Bacterial metabolites: Effects on the development of breast cancer and therapeutic efficacy (Review)
- Authors:
- Published online on: March 4, 2025 https://doi.org/10.3892/ol.2025.14956
- Article Number: 210
Abstract
Introduction
Breast cancer (BC) stands as one of the most commonly diagnosed malignancies worldwide. Mortality rates persist at a high level, ranking BC as the second leading cause of cancer-related deaths among females, with ~~290,000 fatalities annually globally (1). BC is associated with the disruption of several microbiome compartments, such as the gut microbiome, breast tissue microbiome and tumor microbiome, which has been termed oncobiosis (2). The human body harbors diverse communities of commensal and pathogenic bacteria in both the body cavity and the body surface, with >90% of the microbes residing in the gastrointestinal tract. Each individual has a distinct gut microbial composition, and dysbiosis of the gut microbiota has been demonstrated to contribute to the pathogenesis of several diseases, including BC, pancreatic adenocarcinoma, colorectal cancer (CRC), gastric cancer and hepatocellular carcinoma (3–7). The notable role of oncobiosis in the pathogenesis of BC is highlighted by the discovery that antibiotic usage increases susceptibility of mice to BC (8). Additionally, risk factors associated with BC, such as high breast density (9), early onset of menstruation, low levels of physical activity (10), increased body mass index (11), advanced age (12) and high alcohol consumption (10), have also been associated with alterations in the gut microbiome that contribute to the oncobiosis associated with BC.
Numerous bacterial metabolites are either products of microbial metabolism, such as bacteriocins, or compounds derived from the host that have been modified, such as secondary bile acids (BAs), short-chain fatty acids (SCFAs) and amino acid metabolites (13). These metabolites serve as crucial links in the reciprocal interactions between cancer cells and their surrounding microenvironment. The disruption of gut microbial communities, known as dysbiosis, can produce systemic immune responses due to the breakdown of mucosal barriers and the translocation of gut microbiome components to breast tissue via the bloodstream (Fig. 1). Moreover, microbial metabolites are not solely derived from the gut microbiota. Human breast tissue also harbors symbiotic bacterial communities, and these communities and their metabolic products may influence the occurrence and progression of BC (14). Patients with triple-negative breast cancer (TNBC) have the least complex microbial signature, whereas those with luminal BC subtypes have the most complex microbial signature (15). The microbiota can influence cancer progression through several mechanisms, including modulating inflammation, inducing DNA damage and producing metabolites involved in tumorigenesis or tumor suppression (16). In summary, both gut-derived and breast tissue-derived microbiota and metabolites are considered to influence the development, progression and metastasis of BC, and its response to treatment.
In particular, previous studies (17–20) have reported that the efficacy of standard chemotherapeutic, targeted and immunotherapy drugs is influenced by gut microbial metabolites, and these metabolites may be implemented as combination therapies for BC. The impact of different taxa on the immune checkpoint inhibitor (ICI) response is mediated primarily through the release of various bacterial metabolites (21); however, there has been limited research on how these distinct metabolites affect the response to ICIs. The interaction between these metabolites and the host immune system is crucial for understanding how the microbiome can affect ICI efficacy. This also implies that certain metabolites could be used to predict the response to ICIs (22). Although evidence supports various gut metabolites having important effects on BC and its drug treatments, the underlying mechanisms are poorly understood and warrant further investigation.
Research has shown that the gut microbiota composition can impact the effectiveness of and adverse reactions to BC immunotherapy (23,24). However, little is known about the importance of the interactions between microbiota-induced metabolites and drug metabolites. In the present review, the mechanisms and therapeutic potential of several microbial metabolites in BC are reported and discussed.
Effects of microbial metabolites on BC development
Secondary BAs
BAs are synthesized from cholesterol in the liver and released into the small intestine. BAs interfere with glucose, fatty acid and lipid metabolism mainly by activating the farnesoid X receptor and the G protein-coupled bile acid receptor (TGR5), which are crucial for maintaining a healthy gut microbiota, regulating lipid and carbohydrate metabolism, enhancing insulin sensitivity and supporting innate immunity (25). Bacteria ultimately mediate the conversion of BAs into secondary BAs, including deoxycholic acid (DCA) and lithocholic acid (LCA) (26). A small population of intestinal species in the genus Clostridium, including Clostridium scindens, Clostridium hiranonis, Clostridium hylemonae and Clostridium sordelli, are capable of producing secondary bile acids (26). Tang et al (27) and Zhu et al (28) demonstrated that BAs and their metabolites can enter breast tumors through BA transporters, reduce BC aggressiveness and improve BC prognosis. The studies also showed that BAs may interfere with hormonal pathways in the breast tissue. The presence of secondary BAs can promote carcinogenesis through several mechanisms, such as inducing DNA damage, activating β-catenin signaling and increasing cyclooxygenase-2 activity (29). Secondary BAs can also influence tumor development and progression through their immunosuppressive effects (30), and have been shown to have anticancer effects and reduce cancer risk in clinical studies (31).
The contradictory effects of secondary BAs on carcinogenesis are context-dependent and may arise from the following factors. Low concentrations of secondary BAs may exert anticancer effects by activating FXR/TGR5 receptors (25,27,28). Conversely, chronic or high-dose exposure to secondary BAs can promote carcinogenesis (29). The balance between pro- and anti-carcinogenic BAs also depends on microbial composition. Specific Clostridium species (e.g., C. scindens) convert primary BAs to secondary BAs (26). Finaly, receptor signaling dynamics also contribute to the contradictory actions. For example, TGR5 activation in intestinal L cells stimulates GLP-1 secretion, improving insulin sensitivity (anticancer), but in macrophages, chronic TGR5 signaling may drive immunosuppressive M2 polarization (pro-cancer) (25,30).
DCA is formed by the de-hydroxylation of BAs and previous studies have shown that DCA increases the number of metastases generated from 4T1 cell tumors grafted into mouse fat pads by elevating the expression of vascular endothelial growth factor receptor 2 (Flk-1) and reducing the ceramide-mediated apoptosis of BC cells (32). In a previous in vitro study, physiological levels of DCA promoted cell proliferation by inducing AKT phosphorylation and cyclin D1 expression in MCF-7 BC cells, and was cytotoxic at supraphysiological concentrations; the mechanism of DCA involved the induction of apoptosis (33). Cong et al (34) screened a library of gut microbiota-derived metabolites and identified DCA as a negative regulator for CD8+ T cell effector function. The study demonstrated a causal relationship between microbial DCA metabolism and impaired antitumor CD8+ T cell responses in CRC, highlighting the immunosuppressive role of DCA in tumor progression. The dual roles of DCA have important implications for their therapeutic targeting. On one hand, strategies to inhibit the production or activity of DCA could restore antitumor immunity and reduce tumor progression, as suggested by Cong et al (34). On the other hand, modulating BA receptor signaling (e.g., FXR/TGR5 agonists) may offer additional therapeutic avenues (25,27,28). Furthermore, microbiome-directed interventions, such as probiotics or dietary modifications, could reshape the BA pool to favor antitumor effects. However, the precise balance between pro- and anticancer BAs must be carefully considered, as their effects are highly context-dependent.
Clostridium in the large intestine is mainly responsible for the production of LCA (2). In a previous study, patients with early stage BC had reduced serum levels of LCA, which may be associated with a decline in the biosynthetic capacity of LCA within the microbial community (35). Furthermore, serum LCA levels are negatively associated with the Ki67 labelling index in BC cells (36). Therefore, promoting the production of these protective metabolites by the gut microbiota in women with BC may improve their health (37). Moreover, LCA can reduce BC cell proliferation by 10–20%, inhibit tumor infiltration and metastasis, reduce epithelial-mesenchymal transition (EMT) and enhance antitumor immune responses (35). Another study revealed a potential therapeutic role of LCA in BC cells due to its reversal of lipid metabolism dysregulation (38); LCA induced TGR5 expression in MCF-7 and MDA-MB-231 BC cells, and had antiproliferative and proapoptotic effects. Moreover, the expression of the proapoptotic p53 protein increased and the expression of the antiapoptotic protein Bcl-2 decreased in MCF-7 cells after LCA treatment. Furthermore, LCA supplementation reduced vascular endothelial growth factor (VEGF) production by BC cells in an animal model of BC (2). In conclusion, LCA may have a therapeutic effect on BC cells and can be used as an antiproliferative and proapoptotic drug that targets lipid metabolism in these cells (38).
SCFAs
Gut microbes influence cancer growth through the production of SCFAs, such as acetate, butyrate and propionate (39). SCFAs can directly activate G protein-coupled receptors (GPCR) and inhibit histone deacetylases (HDACs) (40), resulting in cell attachment and differentiation, immune cell migration, cytokine production, chemotaxis and programmed cell death (41,42). Although the suppressive function of SCFAs in the development of CRC has been demonstrated (43), SCFAs can also affect various organs through the systemic circulation (29). However, their role in BC needs further study.
Among the SCFAs, propionic acid (SP), which can be produced from succinic acid, inhibits tumor growth and EMT, induces apoptosis in BC cells by binding to the GPCRs, GPR43 and GPR41, and inhibits BC cell proliferation and apoptosis in a dose-dependent manner (30). SP also suppresses the invasion of BC cells overexpressing GPR41 and GPR43 by activating large tumor suppressor kinase 1 and inhibiting extracellular signal-regulated kinase 1/2 (44). A previous study demonstrated the anticancer effects of SP on BC cell growth, programmed cell death, self-digestion processes and the production of substances that prevent oxidative damage (45). SP was shown to inhibit BC cell proliferation and induce apoptosis by inhibiting STAT3, increasing reactive oxygen species (ROS) levels and activating p38; however, these effects were not mediated by the SCFA receptors GPR41 or GPR43. Furthermore, SP suppressed the proliferation of BC cells by inhibiting the expression or activity of cell cycle checkpoint proteins, causing G0/G1 phase arrest and inhibiting DNA synthesis (45). Collectively, these results suggest that SP is a candidate therapeutic drug for BC.
The intestinal concentration of butyrate, the major protective SCFA, serves a crucial role in cell cycle regulation, cell proliferation and apoptosis (29). Owing to its anti-inflammatory properties and ability to induce cell differentiation, trigger cancer cell apoptosis and promote protective histone hyperacetylation, butyrate has shown strong antitumor effects in BC cell lines, including MCF-7, MDA-MB-231 and MDA-MB-453 (42,46), revealing its potential as an anticancer metabolite. Therefore, promoting butyrate production may have beneficial implications for patients with BC or for those at risk of developing BC (37). Previous studies evaluated the effects of butyrate on the proliferation and ultrastructure of BC cells (47,48). The study confirmed that the administration of butyrate resulted in morphological changes to the ultrastructure of MCF-7 cells, suppressed cell proliferation and triggered apoptotic cell death; however, the underlying mechanism of action remains unknown (47,49). Moreover, butyrate can induce calcium influx into MCF-7 cells (42), which activates caspases and other pro-apoptotic pathways, leading to cell death.
Compared with healthy premenopausal women, premenopausal patients with BC were determined to have markedly lower levels of SCFA-producing bacteria in the gut, as well as lower levels of key SCFA-producing enzymes; therefore, these SCFA receptors may be new targets for the treatment of premenopausal patients with BC (50). Overall, SCFAs are promising for BC treatment. Future studies should evaluate the effects of SCFAs on BC to understand their molecular mechanisms of action, as well as how SCFAs affect the efficacy and safety of standard BC drug therapies and BC prognosis.
Amino acid metabolites
Similar to SCFAs and LCA, certain amino acid metabolites play important roles in BC. Several bacterial taxa, such as Clostridium, Bacillus, Lactobacillus, Streptococcus and Proteobacteria species, efficiently metabolize proteins (51). Tryptophan catabolism is inhibited in BC cells and reduced tryptophan catabolism is associated with decreased survival in patients with BC, whereas elevated levels of extracellular tryptophan are linked to worse BC prognosis (52). Tryptophan metabolites may stimulate the immune system by interacting with the aryl hydrocarbon receptor (AHR) to impact human health and disease (52,53).
The microbiome contributes to 4–6% of the overall tryptophan catabolism and generates indole derivatives, including indole propionic acid (IPA) and indoxyl sulfate (IS), both of which have cytostatic effects on BC (54,55). Similar to other indole derivatives, IPA and IS act through the pregnane-X receptor and AHR. The expression of these receptors decreases as BC progresses, and in patients with a poor prognosis, the expression of these receptors is lower. In addition, indole derivatives have strong immunostimulatory effects, and can stimulate an antitumor immune response in BC and alter the microbiome composition (54,55).
The enzyme lysine decarboxylase catalyzes the decarboxylation of lysine to produce cadaverine (CAD), which is also produced by numerous bacterial species (e.g. E. coli, Vibrio sp. and Lactobacillus sp.) in the human microbiome (56). Reduced CAD biosynthesis in the gut has been reported in patients with early BC, leading to reduced production of anticancer bacterial metabolites and resulting in increased BC invasion (39). Kovács et al (56) reported that CAD inhibited BC metastasis and reduced the aggressiveness of the primary tumor. Moreover, CAD amine treatment of BC cell lines reversed EMT, inhibited cell motility and invasion, and rendered cells less stem cell-like by reducing mitochondrial oxidation over the range of serum concentrations tested (100–800 nM). CAD is thus expected to contribute to the development of tumor therapies; however, the mechanism of CAD in BC requires further investigation.
Lipopolysaccharide (LPS)
Metabolites such as LPS, glycans and endotoxins are present in the outer membranes of gram-negative bacteria. The inherent microbiome of the breast and the gut is enriched in gram-negative bacteria. Bacterial LPS was found in CD45+/CD68− cells of a highly inflamed breast tumor, indicating that the colonizing bacteria tune and activate the immune system (57). LPS is closely linked to human immunity. Toll-like receptors (TLRs), which respond to LPS stimulation, have gained significant attention in cancer research due to their role in tumor progression (58). TLRs serve essential roles in the initiation of inflammation and the activation of host innate immune responses against invading microorganisms through their recognition of pathogen-associated molecular patterns (PAMPs). LPS is a type of PAMP that has been reported to enhance the invasiveness of BC cells (2) and stimulate BC cell proliferation (59). The metastatic potential of BC cells is enhanced upon TLR4 and NF-κB activation, making these proteins promising therapeutic targets for metastasis prevention (60). In a previous study, the LPS-mediated activation of TLR4 markedly increased the mRNA expression of matrix metalloproteinases, MMP-2 and MMP-9, and VEGF. The subsequent triggering of the TLR4 downstream protein, myeloid differentiation factor 88, resulting in increased production of interleukins, IL-6 and IL-10, in human BC cells. TLR4 was demonstrated to be overexpressed in human BC tissues and has been associated with lymph node metastasis (61). A study by Li et al (59) revealed that in MCF-7 and MDA-MB-231 BC cells, LPS-stimulated TLR4 pathway activation triggered β-catenin signaling via PI3K/Akt/GSK3β to promote downstream β-catenin target gene transcription during BC metastasis. These findings suggest that TLR4 may be involved in the progression and metastasis of human BC and may be a new therapeutic target.
Nisin
Nisin, the most abundantly produced bacteriocin in the gut, also has important effects on BC and has been demonstrated to be capable of modulating the innate immune system by inducing chemokine secretion and inhibiting LPS-stimulated cytokines in vitro and in vivo (62). The precise mechanism of the anticancer effect of nisin remains unclear. However, it is hypothesized that nisin induces apoptotic cell death by initiating cell cycle arrest and altering intracellular ion levels (e.g., calcium) through membrane disruption and pore formation. This alters the transmembrane potential (42,62) and increases the permeability of the phospholipid bilayer (63).
Previous studies have demonstrated the antitumor potential of nisin in head and neck squamous cell carcinoma in vitro and in vivo (64). In addition, Ahmadi et al (65) demonstrated that nisin induced the intrinsic apoptosis pathway in colon cancer cells. While there is less research on nisin in BC, the in vitro experiments of Avand et al (62) reported for the first time the potent toxicity of nisin to MCF-7 BC cells. Compared with that in normal human umbilical vein endothelial cells (HUVECs), nisin exhibited selective toxicity to cancer cells (62). Overall, nisin is expected to be an anticancer agent; however, its precise anticancer mechanism needs further study.
Pyocyanin
Pyocyanin is a unique extracellular secondary metabolite pigment produced by Pseudomonas aeruginosa that exhibits redox activity and is toxic to mammalian cells (66). Previous research has demonstrated the apoptotic effects of pyocyanin on cancer cell lines, including rhabdomyosarcoma, hepatocellular carcinoma (HepG2) and human pancreatic cancer cell lines (67,68). Abdelaziz et al (69) were the first to report the toxic effects of pyocyanin to MCF-7 human breast adenocarcinoma cells. The study demonstrates that purified pyocyanin may be used in treatment strategies of human breast adenocarcinoma (MCF-7), which results in decreasing the viability of cells by the induction of necrosis and accelerating apoptosis via caspase-3 activation. Pyocyanin has a low molecular weight (210 Da) and exists as a zwitterion that readily diffuses across cell membranes under aerobic conditions, increasing the intracellular ROS levels (66). Excessive ROS leads to caspase-3 activation, which promotes cancer cell death via apoptosis. Therefore, pyocyanin is considered to have potential to be a new and effective alternative treatment for cancer.
Effects of microbial metabolites on BC therapy
There is growing evidence that the gut microbiota influences the effectiveness of cancer therapies and their associated side effects. In the present section, the roles and underlying mechanisms of gut microbial metabolites on the efficacy and side effects of several anticancer treatments are summarized. Understanding these interactions may lead to the development of effective adjuvant therapies to improve the efficacy of anticancer treatments.
SCFAs
Although butyrate has shown notable anticancer effects as a stand-alone therapy, the synergistic effects of this compound combined with conventional anticancer medications have been documented in previous years (70–72). By inhibiting HDACs, butyrate has the potential to enhance the clinical efficacy and mitigate the toxicity associated with standard chemotherapeutic agents (42). A previous study evaluated the antitumor potential of Herceptin in combination with butyrate against HER2-overexpressing BC cells (73). The combination of butyrate and Herceptin markedly increased the growth-inhibitory effect on SKBR3 BC cells compared with the effects of butyrate and Herceptin alone (73). The potential synergistic effect of butyrate with vitamin A on MCF-7 BC cells was investigated in another in vitro study, which revealed that vitamin A enhanced the anti-proliferative effects of butyrate. Cell proliferation inhibition was 34, 10 and 46% following treatment with butyrate, vitamin A and their combination, respectively, suggesting that vitamin A potentiated the inhibitory activities of butyrate (74). Thus, the available evidence suggests that butyrate may offer notable benefits as an adjuvant therapy alongside standard anticancer drugs and has potential far-reaching clinical implications for the management of BC (42).
A clinical cohort study was performed to assess the fecal and plasma concentrations of SCFAs in patients with primary cancer undergoing treatment with the ICIs nivolumab or pembrolizumab. The results indicated that there may be an association between fecal SCFA levels and ICI efficacy. High concentrations of fecal or plasma SCFAs were associated with a response to ICI treatment and longer progression-free survival, suggesting that SCFAs could act as mediators between the gut microbiome and immunotherapy (75). Another study indicated that gut microbiota diversity was notably associated with the response to ICIs therapy in patients with non-small cell lung cancer. Responders (Rs) presented with a marked increase in the abundance of Faecalibacterium in their gut microbiota, along with increased levels of SCFAs, particularly butyrate, acetate and hexanoate. Furthermore, fecal microbiota transplantation from Rs to non-responders enhanced the anticancer effects of ICIs in mice and reduced Ki-67 expression in tumor cells (76). These studies provide evidence regarding how SCFAs affect ICIs therapy efficacy. SCFAs may modulate the response to ICIs by affecting the functions of immune cells and the tumor microenvironment (77). High blood SCFAs levels are associated with resistance to CTLA-4 blockade and a higher proportion of Treg cells (78). SCFAs, particularly butyrate, enhance the differentiation and function of cytotoxic CD8+ T cells by inhibiting HDACs, leading to increased expression of effector molecules such as IFN-γ and granzyme B (42). This promotes antitumor immunity and synergizes with ICIs. The specific mechanism of action needs further investigation.
Trimethylamine N-oxide (TMAO)
TMAO, a gut microbiota metabolite, is derived from phosphatidylcholine, choline, betaine, and L-carnitine. In previous years, TMAO has received increasing attention due to its possible carcinogenic effects (79,80). However, a recent study (81) revealed that TMAO triggers endoplasmic reticulum stress via eukaryotic translation initiation factor 2α kinase 3, which activates caspase 3 and gasdermin E, leading to pyroptosis in tumor cells and enhancing CD8+ T-cell-mediated immunity against TNBC. In addition, high plasma TMAO levels are associated with improved immunotherapy outcomes in patients with advanced TNBC, indicating that TMAO is a potential biomarker for the response to immunotherapy (81). In mice, TMAO combined with anti-programmed cell death protein 1 antibodies inhibited tumor growth to a markedly greater extent than antibody treatment alone, suggesting that TMAO can increase the efficacy of immunotherapy (81). Therefore, as a driver of antitumor immunity, TMAO may enhance the antitumor immune response in BC. However, the effect of TMAO on BC therapy needs further investigation.
Zhou et al (79) demonstrated that TMAO promotes the proliferation and migration of hepatocellular carcinoma cells through the MAPK pathway, which is involved in regulating cell proliferation, differentiation and apoptosis. Given the role of the MAPK pathway in EMT, it is possible that TMAO may also influence EMT in hepatocellular carcinoma cells, but further research is needed to confirm this. Another study revealed that TMAO serves a carcinogenic role in CRC by promoting cell proliferation and angiogenesis (82). Although some studies have reported its carcinogenic effects, a growing body of research has recently focused on its role as a driver of antitumor immunity. TMAO is a significant inducer of inflammatory effects, reconfiguring tumor immune infiltrates to an immune-activated phenotype. Jalandra et al (80) also discussed the possible anticancer mechanisms of TMAO, which include inflammation, oxidative stress, DNA damage and protein misfolding. These studies provide evidence of the potential role of TMAO in immunotherapy efficacy.
The dual role of TMAO in cancer development suggests that its effects are context-dependent and may vary depending on the type of cancer and the specific microenvironment. Therefore, the use of TMAO as a BC treatment option requires careful consideration of its potential pro- and anticancer effects. Further research is needed to fully understand the mechanisms underlying these dual effects and to determine the most effective and safe ways to utilize TMAO in BC treatment.
Pyocyanin
Pyocyanin, a toxin produced and secreted by P. aeruginosa, can induce cancer cell apoptosis and suppress lymphocyte function. Pyocyanin is considered to promote cancer cell death (83) and enhance tumor proliferation inhibition by doxorubicin (84). Pyocyanin has redox activity and is toxic to mammalian cells, making it a potential new anticancer drug. A previous study reported that purified pyocyanin has marked toxicity to MCF-7 BC cells by inhibiting cell growth via the induction of necrosis and accelerating apoptosis induced by caspase-3 activation (69). When used in combination, pyocyanin can enhance the cytotoxic effects of doxorubicin chemotherapy, especially in MDA-MB-231 and MCF-7 BC cells. Compared with the use of doxorubicin alone, the addition of 10% P. aeruginosa culture supernatant markedly increased doxorubicin-induced caspase-7 protein cleavage, indicating that pyocyanin enhances chemotherapy-induced apoptosis (85).
Additionally, since pyocyanin is a potent inducer of ROS, it is reasonable to hypothesize that pyocyanin enhances chemotherapy efficacy via a ROS-dependent mechanism. Compared with normal cells, cancer cells are more sensitive to increases in the ROS content, as cancer cells have a higher baseline level of ROS (86). The aforementioned studies have demonstrated that pyocyanin may be a promising anticancer drug candidate.
Nisin
Nisin has been reported to have specific anticancer activity against MCF-7 breast adenocarcinoma cells and potential synergy with doxorubicin (62). Nisin exhibits highly selective toxicity to MCF-7 cells, with an IC50 value of 5 µM, but is not toxic to noncancerous HUVECs. The combination of nisin and doxorubicin at subinhibitory concentrations resulted in increased cytotoxic activity compared with either agent alone, indicating potential synergistic effects (62). This observation was further supported by another in vitro study demonstrating that the coadministration of nisin and doxorubicin improved treatment outcomes in patients with skin cancer (87). The potential synergistic interactions between nisin and standard chemotherapeutics may lead to improved clinical outcomes for patients with cancer (88). Thus, nisin used alone or in combination with other chemotherapeutic agents could be a potential treatment option for patients with BC. Future research on nisin should prioritize in vitro studies, using multiple BC cell lines and animal studies to understand its potential synergy with standard anticancer therapies and provide more evidence for clinical trials.
L-norvaline
Metabolic profiling has revealed that L-norvaline, a gut-derived metabolite, is key in the diversity of the gut microbiota and has the capacity to modulate disease progression through interactions with the microbiome (89). Notably, Lautropia, Rothia, Centipeda, Corynebacterium and Actinomyces species have exhibited notable associations with L-norvaline biosynthesis (28). As an arginase 1 (ARG1) inhibitor, L-norvaline impedes cancer development by interacting with doxorubicin (28,90,91). ARG1 facilitates the conversion of L-arginine to L-ornithine and urea within M2-type macrophages (92,93). The generated L-ornithine is subsequently metabolized into polyamines by ornithine decarboxylase, a process that can increase cancer cell proliferation and differentiation. Recent research demonstrated that the coadministration of L-norvaline and doxorubicin in an M2 macrophage and BC cell coculture system markedly inhibited cancer cell proliferation (28). This combined therapy has shown superior efficacy over monotherapy with either agent alone, further reducing the proliferative activity of BC cells (28). The potential of bacteriocins to exert anticarcinogenic effects on BC cells highlights their utility as adjuvants in standard BC treatment regimens. Collectively, these discoveries provide novel perspectives for the development of innovative BC therapeutic strategies.
Conclusions and future perspectives
This review examines the types and sources of common metabolites from intestinal and tissue-resident microbiota, their impact on BC development, and their role in modulating tumor therapy responses. The potential roles of secondary BAs, SCFAs, amino acid metabolites and bacteriocins (such as LPS, nisin and pyocyanin) in BC carcinogenesis, progression and metastasis were explored. The impact of bacterial metabolites on the efficacy of BC drug treatments, including chemotherapy, targeted therapy and immunotherapy was also discussed.
Pretreatment analysis of the microbiota provides oncologists with insights into tumor aggressiveness and chemotherapy sensitivity to guide treatment modifications. Currently, breast microbiota research is primarily preclinical and has focused on comparing the microbiota of breast tumor tissue with that of healthy tissue to identify tumor-specific characteristics. Most studies have been performed in vitro, with limited in vivo data and no clinical trials documented. However, both in vitro and in vivo studies are crucial for understanding the complex interactions between the gut metabolites and cancer cells within the host.
Diet, probiotics and prebiotics may markedly impact BC, suggesting they have potential as adjuvants in standard BC treatments (94). Certain Lactobacillus strains have been shown to have effects against BC (95). Prebiotics, which are indigestible fibers, foster the growth of beneficial bacterial in the gut, which can metabolize the conversion of these fibers into phytoestrogens and SCFAs (96). These metabolites, along with others, possess tumor suppressive properties, and exhibit antiestrogenic and antiproliferative effects, which reduce the risk of BC. Investigating the synergistic effects of gut metabolites combined with chemotherapies and immunotherapies could enhance their clinical efficacy and safety.
Acknowledgements
Not applicable.
Funding
The present study was supported by the National Natural Science Foundation of China (grant no. 81700745) and the Shanghai Natural Science Foundation (grant no. 20ZR1456600).
Availability of data and materials
Not applicable.
Authors' contributions
YG and WD wrote the first draft of the manuscript. YG contributed to the study conception, and reviewing and editing of the manuscript. YG, WD, DS, XZ, ZH, CL and YS contributed to manuscript revision. Data authentication is not applicable. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024. View Article : Google Scholar : PubMed/NCBI | |
Kovacs T, Miko E, Ujlaki G, Yousef H, Csontos V, Uray K and Bai P: The involvement of oncobiosis and bacterial metabolite signaling in metastasis formation in breast cancer. Cancer Metastasis Rev. 40:1223–1249. 2021. View Article : Google Scholar : PubMed/NCBI | |
Mikó E, Vida A and Bai P: Translational aspects of the microbiome-to be exploited. Cell Biol Toxicol. 32:153–156. 2016. View Article : Google Scholar : PubMed/NCBI | |
Mikó E, Kovács T, Sebő É, Tóth J, Csonka T, Ujlaki G, Sipos A, Szabó J, Méhes G and Bai P: Microbiome-microbial metabolome-cancer cell interactions in breast cancer-familiar, but unexplored. Cells. 8:2932019. View Article : Google Scholar : PubMed/NCBI | |
Kiss B, Mikó E, Sebő É, Toth J, Ujlaki G, Szabó J, Uray K, Bai P and Árkosy P: Oncobiosis and microbial metabolite signaling in pancreatic adenocarcinoma. Cancers. 12:10682020. View Article : Google Scholar : PubMed/NCBI | |
Kuo WT, Lee TC and Yu LC: Eritoran suppresses colon cancer by altering a functional balance in toll-like receptors that bind lipopolysaccharide. Cancer Res. 76:4684–4695. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chen MC, Chen YL, Wang TW, Hsu HP and Lai MD: Membrane bile acid receptor TGR5 predicts good prognosis in ampullary adenocarcinoma patients with hyperbilirubinemia. Oncol Rep. 36:1997–2008. 2016. View Article : Google Scholar : PubMed/NCBI | |
McKee AM, Kirkup BM, Madgwick M, Fowler WJ, Price CA, Dreger SA, Ansorge R, Makin KA, Caim S, Le Gall G, et al: Antibiotic-induced disturbances of the gut microbiota result in accelerated breast tumor growth. iScience. 24:1030122021. View Article : Google Scholar : PubMed/NCBI | |
Jones GS, Feigelson HS, Falk RT, Hua X, Ravel J, Yu G, Flores R, Gail MH, Shi J, Xu X and Goedert JJ: Mammographic breast density and its association with urinary estrogens and the fecal microbiota in postmenopausal women. PLoS One. 14:e02161142019. View Article : Google Scholar : PubMed/NCBI | |
Wu AH, Tseng C, Vigen C, Yu Y, Cozen W, Garcia AA and Spicer D: Gut microbiome associations with breast cancer risk factors and tumor characteristics: A pilot study. Breast Cancer Res Treat. 182:451–463. 2020. View Article : Google Scholar : PubMed/NCBI | |
Frugé AD, Van Der Pol W, Rogers LQ, Morrow CD, Tsuruta Y and Demark-Wahnefried W: Fecal Akkermansia muciniphila is associated with body composition and microbiota diversity in overweight and obese women with breast cancer participating in a presurgical weight loss trial. J Acad Nutr Diet. 120:650–659. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Yang Y, Su J, Zheng X, Wang C, Chen S, Liu J, Lv Y, Fan S, Zhao A, et al: Age-related compositional changes and correlations of gut microbiome, serum metabolome, and immune factor in rats. Geroscience. 43:709–725. 2021. View Article : Google Scholar : PubMed/NCBI | |
Rahman S, O'Connor AL, Becker SL, Patel RK, Martindale RG and Tsikitis VL: Gut microbial metabolites and its impact on human health. Ann Gastroenterol. 36:360–368. 2023.PubMed/NCBI | |
Neagoe CX, Ionica M, Neagoe OC and Trifa AP: The Influence of microbiota on breast cancer: A review. Cancers. 16:34682024. View Article : Google Scholar : PubMed/NCBI | |
Banerjee S, Tian T, Wei Z, Shih N, Feldman MD, Peck KN, DeMichele AM, Alwine JC and Robertson ES: Distinct microbial signatures associated with different breast cancer types. Front Microbiol. 9:9512018. View Article : Google Scholar : PubMed/NCBI | |
Bhatt AP, Redinbo MR and Bultman SJ: The role of the microbiome in cancer development and therapy. CA Cancer J Clin. 67:326–344. 2017. View Article : Google Scholar : PubMed/NCBI | |
Fernandes MR, Aggarwal P, Costa RGF, Cole AM and Trinchieri G: Targeting the gut microbiota for cancer therapy. Nat Rev Cancer. 22:703–722. 2022. View Article : Google Scholar : PubMed/NCBI | |
Yang L, Li A, Wang Y and Zhang Y: Intratumoral microbiota: Roles in cancer initiation, development and therapeutic efficacy. Signal Transduct Target Ther. 8:352023. View Article : Google Scholar : PubMed/NCBI | |
Cullin N, Antunes CA, Straussman R, Stein-Thoeringer CK and Elinav E: Microbiome and cancer. Cancer Cell. 39:1317–1341. 2021. View Article : Google Scholar : PubMed/NCBI | |
Rao Malla R, Marni R, Kumari S, Chakraborty A and Lalitha P: Microbiome assisted tumor microenvironment: Emerging target of breast cancer. Clin Breast Cancer. 22:200–211. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kovtonyuk LV and McCoy KD: Microbial metabolites and immunotherapy: Basic rationale and clinical indications. Semin Immunol. 67:1017552023. View Article : Google Scholar : PubMed/NCBI | |
Han J, Zhang S, Xu Y, Pang Y, Zhang X, Hu Y, Chen H, Chen W, Zhang J and He W: Beneficial effect of antibiotics and microbial metabolites on expanded Vδ2Vγ9 T cells in hepatocellular carcinoma immunotherapy. Front Immunol. 11:13802020. View Article : Google Scholar : PubMed/NCBI | |
Guo C, Kong L, Xiao L, Liu K, Cui H, Xin Q, Gu X, Jiang C and Wu J: The impact of the gut microbiome on tumor immunotherapy: From mechanism to application strategies. Cell Biosci. 13:1882023. View Article : Google Scholar : PubMed/NCBI | |
Vitorino M, Baptista de Almeida S, Alpuim Costa D, Faria A, Calhau C and Azambuja Braga S: Human microbiota and immunotherapy in breast cancer-a review of recent developments. Front Oncol. 11:8157722021. View Article : Google Scholar : PubMed/NCBI | |
Jia W, Xie G and Jia W: Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol. 15:111–128. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ridlon JM, Kang DJ, Hylemon PB and Bajaj JS: Bile acids and the gut microbiome. Curr Opin Gastroenterol. 30:332–338. 2014. View Article : Google Scholar : PubMed/NCBI | |
Tang W, Putluri V, Ambati CR, Dorsey TH, Putluri N and Ambs S: Liver- and microbiome-derived bile acids accumulate in human breast tumors and inhibit growth and improve patient survival. Clin Cancer Res. 25:5972–5983. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhu Q, Zai H, Zhang K, Zhang X, Luo N, Li X, Hu Y and Wu Y: L-norvaline affects the proliferation of breast cancer cells based on the microbiome and metabolome analysis. J Appl Microbiol. 133:1014–1026. 2022. View Article : Google Scholar : PubMed/NCBI | |
Tsvetikova SA and Koshel EI: Microbiota and cancer: Host cellular mechanisms activated by gut microbial metabolites. Int J Med Microbiol. 310:1514252020. View Article : Google Scholar : PubMed/NCBI | |
Di Modica M, Arlotta V, Sfondrini L, Tagliabue E and Triulzi T: The link between the microbiota and HER2+ breast cancer: The new challenge of precision medicine. Front Oncol. 12:9471882022. View Article : Google Scholar : PubMed/NCBI | |
Jaye K, Li CG, Chang D and Bhuyan DJ: The role of key gut microbial metabolites in the development and treatment of cancer. Gut Microbes. 14:20388652022. View Article : Google Scholar : PubMed/NCBI | |
Krishnamurthy K, Wang G, Rokhfeld D and Bieberich E: Deoxycholate promotes survival of breast cancer cells by reducing the level of pro-apoptotic ceramide. Breast Cancer Res. 10:R1062008. View Article : Google Scholar : PubMed/NCBI | |
Gándola YB, Fontana C, Bojorge MA, Luschnat TT, Moretton MA, Chiapetta DA, Verstraeten SV and González L: Concentration-dependent effects of sodium cholate and deoxycholate bile salts on breast cancer cells proliferation and survival. Mol Biol Rep. 47:3521–3539. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cong J, Liu P, Han Z, Ying W, Li C, Yang Y, Wang S, Yang J, Cao F, Shen J, et al: Bile acids modified by the intestinal microbiota promote colorectal cancer growth by suppressing CD8+ T cell effector functions. Immunity. 57:876–889. 2024. View Article : Google Scholar : PubMed/NCBI | |
Mikó E, Vida A, Kovács T, Ujlaki G, Trencsényi G, Márton J, Sári Z, Kovács P, Boratkó A, Hujber Z, et al: Lithocholic acid, a bacterial metabolite reduces breast cancer cell proliferation and aggressiveness. Biochim Biophys Acta Bioenerg. 1859:958–974. 2018. View Article : Google Scholar : PubMed/NCBI | |
Tang X, Lin CC, Spasojevic I, Iversen ES, Chi JT and Marks JR: A joint analysis of metabolomics and genetics of breast cancer. Breast Cancer Res. 16:4152014. View Article : Google Scholar : PubMed/NCBI | |
Sampsell K, Hao D and Reimer RA: The gut microbiota: A potential gateway to improved health outcomes in breast cancer treatment and survivorship. Int J Mol Sci. 21:92392020. View Article : Google Scholar : PubMed/NCBI | |
Luu TH, Bard JM, Carbonnelle D, Chaillou C, Huvelin JM, Bobin-Dubigeon C and Nazih H: Lithocholic bile acid inhibits lipogenesis and induces apoptosis in breast cancer cells. Cell Oncol (Dordr). 41:13–24. 2018. View Article : Google Scholar : PubMed/NCBI | |
Eslami SZ, Majidzadeh AK, Halvaei S, Babapirali F and Esmaeili R: Microbiome and breast cancer: New role for an ancient population. Front Oncol. 10:1202020. View Article : Google Scholar : PubMed/NCBI | |
Hou H, Chen D, Zhang K, Zhang W, Liu T, Wang S, Dai X, Wang B, Zhong W and Cao H: Gut microbiota-derived short-chain fatty acids and colorectal cancer: Ready for clinical translation? Cancer Lett. 526:225–235. 2022. View Article : Google Scholar : PubMed/NCBI | |
Mirzaei R, Afaghi A, Babakhani S, Sohrabi MR, Hosseini-Fard SR, Babolhavaeji K, Khani Ali Akbari S, Yousefimashouf R and Karampoor S: Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed Pharmacother. 139:1116192021. View Article : Google Scholar : PubMed/NCBI | |
Jaye K, Chang D, Li CG and Bhuyan DJ: Gut metabolites and breast cancer: The continuum of dysbiosis, breast cancer risk, and potential breast cancer therapy. Int J Mol Sci. 23:94902022. View Article : Google Scholar : PubMed/NCBI | |
Liu P, Wang Y, Yang G, Zhang Q, Meng L, Xin Y and Jiang X: The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol Res. 165:1054202021. View Article : Google Scholar : PubMed/NCBI | |
Thirunavukkarasan M, Wang C, Rao A, Hind T, Teo YR, Siddiquee AA, Goghari MAI, Kumar AP and Herr DR: Short-chain fatty acid receptors inhibit invasive phenotypes in breast cancer cells. PLoS One. 12:e01863342017. View Article : Google Scholar : PubMed/NCBI | |
Park HS, Han JH, Park JW, Lee DH, Jang KW, Lee M, Heo KS and Myung CS: Sodium propionate exerts anticancer effect in mice bearing breast cancer cell xenograft by regulating JAK2/STAT3/ROS/p38 MAPK signaling. Acta Pharmacol Sin. 42:1311–1323. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Zhao KN and Vitetta L: Effects of intestinal microbial-elaborated butyrate on oncogenic signaling pathways. Nutrients. 11:10262019. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Hu PC, Ma YB, Fan R, Gao FF, Zhang JW and Wei L: Sodium butyrate-induced apoptosis and ultrastructural changes in MCF-7 breast cancer cells. Ultrastruct Pathol. 40:200–204. 2016. View Article : Google Scholar : PubMed/NCBI | |
Mandal M and Kumar R: Bcl-2 expression regulates sodium butyrate-induced apoptosis in human MCF-7 breast cancer cells. Cell Growth Differ. 7:311–318. 1996.PubMed/NCBI | |
Chopin V, Toillon RA, Jouy N and Le Bourhis X: Sodium butyrate induces P53-independent, Fas-mediated apoptosis in MCF-7 human breast cancer cells. Br J Pharmacol. 135:79–86. 2002. View Article : Google Scholar : PubMed/NCBI | |
He C, Liu Y, Ye S, Yin S and Gu J: Changes of intestinal microflora of breast cancer in premenopausal women. Eur J Clin Microbiol Infect Dis. 40:503–513. 2021. View Article : Google Scholar : PubMed/NCBI | |
Dai ZL, Wu G and Zhu WY: Amino acid metabolism in intestinal bacteria: Links between gut ecology and host health. Front Biosci (Landmark Ed). 16:1768–1786. 2011. View Article : Google Scholar : PubMed/NCBI | |
Roager HM and Licht TR: Microbial tryptophan catabolites in health and disease. Nat Commun. 9:32942018. View Article : Google Scholar : PubMed/NCBI | |
Renga G, Nunzi E, Pariano M, Puccetti M, Bellet MM, Pieraccini G, D'Onofrio F, Santarelli I, Stincardini C, Aversa F, et al: Optimizing therapeutic outcomes of immune checkpoint blockade by a microbial tryptophan metabolite. J Immunother Cancer. 10:e0037252022. View Article : Google Scholar : PubMed/NCBI | |
Sári Z, Mikó E, Kovács T, Boratkó A, Ujlaki G, Jankó L, Kiss B, Uray K and Bai P: Indoxylsulfate, a metabolite of the microbiome, has cytostatic effects in breast cancer via activation of AHR and PXR receptors and induction of oxidative stress. Cancers. 12:29152020. View Article : Google Scholar : PubMed/NCBI | |
Sári Z, Mikó E, Kovács T, Jankó L, Csonka T, Lente G, Sebő É, Tóth J, Tóth D, Árkosy P, et al: Indolepropionic acid, a metabolite of the microbiome, has cytostatic properties in breast cancer by activating AHR and PXR receptors and inducing oxidative stress. Cancers (Basel). 12:24112020. View Article : Google Scholar : PubMed/NCBI | |
Kovács T, Mikó E, Vida A, Sebő É, Toth J, Csonka T, Boratkó A, Ujlaki G, Lente G, Kovács P, et al: Cadaverine, a metabolite of the microbiome, reduces breast cancer aggressiveness through trace amino acid receptors. Sci Rep. 9:13002019. View Article : Google Scholar : PubMed/NCBI | |
Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, Rotter-Maskowitz A, Weiser R, Mallel G, Gigi E, et al: The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 368:973–980. 2020. View Article : Google Scholar : PubMed/NCBI | |
Shcheblyakov DV, Logunov DY, Tukhvatulin AI, Shmarov MM, Naroditsky BS and Gintsburg AL: Toll-like receptors (TLRs): The role in tumor progression. Acta Naturae. 2:21–29. 2010. View Article : Google Scholar : PubMed/NCBI | |
Li J, Yin J, Shen W, Gao R, Liu Y, Chen Y, Li X, Liu C, Xiang R and Luo N: TLR4 promotes breast cancer metastasis via Akt/GSK3β/β-catenin pathway upon LPS stimulation. Anat Rec (Hoboken). 300:1219–1229. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liao SJ, Zhou YH, Yuan Y, Li D, Wu FH, Wang Q, Zhu JH, Yan B, Wei JJ, Zhang GM and Feng ZH: Triggering of toll-like receptor 4 on metastatic breast cancer cells promotes αvβ3-mediated adhesion and invasive migration. Breast Cancer Res Treat. 133:853–863. 2012. View Article : Google Scholar : PubMed/NCBI | |
Yang H, Wang B, Wang T, Xu L, He C, Wen H, Yan J, Su H and Zhu X: Toll-like receptor 4 prompts human breast cancer cells invasiveness via lipopolysaccharide stimulation and is overexpressed in patients with lymph node metastasis. PLoS One. 9:e1099802014. View Article : Google Scholar : PubMed/NCBI | |
Avand A, Akbari V and Shafizadegan S: In vitro cytotoxic activity of a Lactococcus lactis antimicrobial peptide against breast cancer cells. Iran J Biotechnol. 16:e18672018. View Article : Google Scholar : PubMed/NCBI | |
Paiva AD, De Oliveira MD, De Paula SO, Baracat-Pereira MC, Breukink E and Mantovani HC: Toxicity of bovicin HC5 against mammalian cell lines and the role of cholesterol in bacteriocin activity. Microbiology (Reading). 158:2851–2858. 2012. View Article : Google Scholar : PubMed/NCBI | |
Kamarajan P, Hayami T, Matte B, Liu Y, Danciu T, Ramamoorthy A, Worden F, Kapila S and Kapila Y: Nisin ZP, a bacteriocin and food preservative, inhibits head and neck cancer tumorigenesis and prolongs survival. PLoS One. 10:e01310082015. View Article : Google Scholar : PubMed/NCBI | |
Ahmadi S, Ghollasi M and Hosseini HM: The apoptotic impact of nisin as a potent bacteriocin on the colon cancer cells. Microb Pathog. 111:193–197. 2017. View Article : Google Scholar : PubMed/NCBI | |
Hall S, McDermott C, Anoopkumar-Dukie S, McFarland AJ, Forbes A, Perkins AV, Davey AK, Chess-Williams R, Kiefel MJ, Arora D and Grant GD: Cellular effects of pyocyanin, a secreted virulence factor of Pseudomonas aeruginosa. Toxins (Basel). 8:2362016. View Article : Google Scholar : PubMed/NCBI | |
Zhao J, Wu Y, Alfred AT, Wei P and Yang S: Anticancer effects of pyocyanin on HepG2 human hepatoma cells. Lett Appl Microbiol. 58:541–548. 2014. View Article : Google Scholar : PubMed/NCBI | |
Moayedi A, Nowroozi J and Sepahy AA: Cytotoxic effect of pyocyanin on human pancreatic cancer cell line (Panc-1). Iran J Basic Med Sci. 21:794–799. 2018.PubMed/NCBI | |
Abdelaziz AA, Kamer AMA, Al-Monofy KB and Al-Madboly LA: A purified and lyophilized Pseudomonas aeruginosa derived pyocyanin induces promising apoptotic and necrotic activities against MCF-7 human breast adenocarcinoma. Microb Cell Fact. 21:2622022. View Article : Google Scholar : PubMed/NCBI | |
Geng HW, Yin FY, Zhang ZF, Gong X and Yang Y: Butyrate suppresses glucose metabolism of colorectal cancer cells via GPR109a-AKT signaling pathway and enhances chemotherapy. Front Mol Biosci. 8:6348742021. View Article : Google Scholar : PubMed/NCBI | |
Chen M, Jiang W, Xiao C, Yang W, Qin Q, Mao A, Tan Q, Lian B and Wei C: Sodium butyrate combined with docetaxel for the treatment of lung adenocarcinoma A549 cells by targeting Gli1. Onco Targets Ther. 13:8861–8875. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lajkó E, Spring S, Hegedüs R, Biri-Kovács B, Ingebrandt S, Mező G and Kőhidai L: Comparative cell biological study of in vitro antitumor and antimetastatic activity on melanoma cells of GnRH-III-containing conjugates modified with short-chain fatty acids. Beilstein J Org Chem. 14:2495–2509. 2018. View Article : Google Scholar : PubMed/NCBI | |
Chen W, Wei F, Xu J, Wang Y, Chen L, Wang J and Guan X: Trastuzumab enhances the anti-tumor effects of the histone deacetylase inhibitor sodium butyrate on a HER2-overexpressing breast cancer cell line. Int J Mol Med. 28:985–991. 2011.PubMed/NCBI | |
Andrade FO, Nagamine MK, Conti AD, Chaible LM, Fontelles CC, Jordão Junior AA, Vannucchi H, Dagli ML, Bassoli BK, Moreno FS and Ong TP: Efficacy of the dietary histone deacetylase inhibitor butyrate alone or in combination with vitamin A against proliferation of MCF-7 human breast cancer cells. Braz J Med Biol Res. 45:841–850. 2012. View Article : Google Scholar : PubMed/NCBI | |
Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K and Muto M: Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw Open. 3:e2028952020. View Article : Google Scholar : PubMed/NCBI | |
Ren S, Feng L, Liu H, Mao Y and Yu Z: Gut microbiome affects the response to immunotherapy in non-small cell lung cancer. Thorac Cancer. 15:1149–1163. 2024. View Article : Google Scholar : PubMed/NCBI | |
Muradas TC, Freitas RD, Goncalves JI, Xavier FA and Marinowic DR: Potential antitumor effects of short-chain fatty acids in breast cancer models. Am J Cancer Res. 14:1999–2019. 2024. View Article : Google Scholar : PubMed/NCBI | |
Coutzac C, Jouniaux JM, Paci A, Schmidt J, Mallardo D, Seck A, Asvatourian V, Cassard L, Saulnier P, Lacroix L, et al: Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat Commun. 11:21682020. View Article : Google Scholar : PubMed/NCBI | |
Zhou C, Basnet R, Zhen C, Ma S, Guo X, Wang Z and Yuan Y: Trimethylamine N-oxide promotes the proliferation and migration of hepatocellular carcinoma cell through the MAPK pathway. Discov Oncol. 15:3462024. View Article : Google Scholar : PubMed/NCBI | |
Jalandra R, Dalal N, Yadav AK, Verma D, Sharma M, Singh R, Khosla A, Kumar A and Solanki PR: Emerging role of trimethylamine-N-oxide (TMAO) in colorectal cancer. Appl Microbiol Biotechnol. 105:7651–7660. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Rong X, Zhao G, Zhou Y, Xiao Y, Ma D, Jin X, Wu Y, Yan Y, Yang H, et al: The microbial metabolite trimethylamine N-oxide promotes antitumor immunity in triple-negative breast cancer. Cell Metab. 34:581–594.e8. 2022. View Article : Google Scholar : PubMed/NCBI | |
Yang S, Dai H, Lu Y, Li R, Gao C and Pan S: Trimethylamine N-oxide promotes cell proliferation and angiogenesis in colorectal cancer. J Immunol Res. 2022:70438562022. View Article : Google Scholar : PubMed/NCBI | |
Chiba A, Bawaneh A, Velazquez C, Clear KYJ, Wilson AS, Howard-McNatt M, Levine EA, Levi-Polyachenko N, Yates-Alston SA, Diggle SP, et al: Neoadjuvant chemotherapy shifts breast tumor microbiota populations to regulate drug responsiveness and the development of metastasis. Mol Cancer Res. 18:130–139. 2020. View Article : Google Scholar : PubMed/NCBI | |
Groizeleau J, Rybtke M, Andersen JB, Berthelsen J, Liu Y, Yang L, Nielsen TE, Kaever V, Givskov M and Tolker-Nielsen T: The anti-cancerous drug doxorubicin decreases the c-di-GMP content in Pseudomonas aeruginosa but promotes biofilm formation. Microbiology (Reading). 162:1797–1807. 2016. View Article : Google Scholar : PubMed/NCBI | |
Abdelaziz AA, Kamer AMA, Al-Monofy KB and Al-Madboly LA: Pseudomonas aeruginosa's greenish-blue pigment pyocyanin: Its production and biological activities. Microb Cell Fact. 22:1102023. View Article : Google Scholar : PubMed/NCBI | |
Chiba A, Bawaneh A, Velazquez C, Clear KYJ, Wilson AS, Howard-McNatt M, Levine EA, Levi-Polyachenko N, Yates-Alston SA, Diggle SP, et al: Neoadjuvant Chemotherapy shifts breast tumor microbiota populations to regulate drug responsiveness and the development of metastasis. Mol Cancer Res. 18:130–139. 2020. View Article : Google Scholar : PubMed/NCBI | |
Baindara P and Mandal SM: Bacteria and bacterial anticancer agents as a promising alternative for cancer therapeutics. Biochimie. 177:164–189. 2020. View Article : Google Scholar : PubMed/NCBI | |
Rana K, Sharma R and Preet S: Augmented therapeutic efficacy of 5-fluorouracil in conjunction with lantibiotic nisin against skin cancer. Biochem Biophys Res Commun. 520:551–559. 2019. View Article : Google Scholar : PubMed/NCBI | |
Coker OO, Liu C, Wu WKK, Wong SH, Jia W, Sung JJY and Yu J: Altered gut metabolites and microbiota interactions are implicated in colorectal carcinogenesis and can be non-invasive diagnostic biomarkers. Microbiome. 10:352022. View Article : Google Scholar : PubMed/NCBI | |
Gao L, Zhang JH, Chen XX, Ren HL, Feng XL, Wang JL and Xiao JH: Combination of L-Arginine and L-Norvaline protects against pulmonary fibrosis progression induced by bleomycin in mice. Biomed Pharmacother. 113:1087682019. View Article : Google Scholar : PubMed/NCBI | |
Ren X, Wang N, Zhou Y, Song A, Jin G, Li Z and Luan Y: An injectable hydrogel using an immunomodulating gelator for amplified tumor immunotherapy by blocking the arginase pathway. Acta Biomater. 124:179–190. 2021. View Article : Google Scholar : PubMed/NCBI | |
Arlauckas SP, Garren SB, Garris CS, Kohler RH, Oh J, Pittet MJ and Weissleder R: Arg1 expression defines immunosuppressive subsets of tumor-associated macrophages. Theranostics. 8:5842–5854. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yurdagul AJ, Subramanian M, Wang X, Crown SB, Ilkayeva OR, Darville L, Kolluru GK, Rymond CC, Gerlach BD, Zheng Z, et al: Macrophage metabolism of apoptotic cell-derived arginine promotes continual efferocytosis and resolution of injury. Cell Metab. 31:518–533.e10. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wieërs G, Belkhir L, Enaud R, Leclercq S, Philippart de Foy JM, Dequenne I, de Timary P and Cani PD: How probiotics affect the microbiota. Front Cell Infect Microbiol. 9:4542020. View Article : Google Scholar : PubMed/NCBI | |
German R, Marino N, Hemmerich C, Podicheti R, Rusch DB, Stiemsma LT, Gao H, Xuei X, Rockey P and Storniolo AM: Exploring breast tissue microbial composition and the association with breast cancer risk factors. Breast Cancer Res. 25:822023. View Article : Google Scholar : PubMed/NCBI | |
Davani-Davari D, Negahdaripour M, Karimzadeh I, Seifan M, Mohkam M, Masoumi SJ, Berenjian A and Ghasemi Y: Prebiotics: Definition, types, sources, mechanisms, and clinical applications. Foods. 8:922019. View Article : Google Scholar : PubMed/NCBI |