Open Access

Advances in the role of gut microbiota in the regulation of the tumor microenvironment (Review)

  • Authors:
    • Tian Xinyuan
    • Yu Lei
    • Shi Jianping
    • Zhao Rongwei
    • Shi Ruiwen
    • Zhang Ye
    • Zhao Jing
    • Tian Chunfang
    • Cui Hongwei
    • Guan Haibin
  • View Affiliations

  • Published online on: August 22, 2023     https://doi.org/10.3892/or.2023.8618
  • Article Number: 181
  • Copyright: © Xinyuan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As a protector of human health, the gut microbiota plays an important role in the development of the immune system during childhood, and the regulation of dietary habits, metabolism and immune system during adulthood. Dysregulated gut flora is not pathogenic, but it can weaken the protective effect of the immune system and cause various diseases. The tumor microenvironment is a physiological environment formed during tumor growth, which provides nutrients and growth factors necessary for tumor growth. As an important factor affecting the tumor microenvironment, the intestinal microflora affects the development of tumors through the mechanisms of gut and microflora metabolites, gene toxins and signaling pathways. The present article aimed to review the components and mechanisms of action, clinical applications, and biological targets of gut microbiota in the regulation of the tumor microenvironment. The present review provides novel insights for the future use of intestinal flora, to regulate the tumor microenvironment, to intervene in the occurrence, development, treatment and prognosis of tumors.

Introduction

There are numerous microorganisms in the human intestinal tract, the number of which is >10-fold that of human cells, which can be divided into beneficial bacteria for human vitamin synthesis, food digestion and inhibition of the production of toxic substances; harmful bacteria that affect the function of the immune system and produce harmful substances; and neutral bacteria with dual roles, such as Escherichia coli (1). There is a dynamic balance between the intestinal flora and the host, which can maintain the normal physiological activities of the host. When the flora is imbalanced, it can cause inflammation and even diseases. The tumor microenvironment is the internal environment of tumor cells, which is not only limited to tumor cells but also includes various components of the microenvironment and nearby interstitial cells, blood vessels, cytokines and biomolecules (2). The present study conducted a computer search for articles related to the intestinal flora and the tumor microenvironment published in the Wan fang Data Knowledge Service Platform (https://www.wanfangdata.com.cn), CNKI (https://www.cnki.net), China Biomedical Literature Service System (http://www.sinomed.ac.cn/zh/), PubMed (https://pubmed.ncbi.nlm.nih.gov/advanced/) and Medline databases (https://ovidsp.ovid.com/autologin.cgi). The search time limit was from the establishment of each database until February 2023. The literature search revealed that recent studies have shifted their focus from the target of tumor treatment to the tumor microenvironment, and have revealed that the intestinal flora plays an important role in the regulation of the tumor microenvironment (3). The present review describes in detail the specific mechanisms of intestinal microflora affecting tumor microenvironment and introduces the application status and potential biological targets of intestinal microflora in tumor microenvironment intervention, aiming to provide a new direction for disease intervention and treatment.

Gut flora

There is a large number of symbiotic bacteria in the human gut, which dynamically changes under the influence of dietary habits, drug use and specific physiological conditions. The type and abundance of Gut microbiota are affected by genetic, environmental and economic factors as well as living habits, and cohabitation factors are more influential than genetic factors (4). With the continuous development and improvement of modern sequencing technology, genomic technology and in vitro culture technology of intestinal flora, the importance and mechanism of intestinal flora and various diseases have been gradually revealed. Intestinal flora can not only act as an intestinal barrier to resist the invasion of pathogens (5), but also play a role in the occurrence of various diseases such as solid tumors (colorectal, lung, and pancreatic cancer, etc.) and other diseases (leukemia, Alzheimer's disease, etc.), and their development and treatment are inextricably associated (Table I) (616).

Table I.

Types and mechanisms of gut microbiota in different tumors.

Table I.

Types and mechanisms of gut microbiota in different tumors.

Tumor typeCancer typeChange in relevant floraMechanism of action(Refs.)
Solid tumorsColorectal cancerBacteroides fragilis, Escherichia coli, Streptococcs bovis gallolyticus, Enteroccocus faecalis, Fusobacterium nucleatum increaseChanges in flora promote the secretion of inflammatory mediators and produce reactive oxygen species(6)
Liver cancerMicroflora depletionProduces bile acid and affects the liver immune function(7)
Pancreatic cancerHelicobacter pylori, Pseudomonas aeruginosa, BifidobacteriumImmune reaction, inflammation, anti-apoptosis(8)
Thyroid carcinomaIncreased number of bacteria causing cancer and inflammationImpaired intestinal barrier and increased intestinal permeability make it easier for antigens to pass through and stimulate the immune system; microflora affects the supply of essential micronutrients(9)
Lung cancer Firmicutes/Bacteroidetes reductionDecreased circulating SCFAs, thus affecting host system immunity and inflammation(10)
OtherLeukemiaChange in flora abundanceBiological antagonism, immune regulation(11)
diseasesChronic stressChange in floraBrain gut axis and ileum immune regulation(12)
Alzheimer's diseaseHelicobacter pylori infectionAltered level of certain neurotransmitters, proteins and receptors involved in synaptic plasticity(13)
DiabetesSignificantly decreased number of Bifidobacterium, Clostridium and Firmicutes, and increased number of enterococcal fecesLow level of SCFAs leading to intestinal inflammation and insulin resistance(14)
Parkinson's diseaseChronic stress promotes inflammation in the intestinal environment and increases intestinal permeabilityBrain-intestine axis(15)
Bipolar depressionAbundance of Akkermania muciniphila and Citrobacter spp.Brain-intestine axis(16)

[i] SCFAs, short-chain fatty acids.

Tumor microenvironment

The tumor microenvironment is a special biological environment formed by changes in the surrounding tissue structure during tumor growth and development. It was first described as ‘seed and soil’, with tumor cells as seeds, and the appropriate target organ and growth environment called the tumor microenvironment (17). In addition to ‘seed’ tumor cells, the tumor microenvironment also includes immune cells, adipocytes, stromal cells, extracellular matrix and acellular components (cytokines, signaling molecules and chemokines), which together provide nutrition, blood vessels, collagen and signaling molecules to form a complex and dynamic network system that provides support for the occurrence, proliferation, metastasis and immune escape of tumor cells (18). Since tumor cells have the characteristic of malignant proliferation, they consume large quantities of oxygen and nutrients in the soil, which is accompanied by the production of reducing substances (reactive oxygen species). Previous studies have revealed that a hypoxic microenvironment can promote tumor resistance (19), an acidic microenvironment is conducive to tumor cell metastasis (20) and highly reducing substances affect tumor treatment (21). The tumor microenvironment is an important condition to support tumor growth, and in-depth research and effective regulation of it will provide effective means for tumor treatment (Table II) (2227).

Table II.

Tumor microenvironment components.

Table II.

Tumor microenvironment components.

Tumor microenvironment componentsPhysiological functionAssociation with tumor(Refs.)
CAFsStroma cellStimulation of tumor cell proliferation, invasion and metastasis; chemotherapy resistance; inhibition of T cells in tumors; regulation of the inflammatory response and immune system(22)
TANsContain multiple lysosomes and havee strong chemotaxis and phagocytosis capacitiesRecruitment of macrophages and regulation of T cells(23)
TAMsFirst line of defense against microbial infectionPromotion of cancer cell proliferation, angiogenesis, metastasis and immunosuppression(24)
MDSCsMyeloid suppressor cells have heterogeneity and differentiation potential, and usually play an immunosuppressive role in the tumor microenvironmentImmunosuppression promotes tumor cell proliferation and metastasis(25)
DCsAs the main antigen-presenting cells, DCs are the bridge between adaptive and innate immunity Immunosuppression(26)
Hyaluronic acidExtracellular matrixPromotion of tumor cell proliferation, invasion, immune escape and drug resistance(27)

[i] CAFs, cancer-associated fibroblasts; TANs, tumor-associated neutrophils; TAMs, tumor-associated macrophages; MDSCs, myeloid-derived suppressor cells; DCs, dendritic cells.

Gut microbiota regulates the tumor microenvironment

In recent years, with the continuous development of technology and the increase in research, the concept of the tumor biological microenvironment has been proposed, which includes cell metabolites, the immune system, systemic metabolism, body circulation, and intestinal flora related to tumorigenesis, development and metastasis (28). Among these, the intestinal flora plays the most significant regulatory role on the tumor microenvironment, mainly through changes in the flora, brain-gut axis, hypothalamus-pituitary-adrenal axis, gut-liver axis and bacterial translocation, which affect the physiological state of target organs from a long distance. This, in turn, creates a favorable environment for tumor invasion (2931).

Regulation of the components of the tumor microenvironment
Dendritic cells (DCs)

DCs can be divided into conventional DCs and plasmacytoid DCs (pDCs), and the two phenotypes interact to maintain the morphology of DCs and the antigen expression capacity of CD8+ T cells. A previous study has demonstrated that DC antitumor activity is activated under specific conditions, and, after maturation, T cells are stimulated to produce the cytokine IL-2 to convert macrophages in the tumor microenvironment to the M1 phenotype (32). It was revealed that the antitumor function of DCs was enhanced by injecting lipopolysaccharide (LPS) into antibiotic-treated mice in vitro; thus, it was speculated that the microflora may contact or migrate to the tumor site to form an antitumor microenvironment through the bacterial components similar to LPS (33) (Fig. 1).

Figure 1.

Intestinal flora regulates tumor microenvironment components. TAMs: Helicobacter hepaticus can activate TAMs and inhibit the production of IL-10, thus inhibiting the development of breast cancer. The imbalance of gut microbiota directly activates macrophages, macrophages release IL-6, TNF-α and CXCL10. IL-6 and TNF-α accelerate the development of CRC by promoting EMT, while CXCL10 induces T-cell infiltration in the tumor microenvironment and promotes the development of HCC. Intestinal microbiota bacterial diseases induce tumor cells to secrete CTSK, thereby activating macrophages through mTOR-dependent pathways. CTSK stimulates the macrophage secretion of IL-10 and IL-17, thus promoting the invasion and metastasis of CRC cells. Some symbiotic microbiota stimulate macrophages to increase c-Jun phosphorylation in CRC cells through the JNK signaling pathway, thus accelerating CRC cell proliferation. Intestinal microbiota bacterial diseases also induce the production of IL-25 from cluster cells, which promotes EMT and the migration of HCC cells. CAFs: CAFs influence the tumor immune microenvironment by recruiting chemokines and immune factors. MDSCs: IL-17 recruits bone MDSCs into the colon tumor microenvironment of mice colonized with enterotoxin bacteria, which indirectly induce the ectopic production of chemokines and growth factors through direct interaction with IL-17 receptors in CECs, and the expression of IL-17 in submucosa. IL-17 and transformed CECs jointly promote tumor development with angiogenic mediators, namely MMP-9 and VEGF, by inhibiting immune effector cells and activating the STAT3 signaling pathway. Early lack of microbiota in mice enhances the expression of C-X-C motif receptor 2 ligands. A number of gut pathogenic microorganisms or gut microbiota and their synergistic interactions with cytokines activate and proliferate MDSCs in the tumor microenvironment, thus mediating the immune escape of tumor cells. TANs: Helicobacter hepaticus can induce the generation of neutrophils, thereby inducing nitric oxide and TNF-α increased content, which activates the NF-κB signaling pathway and promotes tumor generation. DCs: Lipopolysaccharide and TNF-α stimulate DCs to mature and activate, and subsequently activate T cells to produce IL-2 to form an antitumor microenvironment. TAMs, tumor-associated macrophages; CRC, colorectal cancer; EMT, epithelial-mesenchymal transition; HCC, hepatocellular carcinoma; CTSK, carnosine K; CAFs, cancer-associated fibroblasts; MDSCs, myeloid-derived suppressor cells; CECs, colon epithelial cells; DCs, dendritic cells; TANs, tumor-associated neutrophils; CXCL, C-X-C motif ligand.

Tumor-associated neutrophils (TANs)

TANs can be divided into two phenotypes, tumor suppressor and tumorigenic, with notably high inhibitory and polarized properties. TANs are associated with tumorigenesis, proliferation and immune regulation, and they transform into a pro-angiogenic subtype under the synergistic effect of chemokines. TANs release neutrophil extracellular traps (NETs) to kill harmful microorganisms. NETs activate specific signaling pathways to stimulate dormant cancer cells, restore their proliferative activity, and promote tumor recurrence and metastasis (34). A previous study has shown that Fusobacterium nucleatum can change the composition and phenotype of tumor-associated macrophages (TAMs), TANs and myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment, and can activate the E-cadherin/β-catenin signaling pathway to promote the malignant transformation of epithelial cells (35). Helicobacter hepaticus can stimulate the secretion of nitric oxide and TNF-α from neutrophils to promote the progression of colorectal cancer (CRC) (32) (Fig. 1).

TAMs

TAMs are markedly adaptable, and with subtle changes in the tumor microenvironment, their phenotype would tranform from the antitumor M1 to the M2 one, which promotes tumor development and remodeling (32). TAMs are affected by the combined effects of various microbiota to regulate the progression of breast and colon cancer. Zhou et al (35) compared the fecal microbiota of patients with hepatocellular carcinoma (HCC) and healthy controls, and observed that the abundance of specific flora in patients with HCC was altered, and the authors predicted that tumor cells could alter the intestinal flora to produce TAMs, and reduce the level of antitumor immunity (Fig. 1).

MDSCs

Previous studies have revealed that, under the action of IL-17, MDSCs interact with Bacteroides fragilis (Bf) to indirectly induce ectopic colonic epithelial cells, and to induce the expression of IL-17 in intestinal epithelial tissue. Increased IL-17 expression and activated STAT3 signaling, as well as vascular growth factors and proangiogenic mediators, collectively promote colorectal tumor progression (32).

In addition, F. nucleatum promoted the regeneration of intestinal epithelial tissue by increasing the number of MDSCs in the tumor microenvironment. In the absence of microorganisms, the expression of the MDSC ligand C-X-C motif chemokine receptor 2 was enhanced, exhibiting immunosuppressive and tumor-promoting effects (32) (Fig. 1).

Cancer-associated fibroblasts (CAFs)

CAFs induce chemoresistance in CRC through the synergistic action of hypoxia-inducible factor (HIF)-1α and TGF-β. It was revealed that CAFs can assist in tumor immune escape and resist the action of immunosuppressive drugs. They can induce an inhibitory T-cell microenvironment by recruiting chemokines and immune factors (CCL2, CXCL2, CXCL6, S100A9, IL6) (36) (Fig. 1).

Cytokines

The tumor microenvironment includes inflammatory, immune and hypoxia factors (Table III) (3750). Numerous factors connect different components in the tumor microenvironment through specific signaling pathways, so that different tumor microenvironments are linked together to form a dynamic tumor-promoting or tumor-suppressing microenvironment (Fig. 1).

Table III.

Cytokines in the tumor microenvironment.

Table III.

Cytokines in the tumor microenvironment.

ClassificationNameRole(Refs.)
Inflammatory factorsIL-1βLow concentration of IL-1β induces local inflammatory and protective immune responses, while high concentrations can lead to inflammation-related cancer tissue damage(37)
TNF-αTNF-α can continuously activate NF-κB, which regulates inflammation(38)
IL-6Participates in the inititation of the inflammatory pathway(39)
IL-10Inhibits the inflammatory response(40)
Immune factorsIL-17 Pro-inflammatory(41)
IL-22 Pro-inflammatory(42)
IL-35Antitumor activity(43)
TGF-βSuppresses the immune response(44)
ChemokinesIL-8Cancer-promoting factors promote tumor immune escape(45)
Recombinant chemokine CXCR6Increases immune cell infiltration in tumor tissue(46)
Recombinant human CCL4Achieves antitumor effects by recruiting regulatory T cells and promoting tumor macrophages(47)
Recombinant human CCL3Participates in immune monitoring and tolerance(48)
Recombinant human CXCL5/CXCR2By blocking the bridge between tumor and host cells in the tumor microenvironment, it can improve immune efficacy(49)
Recombinant human CXCL12Antitumor activity(50)

[i] CXCR, C-X-C-motif receptor; CXCL, C-X-C-motif ligand; CCL, C-C-motif ligand.

Regulatory mechanism
Metabolites
Microbial metabolites

Short-chain fatty acids (SCFAs) are the fermentation products of dietary fiber. Ohtani Haraand Hara (51) identified that, in addition to maintaining intestinal homeostasis, intestinal microbiota metabolites could also transport SCFAs to the liver through the portal vein, and produce bile acids that induce DNA damage to remodel the tumor microenvironment and regulate liver function. Huang et al (52) collected feces from patients with liver cancer and healthy individuals for bioinformatics analysis, and observed that bile acids, a metabolite of the gut flora, can affect tumor treatment and prognosis by changing the immune microenvironment. In addition, butyrate and tryptophan metabolites produced by intestinal flora metabolism can affect the adaptive immunity of the body and promote antitumor therapeutic effects (53,54). Cholesterol is metabolized in the gut to produce three metabolites: Bile acids, steroids and vitamin D. Among them, bile acids can modulate the composition of the gut microbiota to affect peripheral and autoimmune immunity, while the metabolic reprogramming of cholesterol in the tumor microenvironment can cause tumor microbiota to change to an immunosuppressive type, thus providing an environment conducive to the proliferation of cancer cells (55) (Fig. 2).

Figure 2.

Mechanisms of intestinal flora-mediated regulation of the tumor microenvironment. Metabolites: i) Metabolites of flora: SCFAs can be transmitted to the liver through the portal vein to produce bile acid and change the tumor microenvironment. Butyrate and tryptophan affect the development of tumors by affecting their immune response. ii) Intestinal metabolites: Intestinal metabolism produces polyphenol analogues, changes the type of gut microbiota, stimulates the production of SCFAs and reshape the tumor microenvironment. Non-hematopoietic components of the intestinal membrane: When intestinal epithelial cells lack ubiquitin ligase, the secretion of intestinal cathelicidin decreases, and epithelial cells die, which causes changes in the gut microbiota and regulates the tumor immune microenvironment. Genotoxins: PKS gene-positive Escherichia coli can destroy the single-stranded DNA of intestinal cells, playing an indirect role in the development of CRC, and colon epithelial cells can cause DNA damage and activate Wnt and NF under the action of fragile bacteroid toxins-κB. STAT3 and other colon epithelial signal transduction pathways affect the self-renewal of colon mucosal epithelial cells and promote tumor formation. Metabolic reprogramming: The tumor microenvironment provides nutrients for malignant proliferation and metastasis of tumor cells by changing the metabolism of different components of the tumor microenvironment. Anaerobic glycolysis of tumor cells produces lactic acid, which can stimulate the production of hyaluronic acid and the expression of CD44, and is conducive to tumor metastasis. The hypoxic acidic tumor microenvironment caused by aerobic glycolysis inhibits the normal metabolism of immune cells and T cell function. Tumor cells mediate the metabolism of tryptophan in T cells by overexpressing indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase in order to transform tryptophan into caninurenine and its metabolites. Lack of tryptophan and accumulation of caninurenine metabolites would inhibit the function of effector T cells. Immune reprogramming: i) Immune escape: Bacteroides fragilis, as an inducer of forkhead box P3+ (a powerful inducer that mediates gastrointestinal immunity and peripheral tolerance), can induce the production of IL-10-mediated mucosal surface tolerance, which can induce the generation of Tregs, while symbiotic microorganisms can promote the escape of pDCs. Both pDCs and Tregs work together to mediate immune escape; ii) reprogramming of immune cells: STING agonists derived from microbiota induce the production of IFN-I through mononuclear phagocytes in tumors, thus forming an antitumor microenvironment. By regulating the natural killer cell-DC crosstalk, the content of IFN-I is regulated, reshaping the tumor microenvironment, and facilitating the response to immune checkpoint blockade. In the absence of PTEN (gene of phosphate and tension homology deleted on chromosome ten) expression, miRNA expression and the proinflammatory bacteria Akkermania muciniphila in the gut microbiota decrease, forming a microenvironment that inhibits tumor growth. When the gut microbiota is altered, the lipopolysaccharide content, the expression of the cathepsin K gene and the risk of colon cancer cell metastasis increase. Due to the loss of miRNA, succinate produced by the microbiota accumulates in the colon, leading to diarrhea in weaned piglets. SCFAs, short-chain fatty acids; CRC, colorectal cancer; STING, stimulator of intereferon genes; Tregs, regulatory T cells; pDCs, plasmacytoid dendritic cells; miRNA, microRNA.

Intestinal metabolites

Radiotherapy and chemotherapy have serious side effects during tumor treatment; thus, an increasing number of experts recommend a diet therapy. Due to their strong antioxidant function, natural polyphenols are often used as targeted regulators for colon cancer prevention and treatment (56). In addition, it was demonstrated that natural polyphenols can not only regulate oxidative stress, cell proliferation, apoptosis and inflammatory inhibition, but can also change the type of gut microbiota that stimulates the production of SCFAs to remodel the tumor microenvironment (56) (Fig. 2).

Non-hematopoietic components of the intestinal membrane

A previous study has shown that the lack of the ubiquitin ligase ring finger protein 5 in intestinal epithelial cells can lead to decreased secretion of intestinal antimicrobial peptides and cell death, which in turn changes the intestinal flora, regulates the activity of lymphoid organs and affects tumor cell invasion (57) (Fig. 2).

Genotoxins

The intestinal flora mediates cancer through genotoxins, such as colicin produced by Escherichia coli, which acts as a DNA alkylating agent to damage host DNA (58) and induce cell senescence. Bf toxin is activated by IL-17 in colonic epithelial cells. NF-kB signal transduction produces a series of inflammatory responses and accelerates the transformation of colitis to colon cancer (59). It has been revealed that genotoxin expression is exacerbated when the gut microbiota is altered (60) (Fig. 2).

Metabolic reprogramming

The tumor microenvironment supports the malignant proliferation of tumor cells by providing nutrients and redox requirements for tumor cell proliferation through aerobic glycolysis, and metabolic reprogramming of fibroblasts, T cells, TAMs and adipocytes (61). A previous study has demonstrated that tumor metabolic reprogramming can mediate tumor immune escape. Lactic acid produced by glycolysis can stimulate tumor cell metastasis, and oxidized compounds highly expressed by tumor cells can accelerate the metabolism of tryptophan to kynurenine by T cells. The lack of tryptophan and the increase in kynurenine alter the function of T cells, rendering them unable to be activated by antigens, thus forming an immunosuppressive microenvironment (62) (Fig. 2).

Immune reprogramming
Immune escape

Gut microbes influence tumor immunotherapy in multiple ways. Some bacteria achieve antitumor efficacy by activating immunity, while some help cancer cells to escape the immune system by mediating immunosuppression (63). Thus, increasing evidence has shown that the clinical treatment effect can be improved by regulating or supplementing microorganisms in vitro (64).

Bf can induce forkhead box P3+ (a potent inducer of gastrointestinal immunity and peripheral tolerance) to induce regulatory T-cell (Treg) generation, while commensal microorganisms can promote the efflux of pDCs. pDCs and Tregs work together to mediate immune escape (65).

Reprogramming of immune cells

Mononuclear phagocytes are highly plastic, and the gut flora interferes with the reprogramming of mononuclear phagocytes in the tumor microenvironment into immunostimulatory monocytes and DCs, making the tumor microenvironment shift to a tumor suppressor environment (66,67). The mechanism is a microbiota-derived IFN-stimulating factor agonist that modulates macrophage polarization and natural killer (NK) cell-DC interactions through monocyte-induced IFN-1. Subsequent in vitro experiments confirmed that IFN-1 increased in mice under a high-fiber diet, and mononuclear phagocytes in the tumor microenvironment were remodeled, the number of DCs increased, and the efficacy of immune blocking agents was improved (68) (Fig. 2).

Signaling pathways and cytokines

The tumor microenvironment mainly includes inflammatory cytokines, immune cytokines and hypoxia cytokines. Research has revealed that, in colon cancer and other diseases, the intestinal flora, and the inflammatory, immune and hypoxic microenvironments cross-talk, are closely related and interact with each other. When the intestinal flora is imbalanced, it leads to the inflammation of epithelial cells, which leads to hypoxia. Increased HIF levels induce an increase in inflammatory (NF-κB) and immune (Th17, IL-17) factors, which aggravates inflammation and leads to cancer. The intestinal flora stimulates the release of TNF-α and VEGF, promotes angiogenesis in the tumor microenvironment, aggravates the hypoxia of the tumor microenvironment, increases the content of HIF, and further aggravates the hypoxia and inflammation of the microenvironment (6972) (Fig. 3).

Figure 3.

Signaling pathways and cytokines. The signal transmission of innate immunity in the intestine is mainly achieved through the recognition of NOD like receptors and TLRs to generate perception. Pathogen related molecular patterns of different microorganisms are recognized by TLRs, TLRs activate NF-κB and stimulate the production of inflammatory factors and chemokines. When the gut microbiota changes abnormally, TLRs will be abnormally activated, producing a series of inflammatory and tumor-promoting reactions. At the same time, they can regulate the invasion and metastasis of cancer cells via MMPs and integrins. Th17 cells can secrete IL-17 and IL-23, which can regulate Th17 cells. Blocking IL-23 secretion can alleviate inflammation. The content of HIF-1α in a hypoxic tumor environment increases, which activates NF-κB. NF-κB can increase macrophage HIF-1α expression in the tumor microenvironment. HIF-1α and TGF-β have synergistic effects. HIF-1α is positively associated with the angiogenic factor VEGF. Th cells mature into Th1, Th2, Treg and Th17 cells. Th1 cells secrete IFN-γ, while Th2 cells and Tregs secrete IL-4, IL-5 and IL-10 for antitumor effects, and Th17 cells secrete IL-17. After gut microbiota depletion, the content of IFN-γ increases and the IL-17 content decreases. PAMPs, pathogen-associated molecular pattern molecules; MMPs, matrix metalloproteinases; TLRs, toll-like receptors; Th, helper T cell; HIF, hypoxia-inducible factor; Treg, regulatory T cell.

Other mechanisms

Phosphatase and tensin homolog (PTEN), as a tumor suppressor gene, can antagonize PI3K-Akt signaling to suppress tumorigenesis (73). Although PTEN deficiency is not sufficient to induce tumorigenesis, it can accelerate tumor progression. Howe et al (73) revealed that the pro-inflammatory Acinetobacter acidophilus was greatly reduced in the microenvironment of PTEN gene-knockout mice. Therefore, it was considered that Adenobacter acidophilus could help to prevent the protumor microenvironment caused by PTEN deficiency and form a preventive tumor microenvironment. The aforementioned study linked genetic changes to the gut microbiota and tumor microenvironment, thus providing new insights for subsequent studies on the role of gut microbiota in shaping the tumor microenvironment.

Cathepsin K (CTSK) mainly acts on bone remodeling and resorption. As the only upregulated metastasis-related signal in colon cancer cells, it has been revealed that intestinal flora dysbiosis leads to increased LPS content, which in turn promotes the expression of the CTSK gene and changes the tumor stromal microenvironment to promote colon cancer cell migration and invasion into the bone (74). As transcriptional regulators, microRNAs (miRNAs) play a significant role in various physiological activities such as immunity and metabolism. Through genomic analysis, it was identified that gut microbiota may reshape the tumor microenvironment by affecting miRNAs, thereby affecting the metastasis and prognosis of CRC (75,76).

In the treatment of diarrhea in piglets, it was identified that diarrhea was caused after weaning. Concurrently, the deletion of specific miRNAs would change the abundance of specific bacteria in the intestinal flora, resulting in increased expression of specific enzymes. Succinic acid is enriched in the intestine and promotes intestinal epithelial tissue fluid. The secretion of fluid causes an inflammatory response leading to diarrhea (77).

In summary, further studies are required to investigate whether the deletion of a certain miRNA can also cause changes in the homeostasis of a target organ (such as inflammatory response, pathway stimulation or immune response) and can help to regulate tumors through interacting with their microenvironment for the treatment and diagnosis of tumors (78).

Clinical application
Probiotics

A reasonable use of probiotics (as a common mean of regulating intestinal flora imbalances) in the treatment of colon cancer can not only change the composition of the flora but also regulate the immune response of the intestinal tract, thereby preventing and treating colon cancer (79). Galunisertib, a TGF-β blocker, was revealed to relieve immunosuppression by enhancing the infiltration of specific effector T cells and promoting DC maturation in the tumor microenvironment, when combined with Bifidobacterium probiotics (80).

Fecal microbiota transplantation (FMT)

As a new therapy, FMT mainly transplants healthy human gut microbiota into patients to remodel and partly restore intestinal homeostasis. In 2013, it was used in the treatment of Clostridium difficile infection. Icreasing evidence has clarified the therapeutic effect of fecal transplantation for other diseases (18). For example, in 2021, allogeneic fecal bacterial transplantation was applied in phase I clinical trials in patients with anti-programmed cell death 1 refractory metastatic melanoma, and it was revealed that it could alter the infiltration and gene expression characteristics of immune cells in the tumor microenvironment (82). In addition, when using trastuzumab to treat a HER2-positive breast cancer mouse model, the researchers observed that allogeneic fecal bacterial transplantation enhanced the efficacy of trastuzumab in blocking cancer cell proliferation and improving immune cell infiltration in the tumor microenvironment (83). In recent years, the concept of autologous fecal transplantation has emerged, which is similar in concept to the preservation of neonatal umbilical cord blood, and implies the rejuvenation of intestinal flora. Although this concept has certain feasibility, its efficacy and safety have yet to be verified (84).

Natural extracts

Numerous studies have shown that natural plant extracts [triterpenoid saponins (85), safflower (86), Astragalus polysaccharides (87), puerarin (88)] and traditional Chinese medicine formulas [SWY (89), Wu Mei Wan (90), and parthenolide (91)] alter the tumor microenvironment by modulating the gut microbiota in vitro. However, their research is currently limited to animal experiments, and have yet to be used in clinical practice (92) (Table IV).

Table IV.

Regulation of intestinal flora by natural extracts.

Table IV.

Regulation of intestinal flora by natural extracts.

PlantExtractFlora changeRegulatory mechanismDisease treatmentSubjects(Refs.)
Carthamus tinctoriusSafflower yellowBarnesiella and Ersipelotrichaceae incertae sedis return to normal levelsImprovement of the immune microenvironmentHepatocellular carcinomaIn vivo test in mice(86)
Astragalus membranaceusAstragalus polysaccharidesRegulation of changes in Bifidobacterium pseudolongum, Lactobacillus johnsonii and Lactobacillus floraImprovement of the metabolism of glutamate and creatine could control tumor growthMelanomaIn vivo test in mice(87)
Pueraria lobataPuerarinRestoring imbalance of firmicutes to acteroidesRegulation of the content of SCFAs and aminoacid metabolites by adjusting the gut/bone axis could improve the bone microenvironmentOsteoporosisIn vivo test in mice(88)
Zingiber officinale Roscoe, Allium tuberosum Rottler ex Spreng., Pyrus Bretschneider Rehder, Nelumbo nucifera Gaertn.Chinese herbal compound extractThe expression of the class Bacilli, the genus Turicibacter, the family Turicibacteae, and the order Turicibacterales return to normal levelsRegulation of metabolism, changes in the tumor microenvironmentEsophageal precancerous lesionIn vivo test in mice(89)
Wumei, Huanglian, Xixin, Guizhi, Dangshen, Fuzi, Huajiao, Ganjiang, Huangbai, DangguiChinese herbal compound extractBacteroidetes and firmicutes imbalance restorationDownregulation of the NF-κB/IL-6/STAT3 signaling pathway to regulate the inflammatory environment of the intestineColorectal cancerIn vivo test in mice(90)
FeverfewParthenolide Alloprevotella displayed high abundanceImproves the Treg/Th17 balance in intestinal mucosa mediated through increased microbiota-derived SCFA productionInflammatory bowel diseaseIn vivo test in mice(91)

[i] Treg, regulatory T cell; Th, helper T cell; SCFA, short-chain fatty acid.

Diet

Diet is the most direct and important factor affecting the intestinal flora. It has been demonstrated that a high-fat diet can change the intestinal flora to accelerate intestinal inflammation through direct or extraintestinal effects, and change the metabolism and tumor immune microenvironment (93). A previous study has also revealed that a high-fat diet increases the sensitivity of the gut to carcinogens (94). Therefore, scientists suggest that a ketogenic and high-fiber diet can be used to regulate intestinal flora metabolism and tumor microenvironment (95). Dietary carrageenan, as a food additive, alters the gut microbiota resulting in SCFA reduction, mucosal thinning and changes in intestinal homeostasis to form a proinflammatory microenvironment. Therefore, it was speculated that this inflammatory response can be reversed by supplementation with probiotics (96). In addition to the direct factor of diet, the intestinal flora of the human body also includes the host environment. Since the growth of intestinal flora requires the body to provide ATP to support its growth and form colonies, factors such as a poor diet, antibiotics and intestinal diseases weaken the control of the body over the flora, thus resulting in changes in flora homeostasis. It is thus possible to quantify the conditions that control the growth of the microbiota, thereby defining the homeostasis and imbalances of the gut microbiota, and regulating microbiota imbalances (97).

Biological targets

The tumor microenvironment, as the place of tumor growth, regulates the occurrence, development and metastasis of tumors. As an important factor influencing the tumor microenvironment, intestinal microbiota has been demonstrated to interact with the tumor microenvironment. Based on extensive literature review, it has been revealed that the role of microorganisms in mediating the tumor microenvironment and then influencing tumor progression is played by a group of bacteria rather than a specific strain. The following is a summary of relevant biological targets, providing potential insights for their clinical applications (Table V).

Table V.

Mechanism of action and diseases of biological targets.

Table V.

Mechanism of action and diseases of biological targets.

Target actionTarget nameMechanism of actionDisease(Refs.)
PreventAnti-inflammatory characteristic bacteria with potential for producing butyric acidChanges in microbiota result in changes in the content of butyric acid, a metabolite of the microbiotaColon cancer(98)
Bile acidFormation of an inflammatory microenvironmentHepatocellular carcinoma risk prediction and grading(99)
DiagnosisNumber of DNA fragments from Fusobacterium nucleatumMicrobiota dictates tolerogenic vs. immunogenic cell death of intestinal epithelial cellsStaging, metastasis, chemotherapy resistance, and prognosis of colon cancer(100)
Increased Leptotrichia and decreased Porphyromonas in salivaGut microbiota can be connected in series with related flora through the mesentery lymphatic pathwayEarly diagnosis of pancreatic cancer(101)
Abundance of AkkermansiaSecretion of interferon by CD8+ T cells causes tumor killingDiagnostic and therapeutic targets of ovarian cancer(102)
TreatmentBeneficial microbiota such as Bifidobacterium, Bacteroides fragilis and Akkermansia muciniphiaRegulation of the tumor immune microenvironmentMay be used as a sensitive target for immunotherapy(103,105)
PrognosisCTSKCTSK secreted by tumors can bind to Toll-like receptor 4 and stimulate M2 polarization of tumor-associated macrophages through the mTOR-dependent pathway, thereby inhibiting the development and metastasis of colorectal cancerInvasion and metastasis of colon cancer(74)
Pseudomonas, Saccharopolysaccharide, Streptomyces and ClostridiumEffects on tumor growth and immune infiltrationPrediction of long-term survival of patients with pancreatic cancer(109)

[i] CRC, colorectal cancer; CTSK, cathepsin K.

Prevention

It has been identified that the induction of SCFA is strain-specific, thus, its inductive ability can be inferred from the abundance of gut flora to predict changes in the tumor microenvironment (98).

Quantitative analysis of the composition of bile acid, the metabolite of gut microbiota, and bile salts in feces can be used to identify the risk index of HCC, and to further combine the composition and category of gut microbiota to grade the risk of HCC. This may be related to the inflammatory environment formed by the gut microbiota and the tumor microenvironment, which can stimulate the occurrence and development of tumors. Therefore, the observation and intervention of gut microbiota can be a good means for the prevention and treatment of HCC (99).

Diagnosis

The quantity of Fusobacterium nucleatum DNA in the intestinal flora has been revealed to be positively associated with tumor stage, metastasis and patient survival. In clinical practice, the level of Fusobacterium nucleatum DNA can be measured for colon cancer tumor staging, metastasis, chemotherapy resistance, sex and prognosis (100). There are differences in the types and abundance of gut microbiota among different diseases, such as Bacteroides, Lachnospiraceae incertae sedis, and Clostridium XIV, which can be used to identify small liver cancer (52).

Pancreatic cancer, as the most lethal malignant tumor, can not be diagnosed by common detection methods in the early, or even in the mid or late stages (101). Yang et al (101) revealed that Leptotrichia increased and Porphyromonas decreased in the saliva of patients with pancreatic cancer, suggesting that it could be used as a marker for early diagnosis. Gut microbiota can enter the pancreas through the mesentery lymphatic pathway to connect different flora, and affect the occurrence and development of pancreatic cancer remotely.

Wang et al (102) revealed that Akkermansia in the intestines of patients with ovarian cancer was significantly reduced by analyzing the abundance of gut microbiota of patients. In addition, when the gut microbiota of these patients was inoculated into mice by fecal bacteria transplantation, the progression of ovarian cancer in mice was accelerated. The addition of Akkermania can significantly inhibit the progression of ovarian cancer in mice. This research has shown that Akkermania restores the integrity of the intestinal mucosa, activates T-cell immune response in the tumor microenvironment, and strengthens immune monitoring. This aforementioned study (102) provided a new direction for the relationship between gut microbiota and the immune microenvironment in ovarian cancer, and also suggests that Akkermania can be used as a new target for diagnosis and treatment of ovarian cancer.

Treatment

A previous study has demonstrated that the intestinal flora inhibits apoptosis, changes epigenetic transplantation, repairs damaged DNA and participates in other mechanisms to generate therapeutic resistance, but it can also be used as a target to manipulate and improve the therapeutic effect of treatments (103).

Traditional radiotherapy is the most effective method to treat tumors. As an important factor in regulating the tumor microenvironment, gut microbes are impacted from the effect of radiotherapy. It has been revealed that there are differences in the sensitivity of different gut microbiota to radiotherapy, but the specific mechanism remains unknown (104). Therefore, the sensitivity of patients to radiotherapy may be evaluated by analyzing the types of intestinal flora, with the intent that the treatment plan can be timely adjusted.

Traditional radiotherapy and chemotherapy are aimed at the tumor cells themselves, using physical rays and chemical drugs to kill them, but drug resistance is prone to occur. It has been revealed that the combination of traditional therapy and immunotherapy can greatly reduce the drug resistance of tumor cells and improve the therapeutic effect. Research has shown that gut microbiota can affect the effectiveness of immunotherapy (105). PD-1/PD-L1 has good efficacy in the treatment of solid tumors, and has been demonstrated that, in in vitro experiments in mouse models, mice with oral microbiota have improved anti-PD-1 efficacy than untreated mice (32). Transplanting fecal bacteria from patients who have responded to anti-PD-1 antibodies into germ-free mice could significantly improve the control effect of T cells on tumors, and have a favorable effect on PD-1/PD-L1 immunotherapy. Immune checkpoint inhibitors (ICIs), as a new treatment method, exhibit favorable curative effects, but some patients are insensitive to them or develop resistance to their long-term use (106). It was shown that patients who responded well to ICIs had a high number of beneficial bacteria in the gut (Bifidobacterium, Bf, Akkermansia muciniphia), which could help to restore and enhance the therapeutic effect of ICI and immunotherapy in patients (79). As the latest targeted therapy, chimeric antigen receptor T-cell immunotherapy (CAR-T) is based on the principle of isolating the T lymphocytes of the patient, expanding them in vitro to make them carry tumor cell antigens, and then infusing them back into the body of the patient, in order to achieve rapid and precise tumor treatment. Through clinical stool sample observation and genome sequencing analysis, it was identified that, in patients with B-cell malignancies, there was a strong association between changes in gut microbiota and clinical treatment outcomes of CAR-T therapy (107).

In addition, studies have demonstrated that microorganisms in tumors, oral microbiota, and other factors can affect the tumor immune microenvironment, thereby affecting tumor immunotherapy. Therefore, understanding the relationship between microorganisms, the tumor microenvironment and diseases provides a new target for better use of microorganisms to treat diseases accurately (108).

Prognosis

Pancreatic cancer is a malignant tumor of the digestive tract with extremely high mortality, because its early diagnosis is difficult. Yang et al (101) revealed that the imbalance of intestinal microbiota is closely related to the incidence and prognosis of pancreatic cancer. In addition, Huang et al (52) revealed that high bile acid metabolism, low levels of Bacteroides, Lachnospiracea incertae sedis, and Clostridium XIVa and content of operational taxonomy unit markers related to bile acid metabolism could be used to predict the postoperative survival time of patients with liver cancer.

As a measure of gut microbiota imbalance and CRC metastasis, CTSK secreted by CRC could accelerate the phenotype transformation of TAMs to M2 by regulating the TLR4-mTOR signaling pathway, thereby accelerating the progression of CRC. Concurrently, it can secrete inflammatory factors to promote cancer cell invasion and metastasis. Therefore, it has been suggested that CTSK may serve as a new prognostic and therapeutic target for CRC (74). In other research, four characteristic microbes in the tumor microbiota (Pseudomonas, Glycopolysaccharides, Streptomyces and Clostridium), which could predict the long-term survival of patients with pancreatic cancer, were also identified. Using donor fecal microbiota transplantation, it was determined that the tumor microbiota may be regulated differently and affect tumor growth and immune infiltration (109).

Conclusions

Overall, the gut microbiota regulates intestinal and distant tumors through changes in the microbiota, brain-gut axis, hypothalamic pituitary adrenal axis, intestinal liver axis and bacterial translocation. It is mainly manifested in the regulation of each component in the tumor microenvironment, to achieve the regulation of the tumor microenvironment. Its regulatory mechanisms include changes in gut and microbiota metabolites, gene toxins, metabolic reprogramming, immune reprogramming, signaling pathways and cytokines. The transplantation of probiotics and fecal microbiota methods in the treatment of tumors in the microenvironment of gut microbiota regulation have matured. However, other treatment methods are still at the theoretical stage and have not been clinically validated, and their adverse effects on the body remain unclear. In addition, the present review also summarized and compared the biological targets of gut microbiota regulating the tumor microenvironment, providing a theoretical basis for future applications in the prevention, diagnosis, treatment and prognosis of tumors (Fig. 4).

At present, most of the specific microbiota and pathway changes in the regulatory mechanism of disease-gut microbiota-tumor microenvironment lack in vivo and in vitro experiments. Numerous studies have demonstrated that substances such as vitamins (81) and lactic acid produced by glycolysis in the tumor microenvironment (110) can change the intestinal flora or tumor microenvironment, however whether these substances mediate the regulation of gut microbiota in the tumor microenvironment has not yet been elucidated, which will open up new directions for future research. As a key cell in tumor immune regulation, tumor-infiltrating myeloid cells, a component of the tumor microenvironment, require modification and activation by m6A methylation (111). Previous studies have revealed that the methylation of intestinal flora can interfere with the expression of the oncogenic gene p53 and activate it. SCFAs promote early onset and metastasis of tumors (112), and it has been shown that quantification of m6A methylation may serve as a potential biological target for pancreatic cancer prognosis (113). Thus, whether the methylation of gut microbiota also regulates tumors by affecting the function of tumor-infiltrating myeloid cells, provides a new objective for future research.

There are complex connections between the gut microbiota and the tumor microenvironment, and the gut microbiota-tumor microenvironment directly affects the prevention, diagnosis, treatment and prognosis of diseases. Therefore, an in-depth study of the association between the gut microbiota and the tumor microenvironment will provide new means for the targeted treatment of clinically common and difficult tumors by regulating the intestinal flora and tumor microenvironment in the future.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Inner Mongolia Autonomous Region Health Science and Technology Plan Project Assignment (grant no. 202201184), the Inner Mongolia Medical University Zhiyuan Talent Program (Good Learning Talent Program) (grant no. ZY0202031), the Program for Young Talents of Science and Technology in Universities of Inner Mongolia Autonomous Region (grant no. NJYT23050) and the Inner Mongolia Autonomous Region ‘Grassland Talent’ project youth innovation and entrepreneurship talent project (grant no. 2022073).

Availability of data and materials

Not applicable.

Authors' contributions

CH and GH conceived and designed this review. TX and YL wrote the first draft. SJ critically revised the review for important intellectual content. ZR, SR, ZY, ZJ and TC contributed in the writing of the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

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

1 

Peng-Xu W, Xin-Ru D, Chen-Hong Z and Hui-Juan Y: Gut microbiota and metabolic syndrome. Chin Med J. 133:808–816. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Freeman JW: Structural biology of the tumor microenvironment. Adv Exp Med Biol. 1350:91–100. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Wong-Rolle A, Wei HK, Zhao C and Jin C: Unexpected guests in the tumor microenvironment: Microbiome in cancer. Protein Cell. 12:426–435. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Gacesa R, Kurilshikov A, Vich Vila A, Sinha T, Klaassen MAY, Bolte LA, Andreu-Sánchez S, Chen L, Collij V, Hu S, et al: Environmental factors shaping the gut microbiome in a Dutch population. Nature. 604:732–739. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Sebastián Domingo JJ and Sánchez Sánchez C: From the intestinal flora to the microbiome. Rev Esp Enferm Dig. 110:51–56. 2018.PubMed/NCBI

6 

Lucas C, Barnich N and Nguyen HTT: Microbiota, inflammation and colorectal cancer. Int J Mol Sci. 18:13102017. View Article : Google Scholar : PubMed/NCBI

7 

Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, Agdashian D, Terabe M, Berzofsky JA, Fako V, et al: Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 360:eaan59312018. View Article : Google Scholar : PubMed/NCBI

8 

Rabelo-Gonçalves EM, Roesler BM and Zeitune JM: Extragastric manifestations of Helicobacter pylori infection: Possible role of bacterium in liver and pancreas diseases. World J Hepatol. 7:2968–2979. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Knezevic J, Starchl C, Tmava Berisha A and Amrein K: Thyroid-gut-axis: How does the microbiota influence thyroid function? Nutrients. 12:17692020. View Article : Google Scholar : PubMed/NCBI

10 

Zheng Y, Fang Z, Xue Y, Zhang J, Zhu J, Gao R, Yao S, Ye Y, Wang S, Lin C, et al: Specific gut microbiome signature predicts the early-stage lung cancer. Gut Microbes. 11:1030–1042. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Gao X, Miao R, Zhu Y, Lin C, Yang X, Jia R, Linghan K, Wan C and Deng J: A new insight into acute lymphoblastic leukemia in children: Influences of changed intestinal microfloras. BMC Pediatr. 20:2902020. View Article : Google Scholar : PubMed/NCBI

12 

Westfall S, Caracci F, Estill M, Frolinger T, Shen L and Pasinetti GM: Chronic Stress-induced depression and anxiety priming modulated by Gut-brain-axis immunity. Front Immunol. 12:6705002021. View Article : Google Scholar : PubMed/NCBI

13 

Angelucci F, Cechova K, Amlerova J and Hort J: Antibiotics, gut microbiota, and Alzheimer's disease. J Neuroinflammation. 16:1082019. View Article : Google Scholar : PubMed/NCBI

14 

Ma Q, Li Y, Li P, Wang M, Wang J, Tang Z, Wang T, Luo L, Wang C, Wang T and Zhao B: Research progress in the relationship between type 2 diabetes mellitus and intestinal flora. Biomed Pharmacother. 117:1091382019. View Article : Google Scholar : PubMed/NCBI

15 

Dodiya HB, Forsyth CB, Voigt RM, Engen PA, Patel J, Shaikh M, Green SJ, Naqib A, Roy A, Kordower JH, et al: Chronic stress-induced gut dysfunction exacerbates Parkinson's disease phenotype and pathology in a rotenone-induced mouse model of Parkinson's disease. Neurobiol Dis. 135:1043522020. View Article : Google Scholar : PubMed/NCBI

16 

Li Z, Lai J, Zhang P, Ding J, Jiang J, Liu C, Huang H, Zhen H, Xi C, Sun Y, et al: Multi-omics analyses of serum metabolome, gut microbiome and brain function reveal dysregulated microbiota-gut-brain axis in bipolar depression. Mol Psychiatry. 27:4123–4135. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Paget S: The distribution of secondary growths in cancer of the breast 1889. Cancer Metastasis Rev. 8:98–101. 1989.PubMed/NCBI

18 

Arneth B: Tumor microenvironment. Medicina (Kaunas). 56:152019. View Article : Google Scholar : PubMed/NCBI

19 

Li Y, Zhao L and Li XF: Hypoxia and the tumor microenvironment. Technol Cancer Res Treat. 20:153303382110363042021. View Article : Google Scholar : PubMed/NCBI

20 

Meng X, Xu Y and Ning X: Tumor microenvironment acidity modulates ROR1 to promote epithelial-mesenchymal transition and hepatocarcinoma metastasis. J Cell Sci. 134:jcs2553492021. View Article : Google Scholar : PubMed/NCBI

21 

Greenwood E: A perfect mismatch. Nat Rev Cancer. 2:76–77. 2002. View Article : Google Scholar

22 

Denton AE, Roberts EW and Fearon DT: Stromal cells in the tumor microenvironment. Adv Exp Med Biol. 1060:99–114. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Russo M and Nastasi C: Targeting the tumor microenvironment: A close up of tumor-associated macrophages and neutrophils. Front Oncol. 12:8715132022. View Article : Google Scholar : PubMed/NCBI

24 

Ngambenjawong C, Gustafson HH and Pun SH: Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv Drug Deliv Rev. 114:206–221. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Yin Z, Li C, Wang J and Xue L: Myeloid-derived suppressor cells: Roles in the tumor microenvironment and tumor radiotherapy. Int J Cancer. 144:933–946. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Patente TA, Pinho MP, Oliveira AA, Evangelista GCM, Bergami-Santos PC and Barbuto JAM: Human dendritic cells: Their heterogeneity and clinical application potential in cancer immunotherapy. Front Immunol. 9:31762018. View Article : Google Scholar : PubMed/NCBI

27 

Spinelli FM, Vitale DL, Sevic I and Alaniz L: Hyaluronan in the tumor microenvironment. Adv Exp Med Biol. 1245:67–83. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Wu M, Bai J, Ma C, Wei J and Du X: The role of gut microbiota in tumor immunotherapy. J Immunol Res. 2021:50615702021. View Article : Google Scholar : PubMed/NCBI

29 

Zhang J, Zhang F, Zhao C, Xu Q, Liang C, Yang Y, Wang H, Shang Y, Wang Y, Mu X, et al: Dysbiosis of the gut microbiome is associated with thyroid cancer and thyroid nodules and correlated with clinical index of thyroid function. Endocrine. 64:564–574. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Quigley EMM: Microbiota-brain-gut axis and neurodegenerative diseases. Curr Neurol Neurosci Rep. 17:942017. View Article : Google Scholar : PubMed/NCBI

31 

Chen Y, Zhou J and Wang L: Role and mechanism of gut microbiota in human disease. Front Cell Infect Microbiol. 11:6259132021. View Article : Google Scholar : PubMed/NCBI

32 

Yang X, Guo Y, Chen C, Shao B, Zhao L, Zhou Q, Liu J, Wang G, Yuan W and Sun Z: Interaction between intestinal microbiota and tumour immunity in the tumour microenvironment. Immunology. 164:476–493. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Maito F, Souza A, Pereira L, Smithey M and Bonorino C: Intratumoral TLR-4 agonist injection is critical for modulation of tumor microenvironment and tumor rejection. Isrn Immunology. 2012:9268172012. View Article : Google Scholar

34 

Albrengues J, Shields MA, Ng D, Park CG, Ambrico A, Poindexter ME, Upadhyay P, Uyeminami DL, Pommier A, Küttner V, et al: Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 361:eaao42272018. View Article : Google Scholar : PubMed/NCBI

35 

Zhou Z, Chen J, Yao H and Hu H: Fusobacterium and colorectal cancer. Front Oncol. 8:3712018. View Article : Google Scholar : PubMed/NCBI

36 

Monteran L and Erez NL: The dark side of fibroblasts: Cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front Immunol. 10:18352019. View Article : Google Scholar : PubMed/NCBI

37 

Zhang W, Borcherding N and Kolb R: IL-1 signaling in tumor microenvironment. Adv Exp Med Biol. 1240:1–23. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Long Q, Huang C, Meng Q, Peng J, Yao F, Du D, Wang X, Zhu W, Shi D, Xu X, et al: TNF patterns and tumor microenvironment characterization in head and neck squamous cell carcinoma. Front Immunol. 12:7548182021. View Article : Google Scholar : PubMed/NCBI

39 

Karakasheva TA, Lin EW, Tang Q, Qiao E, Waldron TJ, Soni M, Klein-Szanto AJ, Sahu V, Basu D, Ohashi S, et al: IL-6 mediates Cross-talk between tumor cells and activated fibroblasts in the tumor microenvironment. Cancer Res. 78:4957–4970. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Batchu RB, Gruzdyn OV, Kolli BK, Dachepalli R, Umar PS, Rai SK, Singh N, Tavva PS, Weaver DW and Gruber SA: IL-10 signaling in the tumor microenvironment of ovarian cancer. Adv Exp Med Biol. 1290:51–65. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Gorczynski RM: IL-17 signaling in the tumor microenvironment. Adv Exp Med Biol. 1240:47–58. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Jiang R and Sun B: IL-22 signaling in the tumor microenvironment. Adv Exp Med Biol. 1290:81–88. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Liu K, Huang A, Nie J, Tan J, Xing S, Qu Y and Jiang K: IL-35 regulates the function of immune cells in tumor microenvironment. Front Immunol. 12:6833322021. View Article : Google Scholar : PubMed/NCBI

44 

Lan Y, Moustafa M, Knoll M, Xu C, Furkel J, Lazorchak A, Yeung TL, Hasheminasab SM, Jenkins MH, Meister S, et al: Simultaneous targeting of TGF-β/PD-L1 synergizes with radiotherapy by reprogramming the tumor microenvironment to overcome immune evasion. Cancer Cell. 39:1388–403.e10. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Jobe NP, Rösel D, Dvořánková B, Kodet O, Lacina L, Mateu R, Smetana K and Brábek J: Simultaneous blocking of IL-6 and IL-8 is sufficient to fully inhibit CAF-induced human melanoma cell invasiveness. Histochem Cell Biol. 146:205–217. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Di Pilato M, Kfuri-Rubens R, Pruessmann JN, Ozga AJ, Messemaker M, Cadilha BL, Sivakumar R, Cianciaruso C, Warner RD, Marangoni F, et al: CXCR6 positions cytotoxic T cells to receive critical survival signals in the tumor microenvironment. Cell. 184:4512–30.e22. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Mukaida N, Sasaki SI and Baba T: CCL4 signaling in the tumor microenvironment. Adv Exp Med Biol. 1231:23–32. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Ntanasis-Stathopoulos I, Fotiou D and Terpos E: CCL3 signaling in the tumor microenvironment. Adv Exp Med Biol. 1231:13–21. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Zhang W, Wang H, Sun M, Deng X, Wu X, Ma Y, Li M, Shuoa SM, You Q and Miao L: CXCL5/CXCR2 axis in tumor microenvironment as potential diagnostic biomarker and therapeutic target. Cancer Commun (Lond). 40:69–80. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Portella L, Bello AM and Scala S: CXCL12 signaling in the tumor microenvironment. Adv Exp Med Biol. 1302:51–70. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Ohtani N and Hara E: Gut-liver axis-mediated mechanism of liver cancer: A special focus on the role of gut microbiota. Cancer Sci. 112:4433–4443. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Huang H, Ren Z, Gao X, Hu X, Zhou Y, Jiang J, Lu H, Yin S, Ji J, Zhou L and Zheng S: Integrated analysis of microbiome and host transcriptome reveals correlations between gut microbiota and clinical outcomes in HBV-related hepatocellular carcinoma. Genome Med. 12:1022020. View Article : Google Scholar : PubMed/NCBI

53 

Gargaro M, Manni G, Scalisi G, Puccetti P and Fallarino F: Tryptophan metabolites at the crossroad of immune-cell interaction via the aryl hydrocarbon receptor: Implications for tumor immunotherapy. Int J Mol Sci. 22:46442021. View Article : Google Scholar : PubMed/NCBI

54 

He Y, Fu L, Li Y, Wang W, Gong M, Zhang J, Dong X, Huang J, Wang Q, Mackay CR, et al: Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8+ T cell immunity. Cell Metab. 33:988–1000.e7. 2021. View Article : Google Scholar : PubMed/NCBI

55 

Xi Y, Yani Z, Jing M, Yinhang W, Xiaohui H, Jing Z, Quan Q and Shuwen H: Mechanisms of induction of tumors by cholesterol and potential therapeutic prospects. Biomed Pharmacother. 144:1122772021. View Article : Google Scholar : PubMed/NCBI

56 

Long J, Guan P, Hu X, Yang L, He L, Lin Q, Luo F, Li J, He X, Du Z and Li T: Natural polyphenols as targeted modulators in colon cancer: Molecular mechanisms and applications. Front Immunol. 12:6354842021. View Article : Google Scholar : PubMed/NCBI

57 

Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA and Knight R: The microbiome and human cancer. Science. 371:eabc45522021. View Article : Google Scholar : PubMed/NCBI

58 

Arthur JC, Perez-Chanona E, Mühlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB, et al: Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 338:120–123. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Boleij A, Hechenbleikner EM, Goodwin AC, Badani R, Stein EM, Lazarev MG, Ellis B, Carroll KC, Albesiano E, Wick EC, et al: The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients. Clin Infect Dis. 60:208–215. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Dutilh BE, Backus L, van Hijum SA and Tjalsma H: Screening metatranscriptomes for toxin genes as functional drivers of human colorectal cancer. Best Pract Res Clin Gastroenterol. 27:85–99. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Nenkov M, Ma Y, Gaßler N and Chen Y: Metabolic reprogramming of colorectal cancer cells and the microenvironment: implication for therapy. Int J Mol Sci. 22:62622021. View Article : Google Scholar : PubMed/NCBI

62 

Cheong JE, Ekkati A and Sun L: A patent review of IDO1 inhibitors for cancer. Expert Opin Ther Pat. 28:317–330. 2018. View Article : Google Scholar : PubMed/NCBI

63 

He Y, Huang J, Li Q, Xia W, Zhang C, Liu Z, Xiao J, Yi Z, Deng H, Xiao Z, et al: Gut Microbiota and tumor immune escape: A new perspective for improving tumor immunotherapy. Cancers (Basel). 14:53172022. View Article : Google Scholar : PubMed/NCBI

64 

Spencer CN, McQuade JL, Gopalakrishnan V, McCulloch JA, Vetizou M, Cogdill AP, Khan MAW, Zhang X, White MG, Peterson CB, et al: Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science. 374:1632–1640. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Ling Z, Shao L and Liu X, Cheng Y, Yan C, Mei Y, Ji F and Liu X: Regulatory T cells and plasmacytoid dendritic cells within the tumor microenvironment in gastric cancer are correlated with gastric microbiota dysbiosis: A preliminary study. Front Immunol. 10:5332019. View Article : Google Scholar : PubMed/NCBI

66 

Nakamura K and Smyth MJ: Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol Immunol. 17:1–12. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J, Chudnovskiy A, Maskey S, Walker L, Finnigan JP, et al: A conserved dendritic-cell regulatory program limits antitumour immunity. Nature. 580:257–262. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Lam KC, Araya RE, Huang A, Chen Q, Di Modica M, Rodrigues RR, Lopès A, Johnson SB, Schwarz B, Bohrnsen E, et al: Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment. Cell. 184:5338–5356.e21. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, Zhang L, Sharma U, Giri B, Garg B, et al: Gut microbiota promotes tumor growth in mice by modulating immune response. Gastroenterology. 155:33–37.e6. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Gagliani N, Hu B, Huber S, Elinav E and Flavell RA: The fire within: Microbes inflame tumors. Cell. 157:776–783. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Tang YA, Chen YF, Bao Y, Mahara S, Yatim S, Oguz G, Lee PL, Feng M, Cai Y, Tan EY, et al: Hypoxic tumor microenvironment activates GLI2 via HIF-1α and TGF-β2 to promote chemoresistance in colorectal cancer. Proc Natl Acad Sci USA. 115:E5990–E5999. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Rius J, Guma M, Schachtrup C, Akassoglou K, Zinkernagel AS, Nizet V, Johnson RS, Haddad GG and Karin M: NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature. 453:807–811. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Howe C, Kim SJ, Mitchell J, Im E, Kim YS, Kim YS and Rhee SH: Differential expression of tumor-associated genes and altered gut microbiome with decreased Akkermansia muciniphila confer a tumor-preventive microenvironment in intestinal epithelial Pten-deficient mice. Biochim Biophys Acta Mol Basis Dis. 1864:3746–3758. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Li R, Zhou R, Wang H, Li W, Pan M, Yao X, Zhan W, Yang S, Xu L, Ding Y and Zhao L: Gut microbiota-stimulated cathepsin K secretion mediates TLR4-dependent M2 macrophage polarization and promotes tumor metastasis in colorectal cancer. Cell Death Differ. 26:2447–2463. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Zhou S, Zhu C, Jin S, Cui C, Xiao L, Yang Z, Wang X and Yu J: The intestinal microbiota influences the microenvironment of metastatic colon cancer by targeting miRNAs. FEMS Microbiol Lett. 369:fnac0232022. View Article : Google Scholar : PubMed/NCBI

76 

Xing SC, Huang CB, Wu RT, Yang YW, Chen JY, Mi JD, Wu YB, Wang Y and Liao XD: Breed differences in the expression levels of gga-miR-222a in laying hens influenced H2S production by regulating methionine synthase genes in gut bacteria. Microbiome. 9:1772021. View Article : Google Scholar : PubMed/NCBI

77 

Zhou X, Liu Y, Xiong X, Chen J, Tang W, He L, Zhang Z, Yin Y and Li F: Intestinal accumulation of microbiota-produced succinate caused by loss of microRNAs leads to diarrhea in weanling piglets. Gut Microbes. 14:20913692022. View Article : Google Scholar : PubMed/NCBI

78 

Li Z, Zhang X, Liu C and Ma J: Non-immune cell components in the gastrointestinal tumor microenvironment influencing tumor immunotherapy. Front Cell Dev Biol. 9:7299412021. View Article : Google Scholar : PubMed/NCBI

79 

Ding S, Hu C, Fang J and Liu G: The protective role of probiotics against colorectal cancer. Oxid Med Cell Longev. 2020:88845832020. View Article : Google Scholar : PubMed/NCBI

80 

Shi L, Sheng J, Wang M, Luo H, Zhu J, Zhang B, Liu Z and Yang X: Combination therapy of TGF-β blockade and commensal-derived probiotics provides enhanced antitumor immune response and tumor suppression. Theranostics. 9:4115–4129. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Yue Y, Ye K, Lu J, Wang X, Zhang S, Liu L, Yang B, Nassar K, Xu X, Pang X and Lv J: Probiotic strain Lactobacillus plantarum YYC-3 prevents colon cancer in mice by regulating the tumour microenvironment. Biomed Pharmacother. 127:1101592020. View Article : Google Scholar : PubMed/NCBI

82 

Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al: Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 371:602–609. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Di Modica M, Gargari G, Regondi V, Bonizzi A, Arioli S, Belmonte B, De Cecco L, Fasano E, Bianchi F, Bertolotti A, et al: Gut microbiota condition the therapeutic efficacy of trastuzumab in HER2-positive breast cancer. Cancer Res. 81:2195–2206. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Ke S, Weiss ST and Liu YY: Rejuvenating the human gut microbiome. Trends Mol Med. 28:619–630. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Chen L, Brar MS, Leung FC and Hsiao WL: Triterpenoid herbal saponins enhance beneficial bacteria, decrease sulfate-reducing bacteria, modulate inflammatory intestinal microenvironment and exert cancer preventive effects in ApcMin/+ mice. Oncotarget. 7:31226–31242. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Fu H, Liu X, Jin L, Lang J, Hu Z, Mao W, Cheng C and Shou Q: Safflower yellow reduces DEN-induced hepatocellular carcinoma by enhancing liver immune infiltration through promotion of collagen degradation and modulation of gut microbiota. Food Funct. 12:10632–10643. 2021. View Article : Google Scholar : PubMed/NCBI

87 

Ding G, Gong Q, Ma J, Liu X, Wang Y and Cheng X: Immunosuppressive activity is attenuated by Astragalus polysaccharides through remodeling the gut microenvironment in melanoma mice. Cancer Sci. 112:4050–4063. 2021. View Article : Google Scholar : PubMed/NCBI

88 

Li B, Liu M, Wang Y, Gong S, Yao W, Li W, Gao H and Wei M: Puerarin improves the bone micro-environment to inhibit OVX-induced osteoporosis via modulating SCFAs released by the gut microbiota and repairing intestinal mucosal integrity. Biomed Pharmacother. 132:1109232020. View Article : Google Scholar : PubMed/NCBI

89 

Shi HJ, Chen XY, Chen XR, Wu ZB, Li JY, Sun YQ, Shi DX and Li J: Chinese medicine formula Siwu-Yin inhibits esophageal precancerous lesions by improving intestinal flora and macrophage polarization. Front Pharmacol. 13:8123862022. View Article : Google Scholar : PubMed/NCBI

90 

Jiang F, Liu M, Wang H, Shi G, Chen B, Chen T, Yuan X, Zhu P, Zhou J, Wang Q and Chen Y: Wu Mei wan attenuates CAC by regulating gut microbiota and the NF-kB/IL6-STAT3 signaling pathway. Biomed Pharmacother. 125:1099822020. View Article : Google Scholar : PubMed/NCBI

91 

Liu YJ, Tang B, Wang FC, Tang L, Lei YY, Luo Y, Huang SJ, Yang M, Wu LY, Wang W, et al: Parthenolide ameliorates colon inflammation through regulating Treg/Th17 balance in a gut microbiota-dependent manner. Theranostics. 10:5225–5241. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Li T, Han L, Ma S, Lin W, Ba X, Yan J, Huang Y, Tu S and Qin K: Interaction of gut microbiota with the tumor microenvironment: A new strategy for antitumor treatment and traditional Chinese medicine in colorectal cancer. Front Mol Biosci. 10:11403252023. View Article : Google Scholar : PubMed/NCBI

93 

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

94 

Jin H and Zhang C: High fat high calories diet (HFD) increase gut susceptibility to carcinogens by altering the gut microbial community. J Cancer. 11:4091–4098. 2020. View Article : Google Scholar : PubMed/NCBI

95 

AlHilli MM and Bae-Jump V: Diet and gut microbiome interactions in gynecologic cancer. Gynecol Oncol. 159:299–308. 2020. View Article : Google Scholar : PubMed/NCBI

96 

Wu W, Zhou J, Xuan R, Chen J, Han H, Liu J, Niu T, Chen H and Wang F: Dietary κ-carrageenan facilitates gut microbiota-mediated intestinal inflammation. Carbohydr Polym. 277:1188302022. View Article : Google Scholar : PubMed/NCBI

97 

Lee JY, Tsolis RM and Bäumler AJ: The microbiome and gut homeostasis. Science. 377:eabp99602022. View Article : Google Scholar : PubMed/NCBI

98 

Peterson CT, Perez Santiago J, Iablokov SN, Chopra D, Rodionov DA and Peterson SN: Short-Chain fatty acids modulate healthy gut microbiota composition and functional potential. Curr Microbiol. 79:1282022. View Article : Google Scholar : PubMed/NCBI

99 

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

100 

Roberti MP, Yonekura S, Duong CPM, Picard M, Ferrere G, Tidjani Alou M, Rauber C, Iebba V, Lehmann CHK, Amon L, et al: Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat Med. 26:919–931. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Yang Q, Zhang J and Zhu Y: Potential roles of the gut microbiota in pancreatic carcinogenesis and therapeutics. Front Cell Infect Microbiol. 12:8720192022. View Article : Google Scholar : PubMed/NCBI

102 

Wang Z, Qin X, Hu D, Huang J, Guo E, Xiao R, Li W, Sun C and Chen G: Akkermansia supplementation reverses the tumor-promoting effect of the fecal microbiota transplantation in ovarian cancer. Cell Rep. 41:1118902022. View Article : Google Scholar : PubMed/NCBI

103 

Sevcikova A, Izoldova N, Stevurkova V, Kasperova B, Chovanec M, Ciernikova S and Mego M: The impact of the microbiome on resistance to cancer treatment with chemotherapeutic agents and immunotherapy. Int J Mol Sci. 23:4882022. View Article : Google Scholar : PubMed/NCBI

104 

Liu J, Liu C and Yue J: Radiotherapy and the gut microbiome: Facts and fiction. Radiat Oncol. 16:92021. View Article : Google Scholar : PubMed/NCBI

105 

Qiu Q, Lin Y, Ma Y, Li X, Liang J, Chen Z, Liu K, Huang Y, Luo H, Huang R and Luo L: Exploring the emerging role of the gut microbiota and tumor microenvironment in cancer immunotherapy. Front Immunol. 11:6122022020. View Article : Google Scholar : PubMed/NCBI

106 

Wang D, Hao H, Li X and Wang Z: The effect of intestinal flora on immune checkpoint inhibitors in tumor treatment: A narrative review. Ann Transl Med. 8:10972020. View Article : Google Scholar : PubMed/NCBI

107 

Smith M, Dai A, Ghilardi G, Amelsberg KV, Devlin SM, Pajarillo R, Slingerland JB, Beghi S, Herrera PS, Giardina P, et al: Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med. 28:713–723. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Ma J, Huang L, Hu D, Zeng S, Han Y and Shen H: The role of the tumor microbe microenvironment in the tumor immune microenvironment: Bystander, activator, or inhibitor? J Exp Clin Cancer Res. 40:3272021. View Article : Google Scholar : PubMed/NCBI

109 

Sethi V, Vitiello GA, Saxena D, Miller G and Dudeja V: The role of the microbiome in immunologic development and its implication for pancreatic cancer immunotherapy. Gastroenterology. 156:2097–2115.e2. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Wei D, Wang L, Zuo X and Bresalier RS: Vitamin D: Promises on the Horizon and Challenges Ahead for Fighting Pancreatic Cancer. Cancers (Basel). 13:27162021. View Article : Google Scholar : PubMed/NCBI

111 

Xiong J, He J, Zhu J, Pan J, Liao W, Ye H, Wang H, Song Y, Du Y, Cui B, et al: Lactylation-driven METTL3-mediated RNA m6A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell. 82:1660–1677.e10. 2022. View Article : Google Scholar : PubMed/NCBI

112 

Kumar H, Lund R, Laiho A, Lundelin K, Ley RE, Isolauri E and Salminen S: Gut microbiota as an epigenetic regulator: Pilot study based on whole-genome methylation analysis. mBio. 5:e02113–14. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Wang L, Zhang S, Li H and Xu Y, Wu Q, Shen J, Li T and Xu Y: Quantification of m6A RNA methylation modulators pattern was a potential biomarker for prognosis and associated with tumor immune microenvironment of pancreatic adenocarcinoma. BMC Cancer. 21:8762021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2023
Volume 50 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xinyuan T, Lei Y, Jianping S, Rongwei Z, Ruiwen S, Ye Z, Jing Z, Chunfang T, Hongwei C, Haibin G, Haibin G, et al: Advances in the role of gut microbiota in the regulation of the tumor microenvironment (Review). Oncol Rep 50: 181, 2023
APA
Xinyuan, T., Lei, Y., Jianping, S., Rongwei, Z., Ruiwen, S., Ye, Z. ... Haibin, G. (2023). Advances in the role of gut microbiota in the regulation of the tumor microenvironment (Review). Oncology Reports, 50, 181. https://doi.org/10.3892/or.2023.8618
MLA
Xinyuan, T., Lei, Y., Jianping, S., Rongwei, Z., Ruiwen, S., Ye, Z., Jing, Z., Chunfang, T., Hongwei, C., Haibin, G."Advances in the role of gut microbiota in the regulation of the tumor microenvironment (Review)". Oncology Reports 50.4 (2023): 181.
Chicago
Xinyuan, T., Lei, Y., Jianping, S., Rongwei, Z., Ruiwen, S., Ye, Z., Jing, Z., Chunfang, T., Hongwei, C., Haibin, G."Advances in the role of gut microbiota in the regulation of the tumor microenvironment (Review)". Oncology Reports 50, no. 4 (2023): 181. https://doi.org/10.3892/or.2023.8618