ABSTRACT
Previously believed to be sterile, the breast microenvironment has been revealed by modern DNA sequencing technologies to harbor a diverse community of microorganisms. The breast tumor microenvironment (TME) has a microbial signature unique to that of other breast pathologies as well as between breast cancer subtypes and stage. Among the plethora of microorganisms identified, Methylobacterium radiotolerans and Sphingomonas yanoikuyae stand out, both elevated in breast cancer tissue and associated with cancer stage. Breast cancer is the most common malignancy affecting women and the second most common cause of cancer-specific death in women worldwide. Gut dysbiosis has recently emerged as a key player, although the exact mechanisms are still unclear. Hypothesized mechanisms include bacterial metabolites inducing genomic instability, imbalances in the local and systemic immune system, the role of gut microbiota in the regulation of estrogen metabolism. Probiotic commensals Akkermansia muciniphila and Bifidobacterium appear to have a protective effect, with evidence of gut wall protection, correlation with less advanced disease and better treatment efficacy and tolerability. This review outlines the relationship between the breast microbiome, the gut microbiome, the ‘estrabolome’, and the immune system in breast cancer. This characterization could make a significant clinical contribution, potentially leading to new methods of primary prevention, better prognostication and prediction, as well as new avenues of treatment.
KEYWORDS: Breast cancer, dysbiosis, microbiome, microbiota, estrabolome
GRAPHICAL ABSTRACT

Created with BioRender.com
Introduction
The breast microbiome is an emerging area of research that explores the presence, diversity, and potential influence of microbial communities within breast tissue. Interest in the human microbiome and its association with disease has seen a sudden increase in the number of publications (>12,000 in 2024 alone). With more than 15% of cancers globally now considered to be attributable to infectious causes,1 establishing the pathogenesis by which dysbiosis propagates tumorigenesis is of increasing relevance. The hype that currently surrounds the trillions of microorganisms (the microbiota) which call our bodies “home” as well as their genetic composition (the microbiome) needs to be carefully considered in light of the available evidence.2 Unlike the well-studied gut microbiome, the breast microbiome is less understood but is gaining attention for its potential role in breast health and disease. Molecular subtyping of breast cancer has critical prognostic and predictive value, routinely assessed by immunohistochemistry for hormone receptor and HER2 status. Estrogen receptor positive breast cancers have the greatest microbial diversity, while triple negative tumors have the least diverse TME.3
Breast cancer has a complex dynamic relationship between the breast microbiome, the gut microbiome, which includes estrogen metabolizing gut bacteria (the ‘estrabolome’) and the immune system. Breast cancer patients can be subdivided by their gut microbiome into four microbial clusters each with distinct gene expression, clinical characteristics and prognosis.4 The unfavorable clusters had high tumor mutational burdens and complex immune environments and interestingly included all molecular subtypes, albeit with a predominance of triple negative breast cancers, which typically have the worst clinical outcomes. Treatment response and outcomes are varied even within the same subgroup, generating interest in novel ways to categorize patients beyond the traditional system for a more tailored approach to treatment.
Gut dysbiosis with loss of diversity has been described in connection with many cancers, including breast, colorectal and lung cancer,5–7 and chronic diseases, with notable bacterial populations Bacteroides, Prevotella and Ruminococcus in chronic disease states.8 There is growing evidence that cancer cells may also be influenced by the pro-carcinogenic effects of microbial dysbiosis, via angiogenesis and increased T-helper 17 (Th17) response.9,10
Proving causality between the gut microbiome and the breast TME is a key topic to be explored in future research for the therapeutic potential of manipulating the microbiome is yet to be unlocked. Doing so could lead to major innovations in the approaches to managing breast cancer, shedding light on the gut microbiome as a prognostic biomarker or even target for precision medicine. In this review, we will outline the key evidence and potential mechanistic features which link the gut microbiome and the breast TME.
Breast cancer
According to data from the Global Cancer Observatory, almost 2.3 million new cases and 700 thousand deaths from breast cancer were estimated in 2022.11 The 5-year overall survival for breast cancer is close to 90% in many developed countries, including the United Kingdom, America and Australia, but as low as 20% for metastatic disease.12,13
Breast cancer has four molecular subtypes: Luminal A, Luminal B, HER2 enriched and basal/triple negative. Whilst this classification is based on gene expression profiling, these equate to a simplified immunohistochemical tumor phenotype dependent on the expression of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and proliferation marker Antigen Kiel 67 (Ki67) (Table 1). These features influence tumor behavior, guide treatment decisions and affect prognosis.14 However, this is further influenced by the complex TME of breast cancer, whereby single cell transcriptomics investigations on the role of tumor infiltrating lymphocytes (TILs) and stromal cells in disease progression and treatment response have been identified.16
Table 1.
| Cancer subtype | Biomarker |
Prevalence (%) | Systemic Treatment | Characteristics | Prognosis | ||
|---|---|---|---|---|---|---|---|
| ER | PR | HER2/Ki67 | |||||
| Luminal A | + | ± | -/low | 40–50 | Hormone therapy e.g. SERM, ER antagonist, AI Ovarian ablation e.g. oophorectomy, LHRH analogues, CDK inhibitors |
|
Best |
| Luminal B | + | ± | +/high | 10–20 | Higher proliferation than luminal A | Worse than luminal A | |
| HER2 | – | – | +/any | 10 | Anti-HER2 therapies Tyrosine kinase inhibitor mTOR inhibitor (everolimus) |
Aggressive | Poor short term |
| TNBC/Basal-like | – | – | –/any | 10–15 | None; chemotherapy |
|
Poor short-term |
AI = aromatase inhibitor; CDK = cyclin-dependent kinase; ER= estrogen receptor; LHRH = luteinizing hormone releasing hormone; mTOR = mammalian target of rapamycin; PR= progesterone receptor SERM = selective ER modulator; TNBC = triple negative breast cancer.
Importantly, the risk of developing breast cancer rises with increased circulating estrogen levels, brought on by metabolism of endogenous hormones and exogenous gut estrogens.17 Linked with increased risk is exposure to xenobiotics (drugs, chemicals, and environmental pollutants) and their impact on the gastrointestinal and mammary microbiota.18
Breast microbiome
The human breast microbiome is unique and distinct from the microbial composition of other organs,19 although an unequivocal profile has yet to be established. Investigation of the breast microbiome has documented a diverse population of bacteria in breast tumors that differs significantly from normal breast tissue, irrespective of age, geographical location, obstetric history and cancer-status. Proteobacteria, Actinobacteria and Firmicutes are the dominant phyla identified, with samples studied including The Cancer Genome Atlas (TCGA) breast cancer data study by Thompson et al.20 and the large tissue sample study by Tzeng et al.20–26 A profile of the human mammary microbiome using 16S rRNA sequencing and culture also reported Proteobacteria as the most common phylum, followed by Firmicutes, Actinobacteria and Bacteroidetes independent of lactation history.25
The composition of the breast microbiome at the phylum level has been validated in multiple subsequent studies. Nejman et al.27 published a landmark publication profiling the microbiome of over 1,500 tumor and normal tissue samples, highlighting the diverse microbiome of breast cancer tissue. Notably, >60% of breast cancer samples were positive for bacterial DNA with an average of 16.4 species identified in each.27 Bacterial species of note include Escherichia coli, Sphingobacteria, Prevotella, Mobiluncus, Brevundimonas Staphylococccus Methylobacterium radiotolerans and Sphingomonas yanoikuyae. Additionally, Bacteriodes20,22,23,25,26 and Bifidobacteria (from phyla Bacteroidetes and Actinobacteria, respectively) were found as dominant genera.28
The breast microbiome may influence tumor behavior through several mechanisms: inflammation, immune modulation, metabolism and hormonal regulation. Still unknown is whether it is local dysbiosis that promotes carcinogenesis or whether the tumor microbial signature is naturally selected by the TME. Current findings highlight unrecognized links between breast dysbiosis and breast cancer. A summary of the major findings in the breast cancer microbiome are detailed in Table 2.
Table 2.
Summary of abundant bacterial species associated with the breast microbiome.
| Reference Chronological order |
Study Size | Breast Sample Type | Method of Analysis | Abundant Bacteria in Breast Cancer Tissue |
|---|---|---|---|---|
| Urbaniak et al. 25 | 60 BC samples Canadian study: 27 BC & adjacent 11 Benign disease 5 healthy Ctls Irish study: 33 BC & adjacent 5 healthy Ctls |
Frozen | 16S rRNA gene seq Bacterial culture |
Proteobacteria: E. coli Canadian BC samples: Acinetobacter (10%), Bacillus (11.4%), Enterobacteriaceae (8.3%) Irish BC samples: Proteobacteria Enterobacteriaceae (30.8%), Staphylococcus (12.7%), Listeria welchimeri (12.1%) |
| Xuan et al.6 | Analysis 1: 20 ER+ tumor 20 adj normal Analysis 2: 39 tumor 39 adj normal 23 healthy controls |
FFPE | 16S DNA pyro-seqencing qPCR |
Abundant phyla in all samples: Proteobacteria, Firmicutes, Actinobacteria, Bacteroidetes Pseudomonas: M. radiotolerans Adjacent tissue: S. yanoikuyae |
| Banerjee et al.29 | 100 TNBC 37 Controls: (17 matched, 20 non-matched) |
FFPE | PathoChip microarray PCR MiSeq |
Arcanobacterium (75%), then Brevundimonas, Sphingobacteria, Providencia, Prevotella, Brucella, Eschherichia, Actinomyces, Mobiluncus & Propionibacteria, Geobacillus, Rothia, Peptinophilus, Capnocytophaga |
| Chan et al.30 | 25 ductal carcinoma 23 healthy ctls |
NAF | 16S rRNA gene amplicon sequencing |
NAF: Firmicutes, Proteobacteria and Bacteroidetes NAF (BC history): Alistipes (phylum Bacteroidetes) Controls: Sphingomonadaceae |
| Hieken et al.23 | 15 invasice BC samples, 13 benign disease no atypia. 28 adj ctls |
Frozen Adjacent normal Adjacent skin |
16S rRNA sequencing | Fusobacteriota: Fusobacterium Atopobium, Gluconacetobacter, Hydrogenophaga, Lactobacillus |
| Urbaniak et al.26 | 45 BC + normal adj 13 Benign breast disease 23 healthy ctls |
Frozen | 16S rRNA amplicon sequencing | Predominant: Firmicutes (phlya Bacillus & Lactococcus), Proteobacteria, Actinobacteria, Bacteroidetes (Prevotella) BC: genus Bacillus, Enterobacteriaceae, Staphylococcus, Comamondaceae, Bacteroidetes. Healthy control: Prevotella, Lactococcus, Streptococcus, Corynebacterium |
| Yazdi et al.31 | 123 sentinel BC-LN + adj normal 5 healthy breast |
Frozen Frozen LN |
RT-PCR |
M. radiotolerans increases in sentinel LN with increasing BC stage. Normal breast tissue: Proteobacteria- Sphingomonas yanoikuyae |
| Thompson et al.20 | 688 tumor tissues 72 adjacent normal |
The Cancer Genome Atlas (TCGA) BC data | 16S rRNA gene seq |
Proteobacteria, Mycobacterium fortuitum & phlei. Adjacent normal: Actinobacteria |
| Wang et al.32 | 57 invasive BC + adj normal 21 healthy ctls |
Frozen | 16S rRNA gene sequencing |
BC HR+: lower abundance of Proteobacteria: Methylobacterium compared to control, increased Alcaligenaceae (unidentified genus) |
| Banerjee et al.21 | 148 BC (50 ER+/PR+, 34 hER2+, 24 triple positive, 40 TN) 20 healthy ctls: non-matched breast tissues |
FFPE | Whole genome & transcriptome amplification PathoChip microarray PCR Sanger seq |
All four BC types: Proteobacteria (genera Brevundimonas, Bartonella), Firmicutes & Actinomyces (sp. Mobiluncus). ER/PR positive: Arcanobacterium, Escherichia Bifidobacterium, Cardiobacterium, Citrobacter, HER2 positive: Streptococcus Triple positive: Bordatella, Campylobacter, Chlamydia, Chlamydophila, Leionela, Pasteurella TNBC least microbially complex; associated with Aerococcus, Arcobacter, Geobacillus, Orientia, Rothia. Prevotella only in BC samples. |
| Costantini et al.22 | 16 BC patients (12 core biopsies, 7 surgical excision) 19 adjacent breast healthy ctls |
Fresh | Microbial gDNA isolation 16S rRNA gene amplicon sequencing |
Proteobacteria, Firmicutes, Actinobacteria, Bacteroidetes; Ralstonia (most abundant genus): Methylobacterium and Ralstonia (phylum Proteobacteria) relatively increased, Sphingomonas decreased |
| Meng et al.28 | 72 IDC 47 ER+; 25 ER- 42 PR+; 30 PR- 22 benign ctls: |
Frozen | 16S rRNA gene amplicon seq | Actinobacteria: Agrococcus, Propionicimonas families Micrococcaceae Caulobacteraceae, Rhodobacteraceae, Nocardioidaceae, Methylobacteriaceae Increasing BC grade: Agrococcus abundance & reduction in Bacteroidaceae |
| Chiba et al.33 | 15 IDC (previous neoadjuv chemo) Ctls: 18 IDC no previous chemo |
Frozen tissue Cell lines: MDA-MB-231 MB-4T1 ZR-75–1, 67NR MCF-7 |
PCR, IHC 16S rRNA seq Microarray staining Western blot Proliferation assay |
IDC Neoadjuvant chemotherapy: Pseudomonas, decrease Prevotella. P. aeruginosa stimulates cell growth: MDA-MB-231, MBF-7 & ZR-75–1 Metastasised tumors: Brevundimonas & Staphylococcus |
| Nejman et al.27 | 355 BC samples 270 ER+; 49 ER- 61 hER2+ 247 hER2- 36 TN; 284 non-TN 256 adj normal |
Frozen FFPE |
16S real-time qPCR 16S rRNA gene amplification CLEM IHC, FISH, IF |
In descending order: Streptococcus infantis, Lactobacillus iners, Corynebacterium and Fusobacterium nucleatum, Staphylococcus cohnii, Paracoccus marcusii, Enterobacter cloacae, Acinetobacter, Staphylococcus aureus |
| Thyagarajan et al.34 | 23 BC samples | Frozen | 16S rRNA amplicon sequencing MiSeq |
In descending order: Ralstonia, Staphylococcus, family Bradyrhizobiaceae, Rubrobacter, Pseudomonas |
| Tzeng et al.24 | 221 BC samples 87 healthy ctls |
Frozen | 16S rRNA gene seq 16S qPCR IHC NanoString |
Pseudomonas, Proteus, Porphyromonas & Azomonas Increased with higher stage: Porphyromonas, Lacibacter, Ezakiella and Fusobacterium. Benign breast tissue: genera Propionibacterium, Finegoldia, Granulicatella, Streptococcus, Anaerococcus, Ruminococcaceae UCG-002, Corynebacterium 1, Alicyclobacillus, Odoribacter, Lactococcus, Esherichica/Shigella |
Adj: adjacent; Chemo: chemotherapy; CLEM: correlative light and electron microscopy; Ctls: controls; FFPE: formalin fixed paraffin embedded; ER+: estrogen receptor positive; FISH: fluorescence in situ hybridization; HER2+: human epidermal growth factor receptor 2; HR: hormone receptor; IDC: invasive ductal carcinoma; IF: immunofluorescence; IHC: immunohistochemistry; LN: lymph nodes; NAF: nipple aspirate fluid; OTU: operational taxonomic unit (sequence identity-based clustering); PCR: polymerase chain reaction; PR+: progesterone receptor positive; Sequencing: seq; TNBC: triple negative breast cancer.
Bacterial species associated with breast cancer
Escherichia coli
Escherichia coli is found abundant in normal breast tissue adjacent to breast tumors in comparison with healthy controls.25,26 E. coli has various virulence factors that promote adhesion, infiltration and survival, as well as a genotoxic effect, including the polyketide synthase (pks) gene.35 Strains of E. coli with the pks pathogenicity island are able to produce colibactin, which can induce DNA damage, contributing to gene instability and carcinogenesis.26
Sphingobacteria, Prevotella (both phylum Bacteroidetes)
A relative abundance of Sphingobacteria and Prevotella has been detected in triple negative breast cancer (TNBC) samples;29 others report reduced numbers in breast cancer tissue relative to healthy control breast tissue, receptor status unspecified,6,26,30
Actinomyces (family Actinomycetaceae), family Propionobacteraceae (both phylum Actinobacteria)
Also elevated in breast cancer tissue are genera Actinomyces, as well as family Propionobacteraceae (both from phylum Actinobacterium) along with Staphylococcus,21,26,29 all gram positive bacteria, which can induce interferon-gamma (IFN-γ) secretion from T -cells and natural killer (NK) cells.36 Traditionally IFN-γ is known to be cytotoxic and inhibitory of tumors,36 with evidence that IFN-γ is lost early in breast cancer.37 IFN-γ is now thought to be able to promote tumor growth and survival in certain environments via immunosuppression, immuno-resistance and upregulation of cell proliferation.38
Staphylococcus and Brevundimonas
Overall, Staphylococcus was found to be more abundant in breast cancer tissue and found by Chiba et al.33 to be more abundant in metastatic disease along with a higher abundance of Brevundimonas.21,25–27,29,33 Brevundimonas concentrations were found to be significantly increased in tumors of all breast cancer subtypes.21
Notably, strains of E. coli and S. epidermidis demonstrated the ability to cause DNA damage by inducing double-stranded DNA breaks in HeLa cells.26
Methylobacterium radiotolerans
Methylobacterium, family Methylobacteriaceae, is reported to be elevated in breast cancer tissue6,22,28 and the sentinel lymph nodes of breast cancer patients, and associated with increasing cancer stage.31 Intratumoral Methylobacterium has been significantly associated with worse prognosis in gastric cancer, with a causal role via reducing CD8+ T cell response at the tumor site.39 In prostate cancer, M. radiotolerans is overrepresented in the tumor microbiome, and negatively associated with staging.40
Contrary to these findings, Wang et al., reported the depletion of Methylobacterium in hormone receptor (HR) positive breast cancer tissue.32 This disparity may be in part due to differences in methodology (e.g. formalin-fixed paraffin embedded vs. fresh vs. frozen tissue, choice of DNA extraction kit). Additionally, heightened invasive potential in breast cancer has been correlated to tumors with a reduced abundance of Methylobacterium.2 Methylobacteriaceae, produces phytohormones, which have an anti-cancer effect by inhibiting proliferation and inducing apoptosis, demonstrated in breast cancer cells in vitro.41,42
Protective bacteria abundant in non-cancerous breast tissue
The breast microbiome may play a crucial role in maintaining breast health, influencing local immune responses and potentially protecting against pathogens. Several studies suggest that a balanced microbiome may support anti-inflammatory and immunomodulatory functions, contributing to tissue homeostasis and defense mechanisms. Prominent bacteria isolated from benign breast tissue, including non-cancerous adjacent breast tissue, compared to cancerous breast tissue include: Corynebacterium, Lactococcus and Streptococcus;24,26 Prevotella;26 Methylobacterium;32 Sphingomonas;6 Escherichia20,24 and Haemophilus.20 Sphingomonadaceae was found more abundant in the nipple aspirate fluid of women without a history of breast cancer compared to that taken from women with a prior history of breast cancer.30 Additionally, Alicyclobacillus, Anaerococcus, Finegoldia, Granulicatella, Odoribacter, Propionibacterium, Ruminococcaceae and Shigella have been reported to be relatively abundant in healthy breast tissue.24
Lactococcus, a genus of lactic acid bacteria, has been shown to be protective against breast cancer by altering the cytotoxic effect of NK cells, which enhance cellular immunity and inhibit tumor growth, demonstrated in both tissue from humans with solid organ tumors and mice breast cancer models.43–45 In mice, oral administration of Lactobacillus was shown to suppress breast cancer growth and metastasis.46
Propionibacterium and Streptococcus are associated with T cell activation genes.24 Propionibacterium, which has anti-tumoral properties, is relatively depleted in breast cancer tissue.24 Propionibacterium induces an anti-cancer effect via activation of NK cells, macrophages, and T cells.47 This genus has also been associated with lower levels of oncogenic growth factors, and its downregulation is speculated to promote tumor growth through inhibition of the adaptive immune response.24 In vivo breast tumor suppressing efficacy has been demonstrated by inhibiting peritumoral angiogenesis and promoting apoptosis.47 Streptococcus has an anti-oxidant effect and is able to neutralize peroxides and reactive oxygen species, protecting cells against DNA damage.48 In addition, Streptococcus has been studied for its anti-tumor effect via promotion of Toll-like receptor (TLR) 4 signaling.49,50 It also produces cadaverine, which inhibits breast cancer invasion and epithelial-to-mesenchymal transition.51,52
Odoribacter (order Bacteroidales), found relatively abundant in benign breast tissue samples,24 produces the short chain fatty acid butyrate, which has been shown to have anti-cancer properties through its anti-inflammatory activity.52
Absolute abundance of S. yanoikuyae is increased in adjacent non-cancerous tissue.6,22 Sphingomonadaceae (particularly genera Sphingomonas, Sphingobium, Novosphingobium, and Sphingopyxis) were shown to degrade aromatic hydrocarbons such as estrogens, which is particularly relevant in HR positive breast cancer.53–56 Sphingomonas is found in breast tumors with increasing breast cancer stage31 with evidence of a protective effect demonstrated in ER positive breast cancers.57 Importantly, S. yanoikuyae expresses glycosphingolipid ligands that activate invariant NK cells,58 which are mediators of tumor immunosurveillance59 and breast cancer metastasis.60
Immunological profile of the breast TME
Breast tumorigenesis attracts a significant influx of immune cells, both adaptive and innate, to the breast TME. The immune profile of the tumor microbiome modulates the local inflammatory response that precedes tumor formation and contributes to progression.61 Disequilibrium between inflammatory and immunosuppressive signals from the immune system is a key player in the development and progression of carcinogenesis.62 Breast tumors demonstrate an influx of immunosuppressive type cells over proinflammatory immune cells.63
Previously thought to have low lymphocyte infiltration and to be immunologically “cold”, breast tumors have recently been found to demonstrate immunological heterogeneity between patients and tumor types.64 Tumor infiltrating lymphocytes (TILs) have an anti-tumoral effect, given their various cytotoxic and cytokine inducing abilities, as well as a role in recognition of tumor antigens. T-helper 1 (Th1) lymphocytes facilitate tumor suppression and upregulation of inflammatory cytokines, such as IFN-y, TNF-α and IL-12, to facilitate tumor cell apoptosis and promote cytotoxic CD8+ T cell activity.65–67 Permeation of TILs by CD8+ T cells is positively correlated with survival and therapeutic efficacy.68,69 Conversely, T-helper 2 (Th2) CD4+ lymphocytes have been associated with pro-tumorigenesis via IL-4 and IL-13 mediated promotion of mammary tumor cell invasion and metastasis.70,71 Other mechanisms described include resting CD4+ T cells being converted into regulatory T (Treg) cells, which support immune escape and have been shown to promote metastasis of breast cancer cells;72 whereas CD8+ T cells inhibit breast tumors via suppression of RANK signaling.73
Treg cells are found in higher numbers in invasive breast cancers than in-situ breast cancers, in ER negative more so than ER positive breast tumors and are associated with higher rates of relapse and poorer survival.74,75 Treg cell infiltration may be useful as a prognostic marker for identifying patients with a favorable benefit-to-risk ratio for undergoing further hormonal therapy beyond the standard dose. Estradiol has been found to promote proliferation of Treg cells76 so it follows that inhibition of estrogen in the TME may also inhibit Treg cell numbers. Treatment of breast cancer patients with the aromatase inhibitor letrozole causes a significant reduction in Treg cells in the TME (p < 0.0001)77 and in ER positive breast cancer patients increases tumor infiltration by B cells and CD4+ T cells.78
TNBC has a poor prognosis with no targeted therapies and few systemic therapeutic options other than chemotherapy.79 High peri-tumoral stromal TIL density is most often seen in TNBC and HER2 positive breast cancers, described to comprise mainly of T cells CD4+ and CD8+ and, CD19+ B cells and NK cells, along with increased PD-L1 expression.80,81 It is associated with improved prognosis and is a positive predictive factor for the efficacy of neoadjuvant chemotherapy.82–84 There is also interest in TIL expression in residual tumor post-neoadjuvant therapy given the growing role of immunotherapy in breast cancer treatment.85 Targeted immunotherapy with programmed death-ligand 1 (PD-L1) or programmed cell death protein 1 (PD-1) monoclonal antibodies is now offered to patients with early and metastatic TNBC, yielding promising results.80,86
Next generation sequencing technology
Gut microbiome 16S gene sequencing vs. Shotgun metagenomics
Current molecular techniques employed to study the microbiome largely involve Next Generation Sequencing (NGS) – either 16S ribosomal RNA (rRNA) sequencing or shotgun metagenomics.87 While 16S rRNA gene sequencing is routine for profiling bacterial gene expression and is useful when sequencing large numbers of samples, it provides limited data on taxonomy to differentiate between species of bacteria.88 The accuracy and reproducibility of bacterial identification by 16S rRNA sequencing is affected by the sampling technique and DNA isolation kit used, particularly its efficacy at cell lysis.89 Recent advancements and novel gene sequencing have provided even more detail about pathogens, such as organism composition and specific immune response. An example is dual RNA sequencing (RNAseq), which allows a parallel transcriptomic analysis of bacterial pathogens and their eukaryotic host cells, and single cell RNAseq, which has enabled identification of cells containing bacteria, and provided information on the interactions between immune cells and microbes during infection.90
Shotgun metagenomics provides taxonomic data on all species present in a sample (i.e. bacteria, viruses, archaea, protozoa, fungi) and information on potential metabolic output through analysis of all genes present, creating a more complete picture of the microbiome.91 On the down side, metagenomics is more expensive and detection is confounded by contaminants during the preparation/DNA extraction phase from kits or laboratory agents.91,92
Direct comparison of the two methods on the same fresh frozen stool samples demonstrate shotgun metagenomics permits for a more extensive characterization of the microbiome and can identify more bacterial species even at low sequencing depths.92 Comparison of oral and gut microbiomes yielded significant community differences with 16S rRNA sequencing analysis at the genus and species level, while shotgun metagenomics demonstrated a larger proportion of shared taxonomy between oral and gut microbiomes with improved resolution at the species level.93
Gut microbiome and the immune response
Microorganisms such as bacteria, archaea, viruses, protozoa and fungi can be found widely throughout the body’s epithelial surfaces. Bacteria from the gastrointestinal tract (GIT) can influence not only the local but also the systemic immune system via the enterohepatic circulation.94,95 It is by this mechanism that distant tumors can be exposed to microorganisms of the GIT.
Remarkably, with approximately 2 kg of microorganisms existing in the gut, the metabolic output of the GIT is equivalent to that of the liver.96 The GIT contains approximately 1000 species of commensal microorganisms distributed along its length,97 generally becoming more abundant distally – with 102–103 bacteria per mL in the stomach and up to 1012 per mL in the colon.98 Their aggregated bacterial genomes comprise 150 times more genes than their host human genome.99
The composition of the gut microbiome is highly variable between individuals and is influenced by factors such as diet, ethnicity, geographical location, drug history, body mass index (BMI), exercise and environmental interactions.95,100 The GIT is exposed to a plethora of external antigens that can trigger immune responses.98 The dominant gut microbes come from four major phyla: Firmicutes, Bacteroidetes, Proteobacteria and Actinobacteria, with approximately 90% of the microbiome represented by the genus Faecalibaterium of phylum Firmicutes, and genera Bacteroides and Alistipes of phylum Bacteroidetes.98,101,102 The Actinobacteria phylum is proportionally less abundant and mainly represented by the Bifidobacterium genus.101
Commensal gut microbes colonize the GIT and are essential for metabolic and immune functions with regions of the GIT affected by specific bacterial density and diversity.96,103 Clostridia species and Bacteroides fragilis, which tend to be found in the colon, are shown to promote production of immune suppressor Treg cells.104,105 Despite innate mechanisms in place at the gut lumen to prevent penetration of gut bacteria, microbes and their metabolic products may still cross the epithelium into the systemic circulation (Figure 1). The breast and GIT are connected via the mucosal immune system, such that infection or immunization of one mucosal organ can induce immunoglobulin A (IgA) production at a distal mucosal site and protect it from the same infection.108 Characterizing the association between breast cancer and gastrointestinal dysbiosis would be of interest in establishing a relationship in pathogenesis.
Figure 1.

Mechanisms of bacterial and metabolite invasion of the colonic mucosa.
Microorganisms on the luminal side of the gut epithelium are equipped with various mechanisms to penetrate the epithelium and migrate into the systemic circulation. Migration can occur via a paracellular pathway for small-diameter organisms, and transcellularly for larger organisms via transcytosis.106 Bacteria can have virulence factors such as: Lipopolysaccharides (LPS), an endotoxin found in gram-negative bacteria, which can activate toll-like receptor 4 (TLR4), CD14 and scavenger receptor-BI (SR-BI) to induce a pro-inflammatory response and allow its transcellular migration across the gut epithelium;106,107 strains of Escherichia coli containing the pks gene cluster and colibactin induces double-stranded DNA breaks and gene instability in host cells.
Some gut bacteria are protective and can produce short chain fatty acids, which have been shown to reduce inflammation and strengthen the intestinal epithelium by increasing production of Treg cells. Treg cells have anti-inflammatory properties and promote differentiation of effector T-helper cells. Proliferation of Treg cells suppresses effector T-cell immune responses, leading to reduced local inflammation. Described to have an immunosuppressive effect, B. fragilis contains polysaccharide A in its capsule, which through ligation with TLR2 mediates the differentiation of T reg cells to produce immunosuppressive IL-10, which in turn suppresses IL-17 production.
Once on the baso-lateral side of the gut epithelium, antigens can be recognized by dendritic cells in Peyer’s patches and mesenteric lymph nodes, which will trigger CD4+ T and B cells to respond. The subsequent systemic reaction means the lymphocytes, with their infection-specific receptors, can reach other organs of the mucosal immune system.108 Once arrived at effector sites, the plasma cells are transported to the apical side of the epithelial cells and release secretory IgA antibodies specific to antigens encountered at the induction site.108
Created with BioRender.com
Diet
Diet has a modifiable role in shaping the composition of the gut microbiome, with most of this variation found to be in the ratio of Prevotella to Bacteriodes, both from the Bacteroidetes phylum. The two genera show an inverse relationship, with Prevotella found more in the gut of hosts with a non-Western diet consisting abundantly of plants, fiber and polysaccharides; whereas Bacteroides is found to dominate in hosts with a Western diet comprising more proteins and fats.109 More specifically, dietary fiber from fruits and vegetables is associated with colonization with Clostridia, while fiber from beans raises the prevalence of Actinobacteria and Bifidobacteriales.110 A plant-rich diet is associated with a lower incidence of colorectal cancer,111 due to its anti-inflammatory effect on Treg cell induction.112
BMI
The association between breast cancer incidence and a higher BMI is well studied. Increased systemic estrogens is seen in obesity, from aromatization of abundant adipose tissue.113 This is particularly the case in post-menopausal women, where the aromatization of androgens and adipose tissue becomes the primary source of estrogens.114 In breast cancer, the percentage of body fat appears to be related to Akkermansia muciniphila abundance along with higher levels of Prevotella, Lactobacillus, and lower levels of Clostridium, Campylobacter and Helicobacter.115
Short chain fatty acids
Intestinal microbiota can directly influence the host immune response via the production of metabolites, such as short chain fatty acids (SCFA),116 amino acids and vitamins.117 Obese and overweight patients have significantly higher levels of fecal SCFA, Firmicutes and a higher Firmicutes to Bacteroidetes ratio than normal-weight patients, independent of diet.118 The Firmicutes to Bacteroidetes ratio is a marker of gut dysbiosis and positively correlated with obesity and inflammation.119 Prominent gut SCFAs, propionate, butyrate and acetate have been shown to reduce inflammation and strengthen the intestinal epithelium.120,121 Major metabolite butyrate has local anti-neoplastic effects and promotes normal colonic growth and differentiation of mucosa.122 Butyrate inhibits the production of inflammatory cytokines, including TNFα, IL-6, IL-8.123 Another SCFA, cadaverine, is thought to be a pro-carcinogenic amine124 with some studies reporting elevated levels in cancer125 and others reporting decreased levels.126 However, in breast cancer, cadaverine appears to be a tumor suppressor, reported to cause lower tumor mass and invasion, less metastases and lower tumor grade.127 Gut commensals are also able to produce SCFA, including Clostridia and Akkermansia species.115,128 Clostridium butyricum has been shown to reduce lymphocyte counts, enhance immunity and improve the ratio of beneficial to pathogenic bacteria in gastric cancer patients.128 Importantly, A. mucinophilia thickens the gut mucosal layer, which impedes bacterial translocation.115
Microbial toxins
The role of bacteria in cancer development and therapy has been well known for almost a century.129 Microbes are thought to promote carcinogenesis by induction of DNA damage via toxins, the most well known being Helicobacter pylori causing gastric cancer.130 Changes in the composition of the gut microbiome can cause inflammation and genotoxic stress via bacterial toxins, which damage the mucosal barrier allowing bacterial infiltration.131
Lipopolysaccharides (LPS), found in the cell wall of gram-negative bacteria, is a virulence factor that promotes inflammation through cytokine stimulation production by binding to TLR4.132 Involved in tumor survival and progression, TLR4 is upregulated in HR positive (MCF-7) and triple negative (MDA-MD-231) human breast cancer cell lines and is linked to increased metastasis.133 LPS has been reported to increase PD-L1 expression in gastric cancers, which when bound to the PD-1 transmembrane receptor inhibits T-cell activation.134 Via induction of the TLR4/NF-KB signaling pathway, LPS has been reported to promote cancer cell adhesion, invasion and metastasis.135–139
Similarly, strains of Bacteroides fragilis, which secrete the B. fragilis toxin (BFT), have been found to cause intestinal inflammation and promote colon tumor cell survival and proliferation. It degrades E-cadherin in the colon, leading to epithelial cell damage, inflammation and increased metastatic potential.140 The toxin can activate signal transducers and activators of transcription (STAT) 3 and T helper 17 (Th17) cell mediated responses leading to an upregulation of IL-17 levels, involved in intestinal inflammation.140 On the other hand, B. fragilis appears to have an opposite effect on the adaptive immune system. It has been described to have an immunosuppressive effect via Treg cells, as it contains polysaccharide A in its capsule, which through ligation with TLR2 mediates the differentiation of Treg cells to produce immunosuppressive IL-10, which in turn suppresses IL-17 production.141,142 B. fragilis has been identified in breast tissue, both benign and cancerous, and nipple aspirate fluid (NAF), and strains producing BFT have been demonstrated in mice models to cause local inflammation and epithelial hyperplasia.143 Additionally, mammary or gut colonization with BFT-bearing B. fragilis increased disease progression and metastases.
Biofilms
Biofilms can form on surfaces in the body from a community of microorganisms that adhere to each other within an extracellular matrix. Recent studies investigating the role of bacterial biofilms in colon carcinogenesis found that colonic biofilms upregulate polyamine metabolites to promote proliferation and tumor growth.144–146 Gut biofilms, suggested to be drivers of gut carcinogenesis, contain bacteria that promote DNA damage in colonic epithelium leading to initiation of tumor formation, although the exact mechanisms and species are still unknown.147–149 A study investigating the presence of bacterial biofilms in malignant breast wounds reported a predominance of anaerobic bacteria, although biofilm formation was not found to be associated with a particular species of bacteria nor clinical signs.150 A symbiotic relationship may exist, whereby biofilms may provide a protective environment for cancer cells allowing them to evade host immune responses.151
Interestingly, it has been proposed that certain drugs used in cancer therapy, such as doxorubicin, promote the formation of biofilms in vitro,152 with the potential to inhibit cancer metastasis or even act as a vector for cancer therapies.129,153
Microbial dysbiosis
Shotgun metagenomic studies exploring stool samples confirmed 38 bacterial species enriched in postmenopausal breast cancer patients and healthy controls, including E. coli, Klebsiella, Prevotella amnii, Enterococcus gallinarum and Actinomycess.119,154 Gut and mammary dysbiosis, resulting in an imbalance and loss of diversity in microbial composition, has been found to be associated not only with breast cancer development but with specific subtypes2,21,117 and responsiveness to therapy.155,156
The gut microbiome plays a critical role in modulating the host immune response. Gut dysbiosis may alter the balance between pro-inflammatory and anti-inflammatory immune responses, potentially impacting tumor surveillance and immune-mediated tumor suppression. The induction of inflammation even in non-cancerous mammary tissue suggests a possible pathway by which early gut dysbiosis is able to remotely contribute to breast carcinogenesis, as mammary inflammation has been linked to increased breast cancer risk.157 Mice transplanted with HR positive breast tumors demonstrated that pre-cancerous gut dysbiosis influences host immunity both systemically and locally, early dysbiosis enhances serum concentrations of cytokines (IL-22 and IL-23) and chemokines (GM-CSF, CCL2 and CXLC2), and increases circulation of cancer cells.158 Within the breast TME, gut dysbiosis promoted fibrosis and inflammation, with significantly upregulated CXCL10 and CCL2 chemokines, and myeloid infiltration both in the tumor and adjacent mammary gland.158
Shi et al.81 looked at TIL abundance in breast tumors and demonstrated a link between the gut microbiome and the TME. Higher abundance of TILs in breast tumors remains associated with a higher gut abundance of genera including Mycobacterium, Rhodococcus, and Catenibacterium, while low tumor abundance of TILs was associated with lower gut concentrations of Methanosphaera and Anaerobiospirillum.81 Also, a higher fecal abundance of Barnesiae (genus Bacteroides), involved in estrogen metabolism, is associated with low TIL abundance.81
Another pathway by which gut dysbiosis can lead to breast tumorigenesis may be via host immune responses to gastrointestinal infection. Infection of mice with Helicobacter hepaticus159 demonstrated increased levels of neutrophils in mammary tissue and amplified risk of breast carcinogenesis, while depletion of neutrophils inhibited tumor growth.160,161 Significant bacterial species identified demonstrating a relationship between breast cancer and the gut microbiome are summarized in Table 3.
Table 3.
Bacterial species possessing the β-glucuronidase enzyme for estrogen metabolism.
| Phylum | Genus | Species | Reference |
|---|---|---|---|
| Actinobacterium | Bifidobacterium | B. adolescentis | Nakamura et al.184 |
| B. breve | Nakamura et al.184; Stringer et al.185 | ||
| B. dentium | McIntosh et al.186 | ||
| B. longum | Nakamura et al.184 | ||
| Bacteroidetes | Bacteroides | B. dorei | McIntosh et al.186; Pollet et al.187 |
| B. fragilis | Pollet et al.187; Stringer et al.185 | ||
| B. ovatus | Gloux et al.188; Pollet et al.187 | ||
| B. uniformis | Nakamura et al.184 Pollet et al.187 |
||
| B. vulgatus | Nakamura et al.184 Pollet et al.187 |
||
| Parabacteroides | P. johnsonii | Gloux et al.188 | |
| P. merdae | Gloux et al.188; Pollet et al.187 | ||
| Firmicutes | Clostridium | C. acetobutylicum | Girbal et al.189 |
| C. asparagiforme | McIntosh et al.186 | ||
| C. bartlettii | Gloux et al.188 | ||
| C. beijerinckii | Girbal et al.189 | ||
| C. clostridioforme | Nakamura et al.184 | ||
| C. coccoides | Luu et al.164 | ||
| C. difficile | Mani et al.190 | ||
| C. hathewayi | McIntosh et al.186 | ||
| C. leptum | Luu et al.164 | ||
| C. paraputrificum | Nakamura et al.184 | ||
| C. perfringens | Hartman et al.191; Leung et al.192; McIntosh et al.186; Nakamura184; Pollet et al.187; Shimizu et al.193 | ||
| Escherichia | E. coli | Leung et al.192; McIntosh et al.186; Nakamura et al.184; Pollet et al.187 | |
| Eubacterium | E. eligens | McIntosh et al.186; Nakamura et al.184; Pollet et al.187 | |
| Faecalibacterium | F. prausnitzii | Dabek et al.194; Gloux et al.188; McIntosh et al.186; Pollet et al.187 | |
| Lactobacillus | L. gasseri | McIntosh et al.186; Russell et al.195 | |
| L. jensenii | McIntosh et al.186 | ||
| L. rhamnosus | McIntosh et al.186 | ||
| Roseburia | R. hominis | Dabek et al.194 | |
| R. intestinalis | Dabek et al.194; McIntosh et al.186 | ||
| R. inulinivorans | Gloux et al.188 | ||
| Ruminococcus | R. gnavus | Beaud et al.196; Gloux et al.188; McIntosh et al.186; Nakamura et al.184 |
Genera that stand out with potentially protective mechanisms against breast cancer include Faecalibacterium (phylum Firmicutes) and Akkermanisia (phylum Verrucomicrobiota). Genus Faecalibacterium has been identified in several breast cancer studies.154,163,164,171 A major commensal of the gut microbiome, F. prausnitzii is a producer of SCFA butyrate and shown to have anti-cancer effects in lung cancer in-vitro.172 Depletion of F. prausnitzii has been associated with Crohn’s disease, ulcerative colitis and colorectal cancer.173,174 In MCF-7 breast cancer cell lines, F. prausnitzii promoted apoptosis while inhibiting cell proliferation and invasion.171
Demonstrating promising probiotic and anti-inflammatory properties, A. muciniphila in feces has been associated with lower rates of metabolic syndrome and inflammation.175,176 It has been shown to increase antimicrobial peptides in the gut and thicken the mucosal layer to prevent bacterial translocation and subsequent systemic inflammation.177 A. mucinophilia is associated with gut microbial diversity in breast cancer patients115 and its abundance positively correlates with smaller breast tumors and node negative disease.170 Frugé et al. found that a higher abundance of A. muciniphila is associated with lower BMI and higher alpha diversity. It is also correlated with significant differences in beta diversity, higher abundance of Prevotella and Lactobacillus; reduced abundance of Clostridium, Campylobacter and Helicobacter, compared to samples with low abundance of A. muciniphila.115
Breast cancer patients have been found to have significantly elevated levels of Clostridiales in their feces, and high total urinary estrogen metabolite levels.154 While Clostridium species, like Faecalibacterium, also produce butyrate and together have been found depleted in the gut of lung cancer patients,178 fecal samples from breast cancer patients with higher staging were enriched in C. coccoides and C. leptum.164 Fuhrman et al. found that fecal levels of Clostridia were positively associated with metabolite-to-patient estrogen ratios, which in turn were positively associated with fecal microbial diversity.114 An earlier study reported a direct correlation between total urine estrogen metabolites, fecal α diversity and abundance of Clostridia.162 It may be the case that the pathogenic hormonal properties of Clostridium outweigh its protective effects in breast oncogenesis.
The gut estrabolome
The association between the gastrointestinal microbiome and breast cancer tumorigenesis is a current area of interest due to the significance of estrogen metabolizing gut bacteria, the genes of which have been referred to as the estrabolome.179 The estrabolome plays an important role in the enterohepatic recirculation of estrogens and systemic estrogen levels. This relationship is illustrated in Figure 2.
Figure 2.

Interactions between the gut microbiome, estrabolome and systemic immune system which may contribute to or modify breast cancer risk.
High estrogen states are a risk factor for breast cancer. Estrogen exposure can come from the systemic circulation or from the gut via the entero-mammary pathway. Estrogen metabolizing bacteria in the gut make up the estrabolome and are affected by various modifiable and non-modifiable risk factors. It interacts via the enterohepatic circulation to excrete or recycle estrogens back through the systemic circulation; on the other hand, gut dysbiosis can alter inflammatory immune cell activity and estrogen receptor signaling to promote neoplastic processes at the breast tumor microenvironment. Interventions to disrupt the complex interactions between the gut and breast microbiome and systemic circulation include antibiotics that alter the composition of β-glucuronidase containing bacteria in the gut and traditional breast cancer therapies.
Created with BioRender.com
Higher estrogen states are a known risk factor for ER positive breast cancer.180 Parent estrogens, estradiol, estrone, and their metabolites are conjugated with glucuronic acid in the liver to facilitate their excretion via urine or bile. Interestingly, higher concentrations of parent estrogens have been linked to increased post-menopausal breast cancer risk, whereas a higher metabolite to parent estrogen ratio is associated with reduced risk, particularly those metabolized via the 2-hydroxylation pathway.181–183
Deconjugation by β-glucuronidase containing bacteria, detailed in Table 4, facilitates the reabsorption of deconjugated estrogens back into the systemic circulation in their active forms. Notable bacteria with the β-glucuronidase enzyme to deconjugate estrogens for reabsorption include Alistipes, Bacteroides, Bifidobacterium, Collinsella, Edwardsiella, Faecalibacterium genera, and Lactobacillus and Roseburia.197 It has been hypothesized that those with a diverse gut microbiome rich in bacteria with deconjugating enzymes are at increased risk of breast cancer, due to enhanced enterohepatic reabsorption of estrogens and overall estrogen burden.179 Microbiome diversity and high β-glucuronidase activity in the gut have indeed been associated with higher concentrations of estrogen metabolites in urine, indicating higher systemic estrogen burden, and lower estrogen concentrations in feces.162,198 Microbial gut diversity is also positively correlated with a higher metabolite to parent estrogen ratio,114 and therefore a lower risk of post-menopausal breast cancer. Indeed, post-menopausal states appear to be associated with lower gut microbial diversity, a microbiome composition more similar to men and less gut β-glucuronidase activity, compared to pre-menopausal women.199 The feces of post-menopausal women with breast cancer have less microbial diversity compared to cancer-free controls, independent of estrogen.154 NAF from breast cancer patients showed different bacterial compositions to normal healthy controls and identified that the cancerous samples expressed higher concentrations of β-glucuronidase.30
Table 4.
Summary of significant bacterial species identified demonstrating a relationship between the gut microbiome and breast cancer.
| Significant Bacteria Identified (Phylum – Genus) |
Study Type | Breast Cancer Study Size | Sample Type | Method of Analysis | Reference |
|---|---|---|---|---|---|
|
Firmicutes: Clostridia |
Cross-sectional | 51 BC (women 19 pre, 7 postmenopausal, 25 men) | Urine Faeces |
LC-MS/MS Realtime kinetics 16S rRNA amplicon pyrosequencing |
Flores et al.162 |
|
Bacteroidetes: −Bacteroides Firmicutes: −Clostridium −Ruminococcus |
Cross-sectional | 60 health postmenopausal women (ages 55–69) | Urine Faeces |
LC-MS/MS 16S rRNA amplicon sequencing |
Fuhrman et al. 114 |
|
Actinobacteria: −Eggerthella −Actinobacteria −Bifidobacterium Firmicutes: −Faecalibacterium spp. prausnitzii −Blautia |
Cross-sectional | 32 BC patients; IDC (81%), ER+/PR+ (80%), HER2+ (15%) |
Faeces | 16S rRNA PCR gene sequencing | Bard et al.163 |
|
Firmicutes: −Clostridium −Faecalibactrium −Ruminococcus −Lachnospiraceae −Dorea |
Case-control | 48 BC postmenopausal pre-treatment 48 Control postmenopausal normal mammograms |
Urine Faeces |
16S rRNA gene sequencing LC-MS/MS |
Goedert et al. 154 |
|
Actinobacteria:Eggerthella Firmicutes: −Bacteroidetes −Blautia −Clostridium −Faecalibacterium |
Cross-sectional | 31 early-stage BC | Faeces | 16S rRNA gene sequencing DNA extraction (PSP Spin Stool DNA kit) |
Luu et al.164 |
|
Actinobacteria:Actinomyces Firmicutes: Enterococcus Proteobacteria: Acinetobacter Citrobacter Erwinia Escherichia Shewanella |
Population based cohort | 48 postmenopausal BC (75% stage 0-I; 88% ER+) 48 Control age-matched postmenopausal with normal mammograms |
Urine Faeces |
16S rRNA gene amplicon sequencing high-performance LC-MS/MS radioimmunoassay |
Goedert et al.165 |
|
Bacteroidetes:Bacteroides Proteobacteria −Pseudomonas −Escherichia Firmicutes: −Clostridium −Staphylococcus |
Case-control | 46 postmenopausal BC 48 Control postmenopausal 28 mice injected with 4T1 cancer cells. MCF-7, 4T1 and primary fibroblast cell lines |
Human feces Mice mammary tissue |
Cell proliferation assays PI assays Scratch assay ECIS, qPCR SDS-PAGE, IHC Western blot |
Mikó et al.166 |
|
Actinobacteria − Actinomyces Bacteroidetes: − Prevotella Firmicutes − Enterococcus Proteobacteria: − Escherichia − Klebsiella − Shewanella − Erwinia |
Case-control | 18 premenopausal BC 44 postmenopausal BC 25 premenopausal controls 46 postmenopausal controls |
Faeces | Shotgun metagenomics | Zhu et al.119 |
|
Bacteroidetes Actinobacteria: Bifidobacterium |
Cross-sectional | 124 invasive BC no chemotherapy currently (46% previous chemo) | Blood Faeces |
16S rRNA gene sequencing Gas chromatography |
Horigome et al.167 |
|
Bacteroidetes – Firmicutes:Anaeotruncus |
Cross-sectional | Mice -mammary injections of PyMT-BO1 and EO771 cell lines, antibiotic cocktail (VNMA) & ampicillin Control: water-treated |
Faeces | 16S rRNA amplicon PCR gene sequencing Flow cytometry NMR spectroscopy |
Kirkup et al.168 |
| Proteobacteria:Escherichia | Case-control | 32 female mice grafted with 4T1 BC cells (16 cadaverine treated, 16 control) Cell lines: MD-MBA-231, SK-BR-3, ZR-75–1, MCF-7 & 4T1 48 postmenopausal BC Control: 48 healthy women |
Murine Mammary tissue Human feces |
Cell proliferation assays Annexin V + PI ECIS assays, IHC RT-qPCR SDS PAGE & Western blot Wound healing assay Matrigel invasion assay TBARS assay Aldefluor assay |
Kovács et al.127 |
|
Proteobacteria: − Citrobacter − Veillonella |
Cross-sectional | 121 BC | Faeces | Metagenomics | Yoon et al.169 |
| Verrucomicrobia:Akkermansia | Longitudinal | 32 BC patients (stage 0 – II, BMI 25; | Faeces | 16S rRNA gene sequencing | Yoon et al.115 |
|
Bacteroidetes: −Verrucomicrobia Actinobacteria Firmicutes: −Faecalibacterium Proteobacteria |
Case-control | 25 BC 25 benign breast disease MCF-7 BC cell line |
Faeces Blood |
16S rDNA sequencing LC-MS metabolomics Chromatography-MS Cell and bacterial culture ELISA, Western blot Cell proliferation, Apoptosis & invasion assays |
Ma et al.40 |
|
Actinobacteria:Collinsella Bacteroidetes:Bacteroides Firmicutes: −Clostridium −Eubacterium Verrucomicrobia:Akkermansia |
Case control |
Cohort 1: 76 pre-chemo– 42% HR+, 34% HER2+, 24% 45 TNBC- pre & post-chemo 54 Ctls: healthy volunteers Cohort 2: 27 BC (17 pre-chemo, 10 post chemo) |
Faeces | Shotgun metagenomics | Terrisse et al.170 |
Annexin V + PI : Annexin V and propidium iodide (PI) labeling of cells is a technique used to identify cell death; BC: breast cancer; Chemo: chemotherapy; ECIS: Electric Cell-substrate Impedance Sensing; ER+: estrogen receptor positive; ELISA: enzyme-linked immunosorbent assay; HER2+: human epithelial growth factor receptor positive; HR+: hormone receptor positive; IDC: invasive ductal carcinoma; IHC: Immunohistochemistry; LC-MS/MS: Liquid chromatography/tandem mass spectrometry; MS: mass spectrometry; NMR: nuclear magnetic resonance spectroscopy; NSCLC: non-small cell lung carcinoma; PCR: polymerase chain reaction; PR+: progesterone receptor positive; RT-qPCR: reverse transcription polymerase chain reaction; TNBC: triple negative breast cancer.
Entero-mammary pathway
The ‘bacterial entero-mammary pathway’, a proposed mechanism by which mononuclear immune cells (i.e. dendritic and CD18 expressing cells) migrate from the GIT to the breast, may be a carrier for gut bacteria to translocate to mammary tissue.200 Well established is the mucosal immune system for the migration of IgA cells between mucosal surfaces, as previously described.
The composition of the breast microbiome may be directly influenced by intestinal bacteria via this pathway and may explain how intestinal bacteria such a Enterobacteriaceae, a large family of gram negative bacteria often found in the gut microbiome, have been found in the breast TME.25,26 Oral ingestion of probiotic Lactobacillus strains (L. fermentum or L. salivarius) to treat mastitis led to isolation of those strains in milk samples, suggesting that there is a mechanism by which gut microorganisms are able to migrate from the gut to mammary tissue.201,202 Detected in human breast milk, L. salivarius and L. gasseri, were shown to cross through a Caco-2 cell monolayer (an in vitro representation of the intestinal epithelial layer) via dendritic cells.203 From the breastmilk of healthy lactating women, bacteria such as Streptococcus, Lactobacillus, Enterococcus, Peptostreptococcus, Staphylococcus, Corynebacterium and occasional Escherichia spp., have been isolated.204
The presence of enteric bacteria in milk, taken with the finding of some rDNA bands common to maternal milk, blood and feces, suggests bacterial translocation via a mononuclear-mediated mechanism.204 In-vivo studies have shown L. reuteri suppresses tumor formation when introduced to the GIT of breast cancer-prone mice with HER2 mutations, by stimulating CD4+ and CD25+ lymphocytes.205 16S rRNA gene sequencing of fecal samples from women with breast cancer detected higher counts of Clostridia and Blautia sp., with increasing grade and stage.163,164 The interplay between enteric microorganisms and the human immune system in the promotion and inhibition of carcinogenesis may be of clinical significance. It has potential as a target for primary prevention of breast cancer, via manipulation of modifiable risk factors, such as diet and antibiotic use.
Estrogen modification
The host’s immune cells are also influenced by the estrabolome. ER signaling has a significant influence on the activity of the innate and adaptive immune systems.206,207 Importantly, 17β-estradiol can cause epigenetic changes and gene transcription by complexing at DNA sites and transcription factors, especially those related to immune cell function.208,209 Proteins ER-α and ER-β, encoded by the ESR1 and ESR2 gene, respectively, are expressed on a variety of innate and adaptive immune cells, including CD4+ and CD8+ Tcells, B-cells, monocytes, NK cells and monocyte-derived dendritic cells, affecting immune cell growth and function.207,210 There is variable ER expression between different cell types with B cells found to have the highest expression of ESR1 and ESR2 RNA, together with plasmacytoid dendritic cells for the latter, while other T cells such as CD4+ and CD8+, monocytes and NK cells have positive but low or moderate expression of those genes.210,211 Estrogens can induce histone modifications affecting processes such as acetylation, phosphorylation and methylation,209 altering the gene expression of thousands of estrogen responsive elements.
Lower physiological estradiol concentrations are usually associated with the production of type 1 IFN, which regulates inflammatory pathways, and hence the production of pro-inflammatory cytokines.212,213 In contrast, higher physiological estrogen levels and ectopic estrogens are generally associated with inhibition of inflammatory pathways,214–216 Thus, in pathological states such as cancer, a TME with a higher local concentration of estrogen would suppress the production of inflammatory cytokines, dampen the immune response and promote the neoplastic process.
Oral antibiotics
Oral antibiotics may be able to modify breast cancer risk via manipulation of the gut estrabolome. Certain antibiotics have been shown to alter the levels of specific bacteria involved in estrogen metabolism, such as Lactobacillus and Bacteroides. Changes in the gut microbiome can lead to increased levels of harmful metabolites or decreased levels of protective ones, thus impacting overall estrogen balance in the body. A large systematic review found that antibiotic exposure, particularly to penicillins, tetracyclines and nitrofurans, moderately increased breast cancer risk.217 Frequent antibiotic use in breast cancer survivors has been associated with an increased risk of second breast cancer events, although without clinically significant results.218 The net effect may be that of increased chronic inflammation and disturbed tissue metabolism.219 There are classes of antibiotics with anti-cancer activity, notably ciprofloxacin and gemifloxacin in TNBC and breast adenocarcinoma, respectively, in vitro.220,221 Incubation of human MDA-MB-231 breast cancer cells with increasing concentrations of ciprofloxacin demonstrated dose and time-dependent cytotoxicity while increasing concentrations of gemifloxacin progressively decreased migration and invasion of MDA-MB-231 and MDA-MB-453 cells.220,221
In contrast, multiple large cohort studies reported a weak relationship between long-term antibiotic use and breast cancer risk.222–226 Numerous studies have reported no association between antibiotic use and breast cancer risk.227–229 Moreover, Zackular et al.230 demonstrated antibiotic therapy to decrease tumor growth by reducing microbial diversity in mice.230
Microbial-based therapy
The gut microbiome has proven to be malleable and dynamic. It can be hypothesized that manipulation of the gut microbiome may be a method of modifying breast cancer risk and optimizing responsiveness to systemic therapies such as chemotherapy or immunotherapy for high-risk disease.
Chemotherapy
Fecal composition may provide crucial information about potential biomarkers for prognosis or drug resistance and may play a role in guiding chemotherapy selection. Higher levels of Blautia obeum in the gut may be protective against colorectal cancer risk231 and is associated with significantly better progression free and overall survival in hepatocellular carcinoma.232 However, genus Slackia (genus of Actinomycetota, in the family Coriobacteriaceae), linked to early colorectal cancer and gastric cancer progression233,234 is associated to poorer survival outcomes. This correlation was found as well in a small study in patients with metastatic HER2 negative breast cancer undergoing capecitabine chemotherapy, reporting longer progression free survival in those with higher fecal levels of Blautia obeum and shorter in those with higher levels of Slackia.235 Of note, it was found that the gut microbiome composition was drastically altered by metronomic capecitabine compared to the routine dose, demonstrating significant differences in fecal microbial composition, function and reduced diversity.235 Species from the family Enterobacteriaceae are shown to inactivate doxorubicin; Klebisella pneumoniae, Escherichia coli, Raoultella planticola.236 Lower levels of A. muciniphila is associated with taxane-induced systemic inflammation and neuropathic pain.237 Abundance of A. muciniphila in the gut of mice injected with TNBC cell line 4T1 has been associated with responsiveness to doxorubicin, while administration of LPS, a component of gram negative bacteria, increased intestinal inflammation and reduced response to doxorubicin.238
Cyclophosphamide has been shown to promote inflammation in mice with breast tumors by disturbing the gut epithelium barrier, allowing the migration of Lactobacillus johnsonii, Lactobacillus murinus and Enterococcus hirae to lymph nodes and triggering production of Th17 and Th1 cells.239 Germ-free and antibiotic-treated mice demonstrated resistance to cyclophosphamide,239 while oral administration of E. hirae improved response to chemotherapy.239,240
Radiotherapy
A gut microbial composition that reduces radiotherapy-related side effects may improve treatment outcomes. The gut microbiome has been revealed to have a role in radiotherapy efficacy and toxicity in the treatment of breast cancer.117 The degree of radiosensitivity may be determined by the immune response of the TME, with evidence that it is influenced by local microbial activity and metabolism.241 Macrophages can be classified as pro-inflammatory (M1) or anti-inflammatory (M2).242 A greater abundance of M1-polarized macrophages has been shown to increase the efficacy of radiotherapy on breast cancer cells in vitro, while M2-polarized macrophages promoted radio-resistance. Rectal microbial diversity has been shown to be predictive for responsiveness to radiotherapy in the treatment of cervical squamous cells carcinoma.243 Ovarian tumors with high infiltration by CD8+ T cells and M1 macrophages had a better prognosis, and a unique intratumoral microbial signature enriched in Achromobacter, Microcella, Devosia, Ancylobacter and Acinetobacter.244
Additionally, gut abundance of Lactobacillus sakei, L. acidophilus, L. casei, L. reuteri and Bifidobacterium spp. have been shown to be protective against radiation-induced enteritis.245–247
Immunotherapy
There is growing evidence that the gut microbiota is involved in the clinical response to immune checkpoint inhibitors (ICIs). Vétizou et al.248 found Bacteroides species, particularly B. thetaiotaomicron and B. fragilis, to have a role in improving the therapeutic efficacy of anti-CTLA-4 ICIs, for the treatment of melanoma and colon cancer in mice.248 On the other hand, the immunosuppressive effect of B. fragilis has been described earlier (section Microbial Toxins). Of note, fecal Bacteroidetes numbers are reported to increase with increasing stage of breast cancer,164 making it a potential target to maximize efficacy of immunotherapy.
Bifidobacterium alleviates the symptoms of anti-CTLA-4 ICI-induced colitis in melanoma-afflicted mice.249 Gopalakrishnan et al.250 found the gut microbiome modulates a response to anti-PD-1 immunotherapy in melanoma patients through enhancing antigen presentation or increasing T cell recruitment to the TME. Responders exhibited a more diverse fecal microbiome with enrichment in Ruminococcaceae, Clostridiales and Faecalibacterium.250 The probiotics Bifidobacterium spp. and A. muciniphila were demonstrated to promote the efficacy of anti-PD-1 therapy against epithelial tumors.251,252
In more recent years, the utility of ICIs in treatment of breast cancer is expanding, demonstrating improved clinical outcomes. A systemic review indicated that specific gut signatures may be predictive of a good response to immunotherapy. For example, patients with a baseline gut abundance of Bacteroides fragilis, Streptococcus and F. prausnitzii had better clinical responses to hormone therapy and pembrolizumab.253 Characterizing the immune profile of the TME will expand the potential of immunotherapy in breast cancer treatment.
Targeted therapy
Clostridiales appears to be significant in the treatment of HER2 positive breast cancers, noting a greater abundance of Clostridiales and less Bacteroidales in the feces of patients who achieved pathological complete response with neoadjuvant trastuzumab.254 The proposed mechanism is that commensal gut bacteria may enhance the response to monoclonal antibody therapy via greater recruitment of immune cells to the TME. Trastuzumab was also more efficacious in the presence of L. lactis or L. paracasei.254
Hormone therapy
The effect of the gut microbiome on hormone therapy efficacy has been observed in hormone dependent cancers. In prostate cancer, dysbiosis involving androgen-producing microbes are able to degrade androgen deprivation therapy.255,256 In ER positive breast cancers, differences were noted in the gut microbial composition of good responders to adjuvant aromatase inhibitor therapy compared to poor responders, with poor responders demonstrating higher gut microbiome diversity and elevated levels of Veillonella.257 Veillonella (phylum Firmicutes) in the gut has been associated with poor prognosis in hematology patients treated with anti-CD19 chimeric antigen receptor (CAR) T cell therapy, and in tumor tissue associated with poor prognosis in lung cancer.258,259
A study of the fecal microbiome of HR positive breast cancer patients reported several bacteria that may be clinically relevant. Fecal enrichment with several members of the family Clostridiaceae was associated with node positive disease and resistance to ICIs in breast cancer patients. On the other hand, Eubacterium rectale, Methanobrevibacter smithii, Coprococcus comes, Coprococcus catus and Collensella aerofaciens were associated with better prognosis; Eubacterium rectale, Eubacterium eligens, Eubacterium ventriosum and Collinsella aerofaciens were shown to inhibit the growth of breast cancer cells in vitro.170
Faecal microbial transplants
Specialized diets, bacterial probiotic “crapsules” or FMTs are hypothesized to nurture a ‘favourable’ gut microbiome, which may have a role in breast cancer treatment or even primary prevention.260,261
Implantation of a favorable microbiome may be the mechanism by which FMTs are able to enhance the immune response. Giving FMTs from epithelial cancer patients who responded well to ICIs to germ free or antibiotic-treated mice facilitated anti-PD-L1 efficacy and increased relative abundance of A. muciniphila.251 A similar study was replicated in renal cell carcinoma patients, whereby feces from good responders to immunotherapy were abundant in A. muciniphila and B. salyersiae, and FMTs given to antibiotic-treated mice induced significant responses to immunotherapy compared to FMTs from non-responders (100% compared to 40%, respectively).262 Melanoma studies have linked an abundance of A. muciniphila, Bifidobacterium longum and Enterococcus faecium with good clinical response to anti-PD1 therapy.250,263 Both oral supplementation of bacteria and FMTs from human responders given to antibiotic-treated or germ-free mice improved response to anti-PD-L1 therapy.250,263
FMTs from breast cancer patients to mice inoculated with AT3 breast cancer cells led to poor response to chemotherapy, contrasting to outcomes in mice given FMTs sourced from healthy volunteers.170 Furthermore, cohabitating both groups of mice improved the effects of chemotherapy on mice treated with FMTs from breast cancer patients. The FMTs from healthy volunteers had more abundance of Eubacterium rectale, Methanobrevibacter smithii, Coprococcus comes and C. catus. This study included patients with a mix of breast cancer subtypes, and subgroup analysis based on HR and HER2 status was not done. This could be important to flesh out in future studies, given the effect the gut microbiome may have on systemic estrogens and immune cells.
The efficacy of trastuzumab in mice with HER2 positive breast tumors was impaired by the administration of antibiotics, an effect that was reversed by FMTs from trastuzumab responsive mice to non-responsive mice.254 Treatment responsive mice had higher serum levels of IL-12, a Th1 cytokine which induces effector T and NK cells, after trastuzumab exposure. The immune-mediated effect of antibiotics on trastuzumab efficacy was supported by the significant decrease in intra-tumoral CD4+ T cells in antibiotic-treated mice. Deceased levels of Lachnospiraceae, Actinobacteria, Turicibacteraceae and Bacteroidetes were found in antibiotic-treated mice, a result which was also found in human patients with HER2 positive breast cancer who were poor responders of trastuzumab in the neoadjuvant setting. While this was a small study of only 24 human patients, the significance of these findings may be of clinical relevance, particularly the importance of Lachnospiraceae and Bacteroidetes which are also less abundant in poor responders of anti-PD1 melanoma treatment.250
Extracellular vesicles
Extracellular vesicles (EVs) are formed when cells break off some of their intracellular material proteins (lipids, genetic material or sugars) and package it in its own cellular membrane. Once independent from the parent-bacteria, EVs can communicate inter-cellularly by macropinocytosis, endocytosis or membrane fusion.264 EVs can transport bacterial intracellular contents around the body in body fluids, becoming a vehicle for communication between different cells and organs.265
Bacteria produce EVs, both in pathological and physiological conditions.265 In the case of pathogenic EVs, as the EV membrane is taken from its parent-bacteria, they also present pathogen-associated molecular patterns and hence can be detected by the body’s immune cells via pattern recognition receptors. This can result in an inflammatory response by induction of the innate and adaptive immune responses.266–270
Intestinal bacterial EVs are able to cross the gut epithelium, interact with local immune cells, as well as disseminate systemically via blood, lymph or the hepatobiliary system,271,272 with or without a compromised gut epithelium.273 Patients with chemotherapy-induced colonic mucositis, HIV or inflammatory bowel disease were found to have higher concentrations of serum EVs,274 likely due to increased gut permeability from a compromised intestinal barrier.275 A study of diabetic patients reported lower serum levels of EVs derived from A. muciniphila, a protective commensal of the gut epithelium.276 Meanwhile, feeding mice EVs derived from A. muciniphila was found to decrease gut permeability.276
A review of studies on the utility of EVs as a biomarker for breast cancer found that while serum levels of EVs were consistently elevated, it was not associated with stage or subtype.277 EVs from metastatic breast cancer cells in vitro increased cell migration and invasion.278,279 Their contents, which have been found to contain oncogenic proteins including Epidermal Growth Factor Receptor and miRNAs, have been suggested as an alternative and more specific biomarker for breast cancer.280–282 Elevated levels of the HER2 protein have been found in EVs produced by breast cancer cells lines expressing the HER2 encoding gene ERBB2.283 Additionally, breast cancer cells resistant to chemotherapy have been demonstrated to be able to transfer their treatment resistance to sensitive cancer cells via EVs.284–286 Hence, peripheral EVs could be a novel strategy to both monitor and promote treatment efficacy.
In recent years, there has been growing interest in the use of EVs as a therapeutic vehicle, mainly in the context of vaccination. An avenue of microbial-based therapy being explored in the field of oncology is FMT, aiming to colonize the gut with favorable bacteria.287 Administration of bacterial EVs could potentially be a better and safer alternative, as they have greater stability across temperatures and do not replicate in vivo while still sharing the same immunogenic membrane and contents derived from its parent-bacteria.288 Notably, the binding of B. fragilis to TLR2 promotes Treg differentiation105 and administration of B. fragilis-derived EVs in mice mimic this same effect.289 Administration of modified E. coli EVs has been shown to stimulate production of CXCL10 and IFN-γ, thus inducing an anti-tumoral immune response.290 There is in vitro evidence of the benefit of probiotic bacterial EVs in treating hepatocellular cancer. EVs from L. rhamnosus was shown to have a cytotoxic effect on the HepG2 liver cancer cell line.291 Incubation of MCF-7 and BT474 breast cancer cell lines with β-galactosidase producing K. pneumoniae and S. aureus-derived EVs enhanced tamoxifen efficacy, via downregulation of cyclin E2, p-ERK and p21.292,293 Similarly, in TBC cell lines MDA-MB-231 and 4T1, bacterial EVs from E. coli have been demonstrated to inhibit tumor cell growth, invasion and migration, decrease Treg cells and promote apoptosis both in in vivo murine models.294–296 Hence, bacterial EVs may have utility as an adjunct to immunotherapeutic and chemotherapy agents.
Conclusion
There is a highly complex dynamic interaction between the gut microbiome, the immune system, estrabolome and the breast TME. Future studies should aim to establish stronger evidence through mechanistic studies to extend our understanding beyond that of the current associations.
The evidence in the literature suggests that both the breast and gut microbiota play a significant role in modulating breast tissue homeostasis. While many genera and species have been named as potentially significant, some of which have associations with other cancers or other disease states, causative links to breast cancer are still largely unknown or poorly defined. There is potential for the application of certain microbial signatures as biomarkers of disease. Species highlighted in this review that may be of particular interest for future studies are M. radiotolerans and S. yanoikuyae in breast tissue, and A. muciniphila in the gut. The microbiome can affect drug responsiveness to systemic chemotherapy, radiotherapy and hormonal therapies as well as modulate the immune system. Elucidating the mechanisms could have massive clinical implications on the future prevention, screening, treatment, and prognostication of breast cancer.
Funding Statement
The author(s) reported that there is no funding associated with the work featured in this article.
Disclosure statement
No potential conflict of interest was reported by the author(s).
References
- 1.Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, Franceschi S.. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Global Health. 2016;4(9):e609–33. doi: 10.1016/S2214-109X(16)30143-7. [DOI] [PubMed] [Google Scholar]
- 2.Fernández MF, Reina-Pérez I, Astorga JM, Rodríguez-Carrillo A, Plaza-Díaz J, Fontana L.. Breast cancer and its relationship with the microbiota. Int J Environ Res Pub Health Public Health. 2018;15(8):1747. doi: 10.3390/ijerph15081747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Banerjee S, Wei Z, Tian T, Bose D, Shih NN, Feldman MD, Khoury T, De Michele A, Robertson ES. Prognostic correlations with the microbiome of breast cancer subtypes. Cell Death Dis. 2021;12(9):831. doi: 10.1038/s41419-021-04092-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Qin W, Li J, Gao N, Kong X, Guo L, Chen Y, Huang L, Chen X, Qi F. Multiomics-based molecular subtyping based on the commensal microbiome predicts molecular characteristics and the therapeutic response in breast cancer. Mol Cancer. 2024;23(1):99. doi: 10.1186/s12943-024-02017-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Van Nhieu JT, Furet JP. Microbial dysbiosis in colorectal cancer (CRC) patients. PLOS ONE. 2011;6(1):e16393. doi: 10.1371/journal.pone.0016393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Xuan C, Shamonki JM, Chung A, DiNome ML, Chung M, Sieling PA, Lee DJ, Takabe K. Microbial dysbiosis is associated with human breast cancer. PLOS ONE. 2014;9(1):e83744. doi: 10.1371/journal.pone.0083744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhuang H, Cheng L, Wang Y, Zhang Y-K, Zhao M-F, Liang G-D, Zhang M-C, Li Y-G, Zhao J-B, Gao Y-N, et al. Dysbiosis of the gut microbiome in lung cancer. Front Cell Infect Microbiol. 2019;9:112. doi: 10.3389/fcimb.2019.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wilkins LJ, Monga M, Miller AW. Defining dysbiosis for a cluster of chronic diseases. Sci Rep. 2019;9(1):1–10. doi: 10.1038/s41598-019-49452-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi KE, Yu G-Y, Österreicher CH, Hung KE, et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 2012;491(7423):254–258. doi: 10.1038/nature11465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Stappenbeck TS, Hooper LV, Gordon JI. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci USA. 2002;99(24):15451–15455. doi: 10.1073/pnas.202604299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J For Clinicians. 2024;74(3):229–263. doi: 10.3322/caac.21834. [DOI] [PubMed] [Google Scholar]
- 12.Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikšić M, Bonaventure A, Valkov M, Johnson CJ, Estève J, et al. Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet. 2018;391(10125):1023–1075. doi: 10.1016/S0140-6736(17)33326-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sundquist M, Brudin L, Tejler G. Improved survival in metastatic breast cancer 1985–2016. Breast. 2017;31:46–50. doi: 10.1016/j.breast.2016.10.005. [DOI] [PubMed] [Google Scholar]
- 14.Jackisch C, Harbeck N, Huober J, von Minckwitz G, Gerber B, Kreipe H-H, Liedtke C, Marschner N, Möbus V, Scheithauer H, et al. 14th St. Gallen international breast cancer conference 2015: evidence, controversies, consensus-primary therapy of early breast cancer: opinions expressed by German experts. Breast Care. 2015;10(3):211–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Perou CM, Børresen-Dale A-L. Systems biology and genomics of breast cancer. Cold S. Harbor Perspectives In Biol. 2011;3(2):a003293. doi: 10.1101/cshperspect.a003293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Beckers RK, Selinger CI, Vilain R, Madore J, Wilmott JS, Harvey K, Holliday A, Cooper CL, Robbins E, Gillett D, et al. Programmed death ligand 1 expression in triple‐negative breast cancer is associated with tumour‐infiltrating lymphocytes and improved outcome. Histopathology. 2016;69(1):25–34. doi: 10.1111/his.12904. [DOI] [PubMed] [Google Scholar]
- 17.Coughlin SS. Epidemiology of breast cancer in women. Breast Cancer Metastasis And Drug Resist. 2019;1152:9–29. [DOI] [PubMed] [Google Scholar]
- 18.Gray JM, Rasanayagam S, Engel C, Rizzo J. State of the evidence 2017: an update on the connection between breast cancer and the environment. Environ Health. 2017;16(1):1–61. doi: 10.1186/s12940-017-0287-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Alpuim Costa D, Nobre JG, Batista MV, Ribeiro C, Calle C, Cortes A, Marhold M, Negreiros I, Borralho P, Brito M. Human microbiota and breast cancer—is there any relevant link?—a literature review and new horizons toward personalised medicine. Front Microbiol. 2021;12:584332. doi: 10.3389/fmicb.2021.584332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Thompson KJ, Ingle JN, Tang X, Chia N, Jeraldo PR, Walther-Antonio MR, Kandimalla KK, Johnson S, Yao JZ, Harrington SC, et al. A comprehensive analysis of breast cancer microbiota and host gene expression. PLOS ONE. 2017;12(11):e0188873. doi: 10.1371/journal.pone.0188873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Banerjee S, Tian T, Wei Z, Shih N, Feldman MD, Peck KN, DeMichele AM, Alwine JC, Robertson ES. Distinct microbial signatures associated with different breast cancer types. Front Microbiol. 2018;9:951. doi: 10.3389/fmicb.2018.00951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Costantini L, Magno S, Albanese D, Donati C, Molinari R, Filippone A, Masetti R, Merendino N. Characterization of human breast tissue microbiota from core needle biopsies through the analysis of multi hypervariable 16S-rRNA gene regions. Sci Rep. 2018;8(1):1–9. doi: 10.1038/s41598-018-35329-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hieken TJ, Chen J, Hoskin TL, Walther-Antonio M, Johnson S, Ramaker S, Xiao J, Radisky DC, Knutson KL, Kalari KR, et al. The microbiome of aseptically collected human breast tissue in benign and malignant disease. Sci Rep. 2016;6(1):30751. doi: 10.1038/srep30751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tzeng A, Sangwan N, Jia M, Liu C-C, Keslar KS, Downs-Kelly E, Fairchild RL, Al-Hilli Z, Grobmyer SR, Eng C. Human breast microbiome correlates with prognostic features and immunological signatures in breast cancer. Genome Med. 2021;13(1):1–17. doi: 10.1186/s13073-021-00874-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Urbaniak C, Cummins J, Brackstone M, Macklaim JM, Gloor GB, Baban CK, Scott L, O’Hanlon DM, Burton JP, Francis KP, et al. Microbiota of human breast tissue. Appl Environ Microbiol. 2014;80(10):3007–3014. doi: 10.1128/AEM.00242-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Urbaniak C, Gloor GB, Brackstone M, Scott L, Tangney M, Reid G, Goodrich-Blair H. The microbiota of breast tissue and its association with breast cancer. Appl Environ Microbiol. 2016;82(16):5039–5048. doi: 10.1128/AEM.01235-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.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. 2020;368(6494):973–980. doi: 10.1126/science.aay9189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Meng S, Chen B, Yang J, Wang J, Zhu D, Meng Q, Zhang L. Study of microbiomes in aseptically collected samples of human breast tissue using needle biopsy and the potential role of in situ tissue microbiomes for promoting malignancy. Front Oncol. 2018;8:318. doi: 10.3389/fonc.2018.00318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Banerjee S, Wei Z, Tan F, Peck KN, Shih N, Feldman M, Rebbeck TR, Alwine JC, Robertson ES. Distinct microbiological signatures associated with triple negative breast cancer. Sci Rep. 2015;5(1):15162. doi: 10.1038/srep15162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chan AA, Bashir M, Rivas MN, Duvall K, Sieling PA, Pieber TR, Vaishampayan PA, Love SM, Lee DJ. Characterization of the microbiome of nipple aspirate fluid of breast cancer survivors. Sci Rep. 2016;6(1):1–11. doi: 10.1038/srep28061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yazdi HR, Movafagh A, Fallah F, Alizadeh Shargh S, Mansouri N, Heidary Pour A, Hashemi M. Evaluation of Methylobacterium radiotolerance and sphyngomonas yanoikoaie in sentinel lymph nodes of breast cancer cases. Asian Pac J Cancer Prev. 2016;17(sup3):279–285. doi: 10.7314/APJCP.2016.17.S3.279. [DOI] [PubMed] [Google Scholar]
- 32.Wang H, Altemus J, Niazi F, Green H, Calhoun BC, Sturgis C, Grobmyer SR, Eng C. Breast tissue, oral and urinary microbiomes in breast cancer. Oncotarget. 2017;8(50):88122. doi: 10.18632/oncotarget.21490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chiba A, Bawaneh A, Velazquez C, Clear KY, 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. Molecular Cancer Res. 2020;18(1):130–139. doi: 10.1158/1541-7786.MCR-19-0451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Thyagarajan S, Zhang Y, Thapa S, Allen MS, Phillips N, Chaudhary P, Kashyap MV, Vishwanatha JK. Comparative analysis of racial differences in breast tumor microbiome. Sci Rep. 2020;10(1):14116. doi: 10.1038/s41598-020-71102-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Allen-Vercoe E, Jobin C. Fusobacterium and Enterobacteriaceae: important players for CRC? Immunol Lett. 2014;162(2):54–61. doi: 10.1016/j.imlet.2014.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hessle C, Andersson B, Wold AE, Moore RN. Gram-positive bacteria are potent inducers of monocytic interleukin-12 (IL-12) while gram-negative bacteria preferentially stimulate IL-10 production. Infect Immun. 2000;68(6):3581–3586. doi: 10.1128/IAI.68.6.3581-3586.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.García-Tuñón I, Ricote M, Ruiz A, Fraile B, Paniagua R, Royuela M. Influence of IFN-gamma and its receptors in human breast cancer. BMC Cancer. 2007;7(1):1–11. doi: 10.1186/1471-2407-7-158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zaidi MR, Merlino G. The two faces of interferon-γ in cancer. Clin Cancer Res. 2011;17(19):6118–6124. doi: 10.1158/1078-0432.CCR-11-0482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Peng R, Liu S, You W, Huang Y, Hu C, Gao Y, Jia X, Li G, Xu Z, Chen Y. Gastric microbiome alterations are associated with decreased CD8+ tissue-resident memory T cells in the tumor microenvironment of gastric cancer. Cancer Immunol Res. 2022;10(10):1224–1240. doi: 10.1158/2326-6066.CIR-22-0107. [DOI] [PubMed] [Google Scholar]
- 40.Ma J, Sun L, Liu Y, Ren H, Shen Y, Bi F, Zhang T, Wang X. Alter between gut bacteria and blood metabolites and the anti-tumor effects of faecalibacterium prausnitzii in breast cancer. BMC Microbiol. 2020;20(1):1–19. doi: 10.1186/s12866-020-01739-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kutschera U. Plant-associated methylobacteria as co-evolved phytosymbionts: a hypothesis. Plant Signaling & Behav. 2007;2(2):74–78. doi: 10.4161/psb.2.2.4073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pollock C, Koltai H, Kapulnik Y, Prandi C, Yarden R. Strigolactones: a novel class of phytohormones that inhibit the growth and survival of breast cancer cells and breast cancer stem-like enriched mammosphere cells. Breast Cancer Res Treat. 2012;134(3):1041–1055. doi: 10.1007/s10549-012-1992-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Carrega P, Bonaccorsi I, Di Carlo E, Morandi B, Paul P, Rizzello V, Cipollone G, Navarra G, Mingari MC, Moretta L, et al. CD56brightperforinlow noncytotoxic human NK cells are abundant in both healthy and neoplastic solid tissues and recirculate to secondary lymphoid organs via afferent lymph. J Immunol. 2014;192(8):3805–3815. doi: 10.4049/jimmunol.1301889. [DOI] [PubMed] [Google Scholar]
- 44.Kassayova M, Bobrov N, Strojný L, Kiskova T, Mikeš J, Demečková V, Orendáš P, Bojková B, Péč M, Kubatka P, et al. Preventive effects of probiotic bacteria Lactobacillus plantarum and dietary fiber in chemically-induced mammary carcinogenesis. Anticancer Res. 2014;34(9):4969–4975. [PubMed] [Google Scholar]
- 45.Kosaka A, Yan H, Ohashi S, Gotoh Y, Sato A, Tsutsui H, Kaisho T. Lactococcus lactis subsp. cremoris FC triggers IFN-γ production from NK and T cells via IL-12 and IL-18. Intl Immunopharmacol. 2012;14(4):729–733. doi: 10.1016/j.intimp.2012.10.007. [DOI] [PubMed] [Google Scholar]
- 46.Aragón F, Carino S, Perdigón G, de Moreno de leblanc A. Inhibition of growth and metastasis of breast cancer in mice by milk fermented with Lactobacillus casei CRL 431. J Appl Psychol Immunotherapy. 2015;38(5):185–196. doi: 10.1097/CJI.0000000000000079. [DOI] [PubMed] [Google Scholar]
- 47.Talib WH, Saleh S, McDowell A. Propionibacterium acnes augments antitumor, anti-angiogenesis and immunomodulatory effects of melatonin on breast cancer implanted in mice. PLOS ONE. 2015;10(4):e0124384. doi: 10.1371/journal.pone.0124384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Koller VJ, Marian B, Stidl R, Nersesyan A, Winter H, Simić T, Sontag G, Knasmüller S. Impact of lactic acid bacteria on oxidative DNA damage in human derived colon cells. Food And Chem Toxicol. 2008;46(4):1221–1229. doi: 10.1016/j.fct.2007.09.005. [DOI] [PubMed] [Google Scholar]
- 49.Marzhoseyni Z, Shojaie L, Tabatabaei SA, Movahedpour A, Safari M, Esmaeili D, Mahjoubin-Tehran M, Jalili A, Morshedi K, Khan H, et al. Streptococcal bacterial components in cancer therapy. Cancer Gene Ther. 2022;29(2):141–155. doi: 10.1038/s41417-021-00308-6. [DOI] [PubMed] [Google Scholar]
- 50.Tano T, Okamoto M, Kan S, Nakashiro K-I, Shimodaira S, Yamashita N, Kawakami Y, Hamakawa H. Growth inhibition and apoptosis by an active component of OK-432, a streptococcal agent, via toll-like receptor 4 in human head and neck cancer cell lines. Oral Oncol. 2012;48(8):678–685. doi: 10.1016/j.oraloncology.2012.02.005. [DOI] [PubMed] [Google Scholar]
- 51.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. 2019;9(1):1–14. doi: 10.1038/s41598-018-37664-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mikó E, Kovács T, Sebő É, Tóth J, Csonka T, Ujlaki G, Sipos A, Szabó J, Méhes G, Bai P. Microbiome—microbial metabolome—cancer cell interactions in breast cancer—familiar, but unexplored. Cells. 2019;8(4):293. doi: 10.3390/cells8040293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Inoue D, Hara S, Kashihara M, Murai Y, Danzl E, Sei K, Tsunoi S, Fujita M, Ike M. Degradation of bis (4-hydroxyphenyl) methane (bisphenol F) by sphingobium yanoikuyae strain FM-2 isolated from river water. Appl Environ Microbiol. 2008;74(2):352–358. doi: 10.1128/AEM.01708-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kertesz MA, Kawasaki A, Stolz A. Aerobic hydrocarbon-degrading alphaproteobacteria: sphingomonadales. Taxon, Genomics And Ecophysiol Hydrocarbon-Degrading Microbes. 2019;4:105–124. [Google Scholar]
- 55.Korsh J, Shen A, Aliano K, Davenport T. Polycyclic aromatic hydrocarbons and breast cancer: a review of the literature. Breast Care. 2015;10(5):316–318. doi: 10.1159/000436956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.White AJ, Bradshaw PT, Herring AH, Teitelbaum SL, Beyea J, Stellman SD, Steck SE, Mordukhovich I, Eng SM, Engel LS, et al. Exposure to multiple sources of polycyclic aromatic hydrocarbons and breast cancer incidence. Environ Intl. 2016;89-90:185–192. doi: 10.1016/j.envint.2016.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lawani-Luwaji EU, Alade T. Sphingomonadaceae: protective against breast cancer? Bull Of The Natl Res Cent. 2020;44(1):1–5. doi: 10.1186/s42269-020-00447-0. [DOI] [Google Scholar]
- 58.Kinjo Y, Wu D, Kim G, Xing G-W, Poles MA, Ho DD, Tsuji M, Kawahara K, Wong C-H, Kronenberg M. Recognition of bacterial glycosphingolipids by natural killer T cells. Nature. 2005;434(7032):520–525. doi: 10.1038/nature03407. [DOI] [PubMed] [Google Scholar]
- 59.Terabe M, Berzofsky JA. NKT cells in immunoregulation of tumor immunity: a new immunoregulatory axis. Trends In Immunol. 2007;28(11):491–496. doi: 10.1016/j.it.2007.05.008. [DOI] [PubMed] [Google Scholar]
- 60.Hix LM, Shi YH, Brutkiewicz RR, Stein PL, Wang C-R, Zhang M, Coonrod SA. CD1d-expressing breast cancer cells modulate NKT cell-mediated antitumor immunity in a murine model of breast cancer metastasis. PLOS ONE. 2011;6(6):e20702. doi: 10.1371/journal.pone.0020702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, Knight R. The microbiome and human cancer. Science. 2021;371(6536):eabc4552. doi: 10.1126/science.abc4552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res. 2015;21(4):687–692. doi: 10.1158/1078-0432.CCR-14-1860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Dieleman S, Aarnoutse R, Ziemons J, Kooreman L, Boleij A, Smidt M. Exploring the potential of breast microbiota as biomarker for breast cancer and therapeutic response. Am J Pathol. 2021;191(6):968–982. doi: 10.1016/j.ajpath.2021.02.020. [DOI] [PubMed] [Google Scholar]
- 64.Gatti-Mays ME, Balko JM, Gameiro SR, Bear HD, Prabhakaran S, Fukui J, Disis ML, Nanda R, Gulley JL, Kalinsky K, et al. If we build it they will come: targeting the immune response to breast cancer. NPJ Breast Cancer. 2019;5(1):1–13. doi: 10.1038/s41523-019-0133-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Haabeth OAW, Lorvik KB, Hammarström C, Donaldson IM, Haraldsen G, Bogen B, Corthay A. Inflammation driven by tumour-specific Th1 cells protects against B-cell cancer. Nat Commun. 2011;2(1):1–12. doi: 10.1038/ncomms1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Showalter L, Czerniecki BJ, Koski GK. Th1 cytokines in conjunction with pharmacological akt inhibition potentiate apoptosis of breast cancer cells in vitro and suppress tumor growth in vivo. Oncotarget. 2020;11(30):2873. doi: 10.18632/oncotarget.27556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Trzonkowski P, Szmit E, Myśliwska J, Dobyszuk A, Myśliwski A. CD4+ CD25+ T regulatory cells inhibit cytotoxic activity of T CD8+ and NK lymphocytes in the direct cell-to-cell interaction. Clin Immunol. 2004;112(3):258–267. doi: 10.1016/j.clim.2004.04.003. [DOI] [PubMed] [Google Scholar]
- 68.Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, Ellis IO, Green AR. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Cin Oncol. 2011;29(15):1949–1955. doi: 10.1200/JCO.2010.30.5037. [DOI] [PubMed] [Google Scholar]
- 69.Seo AN, Lee HJ, Kim EJ, Kim HJ, Jang MH, Lee HE, Kim YJ, Kim JH, Park SY. Tumour-infiltrating CD8+ lymphocytes as an independent predictive factor for pathological complete response to primary systemic therapy in breast cancer. Br J Cancer. 2013;109(10):2705–2713. doi: 10.1038/bjc.2013.634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, Coussens LM. CD4+ T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 2009;16(2):91–102. doi: 10.1016/j.ccr.2009.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Pardoll D. Metastasis-promoting immunity: when T cells turn to the dark side. Cancer Cell. 2009;16(2):81–82. doi: 10.1016/j.ccr.2009.07.007. [DOI] [PubMed] [Google Scholar]
- 72.Olkhanud PB, Damdinsuren B, Bodogai M, Gress RE, Sen R, Wejksza K, Malchinkhuu E, Wersto RP, Biragyn A. Tumor-evoked regulatory B cells promote breast cancer metastasis by converting resting CD4+ T cells to T-Regulatory cells cancer-promoting role of B cells cancer-promoting role of B cells. Cancer Res. 2011;71(10):3505–3515. doi: 10.1158/0008-5472.CAN-10-4316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gómez-Aleza C, Nguyen B, Yoldi G, Ciscar M, Barranco A, Hernndez-Jiménez E, Maetens M, Salgado R, Zafeiroglou M, Pellegrini P, et al. Inhibition of RANK signaling in breast cancer induces an anti-tumor immune response orchestrated by CD8+ T cells. Nat Commun. 2020;11(1):6335. doi: 10.1038/s41467-020-20138-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, Banham AH. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol. 2006;24(34):5373–5380. doi: 10.1200/JCO.2006.05.9584. [DOI] [PubMed] [Google Scholar]
- 75.Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J For Immunother Of Cancer. 2016;4(1):59. doi: 10.1186/s40425-016-0165-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Xiong Y, Zhong Q, Palmer T, Benner A, Wang L, Suresh K, Damico FR, D’Alessio FR. Estradiol resolves pneumonia via ERβ in regulatory T cells. JCI Insight. 2021;6(3). doi: 10.1172/jci.insight.133251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Generali D, Bates G, Berruti A, Brizzi MP, Campo L, Bonardi S, Bersiga A, Allevi G, Milani M, Aguggini S, et al. Immunomodulation of FOXP3+ regulatory T cells by the aromatase inhibitor letrozole in breast cancer patients. Clin Cancer Res. 2009;15(3):1046–1051. doi: 10.1158/1078-0432.CCR-08-1507. [DOI] [PubMed] [Google Scholar]
- 78.Dannenfelser R, Nome M, Tahiri A, Ursini-Siegel J, Vollan HKM, Haakensen VD, Helland Å, Naume B, Caldas C, Børresen-Dale A-L. Data-driven analysis of immune infiltrate in a large cohort of breast cancer and its association with disease progression, ER activity, and genomic complexity. Oncotarget. 2017;8(34):57121. doi: 10.18632/oncotarget.19078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lousberg J, Collignon L, Schroeder H, Jerusalem G. Triple-negative breast cancer: treatment challenges and solutions. Breast Cancer: Targets And Ther. 2016;8:93. doi: 10.2147/BCTT.S69488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Savas P, Salgado R, Denkert C, Sotiriou C, Darcy PK, Smyth MJ, Loi S. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat Rev Clin Oncol. 2016;13(4):228–241. doi: 10.1038/nrclinonc.2015.215. [DOI] [PubMed] [Google Scholar]
- 81.Shi J, Geng C, Sang M, Gao W, Li S, Yang S, Li Z. Effect of gastrointestinal microbiome and its diversity on the expression of tumor‑infiltrating lymphocytes in breast cancer. Oncol Lett. 2019;17(6):5050–5056. doi: 10.3892/ol.2019.10187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Adams S, Goldstein LJ, Sparano JA, Demaria S, Badve SS. Tumor infiltrating lymphocytes (TILs) improve prognosis in patients with triple negative breast cancer (TNBC). Oncoimmunology. 2015;4(9):e985930. doi: 10.4161/2162402X.2014.985930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, Budczies J, Huober J, Klauschen F, Furlanetto J, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018;19(1):40–50. doi: 10.1016/S1470-2045(17)30904-X. [DOI] [PubMed] [Google Scholar]
- 84.Mao Y, Qu Q, Zhang Y, Liu J, Chen X, Shen K, Beck AH. The value of tumor infiltrating lymphocytes (TILs) for predicting response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis. PLOS ONE. 2014;9(12):e115103. doi: 10.1371/journal.pone.0115103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Dieci MV, Radosevic-Robin N, Fineberg S, Van den Eynden G, Ternes N, Penault-Llorca F, Castaneda C. Update on tumor-infiltrating lymphocytes (TILs) in breast cancer, including recommendations to assess TILs in residual disease after neoadjuvant therapy and in carcinoma in situ: a report of the International immuno-oncology biomarker working group on breast cancer. 2018. Paper presented at the Seminars in cancer biology. [DOI] [PubMed]
- 86.Vonderheide RH, Domchek SM, Clark AS. Immunotherapy for breast cancer: what are we missing? In: AACR; 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wensel CR, Pluznick JL, Salzberg SL, Sears CL. Next-generation sequencing: insights to advance clinical investigations of the microbiome. J Clin Invest. 2022;132(7). doi: 10.1172/JCI154944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chakravorty S, Helb D, Burday M, Connell N, Alland D. A detailed analysis of 16S ribosomal RNA gene segments for the diagnosis of pathogenic bacteria. J Microbiol Methods. 2007;69(2):330–339. doi: 10.1016/j.mimet.2007.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Videnska P, Smerkova K, Zwinsova B, Popovici V, Micenkova L, Sedlar K, Budinska E. Stool sampling and DNA isolation kits affect DNA quality and bacterial composition following 16S rRNA gene sequencing using MiSeq illumina platform. Sci Rep. 2019;9(1):1–14. doi: 10.1038/s41598-019-49520-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Saliba A-E, Santos SC, Vogel J. New RNA-seq approaches for the study of bacterial pathogens. Curr Opin Microbiol. 2017;35:78–87. doi: 10.1016/j.mib.2017.01.001. [DOI] [PubMed] [Google Scholar]
- 91.Quince C, Walker AW, Simpson JT, Loman NJ, Segata N. Shotgun metagenomics, from sampling to analysis. Nat Biotechnol. 2017;35(9):833–844. doi: 10.1038/nbt.3935. [DOI] [PubMed] [Google Scholar]
- 92.Laudadio I, Fulci V, Palone F, Stronati L, Cucchiara S, Carissimi C. Quantitative assessment of shotgun metagenomics and 16S rDNA amplicon sequencing in the study of human gut microbiome. Omics: A J Intgr Biol. 2018;22(4):248–254. doi: 10.1089/omi.2018.0013. [DOI] [PubMed] [Google Scholar]
- 93.Maki KA, Kazmi N, Barb JJ, Ames N. The oral and gut bacterial microbiomes: similarities, differences, and connections. Biol Res Nurs. 2021;23(1):7–20. doi: 10.1177/1099800420941606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chen KL, Madak-Erdogan Z. Estrogen and microbiota crosstalk: should we pay attention? Trends In Endocrinol & Metab. 2016;27(11):752–755. doi: 10.1016/j.tem.2016.08.001. [DOI] [PubMed] [Google Scholar]
- 95.Goodman B, Gardner H. The microbiome and cancer. J Pathol. 2018;244(5):667–676. doi: 10.1002/path.5047. [DOI] [PubMed] [Google Scholar]
- 96.Firoozeh F. Gut microbiome and human health. Int J Enteric Pathog. 2019;7(2):30–30. doi: 10.15171/ijep.2019.08. [DOI] [Google Scholar]
- 97.Selber-Hnatiw S, Rukundo B, Ahmadi M, Akoubi H, Al-Bizri H, Aliu AF, Ambeaghen TU, Avetisyan L, Bahar I, Baird A, et al. Human gut microbiota: toward an ecology of disease. Front Microbiol. 2017;8:1265. doi: 10.3389/fmicb.2017.01265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14(10):667–685. doi: 10.1038/nri3738. [DOI] [PubMed] [Google Scholar]
- 99.Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, Fulton RS. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Singh SB, Madan J, Coker M, Hoen A, Baker ER, Karagas MR, Mueller NT. Does birth mode modify associations of maternal pre-pregnancy BMI and gestational weight gain with the infant gut microbiome? Int J Obes. 2020;44(1):23–32. doi: 10.1038/s41366-018-0273-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto J-M, et al. Enterotypes of the human gut microbiome. Nature. 2011;473(7346):174–180. doi: 10.1038/nature09944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Johnson EL, Heaver SL, Walters WA, Ley RE. Microbiome and metabolic disease: revisiting the bacterial phylum bacteroidetes. J Mol Med. 2017;95(1):1–8. doi: 10.1007/s00109-016-1492-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.West CE, Jenmalm M, Prescott S. The gut microbiota and its role in the development of allergic disease: a wider perspective. Clin And Exp Allergy: J Br Soc For Allergy And Clin Immunol. 2015;45(1):43–53. doi: 10.1111/cea.12332. [DOI] [PubMed] [Google Scholar]
- 104.Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, et al. T reg induction by a rationally selected mixture of clostridia strains from the human microbiota. Nature. 2013;500(7461):232–236. doi: 10.1038/nature12331. [DOI] [PubMed] [Google Scholar]
- 105.Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci USA. 2010;107(27):12204–12209. doi: 10.1073/pnas.0909122107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Guerville M, Boudry G. Gastrointestinal and hepatic mechanisms limiting entry and dissemination of lipopolysaccharide into the systemic circulation. Am J physiol-Gastr L. 2016;311(1):G1–G15. doi: 10.1152/ajpgi.00098.2016. [DOI] [PubMed] [Google Scholar]
- 107.Ciesielska A, Matyjek M, Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol Life Sci. 2021;78(4):1233–1261. doi: 10.1007/s00018-020-03656-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.McGhee JR, Fujihashi K. Inside the mucosal immune system. PLOS Biol. 2012;10(9):e1001397. doi: 10.1371/journal.pbio.1001397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Gorvitovskaia A, Holmes SP, Huse SM. Interpreting prevotella and bacteroides as biomarkers of diet and lifestyle. Microbiome. 2016;4(1):15. doi: 10.1186/s40168-016-0160-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Dominianni C, Sinha R, Goedert JJ, Pei Z, Yang L, Hayes RB, Ahn J, Wilson BA. Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLOS ONE. 2015;10(4):e0124599. doi: 10.1371/journal.pone.0124599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.O’keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13(12):691. doi: 10.1038/nrgastro.2016.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic treg cell homeostasis. Science. 2013;341(6145):569–573. doi: 10.1126/science.1241165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Engin A. Obesity-associated breast cancer: analysis of risk factors. Obes And Lipotoxicity. 2017;960:571–606. [DOI] [PubMed] [Google Scholar]
- 114.Fuhrman BJ, Feigelson HS, Flores R, Gail MH, Xu X, Ravel J, Goedert JJ. Associations of the fecal microbiome with urinary estrogens and estrogen metabolites in postmenopausal women. J Clin Endocr Metab. 2014;99(12):4632–4640. doi: 10.1210/jc.2014-2222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Frugé AD, Van der Pol W, Rogers LQ, Morrow CD, Tsuruta Y, 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 And Diet. 2020;120(4):650–659. doi: 10.1016/j.jand.2018.08.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Woo V, Alenghat T. Host–microbiota interactions: epigenomic regulation. Curr Opin Immunol. 2017;44:52–60. doi: 10.1016/j.coi.2016.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Eslami-S Z, Majidzadeh-A K, Halvaei S, Babapirali F, Esmaeili R. Microbiome and breast cancer: new role for an ancient population. Front Oncol. 2020;10:120. doi: 10.3389/fonc.2020.00120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rahat-Rozenbloom S, Fernandes J, Gloor GB, Wolever TM. Evidence for greater production of colonic short-chain fatty acids in overweight than lean humans. Int J Obes. 2014;38(12):1525–1531. doi: 10.1038/ijo.2014.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zhu J, Liao M, Yao Z, Liang W, Li Q, Liu J, Yang H, Ji Y, Wei W, Tan A, et al. Breast cancer in postmenopausal women is associated with an altered gut metagenome. Microbiome. 2018;6(1):136. doi: 10.1186/s40168-018-0515-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Brestoff JR, Artis D. Commensal bacteria at the interface of host metabolism and the immune system. Nat Immunol. 2013;14(7):676–684. doi: 10.1038/ni.2640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Suzuki T, Yoshida S, Hara H. Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability. Br J Nutr. 2008;100(2):297–305. doi: 10.1017/S0007114508888733. [DOI] [PubMed] [Google Scholar]
- 122.Chen J, Zhao K-N, Vitetta L. Effects of intestinal microbial–elaborated butyrate on oncogenic signaling pathways. Nutrients. 2019;11(5):1026. doi: 10.3390/nu11051026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Vinolo MA, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC, Curi R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J Nutritional Biochem. 2011;22(9):849–855. doi: 10.1016/j.jnutbio.2010.07.009. [DOI] [PubMed] [Google Scholar]
- 124.Miller-Fleming L, Olin-Sandoval V, Campbell K, Ralser M. Remaining mysteries of molecular biology: the role of polyamines in the cell. J Mol Biol. 2015;427(21):3389–3406. doi: 10.1016/j.jmb.2015.06.020. [DOI] [PubMed] [Google Scholar]
- 125.Liu R, Li P, Bi CW, Ma R, Yin Y, Bi K, Li Q. Plasma N-acetylputrescine, cadaverine and 1, 3-diaminopropane: potential biomarkers of lung cancer used to evaluate the efficacy of anticancer drugs. Oncotarget. 2017;8(51):88575. doi: 10.18632/oncotarget.19304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Liu R, Li Q, Ma R, Lin X, Xu H, Bi K. Determination of polyamine metabolome in plasma and urine by ultrahigh performance liquid chromatography–tandem mass spectrometry method: application to identify potential markers for human hepatic cancer. Analytica Chimica Acta. 2013;791:36–45. doi: 10.1016/j.aca.2013.06.044. [DOI] [PubMed] [Google Scholar]
- 127.Kovács T, Mikó E, Vida A, Sebo 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. 2019;9(1):1300. doi: 10.1038/s41598-018-37664-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Cao W, Zheng C, Xu X, Jin R, Huang F, Shi M, He Z, Luo Y, Liu L, Liu Z, et al. Clostridium butyricum potentially improves inflammation and immunity through alteration of the microbiota and metabolism of gastric cancer patients after gastrectomy. Front Immunol. 2022;13:1076245. doi: 10.3389/fimmu.2022.1076245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Adnan M, Khan S, Al-Shammari E, Patel M, Saeed M, Hadi S. In pursuit of cancer metastasis therapy by bacteria and its biofilms: history or future. Med Hypotheses. 2017;100:78–81. doi: 10.1016/j.mehy.2017.01.018. [DOI] [PubMed] [Google Scholar]
- 130.McColl KE, El-Omar E. How does H. pylori infection cause gastric cancer? Keio J Med. 2002;51(supplement2):53–56. doi: 10.2302/kjm.51.supplement2_53. [DOI] [PubMed] [Google Scholar]
- 131.Chen J, Pitmon E, Wang K. Microbiome, inflammation and colorectal cancer. Paper presented at the seminars in immunology. 2017. [DOI] [PubMed]
- 132.Rosean CMB, Rutkowski MR. The influence of the commensal microbiota on distal tumor-promoting inflammation. Paper presented at the seminars in immunology. 2017. [DOI] [PMC free article] [PubMed]
- 133.Yang H, Wang B, Wang T, Xu L, He C, Wen H, Yan J, Su H, 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. 2014;9(10):e109980. doi: 10.1371/journal.pone.0109980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Li H, Xia JQ, Zhu FS, Xi ZH, Pan CY, Gu LM, Tian YZ. LPS promotes the expression of PD‐L1 in gastric cancer cells through NF‐κB activation. J Cell Biochem. 2018;119(12):9997–10004. doi: 10.1002/jcb.27329. [DOI] [PubMed] [Google Scholar]
- 135.Afroz R, Tanvir E, Tania M, Fu J, Kamal MA, Khan M. LPS/TLR4 pathways in breast cancer: insights into cell signalling. Curr Med Chem. 2022;29(13):2274–2289. doi: 10.2174/0929867328666210811145043. [DOI] [PubMed] [Google Scholar]
- 136.Hsu RY, Chan CH, Spicer JD, Rousseau MC, Giannias B, Rousseau S, Ferri LE. LPS-induced TLR4 signaling in human colorectal cancer cells increases β1 integrin-mediated cell adhesion and liver metastasis. Cancer Res. 2011;71(5):1989–1998. doi: 10.1158/0008-5472.CAN-10-2833. [DOI] [PubMed] [Google Scholar]
- 137.Huang T, Chen Z, Fang L. Curcumin inhibits LPS-induced EMT through downregulation of NF-κB-Snail signaling in breast cancer cells. Oncol Rep. 2013;29(1):117–124. doi: 10.3892/or.2012.2080. [DOI] [PubMed] [Google Scholar]
- 138.Ikebe M, Kitaura Y, Nakamura M, Tanaka H, Yamasaki A, Nagai S, Wada J, Yanai K, Koga K, Sato N, et al. Lipopolysaccharide (LPS) increases the invasive ability of pancreatic cancer cells through the TLR4/MyD88 signaling pathway. J Surgical Oncol. 2009;100(8):725–731. doi: 10.1002/jso.21392. [DOI] [PubMed] [Google Scholar]
- 139.Li N, Xu H, Ou Y, Feng Z, Zhang Q, Zhu Q, Cai Z. LPS-induced CXCR7 expression promotes gastric cancer proliferation and migration via the TLR4/MD-2 pathway. Diagn Pathol. 2019;14(1):1–10. doi: 10.1186/s13000-019-0780-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Cheng WT, Kantilal HK, Davamani F. The mechanism of Bacteroides fragilis toxin contributes to colon cancer formation. The Malays J Med Sci: MJMS. 2020;27(4):9. doi: 10.21315/mjms2020.27.4.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Jiang F, Meng D, Weng M, Zhu W, Wu W, Kasper D, Walker WA, De Re V. The symbiotic bacterial surface factor polysaccharide a on bacteroides fragilis inhibits IL-1β-induced inflammation in human fetal enterocytes via toll receptors 2 and 4. PLOS ONE. 2017;12(3):e0172738. doi: 10.1371/journal.pone.0172738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Telesford KM, Yan W, Ochoa-Reparaz J, Pant A, Kircher C, Christy MA, Begum-Haque S, Kasper DL, Kasper LH. A commensal symbiotic factor derived from bacteroides fragilis promotes human CD39+ Foxp3+ T cells and treg function. Gut Microbes. 2015;6(4):234–242. doi: 10.1080/19490976.2015.1056973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Parida S, Wu S, Siddharth S, Wang G, Muniraj N, Nagalingam A, Hum CC, Mistriotis P, Hao H, Talbot CC, et al. A procarcinogenic colon microbe promotes breast tumorigenesis and metastatic progression and concomitantly activates notch and β-Catenin axes. Cancer Discov. 2021;11(5):1138–1157. doi: 10.1158/2159-8290.CD-20-0537. [DOI] [PubMed] [Google Scholar]
- 144.Dejea CM, Sears CL. Do biofilms confer a pro-carcinogenic state? Gut Microbes. 2016;7(1):54–57. doi: 10.1080/19490976.2015.1121363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Johnson CH, Dejea CM, Edler D, Hoang LT, Santidrian AF, Felding BH, Ivanisevic J, Cho K, Wick E, Hechenbleikner E, et al. Metabolism links bacterial biofilms and colon carcinogenesis. Cell Metabol. 2015;21(6):891–897. doi: 10.1016/j.cmet.2015.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kumeria T, Maher S, Wang Y, Kaur G, Wang L, Erkelens M, Forward P, Lambert MF, Evdokiou A, Losic D. Naturally derived iron oxide nanowires from bacteria for magnetically triggered drug release and cancer hyperthermia in 2D and 3D culture environments: bacteria biofilm to potent cancer therapeutic. Biomacromolecules. 2016;17(8):2726–2736. doi: 10.1021/acs.biomac.6b00786. [DOI] [PubMed] [Google Scholar]
- 147.Li S, Konstantinov SR, Smits R, Peppelenbosch MP. Bacterial biofilms in colorectal cancer initiation and progression. Trends Mol Med. 2017;23(1):18–30. doi: 10.1016/j.molmed.2016.11.004. [DOI] [PubMed] [Google Scholar]
- 148.Mirzaei R, Mirzaei H, Alikhani MY, Sholeh M, Arabestani MR, Saidijam M, Karampoor S, Ahmadyousefi Y, Moghadam MS, Irajian GR, et al. Bacterial biofilm in colorectal cancer: what is the real mechanism of action? Microbial Pathogenesis. 2020;142:104052. doi: 10.1016/j.micpath.2020.104052. [DOI] [PubMed] [Google Scholar]
- 149.Tjalsma H, Boleij A, Marchesi JR, Dutilh BE. A bacterial driver–passenger model for colorectal cancer: beyond the usual suspects. Nat Rev Microbiol. 2012;10(8):575–582. doi: 10.1038/nrmicro2819. [DOI] [PubMed] [Google Scholar]
- 150.Fromantin I, Seyer D, Watson S, Rollot F, Elard J, Escande MC, De Rycke Y, Kriegel I, Larreta Garde V. Bacterial floras and biofilms of malignant wounds associated with breast cancers. J Clin Microbiol. 2013;51(10):3368–3373. doi: 10.1128/JCM.01277-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Garrett WS. Cancer and the microbiota. Science. 2015;348(6230):80–86. doi: 10.1126/science.aaa4972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Groizeleau J, Rybtke M, Andersen JB, Berthelsen J, Liu Y, Yang L, Tolker-Nielsen T, Kaever V, Givskov M, Tolker-Nielsen T. The anti-cancerous drug doxorubicin decreases the c-di-GMP content in pseudomonas aeruginosa but promotes biofilm formation. Microbiol. 2016;162(10):1797–1807. doi: 10.1099/mic.0.000354. [DOI] [PubMed] [Google Scholar]
- 153.Weitao T. Bacteria form biofilms against cancer metastasis. Med Hypotheses. 2009;72(4):477. doi: 10.1016/j.mehy.2008.11.012. [DOI] [PubMed] [Google Scholar]
- 154.Goedert JJ, Jones G, Hua X, Xu X, Yu G, Flores R, Falk RT, Gail MH, Shi J, Ravel J. Investigation of the association between the fecal microbiota and breast cancer in postmenopausal women: a population-based case-control pilot study. JNCI: J Natl Cancer Inst. 2015;107(8). doi: 10.1093/jnci/djv147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967–970. doi: 10.1126/science.1240527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Viaud S, Daillere R, Boneca I, Lepage P, Langella P, Chamaillard M, Pittet MJ, Ghiringhelli F, Trinchieri G, Goldszmid R, et al. Gut microbiome and anticancer immune response: really hot shot! Cell Death Differ. 2015;22(2):199–214. doi: 10.1038/cdd.2014.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Iyengar NM, Zhou XK, Gucalp A, Morris PG, Howe LR, Giri DD, Morrow M, Wang H, Pollak M, Jones LW, et al. Systemic correlates of white adipose tissue inflammation in early-stage breast cancer. Clinical Cancer Res. 2016;22(9):2283–2289. doi: 10.1158/1078-0432.CCR-15-2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Rosean CB, Bostic RR, Ferey JC, Feng T-Y, Azar FN, Tung KS, Dozmorov MG, Smirnova E, Bos PD, Rutkowski MR. Preexisting commensal dysbiosis is a host-intrinsic regulator of tissue inflammation and tumor cell dissemination in hormone receptor–positive breast cancer. Cancer Res. 2019;79(14):3662–3675. doi: 10.1158/0008-5472.CAN-18-3464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Rao VP, Poutahidis T, Ge Z, Nambiar PR, Boussahmain C, Wang YY, Horwitz BH, Fox JG, Erdman SE. Innate immune inflammatory response against enteric bacteria helicobacter hepaticus induces mammary adenocarcinoma in mice. Cancer Res. 2006;66(15):7395–7400. doi: 10.1158/0008-5472.CAN-06-0558. [DOI] [PubMed] [Google Scholar]
- 160.Deng H, Muthupalani S, Erdman S, Liu H, Niu Z, Wang TC, Fox JG. Translocation of Helicobacter hepaticus synergizes with myeloid-derived suppressor cells and contributes to breast carcinogenesis. Oncoimmunology. 2022;11(1):2057399. doi: 10.1080/2162402X.2022.2057399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lakritz JR, Poutahidis T, Mirabal S, Varian BJ, Levkovich T, Ibrahim YM, Ward JM, Teng EC, Fisher B, Parry N, et al. Gut bacteria require neutrophils to promote mammary tumorigenesis. Oncotarget. 2015;6(11):9387. doi: 10.18632/oncotarget.3328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, Gajer P, Ravel J, Goedert JJ. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. J Transl Med. 2012;10(1):253. doi: 10.1186/1479-5876-10-253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Bard J-M, Luu HT, Dravet F, Michel C, Moyon T, Pagniez A, Nazih H, Bobin‐Dubigeon C. Relationship between intestinal microbiota and clinical characteristics of patients with early stage breast cancer. The FASEB J. 2015;29(S1):914.912. doi: 10.1096/fasebj.29.1_supplement.914.2. [DOI] [Google Scholar]
- 164.Luu TH, Michel C, Bard J-M, Dravet F, Nazih H, Bobin-Dubigeon C. Intestinal proportion of Blautia sp. is associated with clinical stage and histoprognostic grade in patients with early-stage breast cancer. Nutr And Cancer. 2017;69(2):267–275. doi: 10.1080/01635581.2017.1263750. [DOI] [PubMed] [Google Scholar]
- 165.Goedert JJ, Hua X, Bielecka A, Okayasu I, Milne GL, Jones GS, Fujiwara M, Sinha R, Wan Y, Xu X, et al. Postmenopausal breast cancer and oestrogen associations with the IgA-coated and IgA-noncoated faecal microbiota. Br J Cancer. 2018;118(4):471–479. doi: 10.1038/bjc.2017.435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.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 Et Biophys Acta (BBA) - Bioenerget. 2018;1859(9):958–974. doi: 10.1016/j.bbabio.2018.04.002. [DOI] [PubMed] [Google Scholar]
- 167.Horigome A, Okubo R, Hamazaki K, Kinoshita T, Katsumata N, Uezono Y, Xiao JZ, Matsuoka YJ. Association between blood omega-3 polyunsaturated fatty acids and the gut microbiota among breast cancer survivors. BM. 2019;10(7):751–758. doi: 10.3920/BM2019.0034. [DOI] [PubMed] [Google Scholar]
- 168.Kirkup B, McKee A, Makin K, Paveley J, Caim S, Alcon-Giner C, Andrusaite A. Perturbation of the gut microbiota by antibiotics results in accelerated breast tumour growth and metabolic dysregulation. BioRxiv. 2019;1162:553602. [Google Scholar]
- 169.Yoon H-J, Kim H-N, Bang J-I, Lim W, Moon BI, Paik NS, Kim BS, Kim H-L. Physiologic intestinal 18F-FDG uptake is associated with alteration of gut microbiota and proinflammatory cytokine levels in breast cancer. Sci Rep. 2019;9(1):18273. doi: 10.1038/s41598-019-54680-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Terrisse S, Derosa L, Iebba V, Ghiringhelli F, Vaz-Luis I, Kroemer G, Fidelle AM, Christodoulidis S, Segata N, Thomas AM, et al. Intestinal microbiota influences clinical outcome and side effects of early breast cancer treatment. Cell Death Differ. 2021;28(9):2778–2796. doi: 10.1038/s41418-021-00784-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Ma J, Gnanasekar A, Lee A, Li WT, Haas M, Wang-Rodriguez J, Chang EY, Rajasekaran M, Ongkeko WM. Influence of intratumor microbiome on clinical outcome and immune processes in prostate cancer. Cancers. 2020;12(9):2524. doi: 10.3390/cancers12092524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Jafari B, Nejad RAK, Vaziri F, Siadat SD. Evaluation of the effects of extracellular vesicles derived from faecalibacterium prausnitzii on lung cancer cell line. Biologia. 2019;74(7):889–898. doi: 10.2478/s11756-019-00229-8. [DOI] [Google Scholar]
- 173.Lopez-Siles M, Martinez-Medina M, Surís-Valls R, Aldeguer X, Sabat-Mir M, Duncan SH, Flint HJ, Garcia-Gil LJ. Changes in the abundance of faecalibacterium prausnitzii phylogroups I and II in the intestinal mucosa of inflammatory bowel disease and patients with colorectal cancer. Inflammatory Bowel Dis. 2016;22(1):28–41. doi: 10.1097/MIB.0000000000000590. [DOI] [PubMed] [Google Scholar]
- 174.Quévrain E, Maubert M, Michon C, Chain F, Marquant R, Tailhades J, Miquel B, Carlier L, Bermúdez-Humarán LG, Pigneur B, et al. Identification of an anti-inflammatory protein from faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut. 2016;65(3):415–425. doi: 10.1136/gutjnl-2014-307649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Schneeberger M, Everard A, Gómez-Valadés AG, Matamoros S, Ramírez S, Delzenne NM, Gomis R, Claret M, Cani PD. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci Rep. 2015;5(1):16643. doi: 10.1038/srep16643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Wu W, Lv L, Shi D, Ye J, Fang D, Guo F, Li Y, He X, Li L. Protective effect of Akkermansia muciniphila against immune-mediated liver injury in a mouse model. Front Microbiol. 2017;8:1804. doi: 10.3389/fmicb.2017.01804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Delzenne NM, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci, India, Sect B Biol Sci. 2013;110(22):9066–9071. doi: 10.1073/pnas.1219451110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Gui Q, Li H, Wang A, Zhao X, Tan Z, Chen L, Xu K, Xiao C. The association between gut butyrate‐producing bacteria and non‐small‐cell lung cancer. Clinical Laboratory Analy. 2020;34(8):e23318. doi: 10.1002/jcla.23318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Kwa M, Plottel CS, Blaser MJ, Adams S. The intestinal microbiome and estrogen receptor–positive female breast cancer. JNCI: J Natl Cancer Inst. 2016;108(8):1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kerlikowske K, Gard CC, Tice JA, Ziv E, Cummings SR, Miglioretti DL. Risk factors that increase risk of estrogen receptor–positive and –negative breast cancer. J Natl Cancer Inst. 2017;109(5):djw276. doi: 10.1093/jnci/djw276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Dallal CM, Tice JA, Buist DS, Bauer DC, Lacey JV Jr, Cauley JA, Hue TF, LaCroix A, Falk RT, Pfeiffer RM, et al. Estrogen metabolism and breast cancer risk among postmenopausal women: a case–cohort study within B~ FIT. Carcinogenesis. 2014;35(2):346–355. doi: 10.1093/carcin/bgt367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Falk RT, Brinton LA, Dorgan JF, Fuhrman BJ, Veenstra TD, Xu X, Gierach GL. Relationship of serum estrogens and estrogen metabolites to postmenopausal breast cancer risk: a nested case-control study. Breast Cancer Res. 2013;15(2):R34. doi: 10.1186/bcr3416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Fuhrman BJ, Schairer C, Gail MH, Boyd-Morin J, Xu X, Sue LY, Buys SS, Isaacs C, Keefer LK, Veenstra TD, et al. Estrogen metabolism and risk of breast cancer in postmenopausal women. JNCI: J Natl Cancer Inst. 2012;104(4):326–339. doi: 10.1093/jnci/djr531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Nakamura J, Kubota Y, Miyaoka M, Saitoh T, Mizuno F, Benno Y. Comparison of four microbial enzymes in clostridia and bacteroides isolated from human feces. Microbiol And Immunol. 2002;46(7):487–490. doi: 10.1111/j.1348-0421.2002.tb02723.x. [DOI] [PubMed] [Google Scholar]
- 185.Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Keefe DM. Faecal microflora and β-glucuronidase expression are altered in an irinotecan-induced diarrhea model in rats. Cancer Biol & Ther. 2008;7(12):1919–1925. doi: 10.4161/cbt.7.12.6940. [DOI] [PubMed] [Google Scholar]
- 186.McIntosh FM, Maison N, Holtrop G, Young P, Stevens VJ, Ince J, Johnstone AM, Lobley GE, Flint HJ, Louis P. Phylogenetic distribution of genes encoding β‐glucuronidase activity in human colonic bacteria and the impact of diet on faecal glycosidase activities. Environ Microbiol. 2012;14(8):1876–1887. doi: 10.1111/j.1462-2920.2012.02711.x. [DOI] [PubMed] [Google Scholar]
- 187.Pollet RM, D’Agostino EH, Walton WG, Xu Y, Little MS, Biernat KA, Pellock SJ, Patterson LM, Creekmore BC, Isenberg HN, et al. An atlas of β-glucuronidases in the human intestinal microbiome. Structure. 2017;25(7):967–977.e5. doi: 10.1016/j.str.2017.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Gloux K, Berteau O, El Oumami H, Béguet F, Leclerc M, Doré J. A metagenomic β-glucuronidase uncovers a core adaptive function of the human intestinal microbiome. Proc Natl Acad Sci USA. 2011;108(supplement_1):4539–4546. doi: 10.1073/pnas.1000066107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Girbal L, Mortier-Barriere I, Raynaud F, Rouanet C, Croux C, Soucaille P. Development of a sensitive gene expression reporter system and an inducible promoter-repressor system for clostridium acetobutylicum. Appl Environ Microbiol. 2003;69(8):4985–4988. doi: 10.1128/AEM.69.8.4985-4988.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Mani N, Dupuy B, Sonenshein AL. Isolation of RNA polymerase from clostridium difficile and characterization of glutamate dehydrogenase and rRNA gene promoters in vitro and in vivo. J Bacteriol. 2006;188(1):96–102. doi: 10.1128/JB.188.1.96-102.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Hartman AH, Liu H, Melville SB. Construction and characterization of a lactose-inducible promoter system for controlled gene expression in clostridium perfringens. Appl Environ Microb. 2011;77(2):471–478. doi: 10.1128/AEM.01536-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Leung JW, Liu Y-L, Leung PS, Chan RC, Inciardi JF, Cheng AF. Expression of bacterial β-glucuronidase in human bile: an in vitro study. Gastrointestinal Endoscopy. 2001;54(3):346–350. doi: 10.1067/mge.2001.117546. [DOI] [PubMed] [Google Scholar]
- 193.Shimizu T, Ohtani K, Hirakawa H, Ohshima K, Yamashita A, Shiba T, Ogasawara N, Hattori M, Kuhara S, Hayashi H. Complete genome sequence of clostridium perfringens, an anaerobic flesh-eater. Proc Natl Acad Sci USA. 2002;99(2):996–1001. doi: 10.1073/pnas.022493799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Dabek M, McCrae SI, Stevens VJ, Duncan SH, Louis P. Distribution of β-glucosidase and β-glucuronidase activity and of β-glucuronidase gene gus in human colonic bacteria. FEMS Microbiol Ecol. 2008;66(3):487–495. doi: 10.1111/j.1574-6941.2008.00520.x. [DOI] [PubMed] [Google Scholar]
- 195.Russell W, Klaenhammer T. Identification and cloning of gusA, encoding a new β-glucuronidase from Lactobacillus gasseriADH. Appl Environ Microb. 2001;67(3):1253–1261. doi: 10.1128/AEM.67.3.1253-1261.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Beaud D, Tailliez P, Anba-Mondoloni J. Genetic characterization of the β-glucuronidase enzyme from a human intestinal bacterium, ruminococcus gnavus. Microbiology. 2005;151(7):2323–2330. doi: 10.1099/mic.0.27712-0. [DOI] [PubMed] [Google Scholar]
- 197.Edwinson AL, Yang L, Peters S, Hanning N, Jeraldo P, Jagtap P, Simpson JB, Yang T-Y, Kumar P, Mehta S, et al. Gut microbial β-glucuronidases regulate host luminal proteases and are depleted in irritable bowel syndrome. Nat Microbiol. 2022;7(5):680–694. doi: 10.1038/s41564-022-01103-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Mani S. Microbiota and breast cancer. In: Lakshmanaswamy R, editor. Progress in molecular biology and translational science. Vol. 151. New York: Elsevier; 2017. p. 217–229. [DOI] [PubMed] [Google Scholar]
- 199.Peters BA, Lin J, Qi Q, Usyk M, Isasi CR, Mossavar-Rahmani Y, Derby CA, Santoro N, Perreira KM, Daviglus ML, et al. Menopause is associated with an altered gut microbiome and estrobolome, with implications for adverse cardiometabolic risk in the Hispanic community health study/study of latinos. mSystems. 2022;7(3):e00273–00222. doi: 10.1128/msystems.00273-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Rodríguez JM. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv Nutr. 2014;5(6):779–784. doi: 10.3945/an.114.007229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Arroyo R, Martín V, Maldonado A, Jiménez E, Fernández L, Rodríguez JM. Treatment of infectious mastitis during lactation: antibiotics versus oral administration of lactobacilli isolated from breast milk. Clin Infect Dis: Off Publ Infect Dis Soc Am. 2010;50(12):1551–1558. doi: 10.1086/652763. [DOI] [PubMed] [Google Scholar]
- 202.Jimenez E, Fernandez L, Maldonado A, Martin R, Olivares M, Xaus J, Rodriguez J. Oral administration of Lactobacillus strains isolated from breast milk as an alternative for the treatment of infectious mastitis during lactation. Appl Environ Microbiol. 2008;74(15):4650–4655. doi: 10.1128/AEM.02599-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Langa S. Interactions between lactic acid bacteria, intestinal epithelial cells and immune cells. Development of in vitro models. Madrid: Complutense University of Madrid; 2006. [Google Scholar]
- 204.Perez PF, Doré J, Leclerc M, Levenez F, Benyacoub J, Serrant P, Segura-Roggero I, Schiffrin EJ, Donnet-Hughes A. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics. 2007;119(3):e724–e732. doi: 10.1542/peds.2006-1649. [DOI] [PubMed] [Google Scholar]
- 205.Lakritz JR, Poutahidis T, Levkovich T, Varian BJ, Ibrahim YM, Chatzigiagkos A, Mirabal SE, Alm EJ, Erdman SE. Beneficial bacteria stimulate host immune cells to counteract dietary and genetic predisposition to mammary cancer in mice. Int J Cancer. 2014;135(3):529–540. doi: 10.1002/ijc.28702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Chakraborty B, Byemerwa J, Krebs T, Lim F, Chang C-Y, McDonnell DP. Estrogen receptor signaling in the immune system. Endocr Rev. 2023;44(1):117–141. doi: 10.1210/endrev/bnac017. [DOI] [PubMed] [Google Scholar]
- 207.Kovats S. Estrogen receptors regulate innate immune cells and signaling pathways. Cellular Immunol. 2015;294(2):63–69. doi: 10.1016/j.cellimm.2015.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Leitman DC, Paruthiyil S, Vivar OI, Saunier EF, Herber CB, Cohen I, Tagliaferri M, Speed TP. Regulation of specific target genes and biological responses by estrogen receptor subtype agonists. Curr Opinion In Pharmacol. 2010;10(6):629–636. doi: 10.1016/j.coph.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Mann M, Cortez V, Vadlamudi RK. Epigenetics of estrogen receptor signaling: role in hormonal cancer progression and therapy. Cancers. 2011;3(2):1691–1707. doi: 10.3390/cancers3021691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Laffont S, Rouquié N, Azar P, Seillet C, Plumas J, Aspord C, Guéry J-C. X-chromosome complement and estrogen receptor signaling independently contribute to the enhanced TLR7-mediated IFN-α production of plasmacytoid dendritic cells from women. J Immunol. 2014;193(11):5444–5452. doi: 10.4049/jimmunol.1303400. [DOI] [PubMed] [Google Scholar]
- 211.Phiel KL, Henderson RA, Adelman SJ, Elloso MM. Differential estrogen receptor gene expression in human peripheral blood mononuclear cell populations. Immunol Lett. 2005;97(1):107–113. doi: 10.1016/j.imlet.2004.10.007. [DOI] [PubMed] [Google Scholar]
- 212.Panchanathan R, Shen H, Zhang X, Ho S-M, Choubey D, Unutmaz D. Mutually positive regulatory feedback loop between interferons and estrogen receptor-α in mice: implications for sex bias in autoimmunity. PLOS ONE. 2010;5(5):e10868. doi: 10.1371/journal.pone.0010868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Seillet C, Rouquié N, Foulon E, Douin-Echinard V, Krust A, Chambon P, Arnal J-F, Guéry J-C, Laffont S. Estradiol promotes functional responses in inflammatory and steady-state dendritic cells through differential requirement for activation function-1 of estrogen receptor α. J Of Immunol. 2013;190(11):5459–5470. doi: 10.4049/jimmunol.1203312. [DOI] [PubMed] [Google Scholar]
- 214.Papenfuss TL, Powell ND, McClain MA, Bedarf A, Singh A, Gienapp IE, Shawler T, Whitacre CC. Estriol generates tolerogenic dendritic cells in vivo that protect against autoimmunity. J Of Immunol. 2011;186(6):3346–3355. doi: 10.4049/jimmunol.1001322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Relloso M, Aragoneses‐Fenoll L, Lasarte S, Bourgeois C, Romera G, Kuchler K, Corbí AL, Muñoz-Fernández MA, Nombela C, Rodríguez-Fernández JL, et al. Estradiol impairs the Th17 immune response against Candida albicans. J Of Leukocyte Biol. 2011;91(1):159–165. doi: 10.1189/jlb.1110645. [DOI] [PubMed] [Google Scholar]
- 216.Robinson DP, Lorenzo ME, Jian W, Klein SL, Subbarao K. Elevated 17β-estradiol protects females from influenza a virus pathogenesis by suppressing inflammatory responses. PLOS Pathogens. 2011;7(7):e1002149. doi: 10.1371/journal.ppat.1002149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Simin J, Tamimi RM, Engstrand L, Callens S, Brusselaers N. Antibiotic use and the risk of breast cancer: a systematic review and dose-response meta-analysis. Pharmacol Res. 2020;160:105072. doi: 10.1016/j.phrs.2020.105072. [DOI] [PubMed] [Google Scholar]
- 218.Wirtz HS, Buist DS, Gralow JR, Barlow WE, Gray S, Chubak J, Yu O, Bowles EJA, Fujii M, Boudreau DM. Frequent antibiotic use and second breast cancer events antibiotics and second breast cancer events. Cancer Epidemiol, Biomarkers & Prevent. 2013;22(9):1588–1599. doi: 10.1158/1055-9965.EPI-13-0454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Gao Y, Shang Q, Li W, Guo W, Stojadinovic A, Mannion C, Man Y-G, Chen T. Antibiotics for cancer treatment: a double-edged sword. J Cancer. 2020;11(17):5135. doi: 10.7150/jca.47470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Beberok A, Wrześniok D, Rok J, Rzepka Z, Respondek M, Buszman E. Ciprofloxacin triggers the apoptosis of human triple-negative breast cancer MDA-MB-231 cells via the p53/Bax/Bcl-2 signaling pathway. Int J Oncol. 2018;52(5):1727–1737. doi: 10.3892/ijo.2018.4310. [DOI] [PubMed] [Google Scholar]
- 221.Chen T-C, Hsu Y-L, Tsai Y-C, Chang Y-W, Kuo P-L, Chen Y-H. Gemifloxacin inhibits migration and invasion and induces mesenchymal–epithelial transition in human breast adenocarcinoma cells. J Mol Med. 2014;92(1):53–64. doi: 10.1007/s00109-013-1083-4. [DOI] [PubMed] [Google Scholar]
- 222.Boursi B, Mamtani R, Haynes K, Yang Y-X. Recurrent antibiotic exposure may promote cancer formation–another step in understanding the role of the human microbiota? Eur J Public Health Of Cancer. 2015;51(17):2655–2664. doi: 10.1016/j.ejca.2015.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Friedman GD, Oestreicher N, Chan J, Quesenberry CP, Udaltsova N, Habel LA. Antibiotics and risk of breast cancer: up to 9 years of follow-up of 2.1 million women. Cancer Epidem Biomar. 2006;15(11):2102–2106. doi: 10.1158/1055-9965.EPI-06-0401. [DOI] [PubMed] [Google Scholar]
- 224.Kilkkinen A, Rissanen H, Klaukka T, Pukkala E, Heliövaara M, Huovinen P, Männistö S, Aromaa A, Knekt P. Antibiotic use predicts an increased risk of cancer. Intl J Of Cancer. 2008;123(9):2152–2155. doi: 10.1002/ijc.23622. [DOI] [PubMed] [Google Scholar]
- 225.Velicer CM, Heckbert SR, Rutter C, Lampe JW, Malone K. Association between antibiotic use prior to breast cancer diagnosis and breast tumour characteristics (United States). Cancer Cause Control. 2006;17(3):307–313. doi: 10.1007/s10552-005-0445-9. [DOI] [PubMed] [Google Scholar]
- 226.Wirtz HS, Buist DS, Gralow JR, Barlow WE, Gray S, Chubak J, Yu O, Bowles EJA, Fujii M, Boudreau DM. Frequent antibiotic use and second breast cancer events. Cancer Epidem Biomar. 2013;22(9):1588–1599. doi: 10.1158/1055-9965.EPI-13-0454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Garcia Rodriguez L. Use of antibiotics and risk of breast cancer. Am J Epidemiol. 2005;161(7):616–619. doi: 10.1093/aje/kwi087. [DOI] [PubMed] [Google Scholar]
- 228.Tamim H, Hanley J, Hajeer A, Boivin J-F, Collet JP. Risk of breast cancer in relation to antibiotic use. Pharmacoepidemiol Drug Saf. 2008;17(2):144–150. doi: 10.1002/pds.1512. [DOI] [PubMed] [Google Scholar]
- 229.Sørensen HT, Skriver MV, Friis S, McLaughlin J, Blot W, Baron J. Use of antibiotics and risk of breast cancer: a population-based case–control study. Br J Cancer. 2005;92(3):594. doi: 10.1038/sj.bjc.6602313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Zackular JP, Baxter NT, Iverson KD, Sadler WD, Petrosino JF, Chen GY, Schloss PD, Blaser MJ. The gut microbiome modulates colon tumorigenesis. MBio. 2013;4(6):e00692–00613. doi: 10.1128/mBio.00692-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Katsidzira L, Ocvirk S, Wilson A, Li J, Mahachi C, Soni D, DeLany J, Nicholson JK, Zoetendal EG, O’Keefe SJD. Differences in fecal gut microbiota, short-chain fatty acids and bile acids link colorectal cancer risk to dietary changes associated with urbanization among Zimbabweans. Nutrit And Cancer. 2019;71(8):1313–1324. doi: 10.1080/01635581.2019.1602659. [DOI] [PubMed] [Google Scholar]
- 232.Iida N, Mizukoshi E, Yamashita T, Terashima T, Arai K, Seishima J, Kaneko S. Overuse of antianaerobic drug is associated with poor postchemotherapy prognosis of patients with hepatocellular carcinoma. Intl J Of Cancer. 2019;145(10):2701–2711. doi: 10.1002/ijc.32339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Coker OO, Dai Z, Nie Y, Zhao G, Cao L, Nakatsu G, Wu WK, Wong SH, Chen Z, Sung JJY, et al. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut. 2018;67(6):1024–1032. doi: 10.1136/gutjnl-2017-314281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Kharrat N, Assidi M, Abu-Elmagd M, Pushparaj PN, Alkhaldy A, Arfaoui L, Naseer MI, Omri AE, Messaoudi S, Buhmeida A, et al. Data mining analysis of human gut microbiota links Fusobacterium spp with colorectal cancer onset. Bioinformation. 2019;15(6):372. doi: 10.6026/97320630015372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Guan X, Ma F, Sun X, Li C, Li L, Liang F, Li S, Yi Z, Liu B, Xu B. Gut microbiota profiling in patients with HER2-negative metastatic breast cancer receiving metronomic chemotherapy of capecitabine compared to those under conventional dosage. Front Oncol. 2020;10:902. doi: 10.3389/fonc.2020.00902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Yan A, Culp E, Perry J, Lau JT, MacNeil LT, Surette MG, Wright GD. Transformation of the anticancer drug doxorubicin in the human gut microbiome. ACS Infect Dis. 2018;4(1):68–76. doi: 10.1021/acsinfecdis.7b00166. [DOI] [PubMed] [Google Scholar]
- 237.Ramakrishna C, Corleto J, Ruegger PM, Logan GD, Peacock BB, Mendonca S, Yamaki S, Adamson T, Ermel R, McKemy D, et al. Dominant role of the gut microbiota in chemotherapy induced neuropathic pain. Sci Rep. 2019;9(1):1–16. doi: 10.1038/s41598-019-56832-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Bawaneh A, Wilson AS, Levi N, Howard-McNatt MM, Chiba A, Soto-Pantoja DR, Cook KL. Intestinal microbiota influence doxorubicin responsiveness in triple-negative breast cancer. Cancers. 2022;14(19):4849. doi: 10.3390/cancers14194849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971–976. doi: 10.1126/science.1240537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Daillère R, Vétizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, Duong CM, Flament C, Lepage P, Roberti M, et al. Enterococcus hirae and barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45(4):931–943. doi: 10.1016/j.immuni.2016.09.009. [DOI] [PubMed] [Google Scholar]
- 241.McGee HM, Jiang D, Soto-Pantoja DR, Nevler A, Giaccia AJ, Woodward WA. Targeting the tumor microenvironment in radiation oncology: proceedings from the 2018 ASTRO–AACR research workshop proceedings of ASTRO–AACR workshop on radiation and the TME. Clin Cancer Res. 2019;25(10):2969–2974. doi: 10.1158/1078-0432.CCR-18-3781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton J, Ivashkiv L, Lawrence T, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41(1):14–20. doi: 10.1016/j.immuni.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Colbert L, Previs R, Medrano AYD, Eifel P, Jhingran A, Ramondetta L, Hahn SM, Jazaeri A, Frumovitz M, Schmeler K, et al. Rectal microbiome diversity predicts disease response at completion of radiation therapy for squamous cell carcinoma of the cervix. Intl J Of Radiat Oncol Biol Physics. 2017;99(2):S51. doi: 10.1016/j.ijrobp.2017.06.129. [DOI] [Google Scholar]
- 244.Sheng D, Yue K, Li H, Zhao L, Zhao G, Jin C, Zhang L, Jun S-R. The interaction between intratumoral microbiome and immunity is related to the prognosis of ovarian cancer. Microbiol Spectr. 2023;11(2):e03549–03522. doi: 10.1128/spectrum.03549-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.He J-Y, Wang W-Z, Qi H-Z, Ma Y, He S-Y. Use of recombinant lactobacillus sakei for the prevention and treatment of radiation-induced enteritis. Med Hypotheses. 2018;119:37–40. doi: 10.1016/j.mehy.2018.07.024. [DOI] [PubMed] [Google Scholar]
- 246.Huang R, Xiang J, Zhou P. Vitamin D, gut microbiota, and radiation-related resistance: a love-hate triangle. J Exp Clin Cancer Res. 2019;38(1):1–10. doi: 10.1186/s13046-019-1499-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Nascimento M, Caporossi C, Eduardo aguilar-Nascimento J, Michelon castro-Barcellos H, Teixeira Motta R, Reis Lima S. Efficacy of synbiotics to reduce symptoms and rectal inflammatory response in acute radiation proctitis: a randomized, double-blind, placebo-controlled pilot trial. Nutr And Cancer. 2020;72(4):602–609. doi: 10.1080/01635581.2019.1647254. [DOI] [PubMed] [Google Scholar]
- 248.Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CPM, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi: 10.1126/science.aad1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Dubin K, Callahan MK, Ren B, Khanin R, Viale A, Ling L, No D, Gobourne A, Littmann E, Huttenhower C, Pamer EG. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun. 2016;7(1):1–8. doi: 10.1038/ncomms10391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews M, Karpinets T, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. doi: 10.1126/science.aan4236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Routy B, Le Chatelier E, Derosa L, Duong CP, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi: 10.1126/science.aan3706. [DOI] [PubMed] [Google Scholar]
- 252.Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Man Lei Y, Jabri B, Alegre M-L, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science. 2015;350(6264):1084–1089. doi: 10.1126/science.aac4255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Hrubesz G, Leigh J, Ng TL. Understanding the relationship between breast cancer, immune checkpoint inhibitors, and gut microbiota: a narrative review. Transl Breast Cancer Res. 2024;5:31. doi: 10.21037/tbcr-24-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Di Modica M. Gut microbiota and trastuzumab response in HER2-positive breast cancer. United Kingdom: Open University; 2020. [Google Scholar]
- 255.Pernigoni N, Zagato E, Calcinotto A, Troiani M, Mestre RP, Calì B, Attanasio G, Troisi J, Minini M, Mosole S, et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science. 2021;374(6564):216–224. doi: 10.1126/science.abf8403. [DOI] [PubMed] [Google Scholar]
- 256.Terrisse S, Zitvogel L, Kroemer G. Effects of the intestinal microbiota on prostate cancer treatment by androgen deprivation therapy. Microb Cell. 2022;9(12):190–194. doi: 10.15698/mic2022.12.787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Lasagna A, De Amici M, Rossi C, Zuccaro V, Corbella M, Petazzoni G, Comandatore F, Sacchi L, Testa G, Ferraris E, et al. The bio-diversity and the role of gut microbiota in postmenopausal women with luminal breast cancer treated with aromatase inhibitors: an observational cohort study. Pathogens. 2022;11(12):1421. doi: 10.3390/pathogens11121421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Smith M, Dai A, Ghilardi G, Amelsberg KV, Devlin SM, Pajarillo R, Slingerland JB, Beghi S, Herrera PS, Giardina P. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med. 2022;28(4):713–723. doi: 10.1038/s41591-022-01702-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Tsay J-CJ, Wu BG, Sulaiman I, Gershner K, Schluger R, Li Y, Yie T-A, Meyn P, Olsen E, Perez L, et al. Lower airway dysbiosis affects lung cancer progression. Cancer Discov. 2021;11(2):293–307. doi: 10.1158/2159-8290.CD-20-0263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–563. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Preidis GA, Versalovic J. Targeting the human microbiome with antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics era. Gastroenterology. 2009;136(6):2015–2031. doi: 10.1053/j.gastro.2009.01.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Derosa L, Routy B, Fidelle M, Iebba V, Alla L, Pasolli E, Segata N, Desnoyer A, Pietrantonio F, Ferrere G, et al. Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. European Urol. 2020;78(2):195–206. doi: 10.1016/j.eururo.2020.04.044. [DOI] [PubMed] [Google Scholar]
- 263.Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M-L, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–108. doi: 10.1126/science.aao3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.O’donoghue EJ, Krachler AM. Mechanisms of outer membrane vesicle entry into host cells. Cellular Microbiol. 2016;18(11):1508–1517. doi: 10.1111/cmi.12655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Macia L, Nanan R, Hosseini-Beheshti E, Grau GE. Host- and microbiota-derived extracellular vesicles, immune function, and disease development. Int J Mol Sci. 2020;21(1):107. doi: 10.3390/ijms21010107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Cecil JD, O’Brien-Simpson NM, Lenzo JC, Holden JA, Singleton W, Perez-Gonzalez A, Mansell A, Reynolds EC. Outer membrane vesicles prime and activate macrophage inflammasomes and cytokine secretion in vitro and in vivo. Front Immunol. 2017;8:1017. doi: 10.3389/fimmu.2017.01017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Codemo M, Muschiol S, Iovino F, Nannapaneni P, Plant L, Wai SN, Henriques-Normark B, Rappuoli R. Immunomodulatory effects of pneumococcal extracellular vesicles on cellular and humoral host defenses. MBio. 2018;9(2):e00559–00518. doi: 10.1128/mBio.00559-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Davitt CJ, Petersen HE, Kikendall NL, Lavelle EC, Morici LA. Naturally-derived bacterial nano-particles engage diverse innate receptors, driving the activation of dendritic cells and leading to the establishment of potent adaptive immune responses. Am Assoc Immunol. 2016;196(1_Supplement):76.11. doi: 10.4049/jimmunol.196.Supp.76.11. [DOI] [Google Scholar]
- 269.Kim J, Bin BH, Choi EJ, Lee HG, Lee TR, Cho EG. Staphylococcus aureus -derived extracellular vesicles induce monocyte recruitment by activating human dermal microvascular endothelial cells in vitro. Clin And Exp Allergy: J Br Soc For Allergy And Clin Immunol. 2019;49(1):68–81. doi: 10.1111/cea.13289. [DOI] [PubMed] [Google Scholar]
- 270.Perez Vidakovics MLA, Jendholm J, Mörgelin M, Månsson A, Larsson C, Cardell L-O, Riesbeck K, Stevenson FK. B cell activation by outer membrane vesicles—a novel virulence mechanism. PLOS Pathogens. 2010;6(1):e1000724. doi: 10.1371/journal.ppat.1000724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Chronopoulos A, Kalluri R. Emerging role of bacterial extracellular vesicles in cancer. Oncogene. 2020;39(46):6951–6960. doi: 10.1038/s41388-020-01509-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Jones EJ, Booth C, Fonseca S, Parker A, Cross K, Miquel-Clopés A, Hautefort I, Mayer U, Wileman T, Stentz R, et al. The uptake, trafficking, and biodistribution of bacteroides thetaiotaomicron generated outer membrane vesicles. Front Microbiol. 2020;11:57. doi: 10.3389/fmicb.2020.00057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Chen CC, Liu L, Ma F, Wong CW, Guo XE, Chacko JV, Farhoodi HP, Zhang SX, Zimak J, Ségaliny A, et al. Elucidation of exosome migration across the blood–brain barrier model in vitro. Cel Mol Bioeng. 2016;9(4):509–529. doi: 10.1007/s12195-016-0458-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Tulkens J, Vergauwen G, Van Deun J, Geeurickx E, Dhondt B, Lippens L, De Scheerder M-A, Miinalainen I, Rappu P, De Geest BG, et al. Increased levels of systemic LPS-positive bacterial extracellular vesicles in patients with intestinal barrier dysfunction. Gut. 2020;69(1):191–193. doi: 10.1136/gutjnl-2018-317726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet–induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–1481. doi: 10.2337/db07-1403. [DOI] [PubMed] [Google Scholar]
- 276.Chelakkot C, Choi Y, Kim D-K, Park HT, Ghim J, Kwon Y, Jeon J, Kim M-S, Jee Y-K, Gho YS, et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp Mol Med. 2018;50(2):e450–e450. doi: 10.1038/emm.2017.282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Sadovska L, Santos CB, Kalniņa Z, Linē A. Biodistribution, uptake and effects caused by cancer-derived extracellular vesicles. J Educ Chang Circulating Biomarkers. 2015;4:2. doi: 10.5772/60522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Harris DA, Patel SH, Gucek M, Hendrix A, Westbroek W, Taraska JW, Cao J. Exosomes released from breast cancer carcinomas stimulate cell movement. PLOS ONE. 2015;10(3):e0117495. doi: 10.1371/journal.pone.0117495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Menck K, Scharf C, Bleckmann A, Dyck L, Rost U, Wenzel D, Dhople VM, Siam L, Pukrop T, Binder C, et al. Tumor-derived microvesicles mediate human breast cancer invasion through differentially glycosylated EMMPRIN. J Molecular Cell Biol. 2015;7(2):143–153. doi: 10.1093/jmcb/mju047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Eichelser C, Stückrath I, Müller V, Milde-Langosch K, Wikman H, Pantel K, Schwarzenbach H. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget. 2014;5(20):9650. doi: 10.18632/oncotarget.2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Galindo-Hernandez O, Villegas-Comonfort S, Candanedo F, Gonzalez-Vazquez M-C, Chavez-Ocana S, Jimenez-Villanueva X, Sierra-Martinez M, Salazar EP. Elevated concentration of microvesicles isolated from peripheral blood in breast cancer patients. Archives Of Med Res. 2013;44(3):208–214. doi: 10.1016/j.arcmed.2013.03.002. [DOI] [PubMed] [Google Scholar]
- 282.Khan S, Bennit HF, Turay D, Perez M, Mirshahidi S, Yuan Y, Wall NR. Early diagnostic value of survivin and its alternative splice variants in breast cancer. BMC Cancer. 2014;14(1):1–10. doi: 10.1186/1471-2407-14-176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Amorim M, Fernandes G, Oliveira P, Martins‐de‐Souza D, Dias‐Neto E, Nunes D. The overexpression of a single oncogene (ERBB2/HER2) alters the proteomic landscape of extracellular vesicles. Proteomics. 2014;14(12):1472–1479. doi: 10.1002/pmic.201300485. [DOI] [PubMed] [Google Scholar]
- 284.Lv M-M, Zhu X-Y, Chen W-X, Zhong S-L, Hu Q, Ma T-F, Zhang J-H, Chen L, Tang J-H, Zhao J-H. Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein. Tumor Biol. 2014;35(11):10773–10779. doi: 10.1007/s13277-014-2377-z. [DOI] [PubMed] [Google Scholar]
- 285.Ozawa PMM, Alkhilaiwi F, Cavalli IJ, Malheiros D, de Souza Fonseca Ribeiro EM, Cavalli LR. Extracellular vesicles from triple-negative breast cancer cells promote proliferation and drug resistance in non-tumorigenic breast cells. Breast Cancer Res Treat. 2018;172(3):713–723. doi: 10.1007/s10549-018-4925-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Wei Y, Lai X, Yu S, Chen S, Ma Y, Zhang Y, Li H, Zhu X, Yao L, Zhang J. Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res Treat. 2014;147(2):423–431. doi: 10.1007/s10549-014-3037-0. [DOI] [PubMed] [Google Scholar]
- 287.McQuade JL, Daniel CR, Helmink BA, Wargo JA. Modulating the microbiome to improve therapeutic response in cancer. The Lancet Oncol. 2019;20(2):e77–e91. doi: 10.1016/S1470-2045(18)30952-5. [DOI] [PubMed] [Google Scholar]
- 288.Kaparakis-Liaskos S, Ferrero RL. Immune modulation by bacterial outer membrane vesicles. Nat Rev Immunol. 2015;15(6):375–387. doi: 10.1038/nri3837. [DOI] [PubMed] [Google Scholar]
- 289.Shen Y, Torchia MLG, Lawson GW, Karp CL, Ashwell JD, Mazmanian SK. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host & Microbe. 2012;12(4):509–520. doi: 10.1016/j.chom.2012.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Kim OY, Park HT, Dinh NTH, Choi SJ, Lee J, Kim JH, Lee S-W, Gho YS. Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response. Nat Commun. 2017;8(1):1–9. doi: 10.1038/s41467-017-00729-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Behzadi E, Hosseini HM, Fooladi AAI. The inhibitory impacts of lactobacillus rhamnosus GG-derived extracellular vesicles on the growth of hepatic cancer cells. Microbial Pathogenesis. 2017;110:1–6. doi: 10.1016/j.micpath.2017.06.016. [DOI] [PubMed] [Google Scholar]
- 292.An J, Kim JB, Yang EY, Kim HO, Lee W-H, Yang J, Kwon H, Paik NS, Lim W, Kim Y-K, et al. Bacterial extracellular vesicles affect endocrine therapy in MCF7 cells. Med (baltimore). 2021;100(18):e25835. doi: 10.1097/MD.0000000000025835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.An J, Kwon H, Lim W, Moon B-I. Staphylococcus aureus-derived extracellular vesicles enhance the efficacy of endocrine therapy in breast cancer cells. J Clin Med. 2022;11(7):2030. doi: 10.3390/jcm11072030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Chen L, Ma X, Liu W, Hu Q, Yang H. Targeting pyroptosis through lipopolysaccharide-triggered noncanonical pathway for safe and efficient cancer immunotherapy. Nano Lett. 2023;23(18):8725–8733. doi: 10.1021/acs.nanolett.3c02728. [DOI] [PubMed] [Google Scholar]
- 295.Guo Q, Li X, Zhou W, Chu Y, Chen Q, Zhang Y, Li C, Chen H, Liu P, Zhao Z, et al. Sequentially triggered bacterial outer membrane vesicles for macrophage metabolism modulation and tumor metastasis suppression. ACS Nano. 2021;15(8):13826–13838. doi: 10.1021/acsnano.1c05613. [DOI] [PubMed] [Google Scholar]
- 296.Rezaei Adriani R, Mousavi Gargari SL, Bakherad H, Amani J. Anti-EGFR bioengineered bacterial outer membrane vesicles as targeted immunotherapy candidate in triple-negative breast tumor murine model. Sci Rep. 2023;13(1):16403. doi: 10.1038/s41598-023-43762-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
