019;8(6):e1581531.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Franco AT, Israel DA, Washington MK, Krishna U, Fox JG, Rogers AB, et al. Activation of beta-catenin by carcinogenic Helicobacter pylori. Proc Natl.  Acad Sci U S A. 2005;102(30):10646–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Nagy TA, Wroblewski LE, Wang D, Piazuelo MB, Delgado A, Romero-Gallo J, et al. β-Catenin and p120 mediate PPARδ-dependent proliferation induced by Helicobacter pylori in human and rodent epithelia. Gastroenterology. 2011;141(2):553–64.

    Article  CAS  PubMed  Google Scholar 

  146. Brandt S, Kwok T, Hartig R, König W, Backert S. NF-kappaB activation and potentiation of proinflammatory responses by the Helicobacter pylori CagA protein. Proc Natl Acad.  Sci U S A. 2005;102(26):9300–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Deng M, Zhang L, Zheng W, Chen J, Du N, Li M, et al. Correction: Helicobacter pylori-induced NAT10 stabilizes MDM2 mRNA via RNA acetylation to facilitate gastric cancer progression. J Experimental Clin cancer Research: CR. 2023;42(1):98.

    Article  PubMed Central  Google Scholar 

  148. Liu X, Lu R, Wu S, Sun J. Salmonella regulation of intestinal stem cells through the Wnt/beta-catenin pathway. FEBS Lett. 2010;584(5):911–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Lu R, Liu X, Wu S, Xia Y, Zhang YG, Petrof EO, et al. Consistent activation of the β-catenin pathway by Salmonella type-three secretion effector protein AvrA in chronically infected intestine. Am J Physiol Gastrointest Liver Physiol. 2012;303(10):G1113–1125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Lu R, Wu S, Zhang YG, Xia Y, Liu X, Zheng Y, et al. Enteric bacterial protein AvrA promotes colonic tumorigenesis and activates colonic beta-catenin signaling pathway. Oncogenesis. 2014;3(6):e105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Cheng WT, Kantilal HK, Davamani F. The mechanism of Bacteroides fragilis Toxin contributes to Colon Cancer formation. Malaysian J Med Sciences: MJMS. 2020;27(4):9–21.

    Article  Google Scholar 

  152. Snezhkina AV, Krasnov GS, Lipatova AV, Sadritdinova AF, Kardymon OL, Fedorova MS, et al. The dysregulation of Polyamine Metabolism in Colorectal Cancer is Associated with overexpression of c-Myc and C/EBPβ rather than Enterotoxigenic Bacteroides fragilis Infection. Oxidative Med Cell Longev. 2016;2016:2353560.

    Article  Google Scholar 

  153. Pierce JV, Bernstein HD. Genomic Diversity of Enterotoxigenic Strains of Bacteroides fragilis. PLoS ONE. 2016;11(6):e0158171.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Sears CL, Geis AL, Housseau F. Bacteroides fragilis subverts mucosal biology: from symbiont to colon carcinogenesis. J Clin Investig. 2014;124(10):4166–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Lv Y, Ye T, Wang HP, Zhao JY, Chen WJ, Wang X, et al. Suppression of colorectal tumorigenesis by recombinant Bacteroides fragilis enterotoxin-2 in vivo. World J Gastroenterol. 2017;23(4):603–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Rivas-Domínguez A, Pastor N, Martínez-López L, Colón-Pérez J, Bermúdez B, Orta ML. The role of DNA damage response in Dysbiosis-Induced Colorectal Cancer. Cells. 2021;10(8):1934.

  157. Chiu WT, Patrick WK, Asia Pacific Academic Consortium for Public Health APACPH Public Health Recognition Award. Asia Pac J Public Health. 2010;22(2):256–7.

    Article  PubMed  Google Scholar 

  158. Blount ZD. The unexhausted potential of E. coli. eLife. 2015;4:e05826.

  159. Collins D, Hogan AM, Winter DC. Microbial and viral pathogens in Colorectal cancer. Lancet Oncol. 2011;12(5):504–12.

    Article  CAS  PubMed  Google Scholar 

  160. Chassaing B, Darfeuille-Michaud A. The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel Diseases. Gastroenterology. 2011;140(6):1720–8.

    Article  PubMed  Google Scholar 

  161. Croxen MA, Finlay BB. Molecular mechanisms of Escherichia coli pathogenicity. Nat Rev Microbiol. 2010;8(1):26–38.

    Article  CAS  PubMed  Google Scholar 

  162. Arthur JC, Perez-Chanona E, Mühlbauer M, Tomkovich S, Uronis JM, Fan TJ, et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012;338(6103):120–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Gaytán MO, Martínez-Santos VI, Soto E, González-Pedrajo B. Type three Secretion System in attaching and effacing pathogens. Front Cell Infect Microbiol. 2016;6:129.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Dalmasso G, Cougnoux A, Delmas J, Darfeuille-Michaud A, Bonnet R. The bacterial genotoxin colibactin promotes colon Tumor growth by modifying the Tumor microenvironment. Gut Microbes. 2014;5(5):675–80.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Brennan CA, Garrett WS. Fusobacterium nucleatum - Symbiont, opportunist and oncobacterium. Nat Rev Microbiol. 2019;17(3):156–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Nakagaki H, Sekine S, Terao Y, Toe M, Tanaka M, Ito HO, et al. Fusobacterium nucleatum envelope protein FomA is immunogenic and binds to the salivary statherin-derived peptide. Infect Immun. 2010;78(3):1185–92.

    Article  CAS  PubMed  Google Scholar 

  167. Staron MM, Gray SM, Marshall HD, Parish IA, Chen JH, Perry CJ, et al. The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8(+) T cells during chronic Infection. Immunity. 2014;41(5):802–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Boussiotis VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med. 2016;375(18):1767–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Patel SA, Minn AJ. Combination Cancer Therapy with Immune Checkpoint Blockade: mechanisms and strategies. Immunity. 2018;48(3):417–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Gao Y, Bi D, Xie R, Li M, Guo J, Liu H, et al. Fusobacterium nucleatum enhances the efficacy of PD-L1 blockade in Colorectal cancer. Signal Transduct Target Therapy. 2021;6(1):398.

    Article  CAS  Google Scholar 

  172. de Martel C, Georges D, Bray F, Ferlay J, Clifford GM. Global burden of cancer attributable to Infections in 2018: a worldwide incidence analysis. The Lancet Global Health. 2020;8(2):e180–90.

    Article  PubMed  Google Scholar 

  173. 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(6494):973–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Wong-Rolle A, Wei HK, Zhao C, Jin C. Unexpected guests in the Tumor microenvironment: microbiome in cancer. Protein Cell. 2021;12(5):426–35.

    Article  PubMed  Google Scholar 

  175. Panebianco C, Ciardiello D, Villani A, Maiorano BA, Latiano TP, Maiello E, et al. Insights into the role of gut and intratumor microbiota in pancreatic ductal adenocarcinoma as new key players in preventive, diagnostic and therapeutic perspective. Sem Cancer Biol. 2022;86(Pt 3):997–1007.

    Article  CAS  Google Scholar 

  176. 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):3789–3806e3717.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Liang Y, Li Q, Liu Y, Guo Y, Li Q. Awareness of intratumoral bacteria and their potential application in cancer treatment. Discover Oncol. 2023;14(1):57.

    Article  CAS  Google Scholar 

  178. Abbott M, Ustoyev Y. Cancer and the Immune System: the history and background of Immunotherapy. Semin Oncol Nurs. 2019;35(5):150923.

    Article  PubMed  Google Scholar 

  179. Shiao SL, Kershaw KM, Limon JJ, You S, Yoon J, Ko EY, et al. Commensal bacteria and fungi differentially regulate Tumor responses to radiation therapy. Cancer Cell. 2021;39(9):1202–1213e1206.

    Article  CAS  PubMed  Google Scholar 

  180. Geller LT, Straussman R. Intratumoral bacteria may elicit chemoresistance by metabolizing anticancer agents. Mol Cell Oncol. 2018;5(1):e1405139.

    Article  PubMed  Google Scholar 

  181. Choy ATF, Carnevale I, Coppola S, Meijer LL, Kazemier G, Zaura E, et al. The microbiome of Pancreatic cancer: from molecular diagnostics to new therapeutic approaches to overcome chemoresistance caused by metabolic inactivation of gemcitabine. Expert Rev Mol Diagn. 2018;18(12):1005–9.

    Article  CAS  PubMed  Google Scholar 

  182. Thomas H. Pancreatic cancer: intra-tumour bacteria promote gemcitabine resistance in pancreatic adenocarcinoma. Nat Rev Gastroenterol Hepatol. 2017;14(11):632.

    Article  PubMed  Google Scholar 

  183. Corty RW, Langworthy BW, Fine JP, Buse JB, Sanoff HK, Lund JL. Antibacterial use is Associated with an increased risk of hematologic and gastrointestinal adverse events in patients treated with Gemcitabine for Stage IV Pancreatic Cancer. Oncologist. 2020;25(7):579–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Cogdill AP, Gaudreau PO, Arora R, Gopalakrishnan V, Wargo JA. The impact of Intratumoral and gastrointestinal microbiota on systemic Cancer therapy. Trends Immunol. 2018;39(11):900–20.

    Article  CAS  PubMed  Google Scholar 

  185. Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15(8):465–78.

  186. Zhang X, Yu D, Wu D, Gao X, Shao F, Zhao M, et al. Tissue-resident Lachnospiraceae family bacteria protect against colorectal carcinogenesis by promoting Tumor immune surveillance. Cell Host Microbe. 2023;31(3):418–432e418.

    Article  CAS  PubMed  Google Scholar 

  187. Stern C, Kasnitz N, Kocijancic D, Trittel S, Riese P, Guzman CA, et al. Induction of CD4(+) and CD8(+) anti-tumor effector T cell responses by bacteria mediated Tumor therapy. Int J Cancer. 2015;137(8):2019–28.

    Article  CAS  PubMed  Google Scholar 

  188. Shi Y, Zheng W, Yang K, Harris KG, Ni K, Xue L et al. Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling. J Exp Med. 2020;217(5).

  189. Belcheva A, Irrazabal T, Robertson SJ, Streutker C, Maughan H, Rubino S, et al. Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells. Cell. 2014;158(2):288–99.

    Article  CAS  PubMed  Google Scholar 

  190. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Zhang S, Zhang S, Ma X, Zhan J, Pan C, Zhang H, et al. Intratumoral microbiome impacts immune infiltrates in Tumor microenvironment and predicts prognosis in esophageal squamous cell carcinoma patients. Front Cell Infect Microbiol. 2023;13:1165790.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Goto Y, Iwata S, Miyahara M, Miyako E. Discovery of Intratumoral Oncolytic Bacteria Toward Targeted Anticancer Theranostics. Advanced science.  (Weinheim, Baden-Wurttemberg, Germany). 2023;10(20).

    PubMed  Google Scholar 

  193. Quispe-Tintaya W, Chandra D, Jahangir A, Harris M, Casadevall A, Dadachova E, et al. Nontoxic radioactive Listeria(at) is a highly effective therapy against metastatic pancreatic cancer. Proc Natl.  Acad Sci U S A. 2013;110(21):8668–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Loeffler M, Le’Negrate G, Krajewska M, Reed JC. Inhibition of Tumor growth using salmonella expressing Fas ligand. J Natl Cancer Inst. 2008;100(15):1113–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Low KB, Ittensohn M, Luo X, Zheng LM, King I, Pawelek JM, et al. Construction of VNP20009: a novel, genetically stable antibiotic-sensitive strain of tumor-targeting Salmonella for parenteral administration in humans. Methods Mol Med. 2004;90:47–60.

    CAS  PubMed  Google Scholar 

  196. Zhou S, Zhao Z, Lin Y, Gong S, Li F, Pan J, et al. Suppression of pancreatic ductal adenocarcinoma growth by intratumoral delivery of attenuated Salmonella typhimurium using a dual fluorescent live tracking system. Cancer Biol Ther. 2016;17(7):732–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Chen J, Li T, Liang J, Huang Q, Huang JD, Ke Y, et al. Current status of intratumour microbiome in cancer and engineered exogenous microbiota as a promising therapeutic strategy. Volume 145. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie; 2022. p. 112443.

    Google Scholar 

  198. Zheng JH, Nguyen VH, Jiang SN, Park SH, Tan W, Hong SH et al. Two-step enhanced cancer immunotherapy with engineered Salmonella typhimurium secreting heterologous flagellin. Sci Transl Med. 2017;9(376):eaak9537.

  199. Kabwe M, Dashper S, Bachrach G, Tucci J. Bacteriophage manipulation of the microbiome associated with tumour microenvironments-can this improve cancer therapeutic response? FEMS Microbiol Rev. 2021;45(5):fuab017.

  200. Dolgin E. Fighting cancer with microbes. Nature. 2020;577(7792):16–s18.

    Article  Google Scholar 

  201. Zheng DW, Dong X, Pan P, Chen KW, Fan JX, Cheng SX, et al. Phage-guided modulation of the gut microbiota of mouse models of Colorectal cancer augments their responses to chemotherapy. Nat Biomedical Eng. 2019;3(9):717–28.

    Article  CAS  Google Scholar 

  202. Yu H, Li XX, Han X, Chen BX, Zhang XH, Gao S, et al. Fecal microbiota transplantation inhibits Colorectal cancer progression: reversing intestinal microbial dysbiosis to enhance anti-cancer immune responses. Front Microbiol. 2023;14:1126808.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Yang L, Li A, Wang Y, Zhang Y. Intratumoral microbiota: roles in cancer initiation, development and therapeutic efficacy. Signal Transduct Target Therapy. 2023;8(1):35.

    Article  CAS  Google Scholar 

  204. Oh HJ, Kim JH, Bae JM, Kim HJ, Cho NY, Kang GH. Prognostic impact of Fusobacterium nucleatum depends on combined Tumor location and microsatellite instability status in stage II/III colorectal cancers treated with adjuvant chemotherapy. J Pathol Translational Med. 2019;53(1):40–9.

    Article  Google Scholar 

  205. Yang JC, Lu CW, Lin CJ. Treatment of Helicobacter pylori Infection: current status and future concepts. World J Gastroenterol. 2014;20(18):5283–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Zhang H, Liao Y, Zhang H, Wu J, Zheng D, Chen Z. Cytotoxin-associated gene A increases carcinogenicity of helicobacter pylori in colorectal adenoma. Int J Biol Mark. 2020;35(1):19–25.

    Article  CAS  Google Scholar 

  207. Hermida LC, Gertz EM, Ruppin E. Predicting cancer prognosis and drug response from the Tumor microbiome. Nat Commun. 2022;13(1):2896.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge BioRender.com for providing the toolbox that has been used to create all figures.

Funding

This work was supported by the China Postdoctoral Science Foundation (2023M743200), the Henan Medical Science and Technology Joint Building Program (LHGJ20230160), and Dr. Chen Xue was supported by Postdoctoral Science Foundation of Zhengzhou University.

Author information

Authors and Affiliations

Authors

Contributions

JL and XC designed and guided the review. MX wrote the first draft of the manuscript. XG and YL critically revised the manuscript. YL and LY helped with reference collection. DH created the Figs. 1, 2, 3 and 4. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Juan Li or Chen Xue.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

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

Xuan, M., Gu, X., Liu, Y. et al. Intratumoral microorganisms in tumors of the digestive system. Cell Commun Signal 22, 69 (2024). https://doi.org/10.1186/s12964-023-01425-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-023-01425-5

Keywords