Skip to main content

Influence of the gut microbiota on immune cell interactions and cancer treatment

Abstract

The tumour microenvironment represents a novel frontier in oncological research. Over the past decade, accumulating evidence has underscored the importance of the tumour microenvironment (TME), including tumour cells, stromal cells, immune cells, and various secreted factors, which collectively influence tumour growth, invasion, and responses to therapeutic agents. Immune cells within the TME are now widely acknowledged to play pivotal roles in tumour development and treatment. While some perspectives have posited that immune cells within the TME facilitate tumour progression and confer resistance to therapeutic interventions, contrasting conclusions also exist. Affirmative and negative conclusions appear to be context dependent, and a unified consensus has yet to be reached. The burgeoning body of research on the relationship between the gut microbiota and tumours in recent years has led to a growing understanding. Most studies have indicated that specific components of the gut microbiota, such as unique bacterial communities or specific secretory factors, play diverse roles in regulating immune cells within the TME, thereby influencing the prognosis and outcomes of cancer treatments. A detailed understanding of these factors could provide novel insights into the TME and cancer therapy. In this study, we aimed to synthesise information on the interactions between the gut microbiota and immune cells within the TME, providing an in-depth exploration of the potential guiding implications for future cancer therapies.

Introduction

The tumour microenvironment (TME) is composed primarily of tumour cells, stromal cells, immune cells, endothelial cells, and various secreted factors. Its importance lies in the intricate network formed through communication and interactions between diverse cell types and secreted factors surrounding tumour cells. The heterogeneity of cell types and secreted factors within the TME enables modulation of the efficacy of anticancer drugs, thereby influencing treatment outcomes [1]. Notably, the immune cell population in the TME is crucial. These immune cells have been demonstrated to interact with tumour cells, promote tumour growth and metastasis, and confer immune escape properties [2, 3]. Immunotherapeutic strategies targeting immune cells within the TME are emerging as new possibilities for cancer treatment, leading to the development of a variety of therapeutic approaches [4].

The gut microbiota refers to the microbial communities that parasitise the human intestinal tract, and the compositions of these communities are determined by various factors, such as genetics, disease prevalence, and exposure to antibiotics. Alterations in the compositions of these microbial communities not only contribute to the onset of disease but also elicit systemic and local metabolic and immune responses. As research into this subject has become more sophisticated, the relationship between the gut microbiota and tumours has become increasingly evident, conferring implications for the occurrence, progression, and therapeutic responses of tumours [5]. One study showed that alterations in the gut microbiotic community and its associated metabolites could induce hepatocellular carcinoma in mice [6]. The application of modulations to the gut microbiota in cancer therapy has become a burgeoning area of investigation.

Recent research has indicated that existing cancer immunotherapies targeting immune cells do not uniformly improve patient prognosis and that there is considerable variability in treatment responses among individuals [7]. These findings underscore the importance of addressing how to increase the effectiveness of immunotherapies that target immune cells. Considering the impact of the gut microbiota on the TME, particularly on immune cells, it is plausible to explore whether manipulating the gut microbiota or targeting specific microbial alterations could improve the efficacy of immunotherapy. In this study, we aimed to compile and analyse the interactions between the gut microbiota and tumour-associated immune cells, as well as integrating immunotherapeutic approaches, with the intention of providing insights for future enhancements in immunotherapy.

The gut microbiota impacts tumour progression via its associated metabolites

Gut microbiota and colorectal cancer (CRC)

The intestines serve as the primary habitat for the gut microbiome and play a crucial role in promoting or inhibiting the development of intestinal tumours. The primary mechanism by which it exerts this influence is by affecting the progression of CRC through its metabolic products [8]. Sulfate-reducing bacteria, such as Desulfovibrio, can transform primary bile acids into secondary bile acids, such as lithocholic acid and deoxycholic acid, which are associated with carcinogenicity [6]. The enterotoxin fragilysin, which is secreted by Bacteroides fragilis (B. fragilis), stimulates the expression of inflammatory factors, the growth-related oncogene-α, and the oncogene c-Myc, thereby promoting the progression of cancer in the intestines under chronic inflammatory stimulation [9]. Conversely, evidence has suggested that Streptococcus gallolyticus plays a role in promoting tumour activity in colon cells. When cocultured with S. gallolyticus, colon cells exhibit increased expression of β-catenin, c-Myc, and proliferating cell nuclear antigen, key transcription factors involved in cancer development [10]. The cell surface virulence factor Fusobacterium adhesin A (FadA), expressed by Fusobacterium nucleatum (Fn), interacts with the E-cadherin/β-catenin pathway, resulting in the upregulation of expression of transcription factors, oncogenes, and inflammatory genes [11].

The potential of the gut microbiota to modulate cancer suppression has yet to be fully harnessed. Recent evidence has suggested that bacteria such as Faecalibacterium prausnitzii and Eubacterium rectale can participate in the fermentation process to produce butyric acid (BA), a short-chain fatty acid (SCFA) that has various cancer-preventing effects [12, 13]. First, BA can induce the expression of the cyclin-dependent kinase tumour-suppressor protein inhibitor 1 A (p21) gene, inhibit the activator protein-1 (AP-1) signalling pathway, and increase the phosphorylation of c-Fos and ERK1/2 [14, 15]. Second, BA is utilised by colon cell mitochondria, assisting in the maintenance of cellular homeostasis and promoting the proliferation of colonic epithelial cells [16]. Third, GPR109a, an SCFA receptor expressed on immune cells, primarily activates BA-associated ligands and inhibits inflammatory cytokines, thereby suppressing the inflammatory process [17]. The host immune response combats DNA methylation-mediated GPR109a expression silencing through IFNγ signalling, thereby promoting anticarcinogenic effects [18].

Gut microbiota and gastric cancer (GC)

Within the realm of GC research, Helicobacter pylori (H. pylori) has emerged as the gut microbe with the strongest association with this disease, as recognised by the academic community. The bacterium orchestrates an array of immune and inflammatory responses by secreting virulence factors such as cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA), which disrupt numerous cellular signalling pathways [19]. Patients infected with CagA-positive H. pylori demonstrate elevated levels of inflammatory cytokines (such as IFNγ, TNF-α, IL-1, IL-1β, and IL-6) and activation of signalling pathways, including the ERK/MAPK, PI3K/Akt, NF-κB, Wnt/β-catenin, and STAT3 pathways, increasing their GC risk relative to that of uninfected patients. VacA-positive strains of H. pylori induce autophagy, particularly by targeting mitochondria, and manipulate critical cell growth and differentiation pathways, such as by upregulating MAP kinase and ERK1/2 expression, subsequently stimulating vascular endothelial growth factor activity and engaging the Wnt/β-catenin pathway while inhibiting GSK3 expression through PI3K/Akt signalling [15]. Furthermore, H. pylori promotes CpG island methylation in key genes, such as E-cadherin and tumour suppressor genes, significantly increasing the risk of GC. Additionally, the impact of H. pylori extends to dysbiosis of the intestinal microbiota [20]. Infected individuals present increased abundances of Proteobacteria, Spirochaetes, and Acidobacteria and decreased abundances of Actinobacteria, Bacteroidetes and Firmicutes, which are recognised as risk factors for GC development. The gut microbiota in patients with GC is more diverse than that in healthy individuals. However, the connection between this microbial diversity and GC remains to be fully elucidated, necessitating further research on the functions and mechanisms of these intestinal microbes [21].

In addition to the impact of H. pylori on GC, other members of the gut microbiota also play important roles. The genera Lactobacillus and Streptococcus include lactic acid-producing microbes that theoretically support tumour progression; lactic acid can act as a substrate for tumour growth and angiogenesis [22]. These findings suggest a potential oncogenic role for these beneficial bacteria under certain pathological conditions. Furthermore, several members of the phylum Nitrospirae have been implicated in nitrate and nitrite metabolic pathways. These microbes lead to the production of carcinogenic N-nitroso compounds that contribute to the development of GC [23]. This underscores the complex ecosystem within the gut microbiota, in which various microbial species affect cancer risk through distinct metabolic activities. The broader implications of such microbial interactions with host physiology underscore the necessity for comprehensive research regarding their precise roles and mechanisms in gastric carcinogenesis.

The gut microbiota and other cancers

Gut microbiota metabolites have been implicated in the progression of various cancers, primarily through the production of inflammatory mediators that can lead to oncogenesis-related inflammation or accelerate cancer progression. Microbial metabolites disrupt liver metabolic pathways and immune responses, such as the recognition of lipopolysaccharide (LPS) by Toll-like receptor 4 (TLR4) to activate Kupffer cells and stellate cells, thereby promoting hepatocellular carcinoma (HCC) through inflammatory and oncogenic pathways, whereas H. pylori-associated VacA, CagA, and LPS further contribute to HCC by increasing IL-8 and TGF-β1 levels. However, the role of the intestinal microbiota and TLR4 activity in HCC initiation remains controversial. Deoxycholic acid derived from Clostridium causes DNA damage and induces a senescence-associated secretory phenotype (SASP) in hepatic stellate cells, involving inflammatory cytokines and growth factors and thereby contributing to inflammatory and obesity-associated HCC progression [15]. Pathogenic components of H. pylori, such as ammonia and LPS, along with increased levels of inflammatory cytokines, damage the pancreas by activating NF-κB and AP-1 signalling, leading to dysregulated cellular processes, KRAS gene mutations, and persistent STAT3 activation, together promoting pancreatic carcinogenesis and cancer progression. The presence of Pseudomonas aeruginosa can promote pancreatic cancer cell metastasis via Taste receptor 2 member 38 receptor signalling, whereas the presence of Fusobacterium spp., which are present in some pancreatic cancer tissues, is associated with poor prognosis [24]. The gut microbiota significantly influences the development and progression of various cancers, including CRC, GC, and others, through diverse mechanisms involving metabolic products, inflammatory responses, and interactions with host cellular pathways; both tumour-promoting and tumour-suppressing effects have been observed (Fig. 1; Table 1).

Table 1 Gut microbiota involvement in tumour progression
Fig. 1
figure 1

The intricate interactions between the gut microbiota and cancer development. Deoxycholic acid and secondary BAs, such as fragilysin and FadA, contribute to DNA damage, SASP induction, and the upregulation of expression of oncogenic markers such as c-Myc and β-catenin, promoting carcinogenesis. Butyric acid, while generally exerting anti-inflammatory effects via GPR109a and p21, can also enhance inflammation through the AP-1 pathway. Additionally, bacterial toxins from H. pylori, P. aeruginosa, and Fusobacterium trigger activation of inflammatory pathways (NF-κB, AP-1) and genetic mutations (KRAS), leading to cancer development and metastasis. This depiction highlights the multifaceted role of the microbiota in carcinogenesis through various molecular mechanisms. Abbreviations: SASP: Senescence-associated secretory phenotype; FadA: Fusobacterium adhesin A; PCNA: Proliferating cell nuclear antigen; p21: Cyclin-dependent kinase tumour-suppressor protein inhibitor 1 A; AP-1: Activator protein-1; CagA: Cytotoxin-associated gene A; VacA: Vacuolating cytotoxin A; H. pylori: Helicobacter pylori

The gut microbiota induces alterations in the tumour immune microenvironment responses to tumours

The gut microbiota influences cancer progression by modulating T cell activity

Over the past several decades, numerous studies have shown the clear relevance of the microbiota and the TME through various associations. In particular, T cells are crucial components of the adaptive immune system, and their proper activation and differentiation are essential for tumour immunosurveillance [25]. Fn is a common oral anaerobic gram-negative bacterium found in many tumours, especially in CRC, and it inhibits the function of CD4+ and CD8+ T cells by directly binding T cell immunoreceptor with Ig and ITIM domains (TIGIT), an inhibitory receptor expressed on the majority of human tumour-infiltrating leukocytes (TILs), via the fusobacterial fibroblast activation protein-2 (Fap2) [26]. H. pylori induces the development of GC, mediated mainly by CagA and VacA expression [27]. A recent study revealed that H. pylori infection can shift the immune response during the chronic inflammatory phase by replacing CagA-specific gastric CD8+ T cells with CD4+ T cells and changing the tissue-resident memory phenotype of CagA-specific CD8+ T cells [28]. Moreover, H. pylori can induce the expression of programmed death ligand 1 (PD-L1) in gastric epithelial cells via the sonic hedgehog signalling pathway. These changes may help Hp-infected cells escape immunosurveillance and progress to GC cells [29]. Indeed, human clinical cohort studies have shown that H. pylori seropositivity is associated with decreased survival in non-small cell lung cancer patients receiving anti-programmed cell death protein 1 (anti-PD-1) therapy, largely due to a decreased number and activation status of tumour-specific CD8+ T cells [30].

Recent studies have demonstrated that certain bacteria can suppress cancer cells by promoting T-cell activation, and their absence or downregulation may in turn lead to cancer initiation. The administration of broad-spectrum antibiotics reduces the effectiveness of immune checkpoint inhibitors (ICIs) that target cytotoxic T lymphocyte-associated protein 4 or PD-1/PD-L1 [31,32,33], providing preclinical evidence that the abundance of some gut microorganisms may promote T-cell activation. Moreover, Saccharopolyspora, Pseudoxanthomonas, and Streptomyces have been reported to promote the recruitment and activation of CD8+ T cells, which may contribute to antitumour immune responses [34]. As the gut microbiota can promote the maturation of lymphoid organs and the differentiation of immune cells, a recent study also suggested that the microbiota can increase the formation of tertiary lymphoid structures (TLSs) within the TME, which are positive prognostic markers for many types of solid tumours [35, 36]. In a murine model of CRC, introducing Helicobacter hepaticus (H. hepaticus) resulted in a decreased tumour load by triggering the formation of classic TLSs containing germinal centres. Notably, these conventional TLSs housed both H. hepaticus and follicular helper T cells specific to H. hepaticus. This finding implies that within a tumour, H. hepaticus acts as a central focus for the TLS-mediated antitumour immune response [37].

The gut microbiota influences cancer progression by directing macrophage polarisation

Tumour-associated macrophages (TAMs), comprising resident macrophages and circulating monocytes recruited to the TME, have been recognised as key inflammatory cells in the TME [38]. The correlation of TAMs with the prognosis of cancer patients is believed to stem from the heterogeneity of TAMs in both inter- and intratumoural contexts [39]. To delineate the diverse roles of TAMs under various conditions, they are generally categorised into M1-like and M2-like subtypes [40]. M1-like macrophages are activated to promote type 1 helper T (Th1) cell immune responses by producing type 1 proinflammatory cytokines (such as IL-1β, IL-1α, and IL-6), suppress tumour progression, and inhibit type 2 helper T (Th2) responses. In contrast, M2-like macrophages contribute to extracellular matrix production and anti-inflammatory effects, including producing IL-4 and IL-10, which are involved in Th2 immune responses, wound healing promotion, and Th1 response inhibition [41]. The interactions between the gut microbiota and its related products in the TME could lead to diverse changes in tumour progression and prognosis through their interplay with TAMs.

In CRC patients, gut microbiota imbalance promotes high expression of the secretory protein cathepsin K, leading to tumour cells stimulating M2-like macrophage polarisation to induce CRC invasion and metastasis [42]. Fn is a type of human intestinal flora that has been shown to induce M2-like macrophage polarisation and promote CRC metastasis via the miR-1322/CCL20 axis [43]. Peptostreptococcus anaerobius is an anaerobic bacterium that specifically adheres to the mucosa of CRC patients. Although the underlying mechanism is unknown, analyses of tumour-infiltrating immune cell populations have shown that P. anaerobius can increase the number of immune cells, including TAMs, to promote tumour progression [44]. On the other hand, Firmicutes has been shown to have antitumorigenic effects in response to macrophage depletion [45]. Bifidobacterium adolescentis inhibits colorectal tumorigenesis by recruiting and facilitating the infiltration of decorin+ macrophages via the activation of Toll-like receptor 2 (TLR2) and regulation of both primary human macrophages and M1 macrophages through the TLR2/YAP axis [46].

In GC patients, alterations in immune responses and immune evasion by H. pylori are intricately connected with the presence of TAMs [47]. The interaction between H. pylori and macrophages in the TME predominantly involves the induction of M2-like macrophage polarisation, diminishing antigen presentation capabilities, and modulating macrophage secretion factors, collectively fostering the progression and invasion of GC [48]. Propionibacterium acnes triggers M2 polarisation of macrophages via TLR4/PI3K/Akt signalling to promote the migration of GC cells [49].

Some oral-gut microbiota have been found to promote miR-21 expression and reduce the expression of phosphatase and tensin homologues (well-known tumour suppressors), and their loss can induce M2-like macrophage polarisation leading to immune escape by pancreatic ductal adenocarcinoma (PDAC) [50]. In contrast, the gut microbiota metabolic product trimethylamine N-oxide, which has been shown to increase the secretion of the proinflammatory cytokines IL-6 and IL-12p4, leads to the inhibition of PDAC cells after inducing macrophage polarisation towards an M1-like phenotype [51]. In addition, under the influence of Lactobacillus casei and Lactobacillus reuteri, TLR4 is inhibited, promoting M1-like macrophage polarisation, to alleviate PDAC and regulate gut microbial homeostasis [52].

Tumour cells overexpressing antiphagocytic surface proteins such as CD47 and CD24 (known as “do not eat me” signals) are able to evade macrophages [53]. Agents antagonising these “do not eat me” proteins and inducing interactions between tumour cells and macrophages have shown therapeutic potential in patients with various cancers [54]. Therefore, the role of macrophages in tumour treatment is crucial and should not be overlooked.

The gut microbiota and other immune cells in the TME

In the TME, in addition to the primary roles of T cells and macrophages, dendritic cells (DCs) and natural killer (NK) cells also contribute to tumour progression. This summary provides an overview of the gut microbiota and its interactions within the TME.

Secondary BAs, metabolic products of gut microbiota dysbiosis in intestinal tumours, activate the TGR5 receptor, thereby inhibiting NF-κB activation in DCs via the cAMP‒protein kinase A pathway. This inhibition leads to the secretion of inflammatory factors expressed in T cells, thereby inducing T cell differentiation [55]. The presence of SCFAs impairs the ability of DCs to induce the proliferation of CD4+ and CD8+ T cells, potentially owing to the upregulation of IL-10 expression in DCs. Additionally, SCFA-treated DCs have been reported to promote regulatory T cell function while inhibiting effector T cell responses [56]. These results suggest that gut microbiota dysbiosis can enhance tumour cell immune evasion by affecting DC functionality. In addition, in a tumour model of mice colonised with H. hepaticus, an improved cancer prognosis was observed, along with an increased number of NK cells in the tumour tissue [37]. These findings suggest that NK cells may exert their tumour-killing functions following modulation by the gut microbiota and that the gut microbiota-mediated conversion of primary BAs to secondary BAs can regulate the expression of CXCL16 (C-X-C motif chemokine ligand 16) in liver sinusoidal endothelial cells, thereby promoting the accumulation of NK cells and increasing IFNγ production to exert antitumour effects [57]. Clinical studies have also shown that in tumour patients positive for Enterobacter and Enterobacteriaceae, the immune receptor TIGIT has significantly upregulated expression in NK cells, leading to sustained clinical benefits [58].

The interactions between the gut microbiota and the tumour immune microenvironment significantly influence cancer progression by modulating T cell activity, directing macrophage polarisation, and affecting the functions of other immune cells, thereby shaping the body’s immune responses against tumours and impacting therapeutic outcomes (Fig. 2).

Fig. 2
figure 2

The gut microbiota significantly influences the tumour immune microenvironment by modulating various immune cells. The gut microbiota affects T cell activity, promotes CD8+ T cell recruitment, and can inhibit CD4+ and CD8+ T cells through mechanisms such as the fusobacterial Fap2 protein binding to TIGIT. The gut microbiota also directs macrophage polarisation, with imbalances leading to M2-like polarisation and certain metabolites promoting M1-like polarisation. Additionally, it impacts DCs and NK cells, enhancing antitumour responses by modulating their functions and interactions within the TME. Overall, these interactions shape the body’s immune response to cancer and influence therapeutic outcomes. Abbreviations: CCL20: C-C motif chemokine ligand 20; Fap2: Fibroblast activation protein-2; TLR2: Toll-like receptor 2; YAP: Yes-associated protein 1; CXCL6: C-X-C motif chemokine ligand 6; NK cell: Natural killer cell; Treg cell: Regulatory T cell; TIGIT: T cell immunoreceptor with Ig and ITIM domains

Modification of the gut microbiota as cancer therapy

Given that gut and tumour microbiota have been demonstrated to play key roles in cancer development, they are anticipated to become crucial intervention strategies in cancer therapy along with enhancing current therapeutic approaches through targeted reconstruction. Various strategies, such as faecal microbiota transplantation (FMT), targeted microbial therapies, dietary interventions, and phage-based approaches, are being explored in clinical trials to treat, intercept, and prevent cancer.

Faecal microbial transplantation (FMT)

FMT can result in healthier and more diverse microbiota than can a patient’s own preconditioning microbiota, and it has been shown to be safe and effective for restoring microbiota diversity in recipients [59]. Moreover, recipients of autologous FMT exhibit increased numbers of various types of white blood cells, indicating the potential benefits of FMT [60]. Nonetheless, the potential danger of transmitting pathogens or antibiotic-resistant bacteria to immunosuppressed recipients underscores the importance of preserving a patient’s own microbiota.

FMT represents an initial approach for modulating gut microbiota and has been investigated in clinical trials in combination with immune checkpoint blockade (ICB) [31, 33, 61,62,63]. It has shown promise for reversing resistance to ICB therapy, with two recent clinical trials demonstrating its ability to restore the ICB response in melanoma patients resistant to treatment [64, 65]. Successful colonisation of the recipient gut by the donor microbiota, particularly bacteria such as Ruminococcaceae and Bifidobacteriaceae, has been associated with increased immune infiltration in tumours and enriched therapy-associated serum metabolites [64, 65]. Although the pilot studies were small and single-armed, they demonstrated a 36% overall clinical response, which is higher than that observed with other combined treatments for patients resistant to anti-PD1 therapy, and the treatment did not result in additional severe toxicity [64, 65]. Furthermore, even when administered via a single colonoscopy without antibiotic conditioning, FMT altered the microbiota composition for more than a year [64]. Currently, trials are underway to test similar approaches in CRC patients (NCT04729322 and NCT04130763).

Research is now focused on determining whether an ideal FMT donor for cancer trials is a successful cancer responder or healthy individual. Clinical trials investigating a combination of ICB treatment with FMT in complete responders or healthy donors are underway and have shown promising early results. FMT has also shown efficacy beyond ICB treatment, particularly in managing steroid-refractory gastrointestinal graft-versus-host disease after haematopoietic stem cell transplantation. Although FMT trials face challenges, including donor selection and administration protocols [66, 67], they provide valuable insights for developing more effective microbiome-based strategies for cancer treatment and beyond.

Probiotics and microbial consortia

The earliest form of immunotherapy utilised microbial species as antitumour agents. As some bacterial species show proinflammatory properties or the capacity to infiltrate and thrive in a hypoxic TME, they have been used as antitumour agents [68], such as Lactobacillus spp. and Bifidobacterium spp [69]. , Blautia producta [70], Clostridium scindens [71] and Clostridium spp [72]. , which have been reported to have anti-inflammatory or antitumour properties in both patient and mouse models. In certain instances, these bacteria have been genetically engineered to enhance their anticancer efficacy or to serve as a vehicle for delivering tumour-toxic substances [73].

In contrast to FMT, current efforts to modulate the gut microbiota are focused on transplanting specific microbial species or designer microbial consortia to improve patient response to ICB and other cancer treatments. In mouse models, specific gut bacteria such as Akkermansia muciniphila (A. muciniphila) [31], B. fragilis [74], Bifidobacterium [33], Lactobacillus rhamnosus GG [75], Lacticaseibacillus paracasei [76], or combinations of probiotics [77, 78] have been shown to enhance the effectiveness of ICB. In a small, open-label trial, patients with metastatic renal cell carcinoma (RCC) who received CBM588 (containing C. butyricum) alongside ICB had a greater response rate and increased progression-free survival than did those who received ICB alone, suggesting that the addition of bifidogenic bacterial products can enhance clinical outcomes in RCC patients [79]. Although individual probiotic strains have shown potential, bacterial consortia may be more effective for maintaining an ecological balance within the gut microbiota. For example, the oral administration of four Clostridiales strain combinations (Roseburia intestinalis, Eubacterium hallii, F. prausnitzii, and Anaerostipes caccae) to mice increased the number of activated CD8+ T cells within tumours and effectively treated both chemically induced and transplanted colorectal tumours [80]. Another study revealed that the probiotic combination Prohep lowered Th17 (helper T 17) cell numbers in tumours, thereby slowing HCC progression in mice [81]. Some clinical trials have tested the therapeutic potential of the probiotic combination VSL#3 in patients with nonalcoholic fatty liver disease [82] or cirrhosis [83] and have shown that administration of probiotics alleviates the severity of these conditions, which are closely linked to the development of HCC.

Despite the initial success observed, the feasibility and efficacy of this approach remain understudied. Several trials are currently underway to evaluate the therapeutic potential of microbial consortia or targeted microbial strategies in combination with existing cancer treatments (NCT03686202 and NCT05079503). Nevertheless, such approaches have shown significant efficacy in noncancer conditions such as Clostridium difficile-related colitis and are expected to offer distinct advantages over FMT in long-term efforts to optimise gut microbiota modulation for cancer treatment.

Diet and prebiotic strategies

Apart from the aforementioned direct strategies for modulating the gut microbiota, diet plays a crucial role in regulating microbial composition and function. Changes in the diet quickly modify the composition of the gut microbiota and affect the production of bacterial metabolites derived from food fermentation, leading to significant metabolic and immunological consequences [84]. Researchers have tested various dietary strategies, such as high-fibre diets [85, 86], ketogenic diets [87], caloric restriction [88, 89], intermittent fasting, fasting-mimicking diets [90], and fermented foods [84], in both mice and patients to improve cancer treatment [91]. In some cases, these approaches influence immunity by changing the composition of the gut microbiota. These studies are pivotal, as they offer a viable approach to modulate the function of gut microorganisms, either alongside other microorganism-targeting strategies or in conjunction with alternative cancer treatments.

In a mouse model, a low-fibre diet al.tered the microbiota and decreased the abundance of Bifidobacterium spp [85]. and increased the abundance of A. muciniphila [92], leading to a poor response to anti-PD1 therapy. Germ-free mice did not show differential responses based on fibre intake, indicating that dietary fibre affects anticancer immunity through changes in the microbiota [85]. A high-fibre diet in mice promotes tumour immunity by supporting fibre-fermenting Ruminococcaceae spp., which enhances the activation and infiltration of T cells, including ICOS-expressing CD8+ and CD4+ T cells, into tumours [85]. Patients with melanoma undergoing anti-PD1 therapy have better response and survival rates on a fibre-rich diet of more than 20 g fibre/day, with each additional 5 g of fibre reducing the risk of progression or death by 30%85.

Furthermore, there are significant opportunities to leverage prebiotics in cancer treatment. Prebiotics, such as inulin and pectin, are soluble fibres that are naturally present in many vegetables and fruits. The administration of prebiotics enhances the effectiveness of anti-PD-1 antibodies in various mouse models [93]. Mechanistically, although prebiotics cannot be digested by gastrointestinal enzymes, they can be fermented by bacteria, further modifying the composition of the gut microbiota (such as enriching the abundance of Ruminococcaceae spp. and of individual bacteria such as A. muciniphila) and their metabolites (such as SCFAs and cyclic diadenosine monophosphate (cyclic di-AMP)), reinforcing the mucosal barrier, improving epithelial integrity, and regulating the activity of innate immune cells to induce antitumour immunity [94, 95]. These effects of prebiotics can be harnessed for therapeutic potential [96].

Specific bacterial depletion by targeted antibiotics or other methods

Long-term use of broad-spectrum antibiotics has been linked to significant alterations in the gut microbiota and poorer outcomes according to some studies [97, 98]. The median survival of patients who received antibiotics before or immediately after anti-PD-1 therapy was nearly half that of those who did not [31]. Additionally, patients with advanced cancer who received antibiotics before or after immune ICB therapy experienced lower response rates and shorter overall survival and progression-free survival [97, 99]. In general, the use of antibiotics can lead to significant alterations in the composition of the gut microbiota, potentially exerting conflicting effects on the ICB response. Therefore, carefully selected antibiotic regimens can indirectly exert anticancer effects and reduce complications during cancer treatment by targeting oncogenic or pathogenic microorganisms [100,101,102]. More specific antibiotics that modulate the gut microbiota and those of other niches may prove beneficial for cancer patients and disease management.

For example, dietary heme, a metabolite derived from red meat, can induce cytotoxicity in colonic contents, promoting compensatory hyperproliferation and hyperplasia of the epithelium and thereby increasing the risk of CRC. Antibiotics such as ampicillin, metronidazole, and neomycin can mitigate this risk by strengthening the mucus barrier and epithelial integrity [103]. Vancomycin inhibits the growth of primary and metastatic liver cancer in mice by promoting the migration of NKT cells and increasing the production of IFN-γ in the liver [57], leading to the depletion of gram-positive bacteria, especially C. scindens [99]. A clinical trial (NCT03785210) is being undertaken to investigate whether vancomycin enhances the effectiveness of anti-PD1 therapy in patients with primary liver cancer or metastases.

In recent years, to minimise disruption of the commensal microbiota and ensure effective cancer treatment, novel technologies, such as the CRISPR‒Cas9 system, which is delivered by phages [104, 105] and targets specific bacteria at the microbiome‒cancer interface, are essential. An example of this is the CRISPR‒Cas9 system, which enables precise genetic alterations and could provide targeted therapies without disrupting the microbiota [106]. Notably, the administration of irinotecan-loaded dextran nanoparticles covalently linked to azide-modified phages, which target Fn in tumours, enhances the efficacy of chemotherapy against CRC [107]. These approaches are anticipated to proliferate in the future and will have a considerable impact on the therapeutic landscape as this field progresses.

Modulation of the gut microbiota is a promising strategy for cancer therapy. FMT and administration of probiotics enhance the immune response and therapeutic effectiveness in various cancers, such as melanoma, CRC, and RCC (Fig. 3; Table 2). Dietary interventions and prebiotics improve the gut microbiota composition, boosting antitumour immunity. Targeted antibiotics and CRISPR-Cas9 offer precise bacterial depletion, further enhancing treatment outcomes. These approaches highlight the critical role of the gut microbiota in cancer therapy optimisation.

Fig. 3
figure 3

Strategies to combat gut dysbiosis and their benefits. Dysbiosis, an imbalance in the gut microbiota, can be addressed using probiotics, a specific gut-health diet, FMT, and select antibiotics. These interventions aim to restore microbial diversity, increase immune cell numbers, reverse resistance to immune checkpoint blockade (ICB) in clinical trials, and utilise specific antitumour bacterial species. Additionally, combining probiotics with treatment enhances their effectiveness; dietary changes rapidly modify microbiota composition; and selective antibiotics reduce pathogenic bacteria. Clinical trials are also beng performed to evaluate the combined effectiveness of antibiotics and ICB treatments. Abbreviations FMT: Faecal microbiota transplantation

Table 2 Gut microbiota involvement in cancer therapy

Conclusions and future perspectives

The intricate relationship between the gut microbiota and immune cells within the TME has opened new avenues for cancer therapy. Recent research has demonstrated that the gut microbiota significantly affects tumour progression and therapeutic responses by modulating immune cell functions through its metabolic products. For example, the conversion of primary BAs to secondary BAs can regulate immune cell activity and enhance antitumour responses, whereas dysbiosis can lead to immune evasion by tumour cells [6, 55, 57, 108].

Emerging therapeutic strategies such as FMT, administration of probiotics, dietary interventions, and administration of targeted antibiotics show promise in modulating the gut microbiota to improve cancer treatment outcomes. The aim of these approaches is to restore microbial balance, enhance immune cell functionality, and potentially reverse resistance to immunotherapy. Clinical studies have begun to validate these strategies, highlighting their potential to increase the effectiveness of existing cancer treatments.

Gut microbiota dysbiosis can impact energy metabolism, immune homeostasis, gut defence mechanisms, organic compounds, vitamin production, and abnormal hormone regulation [109, 110]. Dysbiosis is also associated with various cancers. This understanding has led to rapid growth in research exploring the clinical potential of regulating the gut microbiota. Numerous studies have investigated the treatment of various cancers through dietary therapy, prebiotics, probiotics, and even FMT, and significant progress has been made, as discussed above. Given that bacterial abundance is significantly elevated in GC patients, bacterial overgrowth in the stomach could be a potential marker for GC [111]. For example, the potential utility of some specific microbial signatures for the early detection and screening of CRC has been identified, and some specific species, such as A. halli, C. difficile, and Fn, serve as specific markers for excess body weight-related CRC [112, 113]. These findings contribute to enhancing the accuracy of GC diagnosis and treatment.

For the diagnosis and treatment of different patients, monitoring the gut microbiota can improve the accuracy of early disease detection. Owing to the ease of sample collection, patient compliance and participation can be greatly improved, and the role of the gut microbiota can significantly reduce the side effects of treatment. Personalised treatment can be achieved by targeting the specific diversity of the gut microbiota in a patient, not only reducing medical costs but also improving treatment efficacy, making it a highly promising therapeutic approach. Future research should focus more on unlocking the potential of the gut microbiota, which could lead to more effective and personalised approaches for disease treatment.

Technological variations are also key factors contributing to the inconsistent results observed in microbiome-related studies [114]. 16 S rRNA sequencing can be used to map the microbiome but is affected by various factors, such as sample collection, contamination, and analysis methods, making comparisons between studies difficult. It is unclear whether the detected genes are from live microbes, and contamination remains a challenge. Studying the microbiota directly in gastric biopsies could provide clearer insights into their true presence and abundance in the stomach. Emerging technologies and methodologies are expanding our understanding of the interactions between the gut microbiota and the TME. For example, whole-metagenome sequencing could enhance the detection of bacterial species, including rare ones, and this technique uses bioinformatics to identify microbial targets that could distinguish chronic gastritis from GC [115]. Owing to high background noise from host RNA, traditional metatranscriptomic methods struggle with these low-microbial samples. To address this, a new workflow combining Kraken 2/Bracken for taxonomic analysis and HUMAnN 3 for functional analysis was developed and tested in one study. The approach to this study was validated via synthetic samples and human gastric tissues, demonstrating its ability to accurately identify microbial species and functions with minimal false positives. This method could enhance understanding of the microbiome in mucosal tissues and its interactions with the host in both health and disease [116].

Recent advancements also aim to minimise disruption of the commensal microbiota while enhancing the effectiveness of cancer treatment. One such technology is the CRISPR‒Cas9 system, which allows for precise genetic modifications, potentially offering targeted therapeutic approaches without adversely affecting the microbiota [106]. A new approach called “probiotic surface coating” technology has been proposed in recent years. This technique involves the use of single-cell coating technology to create a protective outer layer on probiotics, resulting in “armoured probiotics.” By combining functional materials with probiotics based on key surface elements such as charge, adhesion factors, and inherent antigens, various biological and chemical methods have been employed. This technology aims to increase the survival and bioavailability of probiotics in the gastrointestinal tract or affected areas, reduce their biotoxicity and immune rejection, and precisely regulate their activity or biological behaviour, thereby improving the effectiveness of disease treatment [117]. Another novel method involving base editing of bacteria in the mouse gut has been reported. Editing the β-lactamase gene achieved 93% editing efficiency, and the modified bacteria persisted in the intestine for at least 42 days. This technique, which uses nonreplicating DNA vectors, prevents payload spread. This method was also used to edit therapeutic genes in E. coli and Klebsiella pneumoniae in vitro and to modify pathogenic genes in E. coli [118]. Future research should also focus on developing relevant animal models to demonstrate that these novel methods can achieve beneficial outcomes for patients.

The gut microbiota is a highly diverse and complex ecosystem composed of various microorganisms within the human intestinal tract, which is one of the most extensive interfaces. In recent decades, studies oregarding the gut microbiota have highlighted its essential role in supporting overall health and well-being. However, most of the studies are cross-sectional, scarce, and somewhat controversial; thus, they should be interpreted cautiously, particularly when considering causation. To evaluate potential causal relationships, it would be necessary to study bacterial colonisation and its fluctuations in relation to the development of GC longitudinally across different populations worldwide, including analysis of precancerous lesions, different regions in the stomach, and different types of GC. Recently, one study offered a comprehensive analysis of longitudinal multisite microbiome ecology and host dynamics. By utilising date-matched microbiome and host -omics data, we can not only deepen our understanding of the stability and individuality of microbiomes across different body sites but also propose mechanism-generating hypotheses on host‒microbiome interactions within the context of prediabetes [119]. Such research could illuminate the gradual development of microbial dysbiosis preceding GC. The recent advancements in the collection of longitudinal cohorts to study health effects may help address these more complex health questions. Indeed, investigations of longitudinal alterations in the gut mycobiome have revealed that certain gut fungi could serve as noninvasive biomarkers or potential treatments for liver disease progression, particularly from cirrhosis to HCC [120]. Another study investigated the link between changes in gut microbiota dysbiosis and the development of GC. They analysed bacterial DNA from stomach biopsies in a longitudinal study involving 43 participants over at least 5 years and reported that patients with early gastric neoplasia had higher abundance of certain bacteria, such as Proteobacteria and H. pylori, and lower abundance of others, such as Bacteroidetes. This study identified specific bacterial features and functions that could predict progression to EGN with significant accuracy, suggesting that monitoring the microbiome might help in the early detection of GC [121]. More studies are needed to focus on longitudinal analysis of the gut microbiome in the future.

In conclusion, the gut microbiota represents a critical and modifiable factor in cancer therapy. Continued exploration and understanding of its interactions with the immune system will pave the way for innovative treatments, offering hope for improved prognosis and quality of life for cancer patients.

Data availability

Not applicable.

Abbreviations

TME:

The Tumour Microenvironment

CRC:

Colorectal Cancer

B. fragilis:

Bacteroides fragilis

SASP:

Senescence-Associated Secretory Phenotype

Fn:

Fusobacterium Nucleatum

FadA:

Fusobacterium adhesin A

BA:

Butyric Acid

SCFA:

Short-Chain Fatty Acid

PCNA:

Proliferating Cell Nuclear Antigen

p21:

Cyclin-dependent kinase tumour-suppressor protein inhibitor 1 A

AP-1:

Activator Protein-1

CagA:

Cytotoxin-associated gene A

VacA:

Vacuolating cytotoxin A

H. pylori:

Helicobacter pylori

TLR4:

Toll-Like Receptor 4

LPS:

Lipopolysaccharides

HCC:

Hepatocellular Carcinoma

TIGIT:

T cell Immunoreceptor with Ig and ITIM domains

CCL20:

C-C motif Chemokine Ligand 20

Fap2:

Fibroblast activation protein-2

TLSs:

Tertiary Lymphoid Structures

TLR2:

Toll-Like Receptor 2

YAP:

Yes-Associated Protein 1

CXCL6:

C-X-C motif Chemokine Ligand 6

NK cell:

Natural Killer cell

Treg cell:

Regulatory T cell

FMT:

Faecal microbiota transplantation

A. muciniphila:

Akkermansia muciniphila

Th:

T helper

RCC:

Renal Cell Carcinoma

PDAC:

Pancreatic Ductal Adenocarcinoma

E. coli:

Escherichia coli

References

  1. Hanahan D. Hallmarks of Cancer: New dimensions. Cancer Discov. 2022;12:31–46. https://doi.org/10.1158/2159-8290.CD-21-1059.

    Article  PubMed  Google Scholar 

  2. Wu T, Dai Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017;387:61–8. https://doi.org/10.1016/j.canlet.2016.01.043.

    Article  PubMed  Google Scholar 

  3. Lv B, et al. Immunotherapy: reshape the Tumor Immune Microenvironment. Front Immunol. 2022;13:844142. https://doi.org/10.3389/fimmu.2022.844142.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Yost KE, et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 2019;25:1251–9. https://doi.org/10.1038/s41591-019-0522-3.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Chen Y, Liu B, Wei Y, Kuang DM. Influence of gut and intratumoral microbiota on the immune microenvironment and anti-cancer therapy. Pharmacol Res. 2021;174:105966. https://doi.org/10.1016/j.phrs.2021.105966.

    Article  PubMed  Google Scholar 

  6. Zhang X, et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut. 2021;70:761–74. https://doi.org/10.1136/gutjnl-2019-319664.

    Article  PubMed  Google Scholar 

  7. Goliwas KF, Deshane JS, Elmets CA, Athar M. Moving Immune Therapy Forward Targeting TME. Physiol Rev. 2021;101:417–25. https://doi.org/10.1152/physrev.00008.2020.

    Article  PubMed  Google Scholar 

  8. Wong CC, Yu J. Gut microbiota in colorectal cancer development and therapy. Nat Rev Clin Oncol. 2023;20:429–52. https://doi.org/10.1038/s41571-023-00766-x.

    Article  PubMed  Google Scholar 

  9. Valguarnera E, Wardenburg JB. Good gone bad: one toxin away from Disease for Bacteroides fragilis. J Mol Biol. 2020;432:765–85. https://doi.org/10.1016/j.jmb.2019.12.003.

    Article  PubMed  Google Scholar 

  10. Kumar R, Herold JL, Taylor J, Xu J, Xu Y. Variations among Streptococcus gallolyticus subsp. gallolyticus strains in connection with colorectal cancer. Sci Rep. 2018;8:1514. https://doi.org/10.1038/s41598-018-19941-7.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Jiang Y, et al. Banxia Xiexin decoction delays colitis-to-cancer transition by inhibiting E-cadherin/beta-catenin pathway via Fusobacterium nucleatum FadA. J Ethnopharmacol. 2024;328:117932. https://doi.org/10.1016/j.jep.2024.117932.

    Article  PubMed  Google Scholar 

  12. Battat R, et al. Increased primary bile acids with Ileocolonic Resection Impact Ileal inflammation and gut microbiota in inflammatory bowel disease. J Crohns Colitis. 2023;17:795–803. https://doi.org/10.1093/ecco-jcc/jjac173.

    Article  PubMed  Google Scholar 

  13. Lv WQ, et al. Human gut microbiome impacts skeletal muscle mass via gut microbial synthesis of the short-chain fatty acid butyrate among healthy menopausal women. J Cachexia Sarcopenia Muscle. 2021;12:1860–70. https://doi.org/10.1002/jcsm.12788.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Pudlo NA, et al. Symbiotic Human Gut Bacteria with variable metabolic priorities for host mucosal glycans. mBio. 2015;6:e01282–01215. https://doi.org/10.1128/mBio.01282-15.

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  Google Scholar 

  16. Gonzalez-Sarrias A, et al. Phase-II metabolism limits the antiproliferative activity of urolithins in human colon cancer cells. Eur J Nutr. 2014;53:853–64. https://doi.org/10.1007/s00394-013-0589-4.

    Article  PubMed  Google Scholar 

  17. Elangovan S, et al. The niacin/butyrate receptor GPR109A suppresses mammary tumorigenesis by inhibiting cell survival. Cancer Res. 2014;74:1166–78. https://doi.org/10.1158/0008-5472.CAN-13-1451.

    Article  PubMed  Google Scholar 

  18. Singh N, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40:128–39. https://doi.org/10.1016/j.immuni.2013.12.007.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Nabavi-Rad A, et al. The double-edged sword of probiotic supplementation on gut microbiota structure in Helicobacter pylori management. Gut Microbes. 2022;14:2108655. https://doi.org/10.1080/19490976.2022.2108655.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Choi J, et al. Helicobacter pylori Eradication modulates aberrant CpG Island Hypermethylation in gastric carcinogenesis. Korean J Gastroenterol. 2016;68:253–9. https://doi.org/10.4166/kjg.2016.68.5.253.

    Article  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  22. Vinasco K, Mitchell HM, Kaakoush NO, Castano-Rodriguez N. Microbial carcinogenesis: lactic acid bacteria in gastric cancer. Biochim Biophys Acta Rev Cancer. 2019;1872:188309. https://doi.org/10.1016/j.bbcan.2019.07.004.

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  24. Zhang X, Liu Q, Liao Q, Zhao Y. Pancreatic Cancer, gut microbiota, and therapeutic efficacy. J Cancer. 2020;11:2749–58. https://doi.org/10.7150/jca.37445.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Bell HN et al. Microenvironmental ammonia enhances T cell exhaustion in colorectal cancer. Cell Metab 35, 134–149 e136 (2023). https://doi.org/10.1016/j.cmet.2022.11.013

  26. Gur C, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42:344–55. https://doi.org/10.1016/j.immuni.2015.01.010.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Ansari S, Yamaoka Y. Helicobacter pylori virulence factor Cytotoxin-Associated Gene A (CagA)-Mediated gastric pathogenicity. Int J Mol Sci. 2020;21. https://doi.org/10.3390/ijms21197430.

  28. Koch MRA, et al. CagA-specific gastric CD8(+) tissue-resident T cells control Helicobacter pylori during the early infection phase. Gastroenterology. 2023;164:550–66. https://doi.org/10.1053/j.gastro.2022.12.016.

    Article  PubMed  Google Scholar 

  29. Holokai L, et al. Increased programmed death-ligand 1 is an early epithelial cell response to Helicobacter pylori Infection. PLoS Pathog. 2019;15:e1007468. https://doi.org/10.1371/journal.ppat.1007468.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Oster P, et al. Helicobacter pylori infection has a detrimental impact on the efficacy of cancer immunotherapies. Gut. 2022;71:457–66. https://doi.org/10.1136/gutjnl-2020-323392.

    Article  PubMed  Google Scholar 

  31. Routy B, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–7. https://doi.org/10.1126/science.aan3706.

    Article  PubMed  Google Scholar 

  32. Gopalakrishnan V, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103. https://doi.org/10.1126/science.aan4236.

    Article  PubMed  Google Scholar 

  33. Matson V, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–8. https://doi.org/10.1126/science.aao3290.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Riquelme E et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 178, 795–806 e712 (2019). https://doi.org/10.1016/j.cell.2019.07.008

  35. Cabrita R, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–5. https://doi.org/10.1038/s41586-019-1914-8.

    Article  PubMed  Google Scholar 

  36. Mlecnik B, et al. Integrative analyses of Colorectal Cancer Show Immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity. 2016;44:698–711. https://doi.org/10.1016/j.immuni.2016.02.025.

    Article  PubMed  Google Scholar 

  37. Overacre-Delgoffe AE et al. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity 54, 2812–2824 e2814 (2021). https://doi.org/10.1016/j.immuni.2021.11.003

  38. Murray PJ, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41:14–20. https://doi.org/10.1016/j.immuni.2014.06.008.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wu K, et al. Redefining Tumor-Associated macrophage subpopulations and functions in the Tumor Microenvironment. Front Immunol. 2020;11:1731. https://doi.org/10.3389/fimmu.2020.01731.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Gambardella V, et al. The role of tumor-associated macrophages in gastric cancer development and their potential as a therapeutic target. Cancer Treat Rev. 2020;86:102015. https://doi.org/10.1016/j.ctrv.2020.102015.

    Article  PubMed  Google Scholar 

  41. Wynn TA. Type 2 cytokines: mechanisms and therapeutic strategies. Nat Rev Immunol. 2015;15:271–82. https://doi.org/10.1038/nri3831.

    Article  PubMed  Google Scholar 

  42. Li R, et al. Gut microbiota-stimulated cathepsin K secretion mediates TLR4-dependent M2 macrophage polarization and promotes tumor metastasis in colorectal cancer. Cell Death Differ. 2019;26:2447–63. https://doi.org/10.1038/s41418-019-0312-y.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Xu C, et al. Fusobacterium nucleatum promotes colorectal cancer metastasis through miR-1322/CCL20 axis and M2 polarization. Gut Microbes. 2021;13:1980347. https://doi.org/10.1080/19490976.2021.1980347.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Long X, et al. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat Microbiol. 2019;4:2319–30. https://doi.org/10.1038/s41564-019-0541-3.

    Article  PubMed  Google Scholar 

  45. Bader JE, et al. Macrophage depletion using clodronate liposomes decreases tumorigenesis and alters gut microbiota in the AOM/DSS mouse model of colon cancer. Am J Physiol Gastrointest Liver Physiol. 2018;314:G22–31. https://doi.org/10.1152/ajpgi.00229.2017.

    Article  PubMed  Google Scholar 

  46. Lin Y, et al. Bifidobacterium adolescentis induces decorin(+) macrophages via TLR2 to suppress colorectal carcinogenesis. J Exp Clin Cancer Res. 2023;42:172. https://doi.org/10.1186/s13046-023-02746-6.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Zhang X, Arnold IC, Muller A. Mechanisms of persistence, innate immune activation and immunomodulation by the gastric pathogen Helicobacter pylori. Curr Opin Microbiol. 2020;54:1–10. https://doi.org/10.1016/j.mib.2020.01.003.

    Article  PubMed  Google Scholar 

  48. Deng R, et al. Effects of helicobacter pylori on tumor microenvironment and immunotherapy responses. Front Immunol. 2022;13:923477. https://doi.org/10.3389/fimmu.2022.923477.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Li Q, et al. Propionibacterium acnes overabundance in gastric cancer promote M2 polarization of macrophages via a TLR4/PI3K/Akt signaling. Gastric Cancer. 2021;24:1242–53. https://doi.org/10.1007/s10120-021-01202-8.

    Article  PubMed  Google Scholar 

  50. Li R, Hu Y, Hou S. An exploration of oral-gut pathogens mediating Immune escape of pancreatic Cancer via miR-21/PTEN Axis. Front Microbiol. 2022;13:928846. https://doi.org/10.3389/fmicb.2022.928846.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Mirji G, et al. The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer. Sci Immunol. 2022;7:eabn0704. https://doi.org/10.1126/sciimmunol.abn0704.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Zhu Z, et al. Lactobacillus casei combined with Lactobacillus reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis. BMC Cancer. 2023;23:1044. https://doi.org/10.1186/s12885-023-11557-z.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Barkal AA, et al. CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature. 2019;572:392–6. https://doi.org/10.1038/s41586-019-1456-0.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Advani R, et al. CD47 blockade by Hu5F9-G4 and Rituximab in Non-hodgkin’s lymphoma. N Engl J Med. 2018;379:1711–21. https://doi.org/10.1056/NEJMoa1807315.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Hu J, et al. Gut microbiota-mediated secondary bile acids regulate dendritic cells to attenuate autoimmune uveitis through TGR5 signaling. Cell Rep. 2021;36:109726. https://doi.org/10.1016/j.celrep.2021.109726.

    Article  PubMed  Google Scholar 

  56. Yang W, Cong Y. Gut microbiota-derived metabolites in the regulation of host immune responses and immune-related inflammatory diseases. Cell Mol Immunol. 2021;18:866–77. https://doi.org/10.1038/s41423-021-00661-4.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Ma C, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018;360. https://doi.org/10.1126/science.aan5931.

  58. Tsakmaklis A, et al. TIGIT(+) NK cells in combination with specific gut microbiota features predict response to checkpoint inhibitor therapy in melanoma patients. BMC Cancer. 2023;23:1160. https://doi.org/10.1186/s12885-023-11551-5.

    Article  PubMed  PubMed Central  Google Scholar 

  59. DeFilipp Z, et al. Third-party fecal microbiota transplantation following allo-HCT reconstitutes microbiome diversity. Blood Adv. 2018;2:745–53. https://doi.org/10.1182/bloodadvances.2018017731.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Schluter J, et al. The gut microbiota is associated with immune cell dynamics in humans. Nature. 2020;588:303–7. https://doi.org/10.1038/s41586-020-2971-8.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Shaikh FY, et al. Murine fecal microbiota transfer models selectively colonize human microbes and reveal transcriptional programs associated with response to neoadjuvant checkpoint inhibitors. Cancer Immunol Immunother. 2022;71:2405–20. https://doi.org/10.1007/s00262-022-03169-6.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Goc J, et al. Dysregulation of ILC3s unleashes progression and immunotherapy resistance in colon cancer. Cell. 2021;184:5015–30. https://doi.org/10.1016/j.cell.2021.07.029. e5016.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Huang J, et al. Metagenomic and metabolomic analyses reveal synergistic effects of fecal microbiota transplantation and anti-PD-1 therapy on treating colorectal cancer. Front Immunol. 2022;13:874922. https://doi.org/10.3389/fimmu.2022.874922.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Davar D, et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 2021;371:595–602. https://doi.org/10.1126/science.abf3363.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Baruch EN, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371:602–9. https://doi.org/10.1126/science.abb5920.

    Article  PubMed  Google Scholar 

  66. McQuade JL, Daniel CR, Helmink BA, Wargo JA. Modulating the microbiome to improve therapeutic response in cancer. Lancet Oncol. 2019;20:e77–91. https://doi.org/10.1016/S1470-2045(18)30952-5.

    Article  PubMed  Google Scholar 

  67. Ruff WE, Greiling TM, Kriegel MA. Host-microbiota interactions in immune-mediated diseases. Nat Rev Microbiol. 2020;18:521–38. https://doi.org/10.1038/s41579-020-0367-2.

    Article  PubMed  Google Scholar 

  68. Dzutsev A, et al. Microbes and Cancer. Annu Rev Immunol. 2017;35:199–228. https://doi.org/10.1146/annurev-immunol-051116-052133.

    Article  PubMed  Google Scholar 

  69. Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer. 2017;17:271–85. https://doi.org/10.1038/nrc.2017.13.

    Article  PubMed  Google Scholar 

  70. Kim SG, et al. Microbiota-derived lantibiotic restores resistance against Vancomycin-resistant Enterococcus. Nature. 2019;572:665–9. https://doi.org/10.1038/s41586-019-1501-z.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Buffie CG, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517:205–8. https://doi.org/10.1038/nature13828.

    Article  PubMed  Google Scholar 

  72. Feuerstadt P, et al. SER-109, an oral Microbiome Therapy for recurrent Clostridioides difficile infection. N Engl J Med. 2022;386:220–9. https://doi.org/10.1056/NEJMoa2106516.

    Article  PubMed  Google Scholar 

  73. Griffin ME, et al. Enterococcus peptidoglycan remodeling promotes checkpoint inhibitor cancer immunotherapy. Science. 2021;373:1040–6. https://doi.org/10.1126/science.abc9113.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Vetizou M, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–84. https://doi.org/10.1126/science.aad1329.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Si W, et al. Lactobacillus rhamnosus GG induces cGAS/STING- dependent type I interferon and improves response to immune checkpoint blockade. Gut. 2022;71:521–33. https://doi.org/10.1136/gutjnl-2020-323426.

    Article  PubMed  Google Scholar 

  76. Zhang SL, et al. Lacticaseibacillus Paracasei sh2020 induced antitumor immunity and synergized with anti-programmed cell death 1 to reduce tumor burden in mice. Gut Microbes. 2022;14:2046246. https://doi.org/10.1080/19490976.2022.2046246.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Tanoue T, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565:600–5. https://doi.org/10.1038/s41586-019-0878-z.

    Article  PubMed  Google Scholar 

  78. Li Y, et al. Gut microbiota dependent anti-tumor immunity restricts melanoma growth in Rnf5(-/-) mice. Nat Commun. 2019;10:1492. https://doi.org/10.1038/s41467-019-09525-y.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Ebrahimi H, et al. Cabozantinib and Nivolumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat Med. 2024. https://doi.org/10.1038/s41591-024-03086-4.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Montalban-Arques A et al. Commensal Clostridiales strains mediate effective anti-cancer immune response against solid tumors. Cell Host Microbe 29, 1573–1588 e1577 (2021). https://doi.org/10.1016/j.chom.2021.08.001

  81. Li J, et al. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc Natl Acad Sci U S A. 2016;113:E1306–1315. https://doi.org/10.1073/pnas.1518189113.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Alisi A, et al. Randomised clinical trial: the beneficial effects of VSL#3 in obese children with non-alcoholic steatohepatitis. Aliment Pharmacol Ther. 2014;39:1276–85. https://doi.org/10.1111/apt.12758.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Dhiman RK et al. Probiotic VSL#3 reduces liver disease severity and hospitalization in patients with cirrhosis: a randomized, controlled trial. Gastroenterology 147, 1327–1337 e1323 (2014). https://doi.org/10.1053/j.gastro.2014.08.031

  84. Wastyk HC et al. Gut-microbiota-targeted diets modulate human immune status. Cell 184, 4137–4153 e4114 (2021). https://doi.org/10.1016/j.cell.2021.06.019

  85. Spencer CN, et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science. 2021;374:1632–40. https://doi.org/10.1126/science.aaz7015.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Lam KC et al. Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment. Cell 184, 5338–5356 e5321 (2021). https://doi.org/10.1016/j.cell.2021.09.019

  87. Ferrere G, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6. https://doi.org/10.1172/jci.insight.145207.

  88. Levesque S, et al. A synergistic triad of chemotherapy, immune checkpoint inhibitors, and caloric restriction mimetics eradicates tumors in mice. Oncoimmunology. 2019;8:e1657375. https://doi.org/10.1080/2162402X.2019.1657375.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Pietrocola F, et al. Caloric restriction mimetics enhance Anticancer Immunosurveillance. Cancer Cell. 2016;30:147–60. https://doi.org/10.1016/j.ccell.2016.05.016.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Vernieri C, et al. Fasting-mimicking Diet is safe and reshapes metabolism and Antitumor Immunity in patients with Cancer. Cancer Discov. 2022;12:90–107. https://doi.org/10.1158/2159-8290.CD-21-0030.

    Article  PubMed  Google Scholar 

  91. Buque A, et al. Immunoprophylactic and immunotherapeutic control of hormone receptor-positive breast cancer. Nat Commun. 2020;11:3819. https://doi.org/10.1038/s41467-020-17644-0.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Desai MS et al. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 167, 1339–1353 e1321 (2016). https://doi.org/10.1016/j.cell.2016.10.043

  93. Han K, et al. Generation of systemic antitumour immunity via the in situ modulation of the gut microbiome by an orally administered inulin gel. Nat Biomed Eng. 2021;5:1377–88. https://doi.org/10.1038/s41551-021-00749-2.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Beukema M, Faas MM, de Vos P. The effects of different dietary fiber pectin structures on the gastrointestinal immune barrier: impact via gut microbiota and direct effects on immune cells. Exp Mol Med. 2020;52:1364–76. https://doi.org/10.1038/s12276-020-0449-2.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Le Bastard Q, et al. The effects of inulin on gut microbial composition: a systematic review of evidence from human studies. Eur J Clin Microbiol Infect Dis. 2020;39:403–13. https://doi.org/10.1007/s10096-019-03721-w.

    Article  PubMed  Google Scholar 

  96. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol. 2019;16:605–16. https://doi.org/10.1038/s41575-019-0173-3.

    Article  PubMed  Google Scholar 

  97. Ahmed J, et al. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. Oncoimmunology. 2018;7:e1507670. https://doi.org/10.1080/2162402X.2018.1507670.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Mohiuddin JJ, et al. Association of antibiotic exposure with survival and toxicity in patients with Melanoma receiving immunotherapy. J Natl Cancer Inst. 2021;113:162–70. https://doi.org/10.1093/jnci/djaa057.

    Article  PubMed  Google Scholar 

  99. Zhao S, et al. Antibiotics are associated with attenuated efficacy of anti-PD-1/PD-L1 therapies in Chinese patients with advanced non-small cell lung cancer. Lung Cancer. 2019;130:10–7. https://doi.org/10.1016/j.lungcan.2019.01.017.

    Article  PubMed  Google Scholar 

  100. Secombe KR, et al. Antibiotic treatment targeting gram negative bacteria prevents neratinib-induced diarrhea in rats. Neoplasia. 2022;30:100806. https://doi.org/10.1016/j.neo.2022.100806.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Sheahan BJ, Theriot CM, Cortes JE, Dekaney CM. Prolonged oral antimicrobial administration prevents doxorubicin-induced loss of active intestinal stem cells. Gut Microbes. 2022;14:2018898. https://doi.org/10.1080/19490976.2021.2018898.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Rudd P. In search of the gold standard for compliance measurement. Arch Intern Med. 1979;139:627–8.

    Article  PubMed  Google Scholar 

  103. Ijssennagger N, et al. Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation by opening the mucus barrier in colon. Proc Natl Acad Sci U S A. 2015;112:10038–43. https://doi.org/10.1073/pnas.1507645112.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Bikard D, et al. Exploiting CRISPR-Cas nucleases to produce sequence-specific antimicrobials. Nat Biotechnol. 2014;32:1146–50. https://doi.org/10.1038/nbt.3043.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Citorik RJ, Mimee M, Lu TK. Sequence-specific antimicrobials using efficiently delivered RNA-guided nucleases. Nat Biotechnol. 2014;32:1141–5. https://doi.org/10.1038/nbt.3011.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Sadri M, et al. Potential applications of macrophages in cancer immunotherapy. Biomed Pharmacother. 2024;178:117161. https://doi.org/10.1016/j.biopha.2024.117161.

    Article  PubMed  Google Scholar 

  107. Zheng DW, et al. Phage-guided modulation of the gut microbiota of mouse models of colorectal cancer augments their responses to chemotherapy. Nat Biomed Eng. 2019;3:717–28. https://doi.org/10.1038/s41551-019-0423-2.

    Article  PubMed  Google Scholar 

  108. Zhu X et al. Microbial metabolite butyrate promotes anti-PD-1 antitumor efficacy by modulating T cell receptor signaling of cytotoxic CD8 T cell. Gut Microbes. 2023;15(2):2249143. https://doi.org/10.1080/19490976.2023.2249143.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Agus A, Planchais J, Sokol H. Gut microbiota regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe. 2018;23:716–24. https://doi.org/10.1016/j.chom.2018.05.003.

    Article  PubMed  Google Scholar 

  110. Pham VT, Dold S, Rehman A, Bird JK, Steinert RE. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr Res. 2021;95:35–53. https://doi.org/10.1016/j.nutres.2021.09.001.

    Article  PubMed  Google Scholar 

  111. Chen XH, Wang A, Chu AN, Gong YH, Yuan Y. Mucosa-Associated Microbiota in Gastric Cancer tissues compared with non-cancer tissues. Front Microbiol. 2019;10:1261. https://doi.org/10.3389/fmicb.2019.01261.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Pushalkar S, et al. The pancreatic Cancer Microbiome promotes oncogenesis by induction of Innate and Adaptive Immune suppression. Cancer Discov. 2018;8:403–16. https://doi.org/10.1158/2159-8290.CD-17-1134.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Zhu X, et al. Multi-kingdom microbial signatures in excess body weight colorectal cancer based on global metagenomic analysis. Commun Biol. 2024;7:24. https://doi.org/10.1038/s42003-023-05714-0.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Mirzayi C, et al. Reporting guidelines for human microbiome research: the STORMS checklist. Nat Med. 2021;27:1885–92. https://doi.org/10.1038/s41591-021-01552-x.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Jeong S, et al. Microbiome signatures associated with clinical stages of gastric Cancer: whole metagenome shotgun sequencing study. BMC Microbiol. 2024;24:139. https://doi.org/10.1186/s12866-024-03219-2.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Pereira-Marques J, Ferreira RM, Figueiredo C. A metatranscriptomics strategy for efficient characterization of the microbiome in human tissues with low microbial biomass. Gut Microbes. 2024;16:2323235. https://doi.org/10.1080/19490976.2024.2323235.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Zhao R, et al. Single-cell encapsulation systems for probiotic delivery: Armor probiotics. Adv Colloid Interface Sci. 2024;332:103270. https://doi.org/10.1016/j.cis.2024.103270.

    Article  PubMed  Google Scholar 

  118. Brodel AK, et al. In situ targeted base editing of bacteria in the mouse gut. Nature. 2024;632:877–84. https://doi.org/10.1038/s41586-024-07681-w.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Zhou X et al. Longitudinal profiling of the microbiome at four body sites reveals core stability and individualized dynamics during health and disease. Cell Host Microbe 32, 506–526 e509 (2024). https://doi.org/10.1016/j.chom.2024.02.012

  120. Jiang S, et al. Longitudinal gut fungal alterations and potential fungal biomarkers for the progression of primary liver disease. Sci China Life Sci. 2024;67:1183–98. https://doi.org/10.1007/s11427-023-2458-1.

    Article  PubMed  Google Scholar 

  121. Png CW, et al. Mucosal microbiome associates with progression to gastric cancer. Theranostics. 2022;12:48–58. https://doi.org/10.7150/thno.65302.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

First, we would like to express our sincere gratitude to all the team members who contributed to this study. This research could not have been completed without everyone’s collective effort. Additionally, we extend our gratitude to our colleagues at the First Affiliated Hospital of Guangzhou Medical University and Southern Medical University for their literature review support. Your assistance made this research possible.

Author information

Authors and Affiliations

Authors

Contributions

Information collection and Manuscript Writing: Chunxiao Liu, Lingfeng Fu, Yuxin Wang; Concept and Design: Yuxin Wang, Weijun Yang; Revisions and Proofreading: Weijun Yang; Figures and Tables: Chunxiao Liu, Lingfeng Fu, Yuxin Wang.

Corresponding authors

Correspondence to Yuxin Wang or Weijun Yang.

Ethics declarations

Conflict of interest

The authors have no potential conflicts of interest to disclose.

Additional information

Publisher’s note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, C., Fu, L., Wang, Y. et al. Influence of the gut microbiota on immune cell interactions and cancer treatment. J Transl Med 22, 939 (2024). https://doi.org/10.1186/s12967-024-05709-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05709-3

Keywords