Effects of gut microbiome and obesity on the development, progression and prevention of cancer (Review)
- Authors:
- Published online on: November 23, 2023 https://doi.org/10.3892/ijo.2023.5592
- Article Number: 4
Abstract
Introduction
Cancer is a serious public health concern with ~20,000,000 new cases diagnosed and 10,000,000 cases of cancer-related mortality reported worldwide in 2020. According to a World Health Organization (WHO) report, the incidence of cancer is increasing, where it has been predicted that there may be ~28.4 million diagnosed cases worldwide by 2040 (1). Several factors have been reported to contribute to carcinogenesis, although excessive uncontrolled cell division caused by irregular cell signaling is common across most if not all cancer types (2). Hanahan and Weinberg (3) previously proposed six hallmarks of cancer. Specifically, these six hallmarks are resistance to inhibitory growth signals, persistence of proliferative signaling, evasion of cell death, initiation of angiogenesis, invasion and metastasis (3). Subsequently, Hanahan and Weinberg (4) added two more details to this six-hallmark concept in 2011, including dysregulation of cell metabolism and circumvention of immune destruction to the critical characteristics of cancer. A decade later, Hanahan (5) then expanded the hallmarks of cancer to include the importance of non-mutational epigenetic reprogramming, phenotype plasticity and the host-microbiome as recent insights in cancer studies emerged. Among these integrative concepts of cancer, the present review aimed to discuss the association between the microbiome and cancer development, in addition to between the microbiome and malignant progression.
The human body contains trillions of symbiotic microbiomes, including a community of bacteria, viruses, fungi and protozoa, which is collectively known as the human microbiota (6,7). Although the skin, oral cavity, respiratory tract and other body parts all harbor microbiomes, the majority of the microbiome population is predominantly contained within the gastrointestinal tract (8). Specifically, the host-associated microbial community within the gastrointestinal system is called the gut microbiome (1). They have been documented to influence various physiological and pathophysiological processes in the body (9). For example, the metabolites produced by an imbalanced microbiota can result in the pathogenesis of certain neurological conditions. Furthermore, physiological energy homeostasis has been reported to be regulated by microbiota, and microbiota-generated butyrate provides energy to colonocytes and prevents autophagy in the colon (9). In addition, microbiota also disrupt physiological homeostasis, host metabolism and contribute to the immune dysregulation (9).
Over the past decade, high-throughput sequencing and multi-omics methods have contributed to the understanding of gut microbiome diversity, elucidating its functional potential (10). Explorative research in the possible relationship between the gut microbiota and various diseases has revealed that alterations in the microbiome profile of the human gut are associated with a number of diseases, including obesity, cancer, diabetes and neurodegenerative diseases (11). However, the causative role mediated by these microbiomes in various disease conditions remains to be fully characterized. This altered population of microbiota has been documented to be great influencers of the host physiology, in processes including digestion, metabolism, cognition and immunity, though additional as yet unrevealed functions are highly likely (12). Several microbiome studies have previously found that other established factors in cancer, including obesity, inflammation and genotoxicity, are either directly or indirectly associated with the interaction between the human microbiota and the human system and metabolism (13,14).
Association between obesity and cancer
Obesity is becoming a major global issue and already affects millions of individuals. A person is considered obese when their body mass index (BMI) is >30 kg/m2. By this metric, ~650,000,000 adults >18 years old are estimated to be clinically obese worldwide, as reported by the WHO (15). Obesity has been reported to associate the incidence of numerous ailments, including type 2 diabetes, hypertension, cardiovascular disease, osteoarthritis, kidney failure, liver inflammation and cancer. In particular, chronic inflammation and altered phenotype due to obesity are the main driving factors in disease progression (16,17).
Previous studies have reported links between cancer and obesity, specifically in terms of the associated mortality rate. Obesity is an important contributing factor to the development of various types of cancer. According to a previous report, ~20% of cancer cases are associated with obesity (18). In addition, obesity enhances colorectal cancer-associated mortality by 14 and 20% in men and women, respectively (19). According to the International Agency for Research on Cancer, obesity is associated with 13 different types of cancer, including breast, colorectal, endometrial, gall bladder, kidney, gastric cardia, ovarian, liver, pancreatic and thyroid cancer, and adenocarcinoma, meningioma and multiple myeloma (20). However, obesity is not the only isolated factor associated with cancer. Age, family history, resident environment, body fat, alcohol, smoking, sex hormone levels, insulin and nutrient levels have all been observed to contribute to cancer development (21).
The excessive deposition of nutrients into the white adipose tissue causes inflammation in obese individuals. Adipocytes in this specialized type of tissue normally functions to store triglycerides as a long-term source of energy in the form of cytoplasmic lipid droplets. White adipose tissues can also secrete various adipokines, including cytokines, hormones and growth factors, which have a role in obesity-associated disease conditions (22,23). These cytokines and growth factors can activate a number of signaling pathways, including NF-κB, JNK and protein kinase R pathways. Low-grade inflammation is induced upon the stimulation of these signaling pathways, which causes the release of common inflammatory cytokines, including IL-1, IL-6 and TNF-α (24). Hyperplasia and hypertrophy of adipocytes can also aggravate this low-grade inflammatory response by increasing the production of free fatty acids, tissue remodeling and changing the profile of adipokine production (25). Furthermore, overproduction of unfolded proteins and subsequent activation of metabolic pathways by endoplasmic stress in obese individuals can activate inflammatory responses (26). Although white adipocyte tissues serve as the primary area for inflammatory pathway activation, other tissues, such as the liver, brain and pancreas, are also associated with obesity-induced inflammation (27). Due to the reported involvement of adipokines in cancer cell growth, the role of leptin and adiponectin in cancer have been widely studied (28,29). It has been reported that low levels of adiponectin may have a permissive role in triggering the neoplastic growth of ERα-positive breast cancer cells through the MAPK-activating signaling pathway (30). Obesity-associated insulin-like growth factor I and insulin resistance are also involved in the stimulation of several types of cancer, according to data produced from previous in vitro and in vivo analyses (31,32).
According to a previous meta-analysis, malignant melanoma, colon, gallbladder, pancreatic and renal cancer are significantly more common in obese men (33). In obese women, esophageal adenocarcinoma, leukemia, endometrial, colon, gallbladder, pancreatic, postmenopausal breast and renal cancer tended to be more common (Fig. 1). Obese female patients with breast cancer have been reported to show more aggressive characteristics of tumor formation and metastasis (34). In addition, the severity of breast cancer in patients is greatly influenced by age, where women with advancing age are more likely to gain weight and develop obesity. A particularly high risk of breast cancer has been reported in menopausal women between 50 and 60 years old, with weight gain also proposed to be a contributing factor (35). Primary liver cancer is the sixth most common type of cancer globally (36). Several factors have been documented to impact the development of liver cancer. Being overweight is one of the main factors that is associated with the mortality rate of patients with liver cancer and hepatitis viral infection (37,38). Another previous cohort study reported a 17 and 89% liver cancer risk in overweight and obese individuals, respectively, compared with the expected average (39). In addition, this previous study showed that the relative risk of liver cancer for overweight individuals was 1.17 and for obese individuals was 1.89 compared with that in individuals of a healthy weight (39). Liver cancer associated with obesity or excess body weight was reported to be more prevalent in men compared with women, with individuals with excess body weight suffering from liver cirrhosis or hepatitis C viral infection being more prevalent (40).
The increased incidence and prevalence of obesity among populations have elevated the risk of gastrointestinal cancer development. It has been suggested that excess adiposity is a potential cause of gastrointestinal tract, pancreatic, colorectal, esophageal, stomach and gallbladder cancer. The relationship between obesity and gastrointestinal cancer has been proposed to be associated with changes in insulin and insulin-like growth factor I signaling, altered sex hormone metabolism and chronic low-grade inflammation (41). Previous studies have indicated a strong connection between overall fat content and elevated gastro-esophageal reflux, Barrett's esophagus and esophageal adenocarcinoma (42,43). Although reflux can be adjusted in obese individuals, rates of esophageal adenocarcinoma have been shown to be more prevalent whereas waist circumference (a measure of obesity) is associated with the high occurrence of Barrett's esophagus (44). The importance of obesity (specifically central obesity) as a predisposing factor for colorectal cancer, which is the third most common cause of global cancer mortality, has been frequently reported (45,46). The increased deposition of abdominal fat and associated obesity is highly associated with the prevalence of colorectal cancer (47).
Mechanistic role of bacteria in carcinogenesis
The human gut is inhabited by a diverse microbiome, which forms a complex community along the gastrointestinal tract and contributes to the health and well-being of the host. Although it is well-known that these microbiomes interact and maintain a status of symbiosis with the host, alterations in the microbial flora can adversely affect biological functions in the body (4). A recent study by Hou et al (48) on the role of the human microbiota reported the dynamic role of the gut microbiota in host health and its relation to pathogenesis of cancer. The gut microbiome can influence the systemic functioning of the body through the gut-brain axis, colonization resistance and immunomodulation (48). In addition, the strong influence of the human gut microbiota in developing various ailments, including cancer, respiratory, neurophysiological, hepatic and kidney diseases, has been previously reported (Table I).
Cancer is caused by a multitude of factors, including both genome modulation and environmental causes. However, the association between the human microbiota and cancer development has not been considered until the 1990s. Microbiome involvement in carcinogenesis was first detected in gastric cancer in 1994, resulting in the subsequent recognition of Helicobacter pylori as a group 1 carcinogen by the WHO (49,50). The role of the human microbiota in several types of cancer has since been reported, including oral squamous cell carcinoma (51,52), lung cancer (53), breast cancer (54) and genitourinary cancer (55). Several mechanisms contributing to cancer development have been observed to be mediated by the gut microbiome, including the production of microbiota-derived metabolites, immune dysregulation and the modification of genetic and epigenetic factors (56). Previous studies have demonstrated that the intracellular bacteria Chlamydia psittaci can cause ocular adnexal lymphoma by regulating oxidative DNA damage and modulating the NF-κB pathway connected with anti-apoptotic effects (57,58). In addition, the non-motile gram-negative bacteria Porphyromonas gingivalis has been shown to be associated with oral cancer through the epithelial-mesenchymal transition of oral epithelial cells and by inducing MMP-9 and IL-8 expression (59). The pathogenic Mycobacterium tuberculosis can also promote lung cancer development and metastasis by modulating the T-cell immune response by elevating the activity of the programmed cell death protein-1/programmed death-ligand 1 pathway (60). In addition, the intracellular pathogen Chlamydia pneumoniae can promote lung cancer through by provoking the monocytes into secreting TNF-α, IL-8 and superoxide radicals, which promote cellular and DNA damage (61). The gram-negative bacteria Salmonella typhi leads to gallbladder cancer through typhoid toxin-mediated alteration of the cell cycle and DNA damage (62). Other non-motile gram-positive bacteria, including Streptococcus bovis and Streptococcus gallolyticus, have also been documented to contribute to colorectal cancer by degrading anticancer substances, including tannic acid (63). Furthermore, bacteria can trigger the release of inflammatory cytokines, including TNF-α, IL-1β, IL-6 and IL-8, to cause free radical formation, resulting in DNA alterations and cancer (64).
Over the last two decades, there has been an increase in studies investigating the impact of the gut microbiota on cancer. However, the fundamental cause of gut microbiota-induced tumor initiation remains unknown, meaning that it cannot yet be exploited for the treatment of cancer. A number of different hypotheses regarding the impact of the microbiota on cancer prognosis and therapy have been proposed. Microbial metabolites can interrupt cell signaling pathways involved in various processes, such as cell proliferation, division, programmed cell death and interaction with cell from other organs (65). It is also well-known that inflammatory conditions can significantly increase the risk of cancer, representing a pivotal hallmark of the complex process of carcinogenesis. Previous studies have revealed that enteric bacteria can significantly impact the immune system, serving a crucial role in the development of local and systemic inflammation (66,67). Notably, the gut microbiome can inhibit infection by adjusting the niche environment and regulating host immune defense (68). The interaction between cancer cells and other cell types, such as immune cells, myeloid cells and cancer-associated fibroblasts, forms the tumor microenvironment (TME) (69). It is important to note that there is accumulating evidence suggesting an essential link among gut microbiome changes, inflammation and cancer development, in a complex and multifaceted mechanism. High-throughput sequencing has revealed that aspects of bacterial niches can be detected in TME of various types of cancer, which may influence the development of cancer by modulating the immune system (65,68). The mechanistic role of microbes in carcinogenesis is detailed in Table I. In a recent review on the TME in various types of cancer, Chen et al (70) reported that the microbiota tended to differ between cancer and normal tissues (70). A previous comparative case study of gut microbial diversity and composition involving healthy individuals and patients with colon cancer through 16S rDNA sequencing revealed that the levels of beneficial bacteria were decreased, whilst the levels of several harmful bacteria were increased, in the cancer group (71). In another review of the interaction between the microbiota and the immune system, it was described that alterations in the gut microbiome can significantly impact the immune system and related signaling mechanisms through direct cell interaction or microbiota-derived metabolites (72).
Previous studies on the interaction between the gut microbiome and immunity has revealed that the gut microbiome can participate in immune modulation and can promote liver cancer through bile acid metabolism (73,74). Furthermore, assessing the role of microbial metabolites in immune regulation revealed that some of these small to large macromolecules can modulate cell signaling pathways that evoke a positive or negative response in cells (72). Polyamine derivatives found in the microbial metabolites have been reported to activate the Myc oncogene, promoting cancer progression. The c-Myc class of transcription factors are crucial for controlling the expression of genes involved in cell growth, proliferation and differentiation. In addition, butyrate derivatives have been demonstrated to exhibit antitumor proprieties, inhibiting inflammation and carcinogenesis by regulating Wnt and NF-κB signaling pathways (75). The interplay among gut microbiota, inflammation and immune responses can also affect the systemic immune response. These aforementioned observations suggest that the gut microbiome and its metabolites can significantly impact various biological processes, such as cell metabolism and immune regulation, and thus affect health (Fig. 2). Fig. 2 illustrates the intricate interplay between the gut microbiome, cellular processes and cancer progression.
Gut microbiome, obesity and gastric cancer
According to the GLOBOCAN database, gastric cancer is the fourth leading cause of cancer-related mortality worldwide (1,76). Gastric cancer is particularly prevalent in developing countries, with a two-fold higher incidence rate in men compared with that in women (74). Several epidemiological studies have highlighted the robust association between obesity and gastric cancer. In a previous meta-analysis, Lin et al identified an association between increased BMI and gastric cancer. In addition, obesity (BMI, ≥30 kg/m2) was found to be associated with an increased risk of gastric cancer compared with being overweight (BMI, 25.1-30 kg/m2) and normal weight (BMI, 18.5-25 kg/m2) (77).
In 2014, Donohoe et al demonstrated the inter-relationship between adipose and the TME in gastric cancer (44). Specifically, inflammation, hypoxia, energy metabolism and angiogenesis were all found to be key factors associating obesity with gastric cancer, which are commonly dysregulated in the TME and adipose tissue Karczewski et al (41) previously demonstrated that the primary pathophysiological mechanisms connecting obesity and gastric cancer include altered levels of insulin signaling, chronic low-grade inflammation in the adipose tissue and altered steroid hormone production and metabolism. Lee et al (78), in a Health Examinees-Gem study, reported that obesity during early adulthood (20-39 years) was significantly associated with an increased risk of gastric cancer.
The interaction between microorganisms and tumors has garnered widespread attention, with the aim of delineating the features of the complex microbial communities and their possible role in carcinogenesis (13). Numerous studies have discussed the connection between gut microbes and low-grade inflammation, which can regulate the innate and adaptive immune responses, at least in part contributing to the initiation and progression of oncogenic conditions (5,79).
H. pylori-mediated infection can stimulate immune responses and inflammation to regulate gastric cancer (80). The immune response of epithelial and myeloid cells to H. pylori bacteria is regulated by the gastric microbiota, which in turn determines the outcome of the disease. The contact between H. pylori and gastric epithelial and myeloid cells has been shown to induce signaling through innate pathways, leading to changes in cellular homeostasis, and the release of cytokines and chemokines that can trigger the inflammatory responses. H. pylori infection disturbs the balance between the host gastric microbiota and mucosa-related factors leading to inflammatory changes, dysbiosis and subsequently gastric cancer (81). H. pylori is a group 1 carcinogen that can promote gastric carcinogenesis by aggravating inflammation in the gastric mucosa (81,82). The key virulence factors of H. pylori, namely cytotoxin-associated gene (Cag)A and vacuolating toxin A (VacA), are associated with an increased risk of gastric cancer (13,83). Infection with the H. pylori CagA strain results in an increased accumulation of inflammatory cytokines, including IFN-γ, IL-1, IL-6, IL-7, IL-8, IL-10, IL-18 and TNF-α, which stimulates a diverse range of immune cells, including peripheral mononuclear cells, lymphocytes, eosinophils, macrophages, neutrophils and mast cells (13,84). In addition, infection with the Cag+ strain leads to the upregulation of the ERK/MAPK, PI3K/AKT, NF-κB, Wnt/β-Catenin, Sonic Hedgehog and STAT3 signaling pathways, whilst downregulating the activity of tumor suppressor pathways (74,85).
The VacA virulence factor leads to cellular vacuolation and autophagy in human gastric epithelial cells (86,87). VacA has been reported to upregulate MAPK, ERK1/2 and vascular endothelial growth factor expression, whilst also stimulating the Wnt/β-catenin signaling pathway, which is required for cell proliferation and differentiation (88,89). Simultaneously, H. pylori infection can also induce methylation in genes expressing the cell adhesion glycoprotein E-cadherin and various tumor suppressors, such as trefoil factor 2 and fork-head box transcriptional regulator D3, leading to the increased risk of gastric adenocarcinoma (90–92).
Advancements in sequencing technology have improved the understanding into the complexity of the gastric microbiota (13,93). H. pylori-negative individuals harbor distinct microbial populations, including Proteobacteria, Firmicutes and Actinobacteria (94,95). Sequencing-based and quantitative PCR approaches have confirmed that patients with gastric cancer tended to harbor a diverse population of microbiota with a reduced level of Porphyromonas, Neisseria, TM7 group and Streptococcus sinensis, but an enriched level of Klebsiella pneumoniae, Lactobacillus coleohominis and Lachnospiraceae (95,96). The pathogenic components, including the outer-membrane, nickel-binding and BAK proteins of non-H. pylori microbiota, including Helicobacter cinaedi, Lactobacillus coleohominis and Klebsiella pneumoniae, assist their colonization on the gastric mucosal layer, leading to tumorigenesis in the stomach (97).
A high-fat diet large quantities of fatty acids and low amount of vitamins, fibers and minerals are considered the primary cause of obesity (98). Several studies have demonstrated that a high-fat diet can promote gastric cancer through metabolic reprogramming and alteration of intestinal microbes. Arita and Inagaki-Ohara (99) reported that a high-fat diet can increase leptin signaling and promote gastric microbial dysbiosis, leading to intestinal metaplasia. In addition, this previous study found that a high-fat diet given to mice elevated Lactobacillus levels and reduced Bifidobacteria levels. Lactobacillus is a homofermentative probiotic bacterium that converts lactose to lactic acid. Lactic acid is associated with gastric carcinogenesis as it regulates inflammation, metastasis and epithelial-mesenchymal transition (100).
He et al (101) found that 12 weeks of a high-fat diet feeding in C57BL/6 mice resulted in dysbiosis of the gastric microbiota, with a reduced level of community diversity. This high-fat diet also increased the levels of Proteobacteria and Firmicutes, whilst downregulating those of Bacteroidetes and Verrucomicrobiota. Furthermore, enriched levels of Enterobacteriaceae were detected, which are associated with increased plasma and fecal endotoxin production. Endotoxin is known to trigger chronic inflammation and can induce obesity (102). Previously, Xiao et al (103) reported that endotoxin-producing opportunistic pathogens, such as Enterobacteriaceae and Desulfovibrionaceae, can produce lipopolysaccharide (LPS) and lead to metabolic endotoxemia. LPS can enhance C-X-C chemokine receptor type 7 expression in gastric cancer, subsequently regulating the proliferation and migration of gastric cancer cells through the toll-like receptor 4 (TLR4)/myeloid differentiation factor 2 signaling pathway (104). Furthermore, Desulfovibrionaceae can reduce sulfate to hydrogen sulfide (H2S) (105). H2S then activates the fatty acid receptor CD36 in gastric cancer cells to stimulate lipid metabolic reprogramming and gastric cancer metastasis (7).
Gut microbiome, obesity and colorectal cancer
Colorectal cancer is the third most common cancer diagnosed worldwide, with incidence rates 30% higher in men compared with those in women (2). Several epidemiological studies have demonstrated the relationship between obesity and colorectal cancer. Bardou et al (106) reported that obesity is associated with 11% colorectal cancer cases in the European population. Among the list of obesity conditions, visceral fat and abdominal obesity are associated with a higher risk of colorectal cancer compared with subcutaneous obesity. It has also been previously reported that obese individuals exhibit a 20-40% higher risk of colorectal cancer compared with normal-weight individuals (107,108).
Obesity leads to colorectal cancer development through inflammation, metabolic regulation and signaling processes (109). In a meta-analysis on the relationship between BMI and colorectal cancer, Ma et al (110) reported the relevant risk of colorectal cancer to be 1.334 (95% CI, 1.253-1.420) for obese individuals compared with individuals of a healthy weight. In a pooled analysis of eight population-based cohort studies, Matsuo et al (111) also revealed a strong association between obesity and colorectal cancer. The association was stronger in men compared with that in women, with the association higher in the proximal colon compared with the rectum. Recently, Socol et al (112) demonstrated that genetic variation and aberrant signaling in the leptin pathway were associated with obesity and colorectal cancer. Leptin receptor (LEPR) expression is critical for the proliferation of colorectal carcinoma cells (Table II). Higher expression of LEPR has been shown to lead to neoangiogenesis and increased metastatic potential of colorectal carcinoma. By contrast, a lack of LEPR expression reduces tumor proliferation in colorectal carcinoma (113).
A high-fat diet has been shown to promote colorectal cancer progression and metastasis. Niku et al (114) previously demonstrated that a Western diet, containing high fat and low fiber, calcium, vitamin D and folate, acts as a risk factor for the development of colorectal adenoma, through the heterozygous loss of adenomatous polyposis coli and overactivation of the AKT, mTOR and ERK1/2 signaling pathways in C57BL/6 J Min/+ mice. Park et al (115) demonstrated in another study that high-fat diet-related obesity can lead to inflammation-associated colorectal cancer by activating the PI3K/AKT signaling pathway and triggering the expression of IL-12, monocyte chemoattractant protein-1 and TNF-α in the TME.
Several studies have highlighted the altered gut microbiota signatures in colorectal cancer development (6,7). The lower population of the butyrate-producing Clostridium cluster IV and XIV bacteria has previously been associated with colorectal cancer (116). Besides, the population of Firmicutes, Actinobacteria and Lachnospiraceae have also been reported to be increased in pre-malignant colorectal adenoma, whereas Proteobacteria, Enterobacteriaceae and Sutterella species have been shown to be increased during colorectal cancer progression (117). Apart from the bacterial population, temperate phages have also been associated with the development of colorectal cancer. The bacteriophages interact with the host bacteria and stimulate the progression of colorectal cancer by changing the bacterial community structure and regulating the immune microenvironment (118).
Intestinal microbes can regulate polyamine synthesis, LPS production, butyrate metabolism and oxidative phosphorylation to facilitate the occurrence and development of colorectal cancer (119,120). Schulz et al (121) reported that a high-fat diet can lead to small intestinal tumor formation in K-rasG12Dint mice by altering the composition of intestinal microbes. Gaines et al showed that a Western pattern diet can enhance the levels of collagenase-producing microbes, including Proteus mirabilis, Candida parapsilosis and Enterococcus faecalis, in the intestine of the BALB/c mice (122). Colonization by these microbes leads to the transmigration of colorectal cancer cells and promotes colorectal cancer recurrence. Furthermore, the intestinal microbes in obesity can enhance bile acid secretion in the intestine (123). The gram-positive Clostridium converts primary bile acids to secondary bile acids, where the excessive accumulation of secondary bile acids can lead to colorectal cancer (124). In addition, high-fat diet- and Western diet-mediated obesity can reduce the abundance of Parabacteroides in the intestine. Parabacteroides antagonize the TLR4 and AKT signaling pathways (125), which are upregulated during colorectal cancer development and progression. Therefore, antagonizing Parabacteroides in the intestine through high-fat diet-related obesity can promote colorectal cancer. Sánchez-Alcoholado et al demonstrated that obese patients with colorectal cancer exhibit a reduced population of butyrate-producing gut microbes, including Butyricimonas, Roseburia, Faecalibacterium and Ruminococcus, with an abundance of opportunistic pathogens, including Fusobacterium, Desulfovibrio, Clostridium and Enterococcus (120). The enrichment of these proinflammatory pathogens and reduction in butyrate-producing bacteria results in dysbiosis and leads to tumor formation, through elevated levels of the deleterious microbial metabolite trimethylamine N-oxide, the proinflammatory cytokine IL-1β and the intestinal permeability marker Zonulin, in addition to the reduced levels of the anti-inflammatory factor IL-10.
Gut microbiome and liver cancer
Obesity and high-fat-content food are closely associated with the progression of liver cancer. Liver cancer progression is associated with the modified gut microbiome. Previous reports have explored the involvement of the gut microbiome in chronic liver disease and liver cancer (126,127). Liver inflammation causes changes in the gut microbiome, causing microbiome dysbiosis and variations in the intestinal barrier, resulting in a leaky gut (128). In the majority of cases, liver cirrhosis leads to leakiness and dysbiosis, whilst increasing the risk of liver cancer. Patients with liver cirrhosis can also exhibit sudden bacterial peritonitis, contributing to hepatocellular carcinoma (129). Fatty liver disease is also associated with liver carcinoma, even in non-alcoholic patients without cirrhosis (130). Intestinal microbiome dysbiosis in non-alcoholic patients has also been characterized in the pathogenesis of fatty liver disease (130).
Inflammatory bowel disease, type 1 diabetes, autism, cardiovascular diseases, and obesity favor gut microbiome alterations. Gut metabolites produced during gut dysbiosis induced by obesity in the enterohepatic circulation have been reported to promote hepatocellular carcinoma (7,131). A previous study reported that circulation of deoxycholic acid triggers the senescence-associated secretory phenotype in hepatic stellate cells (HSCs), which can result in the secretion of several tumor-promoting factors in the liver of C57BL/6 mice (131,132). The gut microflora-derived bacterial products, such as LPS and bacterial DNA, and endogenous substances, including free fatty acids, trigger the activation of hepatic TLR4, which leads to liver inflammation, fibrosis and cancer (132).
In a previous microbiome analysis of patients and healthy individuals, patients with carcinoma induced by hepatitis B were reported to possess an abundance of Escherichia, Shigella and Enterococcus, whereas decreased levels of Faecalibacterium, Ruminococcus, Ruminoclostridium sp. Clostridium, Corynebacterium, Bacillus, Desulfovibrio and Rhodococcus sp. were observed in non-alcoholic patients with steatohepatitis-associated hepatocellular carcinoma. By contrast, in patients with cirrhosis-related carcinoma, higher levels of Epsilonproteobacteria, Actinobacteria, Clostridia, Fusobacterium and Oribacterium were observed compared with in healthy volunteers (133).
Gut microbiota interventions in cancer: Pre-clinical and clinical studies
Data from model systems, such as rodents, coupled with those from robust clinical trials, have provided evidence on the role of the gut microbiome in the progression of obesity-associated cancer, where insights have been gained on potential therapeutic interventions. Previous pre-clinical and clinical trials have studied the effect of microbial inflammation on the development of the pathophysiology of different types of cancer (14,134). Several pre-biotics have shown anticancer properties against colorectal cancer models. Prenyl flavonoids have been reported to improve the gut microbiome in in vitro models of colorectal cancer and to exert anticancer activity (134). In addition, a high-fat diet (HFD) is known to induce gut dysbiosis. The oral intake of agaro-oligosaccharides (AGO) has been shown to prevent HFD-mediated gut dysbiosis and to thus inhibit colon carcinogenesis in C57BL/6N mice. This previous study reported that phospholipids and bile acids were downregulation in C57BL/6N mice receiving a HFD alone, whereas this downregulation was recovered with the administration of AGO supplements (135). A double-blind crossover study involving healthy human adult volunteers (n=31) reported that polydextrose (PDX) can modulate the composition and function of the colonic microbiota. Besides, PDX was found to be associated with the change in microbial metabolism, including production of butyrate and reduction in metabolic byproducts of bacterial putrefaction including branched-chain fatty acids. This previous study demonstrated that PDX significant reduced the fecal water genotoxicity of volunteers after consumption, thus indicating the potential of PDX for reducing the risk factors associated with colorectal cancer (136). Fermentation of sugar in the colon tends to favor butyrate-producing bacteria, Ruminococcus and Clostridium clusters I, II and IV (136). Administration of the antibiotic cefoxitin in a murine model has been shown to reduce enterotoxigenic Bacteroides fragilis-driven inflammation and colon cancer (13,137). In addition, short-chain fatty acids favor the restoration of intestinal health and the gut microbiome to prevent colon cancer. Several mice and human studies have demonstrated the role of short-chain fatty acid-synthesizing bacteria in the treatment and prevention of cancer (138,139).
In a mouse model, altered intestinal microbiota can in turn alter antitumor immune surveillance, which can increase the risk of liver disease and therefore cancer development. NEMO∆EMOc/Nlrp6−/− mice exhibited the hallmarks of intestinal dysbiosis, as well as aggravated steatohepatitis and increased tumor burden. A significant finding of this previous study was that the loss of intestinal Akkermansia muciniphila could increase the abundance of hepatic monocytic myeloid-derived suppressor cells and T cells associated with the proliferation and expansion of liver cancer cells (140). Understanding the gut-liver axis and microbiome involvement in liver carcinoma development may facilitate the design of effective therapeutic methods. Data from rodent models and clinical trials have suggested using the gut-liver axis as a target for inhibiting liver cancer but not for the complete treatment. Several drugs, such as antibiotics, probiotics, TLR4 antagonists and prokinetics, have been shown to control non-cancerous liver disease and carcinoma progression in rodent models and human patients (141). In a previous retrospective study, patients with liver cirrhosis who received rifaximin exhibited a reduced risk of liver carcinoma development (142). In another study involving patients with liver cirrhosis, administration of different antibiotics and probiotics was found to reduce the development of primary hepatocellular carcinoma and mortality (143). Fecal microbiota transplantation (FMT) is one of the methods used to modulate the gut microbiota and reverse dysbiosis. During this process, a new bacterial population is transferred to the recipient to reverse the dysbiosis that occurred. Microbial species equilibrium is maintained in the gut by introducing fecal transplants from healthy individuals (140). FMT has been reported to improve survival in patients with metastatic gastro-esophageal cancer studied in randomized, double-blind, placebo-controlled pilot trials (143,144). By contrast, the transfer of allogenic FMT from an obese donor to a lean recipient can induce the obese phenotype and its associated metabolic dysfunctions in the lean recipient (145). Another study previously revealed the effectiveness of FMT along with anti-programmed cell death protein 1 in six out of 15 patients with melanoma (146). Several mouse model studies and clinical trials have provided information on FMT and its ability to reverse the intestinal dysbiosis, relieve colon and hepatocellular carcinoma symptoms, whilst facilitating their management (147,148).
Future directions and conclusions
The Gut microbiota aids in the homeostasis of health and disease. The commensal gut microbiome helps to maintain homeostasis by producing beneficial metabolites. However, under the conditions of altered microbiomes induced by various factors, it can promote carcinogenesis. The present review discussed the close association between gut microbes and gastric, colorectal and liver cancer, whilst also discussing potential prevention strategies. It has been observed that microbiome dysbiosis leads to alterations in the profile of essential metabolites, which in turn causes the production of toxins. These metabolites enhance inflammation in the host and can lead to the formation of tumors. Obesity has been reported to serve as a factor associated with the initiation and progression of 13 types of cancer. Therefore, exploring and employing appropriate microbes or microbial-derived molecules is recommended to develop a beneficial gut microbiota that can elicit appropriate immune responses against cancer cells. Probiotics and other cancer therapies are recommended for re-establishing gut microbiota and producing an anti-tumorigenesis environment. In the future, more personalized trials or approaches are sorted to verify the effects of probiotics on different types of cancer by modulating the microbiome.
Acknowledgements
Not applicable.
Funding
RK thanks the Science and Engineering Research Board, Govt. of India (grant no. SERB/2018/001085) for partial financial assistance. HP thanks the ICMR-SRF fellowship (Grant no. 2021-12759) Govt. of India.
Availability of data and materials
Not applicable.
Authors' contributions
RK designed the study. HP, SP, and VA prepared the figures. RK, HP, SP, and VA wrote the manuscript. RK, SB, SP, and MD critically revised the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Deo SVS, Sharma J and Kumar S: GLOBOCAN 2020 report on global cancer burden: Challenges and opportunities for surgical oncologists. Ann Surg Oncol. 29:6497–6500. 2022. View Article : Google Scholar : PubMed/NCBI | |
Siegel RL, Wagle NS, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2023. CA Cancer J Clin. 73:233–254. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI | |
Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI | |
Hanahan D: Hallmarks of cancer: New dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ursell LK, Metcalf JL, Parfrey LW and Knight R: Defining the human microbiome. Nutr Rev. 70:S38–S44. 2012. View Article : Google Scholar : PubMed/NCBI | |
Wang R, Tang R, Li B, Ma X, Schnabl B and Tilg H: Gut microbiome, liver immunology, and liver diseases. Cell Mol Immunol. 18:4–17. 2021. View Article : Google Scholar : PubMed/NCBI | |
Dekaboruah E, Suryavanshi MV, Chettri D and Verma AK: Human microbiome: An academic update on human body site specific surveillance and its possible role. Arch Microbiol. 8:2147–2167. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kuźniar A, Szawica D, Wąsiewicz E, Fularska K and Oleszko M: Human gut microbiome-how intestinal bacteria influence our health. J Educ Health Sport. 1:30–35. 2023. View Article : Google Scholar | |
Zhu X, Li B, Lou P, Dai T, Chen Y, Zhuge A, Yuan Y and Li L: The relationship between the gut microbiome and neurodegenerative diseases. Neurosci Bull. 37:1510–1522. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hoesel B and Schmid JA: The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer. 12:1–15. 2013. View Article : Google Scholar | |
Guinane CM and Cotter PD: Role of the gut microbiota in health and chronic gastrointestinal disease: Understanding a hidden metabolic organ. Ther Adv Gastroenterol. 4:295–308. 2013. View Article : Google Scholar : PubMed/NCBI | |
Meng C, Bai C, Brown TD, Hood LE and Tian Q: Human gut microbiota and gastrointestinal cancer. Genomics Proteomics Bioinformatics. 16:33–49. 2018. View Article : Google Scholar : PubMed/NCBI | |
Bhatt AP, Redinbo MR and Bultman SJ: The role of the microbiome in cancer development and therapy. CA Cancer J Clin. 67:326–344. 2017. View Article : Google Scholar : PubMed/NCBI | |
Boutari C and Mantzoros CS: A 2022 update on the epidemiology of obesity and a call to action: As its twin COVID-19 pandemic appears to be receding, the obesity and dysmetabolism pandemic continues to rage on. Metabolism. 133:1552172022. View Article : Google Scholar : PubMed/NCBI | |
Calle EE, Rodriguez C, Walker-Thurmond K and Thun MJ: Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S adults. N Engl J Med. 17:1625–1638. 2003. View Article : Google Scholar : PubMed/NCBI | |
Martin-Rodriguez E, Guillen-Grima F, Martí A and Brugos-Larumbe A: Comorbidity associated with obesity in a large population: The APNA study. Obes Res Clin Pract. 5:435–447. 2003.PubMed/NCBI | |
Wolin KY, Carson K and Colditz GA: Obesity and cancer. Oncologist. 6:556–565. 2010. View Article : Google Scholar : PubMed/NCBI | |
Amersi F, Agustin M and Ko CY: Colorectal cancer: Epidemiology, risk factors, and health services. Clin Colon Rectal Surg. 3:133–140. 2005. View Article : Google Scholar : PubMed/NCBI | |
Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F and Straif K: Body fatness and cancer-viewpoint of the IARC working group. N Engl J Med. 8:794–798. 2016. View Article : Google Scholar | |
Deslypere JP: Obesity and cancer. Metabolism. 44:24–27. 1995. View Article : Google Scholar : PubMed/NCBI | |
Fasshauer M and Blüher M: Adipokines in health and disease. Trends Pharmacol Sci. 7:461–470. 2015. View Article : Google Scholar : PubMed/NCBI | |
Gregor MF and Hotamisligil GS: Inflammatory mechanisms in obesity. Annu Rev Immunol. 29:415–445. 2011. View Article : Google Scholar : PubMed/NCBI | |
Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, Beguinot F and Miele C: Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci. 20:23582019. View Article : Google Scholar : PubMed/NCBI | |
Amen OM, Sarker SD, Ghildyal R and Arya A: Endoplasmic reticulum stress activates unfolded protein response signaling and mediates inflammation, obesity, and cardiac dysfunction: Therapeutic and molecular approach. Front Pharmacol. 10:9772019. View Article : Google Scholar : PubMed/NCBI | |
Cnop M, Foufelle F and Velloso LA: Endoplasmic reticulum stress, obesity and diabetes. Trends Mol Med. 18:59–68. 2012. View Article : Google Scholar : PubMed/NCBI | |
Kolb R, Sutterwala FS and Zhang W: Obesity and cancer: Inflammation bridges the two. Curr Opin Pharmacol. 29:77–89. 2016. View Article : Google Scholar : PubMed/NCBI | |
Fenton JI, Hord NG, Lavigne JA, Perkins SN and Hursting SD: Leptin, insulin-like growth factor-1, and insulin-like growth factor-2 are mitogens in ApcMin/+ but not Apc+/+ colonic epithelial cell lines. Cancer Epidemiol Biomarkers Prev. 14:1646–1652. 2005. View Article : Google Scholar : PubMed/NCBI | |
VanSaun MN: Molecular pathways: Adiponectin and leptin signaling in cancer. Clin Cancer Res. 19:1926–1932. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hursting SD, Nunez ND, Varticovski L and Vinson C: The obesity-cancer link: Lessons learned from a fatless mouse. Cancer Res. 67:2391–2393. 2007. View Article : Google Scholar : PubMed/NCBI | |
Cohen DH and LeRoith D: Obesity, type 2 diabetes, and cancer: The insulin and IGF connection. Endocr Relat Cancer. 19:F27–F45. 2012. View Article : Google Scholar : PubMed/NCBI | |
Amin MN, Hussain MS, Sarwar MS, Moghal MM, Das A, Hossain MZ, Chowdhury JA, Millat MS and Islam MS: How the association between obesity and inflammation may lead to insulin resistance and cancer. Diabetes Metab Syndr. 13:1213–1224. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dobbins M, Decorby K and Choi BCK: The association between obesity and cancer risk: A meta-analysis of observational studies from 1985 to 2011. ISRN Prev Med. 2013:1–16. 2013. View Article : Google Scholar : PubMed/NCBI | |
Jiralerspong S and Goodwin PJ: Obesity and breast cancer prognosis: Evidence, challenges, and opportunities. J Clin Oncol. 34:4203–4216. 2016. View Article : Google Scholar : PubMed/NCBI | |
Osman MA and Hennessy BT: Obesity correlation with metastases development and response to first-line metastatic chemotherapy in breast cancer. Clin Med Insights Oncol. 9:105–112. 2015. View Article : Google Scholar : PubMed/NCBI | |
Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, Laversanne M, McGlynn KA and Soerjomataram I: Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 77:1598–1606. 2022. View Article : Google Scholar : PubMed/NCBI | |
El-Serag HB and Mason AC: Risk factors for the rising rates of primary liver cancer in the United States. Arch Intern Med. 160:3227–3230. 2000. View Article : Google Scholar : PubMed/NCBI | |
Ohishi W, Fujiwara S, Cologne JB, Suzuki G, Akahoshi M, Nishi N, Tsuge M and Chayama K: Impact of radiation and hepatitis virus infection on risk of hepatocellular carcinoma. Hepatology. 53:1237–1245. 2011. View Article : Google Scholar : PubMed/NCBI | |
Larsson S and Wolk A: Overweight, obesity and risk of liver cancer: A meta-analysis of cohort studies. Br J Cancer. 97:1005–1008. 2007. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Wang X, Wang J, Yan Z and Luo J: Excess body weight and the risk of primary liver cancer: An updated meta-analysis of prospective studies. Eur J Cancer. 48:2137–2145. 2012. View Article : Google Scholar : PubMed/NCBI | |
Karczewski J, Begier-Krasińska B, Staszewski R, Popławska E, Gulczynska-Elhadi K and Dobrowolska A: Obesity and the risk of gastrointestinal cancers. Dig Dis Sci. 64:2740–2749. 2019. View Article : Google Scholar : PubMed/NCBI | |
El-Serag HB, Ergun GA, Pandolfino J, Fitzgerald S, Tran T and Kramer JR: Obesity increases oesophageal acid exposure. Gut. 56:749–755. 2007. View Article : Google Scholar : PubMed/NCBI | |
Long E and Beales IL: The role of obesity in oesophageal cancer development. Ther Adv Gastroenterol. 7:247–268. 2014. View Article : Google Scholar : PubMed/NCBI | |
Donohoe CL, O'Farrell NJ, Doyle SL and Reynolds JV: The role of obesity in gastrointestinal cancer: Evidence and opinion. Ther Adv Gastroenterol. 7:38–50. 2014. View Article : Google Scholar : PubMed/NCBI | |
Pati S, Irfan W, Jameel A, Ahmed S and Shahid RK: Obesity and cancer: A current overview of epidemiology, pathogenesis, outcomes, and management. Cancers (Basel). 15:4852023. View Article : Google Scholar : PubMed/NCBI | |
Rawla P, Sunkara T and Barsouk A: Epidemiology of colorectal cancer: Incidence, mortality, survival, and risk factors. Prz Gastroenterology. 14:89–103. 2019. | |
Soltani G, Poursheikhani A, Yassi M, Hayatbakhsh A, Kerachian M and Kerachian MA: Obesity, diabetes and the risk of colorectal adenoma and cancer. BMC Endocr Disord. 19:1132019. View Article : Google Scholar : PubMed/NCBI | |
Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, Zhu D, Koya JB, Wei L, Li J and Chen ZS: Microbiota in health and diseases. Signal Transduct Target Ther. 7:1352022. View Article : Google Scholar : PubMed/NCBI | |
Barra WF, Sarquis DP, Khayat AS, Khayat BCM, Demachki S, Anaissi AKM, Ishak G, Santos NPC, Dos Santos SEB, Burbano RR, et al: Gastric cancer microbiome. Pathobiology. 88:156–169. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chattopadhyay I, Verma M and Panda M: Role of oral microbiome signatures in diagnosis and prognosis of oral cancer. Technol Cancer Res Treat. 18:15330338198673542019. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Dai D, Jin W, Huang Y, Zhang Y, Chen Y, Wang W, Lin W, Chen X, Zhang J, et al: Microbiota and metabolites alterations in proximal and distal gastric cancer patients. J Transl Med. 20:4392022. View Article : Google Scholar : PubMed/NCBI | |
Irfan M, Delgado RZR and Frias-Lopez J: The oral microbiome and cancer. Front. Immunol. 11:5910882020.PubMed/NCBI | |
Zhao Y, Liu Y, Li S, Peng Z, Liu X, Chen J and Zheng X: Role of lung and gut microbiota on lung cancer pathogenesis. J Cancer Res Clin Oncol. 147:2177–2186. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ruo SW, Alkayyali T, Win M, Tara A, Joseph C, Kannan A, Srivastava K, Ochuba O, Sandhu JK, Went TR, et al: Role of gut microbiota dysbiosis in breast cancer and novel approaches in prevention, diagnosis, and treatment. Cureus. 26:e174722021.PubMed/NCBI | |
Nicolaro M, Portal DE, Shinder B, Patel HV and Singer EA: The human microbiome and genitourinary malignancies. Ann Transl Med. 8:12452020. View Article : Google Scholar : PubMed/NCBI | |
Allen J and Sears CL: Impact of the gut microbiome on the genome and epigenome of colon epithelial cells: Contributions to colorectal cancer development. Genome Med. 25:112019. View Article : Google Scholar | |
Collina F, Chiara AD, Renzo AD, Rosa GD, Botti G and Franco R: Chlamydia psittaci in ocular adnexa MALT lymphoma: A possible role in lymphomagenesis and a different geographical distribution. Infect Agent Cancer. 7:82012. View Article : Google Scholar : PubMed/NCBI | |
Tang T, Wu H, Chen X and Chen L, Liu L, Li Z, Bai Q, Chen Y and Chen L: The hypothetical inclusion membrane protein CPSIT_0846 regulates mitochondrial-mediated host cell apoptosis via the ERK/JNK signaling pathway. Front Cell Infect Microbiol. 11:6074222021. View Article : Google Scholar : PubMed/NCBI | |
Olsen I and Yilmaz Ö: Possible role of Porphyromonas gingivalis in orodigestive cancers. J Oral Microbiol. 11:15634102019. View Article : Google Scholar : PubMed/NCBI | |
Cao S, Li J, Lu J, Zhong R and Zhong H: Mycobacterium tuberculosis antigens repress Th1 immune response suppression and promotes lung cancer metastasis through PD-1/PDl-1 signaling pathway. Cell Death Dis. 10:442019. View Article : Google Scholar : PubMed/NCBI | |
Littman AJ, Jackson LA and Vaughan TL: Chlamydia pneumoniae and lung cancer: Epidemiologic evidence. Cancer Epidemiol Biomarkers Prev. 14:773–778. 2005. View Article : Google Scholar : PubMed/NCBI | |
Di Domenico EG, Cavallo I, Pontone M, Toma L and Ensoli F: Biofilm producing Salmonella Typhi: Chronic colonization and development of gallbladder cancer. Int J Mol Sci. 18:18872017. View Article : Google Scholar : PubMed/NCBI | |
Oehmcke-Hecht S, Mandl V, Naatz LT, Dühring L, Köhler J, Kreikemeyer B and Maletzki C: Streptococcus gallolyticus abrogates anti-carcinogenic properties of tannic acid on low-passage colorectal carcinomas. Sci Rep. 10:47142020. View Article : Google Scholar : PubMed/NCBI | |
Abdulamir AS, Hafidh RR and Bakar FA: The association of Streptococcus bovis/gallolyticus with colorectal tumors: The nature and the underlying mechanisms of its etiological role. J Exp Clin Cancer Res. 30:112011. View Article : Google Scholar : PubMed/NCBI | |
Zhou X, Kandalai S, Hossain F and Zheng Q: Tumor microbiome metabolism: A game changer in cancer development and therapy. Front Oncol. 12:9334072022. View Article : Google Scholar : PubMed/NCBI | |
Johansson P, Eckstein A and Küppers R: Biology of ocular adnexal marginal zone lymphomas. Cancers (Basel). 14:12642022. View Article : Google Scholar : PubMed/NCBI | |
Yoo JY, Groer M, Dutra SVO, Sarkar A and McSkimming DI: Gut microbiota and immune system interactions. Microorganisms. 8:15872020. View Article : Google Scholar : PubMed/NCBI | |
Ge Y, Wang X, Guo Y, Yan J, Abuduwaili A, Aximujiang K, Yan J and Wu M: Gut microbiota influence tumor development and Alter interactions with the human immune system. J Exp Clin Cancer Res. 40:422021. View Article : Google Scholar : PubMed/NCBI | |
Kovács T, Mikó E, Ujlaki G, Sári Z and Bai P: The microbiome as a component of the tumor microenvironment. Tumor Microenviron. 2020:137–153. 2020. View Article : Google Scholar | |
Chen Y, Wu FH, Wu PQ, Xing HY and Ma T: The role of the tumor microbiome in tumor development and its treatment. Front Immunol. 13:9358462023. View Article : Google Scholar : PubMed/NCBI | |
He T, Cheng X and Xing C: The gut microbial diversity of colon cancer patients and the clinical significance. Bioengineered. 12:7046–7060. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zheng D, Liwinski T and Elinav E: Interaction between microbiota and immunity in health and disease. Cell Res. 30:492–506. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chénard T, Prévost K, Dubé J and Massé E: Immune system modulations by products of the gut microbiota. Vaccines (Basel). 8:4612020. View Article : Google Scholar : PubMed/NCBI | |
Rawla P and Barsouk A: Epidemiology of gastric cancer: Global trends, risk factors and prevention. Gastroenterol Rev Gastroenterol. 14:26–38. 2018. View Article : Google Scholar | |
Hanus M, Parada-Venegas D, Landskron G, Wielandt AM, Hurtado C, Alvarez K, Hermoso MA, López-Köstner F and la Fuente MD: Immune system, microbiota, and microbial metabolites: The unresolved triad in colorectal cancer microenvironment. Front Immunol. 12:6128262021. View Article : Google Scholar : PubMed/NCBI | |
Ilic M and Ilic I: Epidemiology of stomach cancer. World J Gastroenterol. 28:1187–1203. 2022. View Article : Google Scholar : PubMed/NCBI | |
Lin XJ, Wang CP, Liu XD, Yan KK, Li S, Bao HH, Zhao LY and Liu X: Body mass index and risk of gastric cancer: A meta-analysis. Jpn J Clin Oncol. 44:783–791. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lee HW, Huang D, Shin WK, de la Torre K, Yang JJ, Song M, Shin A, Lee JK and Kang D: Obesity at early adulthood increases risk of gastric cancer from the health Examinees-Gem (HEXA-G) study. PLoS One. 17:e02608262022. View Article : Google Scholar : PubMed/NCBI | |
Cani PD and Jordan BF: Gut microbiota-mediated inflammation in obesity: A link with gastrointestinal cancer. Nat Rev Gastroenterol Hepatol. 15:671–682. 2018. View Article : Google Scholar : PubMed/NCBI | |
Doorakkers E, Lagergren J, Engstrand L and Brusselaers N: Eradication of helicobacter pylori and gastric cancer: A systematic review and meta-analysis of cohort studies. J Natl Cancer Inst. 108:djw1322016. View Article : Google Scholar : PubMed/NCBI | |
Toh JWT and Wilson RB: Pathways of gastric carcinogenesis, helicobacter pylori virulence and interactions with antioxidant systems, vitamin C and phytochemicals. Int J Mol Sci. 21:64512020. View Article : Google Scholar : PubMed/NCBI | |
Colotta F, Allavena A, Sica C, Garlanda C and Mantovani A: Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis. 30:1073–1081. 2009. View Article : Google Scholar : PubMed/NCBI | |
Khatoon J, Rai RP and Prasad KN: Role of helicobacter pylori in gastric cancer: Updates. World J Gastrointest Oncol. 8:147–158. 2016. View Article : Google Scholar : PubMed/NCBI | |
Moyat M and Velin D: Immune responses to Helicobacter pylori infection. World J Gastroenterol. 20:5583–5593. 2014. View Article : Google Scholar : PubMed/NCBI | |
Udhayakumar G, Jayanthi V, Devaraj N and Devaraj H: Interaction of MUC1 with β-catenin modulates the Wnt target Gene cyclinD1 in H. pylori-induced gastric cancer. Mol Carcinog. 46:807–817. 2007. View Article : Google Scholar : PubMed/NCBI | |
Hotchin NA, Cover TL and Akhtar N: Cell vacuolation induced by the VacA cytotoxin ofhelicobacter pylori is regulated by the Rac1 GTPase. J Biol Chem. 275:14009–14012. 2000. View Article : Google Scholar : PubMed/NCBI | |
Yahiro K, Akazawa Y, Nakano M, Suzuki H, Hisatune J, Isomoto H, Sap J, Noda M, Moss J and Hirayama T: Helicobacter pylori VacA induces apoptosis by accumulation of connexin 43 in autophagic vesicles via a Rac1/ERK-dependent pathway. Cell Death Discov. 1:150352015. View Article : Google Scholar : PubMed/NCBI | |
Caputo R, Tuccillo C, Manzo BA, Zarrilli R, Tortora G, Blanco CD, Ricci V, Ciardiello F and Romano M: Helicobacter pylori VacA toxin up-regulates vascular endothelial growth factor expression in MKN 28 gastric cells through an epidermal growth factor receptor-, cyclooxygenase-2-dependent mechanism1. Clin Cancer Res. 9:2015–2021. 2003.PubMed/NCBI | |
Song X, Xin N, Wang W and Zhao C: Wnt/β-catenin, an oncogenic pathway targeted by H. pylori in gastric carcinogenesis. Oncotarget. 6:35579–35588. 2015. View Article : Google Scholar : PubMed/NCBI | |
Muhammad JS, Eladl MA and Khoder G: Helicobacter pylori-induced DNA methylation as an epigenetic modulator of gastric cancer: Recent outcomes and future direction. Pathogens. 8:232019. View Article : Google Scholar : PubMed/NCBI | |
Peterson AJ, Menheniott TR, O'Connor L, Walduck AK, Fox JG, Kawakami K, Minamoto T, Ong EK, Wang TC, Judd LM and Giraud AS: Helicobacter pylori infection promotes methylation and silencing of trefoil factor 2, leading to gastric tumor development in mice and humans. Gastroenterology. 139:2005–2017. 2010. View Article : Google Scholar : PubMed/NCBI | |
Sato F and Meltzer SJ: CpG island hypermethylation in progression of esophageal and gastric cancer. Cancer. 106:483–493. 2006. View Article : Google Scholar : PubMed/NCBI | |
Smet A, Kupcinskas J, Link A, Hold GL and Bornschein J: The role of microbiota in gastrointestinal cancer and cancer treatment: Chance or curse? Cell Mol Gastroenterol Hepatol. 13:857–874. 2022. View Article : Google Scholar : PubMed/NCBI | |
Engstrand L and Lindberg M: Helicobacter pylori and the gastric microbiota. Best Pract Res Clin Gastroenterol. 27:39–45. 2013. View Article : Google Scholar : PubMed/NCBI | |
Aviles-Jimenez F, Vazquez-Jimenez F, Medrano-Guzman R, Mantilla A and Torres J: Stomach microbiota composition varies between patients with non-atrophic gastritis and patients with intestinal type of gastric cancer. Sci Rep. 4:42022014. View Article : Google Scholar : PubMed/NCBI | |
Dias-Jácome E, Libânio D, Borges-Canha M, Galaghar A and Pimentel-Nunes P: Gastric microbiota and carcinogenesis: The role of non-Helicobacter pylori bacteria-A systematic review. Rev Esp Enferm Dig. 108:530–540. 2016. View Article : Google Scholar : PubMed/NCBI | |
De Witte C, Schulz C, Smet A, Malfertheiner P and Haesebrouck F: Other Helicobacters and gastric microbiota. Helicobacter. 21:62–68. 2016. View Article : Google Scholar : PubMed/NCBI | |
Tong Y, Gao H, Qi Q, Liu X, Li J, Gao J, Li P, Wang Y, Du L and Wang C: High fat diet, gut microbiome and gastrointestinal cancer. Theranostics. 11:5889–5910. 2021. View Article : Google Scholar : PubMed/NCBI | |
Arita S and Inagaki-Ohara K: High-fat-diet-induced modulations of leptin signaling and gastric microbiota drive precancerous lesions in the stomach. Nutrition. 67–68. 1105562019. | |
Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, et al: Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 513:559–563. 2014. View Article : Google Scholar : PubMed/NCBI | |
He C, Cheng D, Peng C, Li Y, Zhu Y and Lu N: High-fat diet induces dysbiosis of gastric microbiota prior to gut microbiota in association with metabolic disorders in mice. Front Microbiol. 9:6392018. View Article : Google Scholar : PubMed/NCBI | |
Kim KA, Gu W, Lee IA, Joh EH and Kim DH: High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One. 7:e477132012. View Article : Google Scholar : PubMed/NCBI | |
Xiao S, Fei N, Pang X, Shen J, Wang L, Zhang B, Zhang M, Zhang X, Zhang C, Li M, et al: A gut microbiota-targeted dietary intervention for amelioration of chronic inflammation underlying metabolic syndrome. FEMS Microbiol Ecol. 87:357–367. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li N, Xu H, Ou Y, Feng Z, Zhang Q, Zhu Q and Cai Z: LPS-induced CXCR7 expression promotes gastric Cancer proliferation and migration via the TLR4/MD-2 pathway. Diagn Pathol. 14:32019. View Article : Google Scholar : PubMed/NCBI | |
Zhang XHF, Giuliano M, Trivedi MV, Schiff R and Osborne CK: Metastasis dormancy in estrogen receptor-positive breast cancer. Clin Cancer Res. 19:6389–6397. 2013. View Article : Google Scholar : PubMed/NCBI | |
Bardou M, Barkun AN and Martel M: Obesity and colorectal cancer. Gut. 62:933–947. 2013. View Article : Google Scholar : PubMed/NCBI | |
Pischon T and Nimptsch K: Obesity and risk of cancer: An introductory overview in obesity and cancer. Recent Results Cancer Res. 208:1–15. 2016. View Article : Google Scholar : PubMed/NCBI | |
Torre LA, Siegel RL, Ward EM and Jemal A: Global cancer incidence and mortality rates and trends-an update. Cancer Epidemiol Biomarkers Prev. 25:16–27. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chaplin A, Rodriguez RM, Segura-Sampedro JJ, Ochogavía-Seguí A, Romaguera D and Barceló-Coblijn G: Insights behind the relationship between colorectal cancer and obesity: Is visceral adipose tissue the missing link? Int J Mol Sci. 23:131282022. View Article : Google Scholar : PubMed/NCBI | |
Ma Y, Yang Y, Wang F, Zhang P, Shi C, Zou Y and Qin H: Obesity and risk of colorectal cancer: A systematic review of prospective studies. PLoS One. 8:e539162013. View Article : Google Scholar : PubMed/NCBI | |
Matsuo K, Mizoue T, Tanaka K, Tsuji I, Sugawara Y, Sasazuki S, Nagata C, Tamakoshi A, Wakai K, Inoue M, et al: Association between body mass index and the colorectal cancer risk in Japan: Pooled analysis of population-based cohort studies in Japan. Ann Oncol. 23:479–490. 2012. View Article : Google Scholar : PubMed/NCBI | |
Socol CT, Chira A, Martinez-Sanchez MA, Nuñez-Sanchez MA, Maerescu CM, Mierlita D, Rusu AV, Ruiz-Alcaraz AJ, Trif M and Ramos-Molina B: Leptin signaling in obesity and colorectal cancer. Int J Mol Sci. 23:47132022. View Article : Google Scholar : PubMed/NCBI | |
Milosevic VS, Vukmirovic FC, Krstic MC, Zindovic MM, Stojanovic DL and Jancic SA: Involvement of leptin receptors expression in proliferation and neoangiogenesis in colorectal carcinoma. J BUON. 20:100–108. 2015.PubMed/NCBI | |
Niku M, Pajari AM, Sarantaus L, Päivärinta E, Storvik M, Heiman-Lindh A, Suokas S, Nyström M and Mutanen M: Western diet enhances intestinal tumorigenesis in Min/+ mice, associating with mucosal metabolic and inflammatory stress and loss of Apc heterozygosity. J Nutr Biochem. 39:126–133. 2017. View Article : Google Scholar : PubMed/NCBI | |
Park S, Koh E, Koo JS, Kim SI, Park BW and Kim KS: Lack of both androgen receptor and forkhead box A1 (FOXA1) expression is a poor prognostic factor in estrogen receptor-positive breast cancers. Oncotarget. 8:82940–82955. 2017. View Article : Google Scholar : PubMed/NCBI | |
Gheorghe AS, Negru ȘM, Preda M, Mihăilă RI, Komporaly IA, Dumitrescu EA, Lungulescu CV, Kajanto LA, Georgescu B, Radu EA and Stănculeanu DL: Biochemical and metabolical pathways associated with microbiota-derived butyrate in colorectal cancer and omega-3 fatty acids implications: A narrative review. Nutrients. 14:11522022. View Article : Google Scholar : PubMed/NCBI | |
Mori G, Rampelli S, Orena BS, Rengucci C, De Maio G, Barbieri G, Passardi A, Gardini AC, Frassineti GL, Gaiarsa S, et al: Shifts of faecal microbiota during sporadic colorectal carcinogenesis. Sci Rep. 8:103292018. View Article : Google Scholar : PubMed/NCBI | |
Hannigan GD, Duhaime MB, Ruffin MT IV, Koumpouras CC and Schloss PD: Diagnostic potential and interactive dynamics of the colorectal cancer virome. mBio. 9:e02248–e02218. 2018. View Article : Google Scholar : PubMed/NCBI | |
Fang Y, Yan C, Zhao Q, Xu J, Liu Z, Gao J, Zhu H, Dai Z, Wang D and Tang D: The roles of microbial products in the development of colorectal cancer: A review. Bioengineered. 12:720–735. 2021. View Article : Google Scholar : PubMed/NCBI | |
Sánchez-Alcoholado L, Ramos-Molina B, Otero A, Laborda-Illanes A, Ordóñez R, Medina JA, Gómez-Millán J and Queipo-Ortuño MI: The role of the gut microbiome in colorectal cancer development and therapy response. Cancers (Basel). 12:14062020. View Article : Google Scholar : PubMed/NCBI | |
Schulz MD, Atay C, Heringer J, Romrig FK, Schwitalla S, Aydin B, Ziegler PK, Varga J, Reindl W, Pommerenke C, et al: High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature. 514:508–512. 2014. View Article : Google Scholar : PubMed/NCBI | |
Gaines S, van Praagh JB, Williamson AJ, Jacobson RA, Hyoju S, Zaborin A, Mao J, Koo HY, Alpert L, Bissonnette M, et al: Western diet promotes intestinal colonization by collagenolytic microbes and promotes tumor formation after colorectal surgery. Gastroenterology. 158:958–970.e2. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zeng H, Umar S, Rust B, Lazarova D and Bordonaro M: Secondary bile acids and short chain fatty acids in the colon: A focus on colonic microbiome, cell proliferation, inflammation, and cancer. Int J Mol Sci. 20:12142019. View Article : Google Scholar : PubMed/NCBI | |
Schramm C: Bile acids, the microbiome, immunity, and liver tumors. N Engl J Med. 379:888–890. 2018. View Article : Google Scholar : PubMed/NCBI | |
Koh GY, Kane A, Lee K, Xu Q, Wu X, Roper J, Mason JB and Crott JW: Parabacteroides distasonis attenuates toll-like receptor 4 signaling and Akt activation and blocks colon tumor formation in high-fat diet-fed azoxymethane-treated mice. Int J Cancer. 143:1797–1805. 2018. View Article : Google Scholar : PubMed/NCBI | |
Gupta H, Youn GS, Shin MJ and Suk KT: Role of gut microbiota in hepatocarcinogenesis. Microorganisms. 7:1212019. View Article : Google Scholar : PubMed/NCBI | |
Trivedi Y, Bolgarina Z, Desai HN, Senaratne M, Swami SS, Aye SL and Mohammed L: The role of gut microbiome in hepatocellular carcinoma: A systematic review. Cureus. 15:e438622023.PubMed/NCBI | |
Plaza-Díaz J, Solís-Urra P, Rodríguez-Rodríguez F, Olivares-Arancibia J, Navarro-Oliveros M, Abadía-Molina F and Álvarez-Mercado AI: The gut barrier, intestinal microbiota, and liver disease: Molecular mechanisms and strategies to manage. Int J Mol Sci. 21:83512020. View Article : Google Scholar : PubMed/NCBI | |
Schwabe RF and Greten TF: Gut microbiome in HCC-Mechanisms, diagnosis and therapy. J Hepatol. 72:230–238. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chu H, Williams B and Schnabl B: Gut microbiota, fatty liver disease, and hepatocellular carcinoma. Liver Res. 2:43–51. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, et al: Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 499:97–101. 2013. View Article : Google Scholar : PubMed/NCBI | |
Roh YS and Seki E: Toll-like receptors in alcoholic liver disease, non-alcoholic steatohepatitis and carcinogenesis. J Gastroenterol Hepatol. 28:38–42. 2013. View Article : Google Scholar : PubMed/NCBI | |
Bartolini I, Risaliti M, Tucci R, Muiesan P, Ringressi MN, Taddei A and Amedei A: Gut microbiota and immune system in liver cancer: Promising therapeutic implication from development to treatment. World J Gastrointest Oncol. 13:1616–1631. 2021. View Article : Google Scholar : PubMed/NCBI | |
Allsopp P, Possemiers S, Campbell D, Gill C and Rowland I: A comparison of the anti-cancer properties of isoxanthohumol and 8-prenylnaringenin using in vitro models of colon cancer. Biofactors. 39:441–447. 2013. View Article : Google Scholar : PubMed/NCBI | |
Higashimura Y, Naito Y, Takagi T, Uchiyama K, Mizushima K, Ushiroda C, Ohnogi H, Kudo Y, Yasui M, Inui S, et al: Protective effect of agaro-oligosaccharides on gut dysbiosis and colon tumorigenesis in high-fat diet-fed mice. Am J Physiol Gastrointest Liver Physiol. 310:G367–G375. 2016. View Article : Google Scholar : PubMed/NCBI | |
Costabile A, Fava F, Röytiö H, Forssten SD, Olli K, Klievink J, Rowland IR, Ouwehand AC, Rastall RA, Gibson GR and Walton GE: Impact of polydextrose on the faecal microbiota: A double-blind, crossover, placebo-controlled feeding study in healthy human subjects. Br J Nutr. 108:471–481. 2012. View Article : Google Scholar : PubMed/NCBI | |
Shields CE, Van Meerbeke SW, Housseau F, Wang H, Huso DL, Casero RA Jr, O'Hagan HM and Sears CL: Reduction of murine colon tumorigenesis driven by enterotoxigenic bacteroides fragilis using cefoxitin treatment. J Infect Dis. 214:122–129. 2016. View Article : Google Scholar | |
Hou H, Chen D, Zhang K, Zhang W, Liu T, Wang S, Dai X, Wang B, Zhong W and Cao H: Gut microbiota-derived short-chain fatty acids and colorectal cancer: Ready for clinical translation? Cancer Lett. 526:225–235. 2022. View Article : Google Scholar : PubMed/NCBI | |
Son MY and Cho HS: Anticancer effects of gut microbiota-derived short-chain fatty acids in cancers. J Microbiol Biotechnol. 33:849–856. 2023. View Article : Google Scholar : PubMed/NCBI | |
Schneider KM, Mohs A, Gui W, Galvez EJC, Candels LS, Hoenicke L, Muthukumarasamy U, Holland CH, Elfers C, Kilic K, et al: Imbalanced gut microbiota fuels hepatocellular carcinoma development by shaping the hepatic inflammatory microenvironment. Nat Commun. 13:39642022. View Article : Google Scholar : PubMed/NCBI | |
Yu LX and Schwabe RF: The gut microbiome and liver cancer: Mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol. 14:527–539. 2017. View Article : Google Scholar : PubMed/NCBI | |
Patel AH, Li Y, Minacapelli CD, Catalano K and Rustgi V: Reduction in gastrointestinal cancers in cirrhotic patients receiving rifaximin vs lactulose only therapy for hepatic encephalopathy. Cureus. 15:e352592023.PubMed/NCBI | |
Ting NLN, Lau HCH and Yu J: Cancer pharmacomicrobiomics: Targeting microbiota to optimise cancer therapy outcomes. Gut. 71:1412–1425. 2022. View Article : Google Scholar : PubMed/NCBI | |
de Clercq NC, van den Ende T, Prodan A, Hemke R, Davids M, Pedersen HK, Nielsen HB, Groen AK, de Vos WM, van Laarhoven HWM and Nieuwdorp M: Fecal microbiota transplantation from overweight or obese donors in cachectic patients with advanced gastroesophageal cancer: A randomized, double-blind, placebo-controlled, phase II study. Clin Cancer Res. 27:3784–3792. 2021. View Article : Google Scholar : PubMed/NCBI | |
Napolitano M and Covasa M: Microbiota transplant in the treatment of obesity and diabetes: Current and future perspectives. Front Microbiol. 11:5903702020. View Article : Google Scholar : PubMed/NCBI | |
Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al: Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 371:595–602. 2021. View Article : Google Scholar : PubMed/NCBI | |
Xu H, Cao C, Ren Y, Weng S, Liu L, Guo C, Wang L, Han X, Ren J and Liu Z: Antitumor effects of fecal microbiota transplantation: Implications for microbiome modulation in cancer treatment. Front Immunol. 13:9494902022. View Article : Google Scholar : PubMed/NCBI | |
Zhao LY, Mei JX, Yu G, Lei L, Zhang WH, Liu K, Chen XL, Kołat D, Yang K and Hu JK: Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications. Signal Transduction Target Ther. 8:2012023. View Article : Google Scholar : PubMed/NCBI |