Skip to main content
Erschienen in: Cancer and Metastasis Reviews 4/2021

Open Access 30.12.2021

The involvement of oncobiosis and bacterial metabolite signaling in metastasis formation in breast cancer

verfasst von: Tünde Kovács, Edit Mikó, Gyula Ujlaki, Heba Yousef, Viktória Csontos, Karen Uray, Peter Bai

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 4/2021

Abstract

Breast cancer, the most frequent cancer in women, is characterized by pathological changes to the microbiome of breast tissue, the tumor, the gut, and the urinary tract. Changes to the microbiome are determined by the stage, grade, origin (NST/lobular), and receptor status of the tumor. This year is the 50th anniversary of when Hill and colleagues first showed that changes to the gut microbiome can support breast cancer growth, namely that the oncobiome can reactivate excreted estrogens. The currently available human and murine data suggest that oncobiosis is not a cause of breast cancer, but can support its growth. Furthermore, preexisting dysbiosis and the predisposition to cancer are transplantable. The breast’s and breast cancer’s inherent microbiome and the gut microbiome promote breast cancer growth by reactivating estrogens, rearranging cancer cell metabolism, bringing about a more inflammatory microenvironment, and reducing the number of tumor-infiltrating lymphocytes. Furthermore, the gut microbiome can produce cytostatic metabolites, the production of which decreases or blunts breast cancer. The role of oncobiosis in the urinary tract is largely uncharted. Oncobiosis in breast cancer supports invasion, metastasis, and recurrence by supporting cellular movement, epithelial-to-mesenchymal transition, cancer stem cell function, and diapedesis. Finally, the oncobiome can modify the pharmacokinetics of chemotherapeutic drugs. The microbiome provides novel leverage on breast cancer that should be exploited for better management of the disease.
Hinweise

Publisher's note

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

1 Introduction

Dysbiosis is an abnormal adaptation of the microbiome, characterized by abnormal microbial composition and function. Neoplastic diseases are characterized by dysbiosis that is coined oncobiosis [1]. The microbiome that is undergoing oncobiotic transformation is termed the oncobiome. Oncobiosis occurs in multiple neoplasias, including breast cancer, and oncobiosis may have a pathogenic role in these cancers [28]. In this review, we will dissect the microbiome-elicited pathways and discuss how these pathways protect against metastasis formation in breast cancer.
Breast cancer is the most frequent cancer among women and is the leading cause of cancer-related deaths in women [9, 10]. Nevertheless, in developed countries, the 5-year survival of breast cancer is above 80% due to population-wide screening programs and the consequent early identification [11]. Although several risk factors were identified that increase the risk for breast cancer, most newly diagnosed patients have no obvious risk factors [12]. The risk for breast cancer increases with age, and most breast cancer patients are diagnosed in their 50 s after menopause. Extended exposure to female hormones due to hormone-replacement therapy, early menarche, and late menopause are risk factors for breast cancer [12]. BRCA1 and BRCA2 genes were identified as genetic risk factors for breast cancer, although mutation carriers represent a minority among breast cancer patients [13]. A family history of breast cancer or neoplasias is also a risk factor for breast cancer [12] and dense breast [14, 15]. Successful pregnancies, lactation, and physical activity are protective factors against the disease [12, 16]. For further information, we refer the readers to the relevant guidelines [1720]. We reference the latest versions of the guidelines. Nevertheless, we ask the readers to check whether the guidelines were updated at the time our paper is being read.

2 Oncobiosis in breast cancer

The first mention of the possible pathological role of the oncobiome in breast cancer dates back to 1971 [21]. The causative role of oncobiosis in the pathogenesis of breast cancer is underscored by the observations that antibiotic use increases the risk for breast cancer in mice [2224], and the majority of studies suggest an increased risk in humans, also [2533] (it should be noted that [34] and [35] found no association between antibiotic exposure and breast cancer risk). In further support of the pathological role of the microbiome, prebiotics [36], probiotics [3745], and diverse nutrition [4649] reduce the risk of breast cancer. Furthermore, risk factors of breast cancer, such as high-density breast [50], early menarche [51], low physical activity [51], increases in BMI [51, 52], age [53], and alcohol consumption [54], are also associated with microbiome changes culminating in breast cancer–associated oncobiosis.
Multiple microbial compartments undergo oncobiotic transformation during breast cancer, including breast tissue [55, 56], milk ducts [57], the inherent microbiome of the breast carcinoma [54, 5872], the distal gut [51, 52, 7389], and the urinary microbiome [54, 90]. However, no differences in the microbiome of the nipple [57, 69] and the oral microbiome [54] between healthy individuals and breast cancer patients have been detected. Besides bacteria, viruses (parapoxviruses [91], human papillomavirus [92], Herpesviridae, Retroviridae, Parapoxviridae, Polyomaviridae, Papillomaviridae [64]), fungi, and parasites were identified in breast cancer tissue [64, 65, 70], although these signatures are not ubiquitous in all individuals. Of note, microbiome signatures in the oncobiome correlate with survival in breast cancer, which underlines the importance of oncobiotic transformation in regulating breast cancer behavior [70]. The microbiome is now considered a component of the tumor microenvironment [93].
In the following chapters, we will discuss changes in the microbiome of different compartments. In each compartment, the results can be contradictory, so we identified common elements that are discussed in the chapters discussing the respective compartments. The findings of the individual studies are assembled in Table 1. The bilateral mechanistic connections between the host and the microbiome [94] and oncobiosis support of cancer cells (Fig. 1) are discussed in the following chapters.
Table 1
Changes to the gut microbiome in breast cancer
Patient cohort
Mode of analysis
Changes to the microbiome, biomarker observations, diversity
Reference
Changes to the breast tissue microbiome
Breast tissue obtained from surgery of benign tumors (n = 13), cancerous (n = 45), and healthy breast tissue (n = 23)
16S rRNA gene sequencing
The bacterial composition of the healthy breast tissue and breast cancer tissue is different. Higher abundances of Prevotella, Lactococcus, Streptococcus, Corynebacterium, and Micrococcus in healthy breast tissue and Bacillus, Staphylococcus, unclassified Enterobacteriaceae, unclassified Comamonadaceae, and unclassified Bacteroidetes in breast cancer tissue
[55]
15 malignant cancer (stages I and II) and 13 benign atypia patients
16S rRNA gene sequencing
No significant differences in alpha diversity values, but beta diversity differs between the breast tissue of malignant and benign breast tissue. Fusobacterium, Atopobium, Hydrogenophaga, Gluconacetobacter, and Lactobacillus abundance increased in the tissue of the malignant cases
[56]
141 breast tissue samples from BC patients
Enterococcus abundance plays a vital role in regional recurrence
[95]
Changes to the milk duct microbiome
Nipple aspirate fluid from breast cancer surviving patients (n = 6) and healthy controls (n = 9)
16S rRNA gene sequencing
Beta diversity, but not the alpha diversity, is different between breast cancer patients and healthy controls. Alistipes was present only in the nipple aspirate from breast cancer patients, while unclassified Sphingomonadaceae genus was enriched in the nipple aspirate of healthy controls
[57]
Changes to the breast carcinoma microbiome
Percutan needle biopsy from 22 benign and 72 malignant breast cancer patients
16S rRNA gene sequencing
Slightly higher alpha diversity in patients with malignant disease. Proteobacteria increased in malignant cases. The genus Propionicimonas and the families Micrococcaceae, Caulobacteraceae, Rhodobacteraceae, Nocardioidaceae, and Methylobacteriaceae increased in the malignant disease group
[58]
8 normal breast tissues, 64 breast tumors, in 11 cases paired non-cancerous adjacent tissue
16S rRNA gene sequencing
Alpha diversity and beta diversity indices were lower in the tumor tissue. Clostridia, Bacteroidia, WPS_2, Ruminococcaceae, Fusobacteria, and Spirochetes increased, while Pseudomonadaceae, Sphingomonadaceae, Caulobacteraceae, Thermi, and Actinobacteria decreased in tumors. Streptococcaceae and Ruminococcus were abundant in TNBC tumors; Xanthomonadales in Luminal A; Clostridium in Luminal B; Akkermansia. Ruminococcaceae and genus Hyphomicrobium were more abundant in stage I Her2 + tumors, Sporosarcina in stage II, Bosea in stage III + IV
[59]
221 breast cancer specimens, breast tissue from 18 individuals predisposed to breast cancer, and 69 controls
16S rRNA gene sequencing
Alpha diversity values were lower in tumors and the breast tissue of the risk population. Widespread association with stage, lobular or ductal origin, and hormone receptor positivity
[60]
Cancerous tissue and adjacent healthy tissue from 16 breast cancer patients
16S rRNA gene sequencing
No significant differences in alpha diversity. No difference between the cancerous and the adjacent healthy tissues
[61]
BC tumor and adjacent normal tissue from 6 + 10 TNBC WNH and 7 TNBC BNH, 7 TPBC WNH, and 3 TPBC BNH
16S rRNA gene sequencing
In triple-positive and triple-negative breast cancer from black non-Hispanic alpha indices decrease, in white non-Hispanic women alpha indices increase
Fusobacteria and Streptococcus abundance increase in the tumor. There is a difference between the microbiome composition of triple-positive and triple-negative breast cancers
[63]
10 archived breast cancer tumor tissue, 10 freshly excised normal breast tissue, 8 of them from both breasts
16S rRNA gene sequencing
Ruminococcaceae, Akkermansia, Verrucomicrobia increased, while Sutterella, Acinetobacter Bacteroides, Cyanobacteria, Proteobacteria, Synergistetes, and Tenericutes decreased in the tumor tissue. Alpha index decreased in the tumor tissue
[62]
44 BC patients and 20 controls. Significant age and body mass index difference between cohorts
16S rRNA gene sequencing
No significant difference in alpha diversity. Methylobacterium decreased in cancer patients and was drastically reduced when invasion was reported. In cancer patients, levels of gram-positive organisms including Corynebacterium, Staphylococcus, Actinomyces, and Propionibacteriaceae increased
[54]
100 TNBC, 17 matched, 20 non-matched controls
PathoChip technology
Higher association of Brevundimonas diminuta, Arcanobacterium haemolyticum, Peptoniphilus indolicus, Prevotella nigrescens, Propiniobacterium jensenii, and Capnocytophaga canimorsus and a set of viruses and fungi are associated with TNBC samples compared to normal tissues
[64]
50 BRER, 34 BRHR, 24 BRTP, 40 TNBC, 20 controls
PathoChip technology
BRER is characterized by Arcanobacterium, Bifidobacterium, Cardiobacterium, Citrobacter, and Escherichia species; BRTP is characterized by Bordetella, Campylobacter, Chlamydia, Chlamydophila, Legionella, and Pasteurella; BRHR is characterized by Streptococcus; TNBC is characterized by Arcobacter, Geobacillus, Orientia, and Rothia
[65]
Healthy (age-matched) (n = 23), paired normal (n = 39), and tumor tissue (n = 39)
16S rRNA gene sequencing
Bacterial copy number decreased in tumors and with increased grade. Sphinomonadaceae family, Sphingomonas species decreased, while the Methylobacteriaceae family, Methylobacterium species increased in tumors
[66]
668 breast tumor and 72 non-cancerous breast tumor sequences from the TCGA data portal
Salmonella enterica, Escherichia coli, Bacillus alcalophilus, Brachybacterium muris, Plesonius fermentans, Mycobacterium phlei, and Acinetobacter radioresistens increased, while Microbacterium barkeri, Acinetobacterium baumannii, Ralstonia pickettii, Lactobacillus rossiae, and Mycobacterium fortuitum decreased in the tumors
[68]
256 normal tissue and 355 breast tumors
Bacterial LPS and 16S RNA were detected in breast cancer cells in breast tumors. The microbiome of the breast tumors was richer than other tumors assessed and in normal adjacent breast tissue. Proteobacteria, Firmicutes, and Actinobacteria can be found in breast tumors. Differences in ER + and ER- breast tumors
[67]
21 female and 2 male BC patients
16S rRNA gene sequencing
Compared to normal breast tissue, the abundance of Proteobacteria increased in tumor tissue
[69]
95–105 FFPE samples for each BC subtype, 86 controls
Pathochip
Large set of viruses, parasites, and fungi were detected in FFPE sections of breast cancer. The least of these were detected in TNBC cases
[70]
16 healthy controls, 32 breast cancer patients
16S rRNA gene sequencing
The abundance of Corynebacterium, Prevotella, and Gammaproteobacteria (unclassified) decreased, while Acinetobacter increased in BC tissue
[71]
Changes to the gut microbiome
48 postmenopausal BC patients (most stages 0–I), vs. 48 control patients
16S rRNA gene sequencing
Breast cancer patients had a higher abundance of Clostridium, Faecalibacterium, and Ruminococcus (all Clostridiales) and a lower abundance of Dorea and Lachnospiraceae
Lower number of observed species, Chao1 and PD whole tree indices in breast cancer patients
[73]
30 BC and 36 control patients
Classical bacterial culture
Fecal bile acid levels were lower in breast cancer patients. Bacterial nuclear dehydrogenating activity increased in breast cancer patients suggesting increases in Clostridia abundance in feces
[82]
18 premenopausal BC patients, 25 premenopausal controls, 44 postmenopausal BC patients, 46 postmenopausal healthy controls
Comprehensive shotgun sequencing
Species number, chao1, and JSD values were higher in postmenopausal cancer patients than in controls. Widespread taxonomical changes in BC patients
[75]
379 BC patients, 102 non-malignant breast disease, 414 population-based controls
16S rRNA gene sequencing
Alpha diversity indices correlate negatively with the odds for BC, BC grade, and subtype. No difference in the microbiome of malignant and non-malignant patients, but differ when compared to controls. Bacteroides, Flavonifractor, and Ruminococcaceae strongly and positively associated with BC, while Rombutsia, Coprococcus 2, Christensenellaceae R-7 group, Dorea, [Eubacterium] coprostanoligenes group, Pseudobutyrivibrio, and Lachnospira negatively associated with BC
[74]
32 overweight stage 0–II BC patients
16S rRNA gene sequencing
Akkermansia high abundance patients had higher alpha diversity compared to low abundance patients
[52]
31 female BC patients
16S rRNA gene sequencing
Total bacterial count decreased in overweight patients. The abundance of Firmicutes, Faecalibacterium prausnitzii, Blautia sp., and Eggerthella lenta decreased in overweight patients. Blautia sp. abundance increased as a function of tumor grade. Bacteroidetes, Clostridium coccoides, Clostridium leptum, Faecalibacterium prausnitzii, and Blautia sp. increased in stage II–III patients compared to stage 0–I patients
[77]
37 incident BC patients
16S rRNA gene sequencing
Early menarche patients had lower Chao1 and OTU indices and lower Firmicutes abundance. Lower OTU, Chao1, and Shannon indices; lower Blautia, Coprococcus, Ruminococcus, and Oscillospira; and higher Firmicutes abundance in Her2 − cases compared to Her2 + cases. Clostridium and Vellionella increased in grade III patients. High total body fat was associated with lower Chao1 and OTU indices
[51]
48 postmenopausal BC case patients (most stages 0–I), vs 48 control patients (same as [73])
qPCR of specific loci
Abundance of DNA coding for the baiH gene of Clostridium sordelli, Bacteroides thetaiotaomicron, Escherichia coli, Pseudomonas putida, and Staphylococcus haemolyticus decreased in breast cancer patients, the most pronounced changes were detected in in situ carcinoma patients
[78]
48 postmenopausal BC case patients (most stages 0–I), vs 48 control patients (same as [73])
qPCR of specific loci
Abundance of DNA coding for the CadA gene of Escherichia coli and the LdcC gene of Escherichia coli, Enterobacter cloacae, and Hafnia alvei decreased in breast cancer patients, the most pronounced changes were detected in in situ carcinoma patients
[79]
3 control and 4 stage I BC patients
Western blotting
Fecal expression of Escherichia coli LdcC protein was lower in stage I patients
48 postmenopausal BC case patients (most stages 0–I), vs 48 control patients (same as [73])
qPCR of specific loci
Abundance of DNA coding for the TnaA gene of Alistipes shahii, Providencia rettegri, Bacteroides xylanisolvens, and Clostridium sp. decreased in breast cancer patients, the most pronounced changes were detected in in situ carcinoma patients
[80]
35 BC patients
Western blotting
Fecal expression of Escherichia coli TnaA was higher in patients with tumor-infiltrating lymphocyte (TIL) ratio over 20% compared to that of those below. Fecal E. coli TnaA expression correlated with TIL percentage
35 BC cases
Western blotting
Fecal expression of TnaA Escherichia coli LdcC protein was lower in lobular cases
[81]
48 postmenopausal BC cases, 48 control
16S rRNA gene sequencing
Lower alpha diversity in breast cancer patients. Lower alpha diversity among IgA-coated bacteria
[83]
124 BC survivor patients
16S rRNA gene sequencing
Abundance of Actinobacteria (Bifidobacterium) was associated with increased levels of DHA. Abundance of Bacteroidetes negatively correlated with EPA levels that were abrogated in patients receiving chemotherapy
[85]
30 controls vs. 25 BC cases
16S rRNA gene sequencing
In breast cancer patients, Bacteroidetes phylum, Clostridium cluster IV, Clostridium cluster XIVa, and Blautia sp. decreased, and Firmicutes phylum increased. No difference in the total number of bacteria. Alpha diversity increased in patients
[84]
200 BC patients (stages I–II) and 67 controls
16S rRNA gene sequencing
Alpha diversity lower in premenopausal patients, no difference in the postmenopausal cohort; beta diversity is different. Bacteroidetes proportions increased in BC patients
[87]
76 BC patients (35 stage II/III, 21 stage I), 336 healthy volunteers
Comprehensive shotgun sequencing
52 units, mostly at the species level, decreased, while 38 units increased in breast cancer patients compared to healthy volunteers. 11 species increased in stage II/III patients, while 21 species increased in stage I patients
[86]
83 invasive BC patients, 19 patients with benign breast tumors
16S rRNA gene sequencing
No difference in alpha and beta diversity indices. The abundance of Clostridium, Faecalibacterium, Lachnospira, Erysipelotrichaceae, Romboutsia, Fusicatenibacter, Xylophilus, and Arcanobacterium decreased, while the abundance of Citrobacter increased in malignant BC patients. Distinct patterns identified BC subtypes and a microbial pattern associated with highly proliferative tumors
[89]
Changes to the urinary microbiome
44 BC patients and 20 controls. Significant age and body mass index difference between cohorts
16S rRNA gene sequencing
Cancer patients had significantly higher Shannon index. Peri/postmenopausal urinary microbiome had higher Shannon index compared to premenopausal samples. Corynebacterium, Staphylococcus, Actinomyces, and Propionibacteriaceae abundance increased in cancer patients
[54]
220 controls and 127 BC patients
16S rRNA gene sequencing of the bacterial extracellular vesicles
The abundance of Bacteroides and Ruminococcaceae-derived extracellular vesicles were higher in the breast cancer group, while Clostridiales and Pseudomonas-derived extracellular vesicles were more abundant in healthy controls
[90]
Abbreviations: BRHR, human epidermal growth factor receptor 2 positive; BMI, body mass index; BNH, African American as Black non-Hispanic; BRER, estrogen or progesterone receptor positive; BRTP, estrogen, progesterone, and HER2 receptor positive; EPA, eicosapentaenoic acid; FFPE, formalin-fixed paraffin-embedded; HER2, epidermal growth factor receptor 2; IDC, invasive ductal carcinoma; ILC, invasive lobular carcinoma; yrs, years; TIL, tumor-infiltrating lymphocyte; TNBC, triple-negative breast cancer; TPBC, triple-positive breast cancer; rRNA, ribosomal RNA; WNH, White non-Hispanic

3 Interactions between the oncobiome, tumors, and tumor cells

3.1 Tumor colonization

The breast tissue has its own microbiome that has higher diversity than that of the vagina, but has lower diversity than that of the oral cavity or the gut [96]. The microbiomes of the breast skin and the inner breast tissues are different [56]. The microbiome of the exterior surface of the breast (e.g., the nipple) does not change in breast cancer [57]. Nevertheless, the composition of the microbiome of the milk ducts changes in the presence of a malignant process, evidenced in nipple aspirates [57].
Next-generation sequencing methods identified bacterial DNA in breast tumors [66] that was confirmed later by alternative methods [64, 65, 67]. Breast tumor has a higher bacterial load than melanoma, lung tumors, ovarian cancer, and glioblastoma and has similar counts to pancreas and bone cancers [67]. The carcinoma tissue is colonized by bacteria [54, 5860, 62, 65, 67, 68, 70], and differences were detected among the subtypes of breast cancer as a function of hormone receptor status [54, 59, 60, 6365, 70], HER2 receptor status [65], invasiveness [54], grade [54, 58, 60, 66, 70], stage [59, 60, 70], and immunological signatures [60]. Tumor-associated bacteria are culturable and class among Proteobacteria, Firmicutes, and Actinobacteria [67]. In a fraction of breast cancer cases, intracellular and perinuclear bacteria were identified [67]. Furthermore, fungi, parasites, and viruses [64, 70, 91, 92] were detected in tumor tissue. Racial differences were identified in the breast tissue and tumor microbiome [59, 63]. Of note, certain components of the intratumoral and tissular microbiome correlate with patient survival [70].
What are the functional contributions of the breast microbiome to tumorigenesis and tumor progression? As LPS + , Gram negative bacteria were detected intratumorally [67], intratumoral bacteria likely have a key role in the local immune response (see in a later chapter). In agreement with this concept, the expression of bacterial LPS biosynthetic genes were upregulated in nipple fluid aspirate [57]. Lipoteichoic acid (LTA), a marker for Gram positive bacteria, was absent in breast cancer [67].
Imputed pathway analysis revealed relevant functions for the breast microbiome (Table 2). As with other features, there are discrepancies between the studies. Nevertheless, the identified pathways can be classified into logical categories that can be linked to tumorigenesis. Furthermore, certain predicted metabolic pathways are similar to the pathway associated with carcinogenesis in breast tissue [62].
Table 2
Changes to the function of the oncobiome in breast cancer deducted from imputed pathway analyses
Bacterial pathway
Study
Stage I tumors were enriched in energy metabolism, fat digestion, and absorption
[59]
Stage II tumors are enriched in phosphotransferase system proteins
Increased in benign cases:
[56]
Cysteine metabolism
Methionine metabolism
Glycosyltransferases
Fatty acid biosynthesis
Branched dibasic acid metabolism
Increased in malignant cases:
Drug metabolism (other enzymes)
Inositol phosphate metabolism
Upregulated in breast cancer cases:
[67]
Dermatan sulfate degradation
Indole acetate biosynthesis
L-ascorbate biosynthesis II (L-glucose pathway)
Mycothiol biosynthesis
Upregulated in breast cancer cases:
[55]
Colibactin biosynthesis
Upregulated in breast cancer cases:
[57]
Flavone and flavonol biosynthesis (incl. beta-glucuronidase)
Isoflavonoid biosynthesis
Flavonoid biosynthesis
Steroid hormone biosynthesis
Synthesis and degradation of ketone bodies
Tryptophan metabolism
Sulfur metabolism
Lipopolysaccharide biosynthesis
Sphingolipid metabolism
Polycyclic aromatic hydrocarbon degradation
Glycine, serine, and threonine biosynthesis
Oxidative phosphorylation
Benzoate degradation
Phenylalanine biosynthesis
Peptidoglycan biosynthesis
Linoleic acid biosynthesis
Nitrogen metabolism
Upregulated in breast cancer cases:
[62]
Base excision repair
Th17 cell differentiation
Choline metabolism
Central carbon metabolism
Necroptosis, microRNAs involved in carcinogenesis
Proteoglycans involved in carcinogenesis
Signaling pathways including IL-17, PI3K-Akt, HIF-1, and AMPK
DNA coding for beta-glucuronidase enzymes (KEGG ortholog Beta-Glucuronidase K01195), responsible for conjugated estrogen reactivation, was elevated in the nipple aspirate fluid from breast cancer patients [57], suggesting a possible local reactivation of estrogens. In patients, Roseburia, Rikenellaceae, Bacteroides uniformis, Paenibacillus amylolyticus, and Ellin6075 were the OTUs contributing to increased beta-glucuronidase abundance [57]. The same study showed an increase in the abundance of genes coding for steroid hormone biosynthesis [57].
Breast cancer cells are characterized by lower oxidative stress than the parent terminal lobular-ductal unit (TLDU) tissue [97]. The low level of oxidative and nitrosative stress is a key feature of successful tumor growth [98, 99]. Tissular and intratumor bacteria can support low tumoral oxidative and nitrosative stress by upregulating L-ascorbate biosynthesis II (L-glucose pathway) [67]. This is further strengthened by increased mycothiol biosynthesis in ER + tumors [67]. Mycothiol is used by bacteria to detoxify reactive oxygen species [100]. In addition to oxidative/nitrosative stress, the physical presence of bacteria can also induce DNA damage, at least in part, due to the overexpression of colibactin [55]. Among Enterobacteriaceae, Escherichia coli or Staphylococcus can produce a toxin, colibactin [55, 101, 102]. In agreement with this concept, Klann and colleagues [62] demonstrated a correlation between the expression of base excision repair genes and the bacterial colonization of the breast tissue.
Metabolic changes were described in the imputed pathway analyses. Of note, some reports show contradictory results. Changes affect core metabolic pathways, including anaerobic respiration [67], oxidative phosphorylation [57], and central carbon metabolism [62]; stage I tumors were enriched in genes of energy metabolism [59]. Another set of changes affected lipid metabolites, such as sphingolipid metabolism [57], synthesis and degradation of ketone bodies [57], linoleic acid biosynthesis [57], and choline metabolism [62]. In benign cases, fatty acid biosynthesis and branched dibasic acid metabolism increased [56], while in stage I tumors, genes of fat digestion and absorption were enriched [59]. A large set of amino acid metabolic pathways were affected (tryptophan glycine, serine, threonine, phenylalanine biosynthesis [57]), as well as nitrogen metabolism [57]. In benign cases, cysteine and methionine metabolism genes were enriched [56]. Changes were described in detoxification processes, such as polycyclic aromatic hydrocarbon degradation [57] and benzoate degradation [57] in the oncobiome. In malignant cases, local, oncobiome-mediated drug metabolism may increase [56]. Interestingly and importantly, signal transduction pathways such as PI3K-Akt [62], HIF-1 [62] and the AMPK pathway [62], microRNAs involved in carcinogenesis [62], and inositol phosphate metabolism [56] changed in the breast cancer oncobiome. In stage 2 tumors, the microbiome was enriched in phosphotransferase system protein genes [59]. Furthermore, sulfur metabolism genes changed in the breast cancer oncobiome [57] and glycosyltransferases increased in benign cases [56]. Breast cancer is characterized by changes in cell metabolism that is actionable for disease treatment and management [103112]. Furthermore, the intricate supportive metabolic circuit between cancer cells and non-cancerous stroma cells can facilitate tumor growth and lead to worse clinical outcomes [103105, 113]. Giallourou and colleagues [71] showed that breast bacteria interfere with the biosynthesis of ceramide, cholesterol, oxidized cholesteryl esters, diacylglycerol, lysophosphatidylcholine, phosphatidylethanolamines, and phosphatidylcholines to modulate the lipid composition of tumors. Of note, cholesterol and lipid homeostasis also play a role [4, 114116]. The widespread changes to the oncobiome metabolism suggest that the breast cancer oncobiome participates in the metabolic support of rapidly dividing breast cancer cells.
In terms of cell death and cell division, the breast cancer oncobiome is associated with necroptosis [62]. Haemophilus influenzae is associated with mitotic spindle formation and G2M checkpoint regulation [68]. The breast cancer oncobiome is also associated with movement and metastasis-related processes, such as dermatan sulfate degradation [67], peptidoglycan biosynthesis [57], and proteoglycan homeostasis [62]. Listeria fleischmannii in the breast cancer oncobiome is associated with epithelial-to-mesenchymal transition [68].
Tzeng and colleagues have shown that elements of the microbiome covaried with different markers of bad clinical outcomes. Namely, Staphylococcus negatively covaried with TRAF4, Pelomonas positively covaried and Bradyrhizobium negatively covaried with VEGF-A, and Propionibacterium negatively covaried with PDGF-AA and PDGF-BB. In addition to these data, circumstantial data suggest bacterial secretory proteins [41] are probably also involved in communication between the microbiome and tumor tissue.

3.2 Bacterial metabolite signaling and the oncobiosis of the gastrointestinal tract and urinary tract

The gut microbiome undergoes oncobiotic transformation in breast cancer [21, 51, 52, 7382]. In terms of diversity indices, Goedert and colleagues [73] and Byrd and colleagues [74] reported lower or trends towards lower diversity indices in three different cohorts, while Zhu and colleagues [75] and Bobin-Dubigeon and co-workers [84] reported increased alpha diversity indices. Howe and co-workers [76] reported increases in alpha diversity indices in a Pten-deficient mouse model backing the observations of Zhu and colleagues [75] and Bobin-Dubigeon and co-workers [84]. No differences in alpha diversity were reported in [87] and [89]. However, the multiple observations make lower alpha diversity more likely. As noted earlier, risk factors for breast cancer lead to decreases in diversity, such as high-density breast [50], early menarche [51], low physical activity [51], and increases in BMI [51, 52]. Furthermore, antibiotic overdose, which leads to lower diversity, increases the risk for breast cancer [22, 23, 2532], while probiotics that increase diversity have a protective effect [3745].
Characteristic changes in the microbiome were observed between clinical stages [51, 7779, 81] and grades [51, 89], MIB positivity [51], receptor status [51, 89], and proliferative capacity [89]. The most drastic changes were observed among in situ carcinoma and stage I patients, which were gradually rediversified in subsequent stages [7779, 81]. Characteristic changes in taxa between patients and controls include Clostridiales [51, 73, 77, 82], Blautia [51, 77], and Akkermansia muciniphila [52, 76].
The gut microbiome is distant from the breast tumor; hence, signaling pathways are needed to connect the two distant compartments. Multiple pathways cross-connect the oncobiome and the tumor tissue. The direct immunomodulatory effects of the microbiome will be discussed in the next chapter.
Intestinal bacteria expressing beta galactosidases (gus and BC genes [117119]) can deconjugate conjugated estrogens. The gus gene is widespread among bacteria, while changes in BC include Bacteroidetes and Firmicutes [119]. Collinsella, Edwardsiella, Alistipes, Bacteroides, Bifidobacterium, Citrobacter, Clostridium, Dermabacter, Escherichia, Faecalibacterium, Lactobacillus, Marvinbryantia, Propionibacterium, Roseburia, and Tannerella were shown to express β-glucuronidases [120]. Goedert and colleagues provided strong evidence for the involvement of Clostridiales in estrogen reactivation in breast cancer patients [73, 121, 122]. The oncobiome has increased capacity to reactivate estrogens [21, 50, 73, 74, 83, 121124] enabling their reuptake and supporting the growth of estrogen-dependent, estrogen receptor–positive (ER +) breast tumors. Of note, the capacity for estrogen reactivation was identified by pathway analysis in the breast and nipple aspirate microbiomes [57].
Bioactive metabolites, synthesized by the microbiome or the oncobiome, can act in a similar fashion to hormones and can link up the microbiome and the distantly located cancer cells [24, 125]. As the gut microbiome is the biggest in the body in terms of the number of bacteria, its metabolic capacity is considerable. The biosynthetic capacity of the oncobiome is suppressed compared to the eubiome [126, 127]. Multiple bioactive bacterial metabolites were identified that can modulate the behavior of breast cancer cells (Table 3). The importance of changes to the metabolome in breast cancer is further highlighted by the large number of metabolomic studies that point towards the role of the metabolome in breast cancer incidence and evolution [128132]. The bioactive bacterial metabolites are very chemically diverse. We will briefly highlight the most important bacterial metabolites that have cytostatic features. A set of bacterial toxins contributes to the oncogenic property of the oncobiome (Table 4) similar to other neoplasias [133135].
Table 3
Bioactive microbial metabolites in breast cancer
Metabolite group
Made from
Producing bacteria
Relevant enzyme(s)
Receptor
Effect
 
Ref
 
Ref
 
Ref
 
Ref
Reactivated estrogens
Conjugated estrogens
Firmicutes
Collinsella
Edwardsiella
Alistipes
Bacteroides
Bifidobacterium
Citrobacter
Clostridium
Dermabacter
Escherichia
Faecalibacterium
Lactobacillus
Marvinbryantia
Propionibacterium
Roseburia
Tannerella
[73, 117119, 121, 122]
β-glucuronidase (gus/BC)
[117119]
ERα
ERβ mER (mERα, mERβ, GPER, GPRC6, ER-X, Gq-mER)
[136, 137]
OXPHOS, tamoxifen resistance, metastasis, aggressivity, hormone-induced apoptosis, EMT, proliferation, metastasis
[138146]
Short-chain fatty acids
Acetate
Butyrate
Formate
Lactate
Propionate
Pyruvate
Non-digestible carbohydrates, branched
Chain amino acids
Akkermansia muciniphila
Lachnospiraceae
Ruminococcus obeum
Roseburia inulinivorans
Bacteroidetes
Negativicutes sp.
Faecalibacterium
Prausnitzii
Eubacterium rectale
Roseburia faecis
Eubacterium hallii
SS2/1
Odoribacter
Anaeotruncus
[23, 147149]
Thioesterases, phosphate acetyltransferase, acetate kinase, phosphate butyryltransferase, butyrate kinase, lactate dehydrogenase
[150]
FFAR
HDAC
AHR
[151153]
OXPHOS (direct energy substrates), apoptosis, HDAC inhibition, macrophage antimicrobial activity
[154158]
Secondary bile acids
LCA
DCA
UDCA
CDCA
CA
7-keto-litocholic acid
Clostridium
Enterococcus
Bifidobacterium
Lactobacillus
Streptococcus
Eubacterium
Listeria
Bacteroides
Methanobrevibacter
Methanosphera
Escherichia
Ruminococcus
[159166]
Bile salt hydrolases (BSH), 7α/β
-hydroxysteroid, dehydroxylase (baiH)
[167]
TGR5
FXR
SHP
[168170]
Apoptosis, proliferation, VEGF production, OXPHOS, antitumor immunity, EMT, fatty acid biosynthesis, movement, metastasis formation, increased oxidative and nitrosative stress
[78, 97, 171]
Biologically active amines
Cadaverine
L-lysine
Shigella flexneri
Shigella sonnei
Escherichia coli
Streptococci
[172, 173]
Lysine decarboxylase (LdcC, CadA)
[172, 173]
TAAR1, 2, 3, 5, 8, 9
[174]
OXPHOS, CSC, movement, invasion, EMT, metastasis formation
[79]
Indole derivatives
Indoxyl sulfate
Indolepropionic acid
Tryptophan
Providencia rettgeri
Alistipes shahii
Bacteroides xylanisolvens
Clostridium
Lactobacillus reuteri
[81, 175, 176]
TnaA
SULT1, Cyp2e1
[176]
[176]
AHR
PXR
[176]
[81, 175]
OXPHOS, CSC, movement and proliferation, invasion, EMT, metastasis formation, antitumor immunity
[81, 175]
Abbreviations: CA, cholic acid; CDCA, chenodeoxycholic acid; CSC, cancer stem cell; EMT, epithelial-to-mesenchymal transition; ER, estrogen receptor; FFAR, free fatty acid receptor; FXR, farnesyl X receptor; HDAC, histone deacetylase; LPA, lysophosphatidic acid; LPS, lysophospholipids; OXPHOS, oxidative phosphorylation; TAAR, trace amine-related receptor; TGR5/GPBAR1, G protein-coupled bile acid receptor 1; VEGF, vascular endothelial growth factor
Table 4
Structural and secreted bacterial toxins supporting breast cancer
Metabolite group
Made from
Producing bacteria
Relevant enzyme(s)
Receptor
Effect
 
Ref
 
Ref
 
Ref
 
Ref
LPS
Lipid A + core oligosaccharide + O-specific polysaccharide
Escherichia coli
Salmonella enterica
Vibrio cholera
Pseudomonas
Pantoea
[177]
Lpx
[178, 179]
TLR2
TLR4
[177, 180]
Apoptosis, migration and metastases, EMT and β-catenin signaling, invasiveness
[181183]
Lysophospholipids (LPS)
Lysophosphatidic acid (LPA)
Phospholipid
Vibrio cholerae
Helicobacter pylori
Yersinia pseudotuberculosis
[184]
Phospholipase A2
Exogenous lipase
[184]
LPAR1-5
[185, 186]
Proliferation, migration, metastasis, stress fiber and focal adhesion formation
[187189]
Colibactin
Precolibactin
Escherichia coli
Klebsiella pneumoniae
Enterobacter aerogenes
Citrobacter koseri
[190]
ClbA-S
[190]
Unknown
 
Unknown
 
Lithocholic acid (LCA) is a secondary bile acid derived from primary bile acids. Mostly Clostridia in the large intestines are responsible for the production of LCA and secondary bile acids in general [82]; however, other taxons are also involved [191]. The enzymes involved in secondary bile acid production are assembled in the bile acid inducible (bai) operon in bacteria [191]. The production of LCA from its precursors involves the deconjugation of primary bile acids and the oxidation, epimerization, and dehydroxylation of the gonane core [2, 4, 167, 191, 192]. Secondary bile acids, such as LCA, have pleiotropic roles in the microbiome, including regulation of the microbiome composition [193200], facilitation of bacterial translocation into tissues [201], and quorum sensing [202, 203].
The bile acids in the breast are of the gut origin [204, 205]. Total bile acid levels in the serum of healthy individuals are lower than 5 µM. LCA serum reference concentrations are low (30–50 nM). However, tissue levels in the breast may be substantially higher [78, 206]. In breast cancer, both the hepatic synthesis of primary bile acids and the bacterial conversion to secondary bile acids in the large intestine are suppressed, and this suppression is the most dominant in in situ carcinoma and stage I patients [78, 82]. In good agreement with this concept, serum LCA levels negatively correlate with the Ki67 labeling index in breast cancer [207]. The composition of the serum bile acid pool in patients with benign breast disease is different from breast cancer patients; breast cancer patients had higher serum chenodeoxycholic acid levels and lower dihydroxytauro-conjugated bile acids (Tdi-1) and sulfated dihydroxyglyco-conjugated bile acids (Gdi-S-1) [208]. Another secondary bile acid deoxycholic acid (DCA) may act as a procarcinogenic agent [209, 210] and may be responsible for the procarcinogenic character of secondary bile acids [211].
A multitude of receptors is involved in bile acid signaling, including Takeda G protein-coupled receptor 5 (TGR5) and farnesoid X receptor (FXR), which are important for the current discussion. One study [212] suggested the use of bile acid-tamoxifen conjugates for breast cancer therapy.
Multiple amino acid catabolic products derived from lysine and tryptophan have cytostatic properties in breast cancer. Indole derivatives are made from tryptophan, while lysine decarboxylation yields cadaverine.
The microbiome accounts for 4–6% of tryptophan catabolism to yield indol derivatives [213], of which indolepropionic acid (IPA) and indoxyl sulfate (IS) have cytostatic properties in breast cancer [81, 175]. The serum reference concentration of IPA is submicromolar [176, 214, 215], while IS concentrations are low micromolar [216]. The bacterial enzyme responsible for IPA and IS biosynthesis, called tryptophanase (TnaA), can be found in the tryptophanase operon [176]. Tryptophanase expression is widespread among bacteria [217, 218]. IPA and IS, similar to other indole derivatives, can activate the aryl hydrocarbon receptor (AHR) and bind to the pregnane-X receptor (PXR) receptor [219221]. Indole derivatives have a strong immunostimulatory effect [222224]; IPA and IS can induce antitumor immunity in breast cancer [81, 175] and modulate the composition of the microbiome [219, 225227]. Evidence from human studies supports the role of tryptophan and indole metabolism in breast cancer. Elevated extracellular tryptophan levels decrease survival in breast cancer (Table S8 [228]). Ki67 positivity negatively correlates with 3-indoxyl sulfate levels ([207] Additional file 9, Table S8 line 130), and 3-indoxyl sulfate levels are downregulated in both estrogen receptor–positive and negative cases ([207] Additional file 3, Table S3 line 44). TnaA DNA is downregulated in the breast cancer microbiome, and the most drastic changes were observed in in situ and stage I cases [81].
In the human body, cadaverine can be of bacterial, human, or nutritional origin. Nevertheless, bacterial cadaverine production seems to dominate [79]. In bacteria, the LdcC and CadA genes are responsible for cadaverine biosynthesis [172, 229], while diamino-oxidase eliminates cadaverine [230]. The capacity for cadaverine biosynthesis was identified in a number of bacteria [231233]. Human serum reference concentration of cadaverine is submicromolar (100–800 nM) [234, 235]. Cadaverine can activate trace amine-associated receptors (TAAR1, 2, 3, 5, 8, 9), and these receptors are associated with breast cancer [79, 174]. Fecal TnaA protein content was reduced in E-cadherin-negative breast cancer cases compared to E-cadherin-positive cases [80].
Short-chain fatty acids (SCFAs), such as formate, acetate, propionate, butyrate, and lactate, are generated by a large set of bacterial species from non-digestible carbohydrates and a minor fraction from amino acids [236]. SCFAs are formed at multiple points in bacterial metabolism and are then released into the environment. Therefore, a wide circle of bacteria can synthesize SCFAs. The reference concentration of SCFAs in the human serum is in the range of 10–100 µM [237239] and may reach up to 1 mM locally [240]. The receptors for SCFAs are the free fatty acid receptors (FFARs) and AHR [151].
Oncobiosis-related changes to bacterial metabolite production support oncogenesis and not tumor initiation through multi-faceted effects. In certain cases, as for SCFAs, effects are context-dependent; SCFAs can have positive (e.g., [241]) or negative (e.g., [242]) effects in breast cancer. Many studies used metabolites in superphysiological concentrations that may render the interpretation of these studies difficult [243246]. In superphysiological concentrations, metabolites can induce cell death [171, 244247] and other features (e.g., in superphysiological concentrations, LCA blocked fatty acid biosynthesis [171] or induced cell death and the expression of multidrug resistance proteins [247]) that is usually mediated by secondary receptors, such as FXR in the case of LCA [247]. The metabolites assessed in the context of breast cancer were ineffective on untransformed primary human fibroblasts at their reference concentration, suggesting selectivity towards transformed cells [78, 79, 81, 97, 175].
Metabolites elicit effects by reducing oxidative and nitrosative stress through induction of NRF2 expression and its downstream effector genes and suppressing inducible nitric oxide synthase (iNOS). These effects may also involve the catechol-quinone metabolites of estrone and estradiol [81, 97, 175, 248]. Furthermore, the oncobiome contributes to the induction of Warburg metabolism in tumor cells [78, 79, 81, 97, 175]. These basic processes support epithelial-to-mesenchymal transition [78, 79, 81, 249], migration and invasion [79, 81], increased aldehyde dehydrogenase-1 (ALDH1)–positive cancer stem cells [79, 81], and suppression of tumor immunity [22, 24, 78, 81, 175, 250, 251]. These processes together support metastasis formation [24, 78, 79, 250, 251] and disease recurrence [29]. In addition, most SCFAs can act as energy sources in cells [233] and may inhibit histone deacetylases to modulate epigenetics [152, 154157, 241, 252257]. LCA supplementation can reduce VEGF production by the implanted breast cancer cell in an animal model of breast cancer [78]. Furthermore, the oncobiome correlates with omega-3 polyunsaturated fatty acid homeostasis [85].
Our current understanding indicates that these metabolites only represent the tip of the iceberg. Multiple studies [75, 87, 89] identified imputed pathways that were differentially regulated in the oncobiome and eubiome in breast cancer patients (Table 5). In addition to the metabolic changes, a large set of transport systems are dysregulated in bacteria [75]. These findings highlight the widespread effects of bacterial bioactive metabolite production.
Table 5
Imputed metabolic pathways dysregulated in the gut oncobiome of breast cancer patients
Induced in BC patients
Decreased in BC patients
Ref
Premenopausal patients
Beta oxidation
Pyridoxal biosynthesis
Pentose phosphate pathway (oxidative)
Heparane sulfate degradation
Entner-Duodoroff pathway
Premenopausal patients
Uridine monophosphate biosynthesis
Reductive pentose phosphate cycle (ribulose5P → glyceraldehyde3P)
Pyruvate oxidation to acetyl-CoA
Phosphatidylethanolamie biosynthesis
Inosine monophosphate biosynthesis
Glycolysis
GABA biosynthesis
Formaldehyde assimilation, serine pathway
F-type ATPase
Dicarboxylate pathway
Pantothenate biosynthesis
C5 isopernoid biosynthesis, non-mevalonate pathway
C1-unit interconversion
[75]
Postmenopausal patients
Ubiquinone biosynthesis
Jasmonic acid biosynthesis
Beta oxidation
LPS biosynthesis
Glyoxylate cycle
 
[75]
Meta cleavage pathway of aromatic compounds
Aromatic biogenic amine degradation
Androstenedione degradation
 
[87]
LPS biosynthesis
Ubiquinone and other terpenoid-quinone biosynthesis
Folate biosynthesis
Aminobenzoate degradation
Biotin metabolism
Glutathione metabolism
Penicillin and cephalosporin biosynthesis
D-Arginine and D-ornithine metabolism
N-glycan biosynthesis
Isoquinoline alkaloid biosynthesis
Styrene degradation
TCA cycle
Geraniol degradation
Indole alkaloid biosynthesis
Glycolysis/gluconeogenesis
Glycerophospholipid metabolism
[89]
BC, breast cancer; CoA, coenzyme A; GABA, gamma-aminobutyric acid; LPS, lipopolysaccharide; TCA, tricarboxylic acid cycle
As mentioned earlier, the suppression of the biosynthetic capacity of the microbiome is most pronounced in the early stages of the disease [79, 81, 97]. Nevertheless, studies on the expression of receptors for the metabolites suggest that receptors on the surface of tumor cells are downregulated as tumor stage or grade increases [81, 97]. The receptors for microbiome-derived metabolites and the components of the downstream signaling pathways in the tumor correlate with the receptor status, grade, and stage. The intratumoral expression of AHR, PXR, and TGR5 decreases in TNBC cases compared to ER + cases [81, 97]. These findings were mirrored in relapse-free survival rates, where high expression of the receptors and their downstream signaling components provided better survival for patients that was abrogated in TNBC cases [81, 97]. In Figs. 2 and 3, we show that a subset of TAAR receptors and FFAR1 receptors have similar properties. Similarly, in LCA-elicited downstream signaling events, PXR and AHR receptor expression decreased as a function of stage, grade, or high mitotic rate [81, 97].
The impact of changes in the urinary microbiome is unexplored. However, recently, An and colleagues showed that bacterial extracellular vesicles can be isolated from the urine of healthy subjects and breast cancer patients and these extracellular vesicles can affect breast cancer cells differently [90].

3.3 Antitumor immune responses

In general, the diverse status of the microbiome supports normal immune responses that are crucial for antitumor immunity [259261]. Both the breast and the gut oncobiome have altered interactions with the immune system that we will review here. Whether changes to the bacterial community lead to changes to the immune system or the immune system causes changes to the microbiome remains an open question. Furthermore, myeloid and lymphoid infiltrations in tumors have differential effects; lymphoid infiltration is generally considered to have antitumor effects, while neutrophils are considered proneoplastic [262, 263]. We will discuss the microbiome-mediated changes to the immune system separately for the breast microbiome and the gut microbiome.
Before going into details, we would like to briefly introduce two bacterial immunogenic toxins, which are classified as pathogen-associated molecular patterns (PAMP). First, lipopolysaccharides (LPS) (Table 4), lypoglycans, or endotoxins are constituents of the outer membrane of Gram negative bacteria [177]. Toll-like receptor 2 and 4 respond to LPS stimulation [177, 180]. Second, lysophosphatids (Table 4) are generated in reactions related to bacterial membrane homeostasis [184, 264]. Cells of the host organism can also generate lysophosphatids. Gram negative bacteria tend to generate lysophosphatids [184, 264]. Endogenous phospholipase A2 or other exogenous lipases can generate lysophosphatids [184]. Lysophosphatids exert their effects through lysophosphatidic acid receptors (LPAR1-6) [185].

3.3.1 The breast’s inherent microbiome and immune responses

The inherent microbiome of the breast and the tumor is enriched in Gram negative bacteria [67], while lipotheichoic acid, specific for Gram positive bacteria, is absent in tumors [67]. These culturable bacteria are from Proteobacteria, Firmicutes, and Actinobacteria [67]. Alongside these findings, LPS was detected in tumor samples [67]. Bacterial LPS and 16S rRNA were demonstrated in CD45 + /CD68 − cells of a highly inflamed breast tumor, indicating that the colonizing bacteria tune and activate the immune system [67]. In the nipple fluid aspirate of breast cancer patients, the genes for LPS and lysophosphatid biosynthesis were upregulated in an imputed pathway analysis [57]. Similarly, imputed pathway analysis genes related to Th17 cell differentiation were enriched in the breast microbiome of breast cancer patients [62]. Further underlining these observations, bacterial peptidoglycan rendered breast cancer cells more invasive through activation of TLR2 receptors [261]. High intratumoral (antibacterial) inflammation can be boosted or sustained by viral infection [64, 91, 92]. In fact, human papillomavirus infection induces Stat3-activation and IL-17 expression in breast cancer patients [92]. Van der Merwe and colleagues [265] identified Fusobacterium nucleatum as a major species in the breast microbiome that overgrows in breast cancer patients, exerts an immunosuppressive phenotype, and activates the TLR4 receptor leading to immunosuppression.
Nevertheless, two studies [60, 66] refuted these observations. These studies found an association between microbiome components and inflammatory signaling. Namely, Methylibium and Enhydrobacter positively covaried with TLR signaling (TLR3, TLR4, IRAK1) in healthy control networks [60] and Streptococcus positively associated with CD6, LAG3, SH2D1A, and TIGIT expression and with T cell abundance in healthy control tissue [66]. However, in the tumor tissue, these associations [60] were lost, and the expression of antibacterial response genes (TLR2, TLR5, TLR9, NOD1, NOD2, CARD6, CARD9, TRAF6, borderline significant NFκB, BPI, IL-12A, MPO, and PRNT3) was lost [66]. Furthermore, in tumor tissue, Methylibium showed significant negative correlations with ICOS and TBX21 expression and with T cell abundance [66]. These data [60, 66] contradict the data presented by other studies [57, 62, 67, 92, 261], and this discrepancy has not been explained yet.

3.3.2 The gut microbiome and immune responses

Immune responses likely play a role in the oncobiotic transformation of the gut microbiome, as multiple species, such as Ruminococcus and Alistipes, were opsonized in the stool of breast cancer patients [83]. On the other hand, the gut microbiome plays a pivotal role in fine-tuning the host’s immune system. In a large-cohort fecal microbiome study, multiple taxa immune-related functions were strongly associated with breast cancer [74]. In murine models, Lactobacillus acidophilus [39] or Helicobacter hepaticus [266] were associated with immune function in breast cancer. Oral gavage of Helicobacter hepaticus promotes mammary tumorigenesis that is dependent on the recruitment of neutrophils to the tumor; depletion of neutrophils using a monoclonal antibody abolished the promoter effect of Helicobacter hepaticus [266]. Oral treatment of mice with Lactobacillus acidophilus improved the immune response against the experimental tumor, reduced tumor growth, and tuned the immune response towards a Th1-type response [39]. Dysbiosis in the gut is associated with enhanced macrophage infiltration (CD11b + cells) to the breast tissue, of which the majority were M2-polarized (tolerogenic) macrophages [24]. Dysbiosis enhanced the mammary content of GM-CSF, IL23, IL22, CXCL1, and CCL2 in pretumor breast tissue that was further exacerbated in mice bearing breast tumors [24]. Mast cells also play a role in the oncobiome-induced immune effects [22].
Multiple gut-derived bacterial metabolites possess immunomodulatory roles in breast tumors. Lithocholic acid, indolepropionic acid, and indoxyl sulfate are produced in the intestines and can regulate tumor immune response against cancer cells in experimental models of breast cancer [78, 81, 175]. Butyrate may have immunomodulatory roles at multiple levels that were not directly assessed in breast cancer [267].

4 The role of oncobiosis in metastasis formation, survival, and recurrence in breast cancer

Reports unanimously show that dysbiosis supports invasion and metastasis formation [24, 54, 78, 79, 250, 251]. However, reports concerning the association of species with node positivity or metastasis formation are divergent. In the breast microbiome, Methylobacterium decreased in cases with lymphovascular invasion [54]. In another study [60], node positivity was associated with Acinetobacter and Bacteroides and negatively associated with Achromobacter. Lymphovascular invasion was positively associated with Lactobacillus and negatively associated with Alkanindiges abundances. Reduced Oblitimonas abundance was associated with both lymphovascular invasion and node-positive status. Brevundimonas abundance increased in patients developing distant metastases [250].
What are the modalities through which oncobiosis can support metastasis formation? The oncobiome-elicited anti-metastatic and anti-recurrence effects on breast cancer are multi-pronged (Fig. 4). The breast cancer microbiome is associated with extracellular matrix degradation (e.g., dermatan sulfate degradation [67], suppression of peptidoglycan biosynthesis [57], and proteoglycans homeostasis [62]) that can support cellular movement within the tissue. The physical presence of bacteria can promote breast cancer cell invasiveness by activating toll-like receptor 2 on cancer cells via bacterial peptidoglycan [261]. The oncobiosis of the GI tract can also support cellular movement. For example, cadaverine, a cytostatic bacterial metabolite of the GI tract, can suppress breast cancer cell movement in in vitro assays and suppress matrix metalloproteinase-9 expression [79]. Furthermore, fecal TnaA protein content, responsible for bacterial cadaverine biosynthesis, is reduced in E-cadherin negative breast cancer cases compared to E-cadherin positive cases [80].
Improved tissular displacement of cancer cells accompanies the process of epithelial-to-mesenchymal transition (EMT). Intratumoral Listeria fleischmannii is associated with EMT [68]. Cytostatic bacterial metabolites (lithocholic acid, cadaverine, indoxyl sulfate, indolepropionic acid), which are lost in breast cancer, can suppress EMT [78, 79, 81, 175]. These metabolites can improve the expression of E-cadherin and ZO-1 and mesenchymal markers (Vim, Fgfbp1, Tgfb3, MMP9, SnaiI, β-catenin) [78, 79, 81, 249]. Altogether, these processes support migration and invasion [79, 81].
A very important contribution from Buchta Rosean [24] provided evidence that preexisting dysbiosis of the GI tract, induced by antibiotic treatment, supports tumor growth and metastasis formation in a murine model of breast cancer. Gut dysbiosis/oncobiosis induces an inflammatory response in the breast tumor, meloid cell infiltration to tumors, tumor fibrosis, and tumor dissemination. These proneoplastic, prometastatic traits can be induced in mice by transfer of fecal content from tumor-bearing mice.
In addition to cellular movement and EMT regulation, bacterial metabolites from the GI tract, such as lithocholic acid [78, 97] cadaverine [79], indolepropionic acid [81], and indoxyl sulfate [175], can suppress cellular proliferation. Furthermore, lithocholic acid reduces VEGF expression in experimental tumors [78]. These processes decrease the likelihood of tumor growth and blood or lymph vessel infiltration. The synthesis of these bioactive metabolites is suppressed in breast cancer patients [78, 79, 81]. An interesting observation from Absil and colleagues suggests that FXR, a bile acid receptor, plays a major role in setting the osteotropism of breast cancer cells [268].
The microbiome apparently has an impact on the neutrophil-to-lymphocyte ratio, which is a prognostic factor in breast cancer patients [263, 269]. Higher proportions of lymphocytes support antitumor immunity and reduce metastatic behavior, while higher neutrophil ratios support cancer progression and metastasis formation. As noted above, orally administered Helicobacter hepaticus promotes mammary tumorigenesis through neutrophil recruitment to tumors [266]. In contrast, among the bacterial metabolites, lithocholic acid [78] and indolepropionic acid [81] can induce higher proportions of tumor-infiltrating lymphocytes.
The composition of the oncobiome changes as a function of disease stage [60, 78, 79, 81] in breast cancer and, hence, is associated with survival. Antibiotic overdosing, which induces oncobiosis and supports the development of breast cancer, can increase the frequency of disease recurrence [29]. Components of the intratumoral and tissular microbiome correlate with patient survival [70, 72]. Based on the assessment of over 1000 archived breast cancer tissue samples, 46 bacteria were identified as risk factors (including Leptospira, Desulfotalea, Archangium, Dicipivirus, Halosimplex, Spo1virus, Candidatus_Amoebophilus, Roseibium, and Arcticibacter), while 48 bacteria were identified as favorable factors (including Gordonia, Planktothricoides, Lachnoclostridium, Bafinivirus, Actinomadura, and Methanothermus) [72]. Mao and colleagues [72] identified bacterial patterns in breast cancer tissue that strongly correlate with prognosis; poor and good prognosis patients could be separated based on these bacterial patterns (AUC > 0.8). Terrisse et al. [86] demonstrated that stool/fecal transfer of the human microbiome into tumor-bearing mice transformed the properties of the mouse tumors to the human counterpart (e.g., rapidity of progression).
Oncobiosis is also associated with disease recurrence. Kim et al. reported that when comparing patients with high and low risk of regional recurrence of breast cancer, bacterial pentose and glucuronate interconversions and Enterococcus were the main discriminating factors [95]. In support of these observations, antibiotic overdosing can increase the likelihood of disease recurrence [29].
Although little is known about the processes supporting disease recurrence, aldehyde dehydrogenase-1 (ALDH1) positive cancer stem cells are likely to have a supportive role. Bacterial metabolites (cadaverine and indolepropionic acid) produced by the healthy gut eubiome can reduce the proportions of ALDH1 + cancer stem cells in cultured cells [79, 81], leading to reduced recurrence and therapy resistance (Fig. 4). In breast cancer patients, the production of these metabolites is suppressed, supporting the expansion of ALDH1 + cancer stem cells [79, 81]. As noted earlier, widespread metabolic rearrangements were identified in the cancer tissue oncobiome [5557, 59, 62, 67] and the gut oncobiome that encompass elements of Warburg metabolism [75, 78, 79, 81, 87, 89, 175]. Warburg-type metabolism may be a molecular mechanism through which oncobiosis supports ALDH1 + cancer stem cell formation.

5 The role of the breast cancer oncobiome in chemotherapy

Neoadjuvation is a chemotherapeutic procedure aimed at shrinking tumor size to enable surgical excision. The microbiome metabolizes chemotherapeutic drugs, including those used in the (neoadjuvant) chemotherapy of breast cancer [270]. Hence, the microbiome can fundamentally change the pharmacokinetics and pharmacodynamics of these drugs (reviewed in [3, 4, 270, 271]). Not surprisingly, antibiotics modulate the pharmacokinetic and pharmacodynamic properties of chemotherapeutic drugs and the therapeutic outcome [272]. Pseudomonas aeruginosa-conditioned medium affected breast cancer cell proliferation and doxorubicin-induced cell death, highlighting the role of secreted bacterial metabolites and toxins [250], or possibly bacterial extracellular vesicles [90].
Most chemotherapy agents have antimicrobial effects and, thus, affect the microbiome [4]. In line with that concept, neoadjuvation changes the composition of the microbiome [86, 88, 250]. In terms of diversity, the reports show differences between compartments. Chiba et al. [250] reported decreased alpha diversity in breast microbiome in patients undergoing neoadjuvation compared to those undergoing surgery without neoadjuvation. With regard to the gut microbiome, Terrisse et al. [86] reported increases in alpha diversity between stool samples from the same patient undergoing neoadjuvant chemotherapy and after the completion of neoadjuvant chemotherapy. Beta diversity values were able to separate patients according to tumor size, grade, auxiliary node involvement, and TNM stage [86]. Wu and colleagues confirmed increases in alpha diversity of the gut microbiome upon neoadjuvation [88].
Chiba et al. [250] showed that Pseudomonas species increased, while Prevotella decreased in tumors undergoing neoadjuvation in the breast microbiome. Wu and colleagues [88] reported changes to Bacteroidetes (g_Alistipes), Firmicutes (g_Clostridium, g_Eubacterium, g_Bilophyla), and Proteobacteria (g_Haemophylus) in the gut microbiome. Terrisse et al. [86] reported a large set of biochemical pathways that were differentially regulated between the pre- and postneoadjuvant samples in the gut microbiome. Most of these pathways were involved in amino acid and nitrogen metabolism [86].

6 Conclusions

The involvement of the microbiome in breast cancer is compelling. From a clinical perspective, it is important to understand that oncobiosis has to be preexisting [24]. In other words, dysbiosis is a factor that supports carcinogenesis but is unlikely to be the cause of the disease. The interactions between the different pools of the microbiome and breast cancer cells are multi-pronged. The microbiome plays a pivotal role in preventing the metastatic spread of breast cancer [24, 78, 79, 81, 175]. Furthermore, the oncobiome interacts with chemotherapy [86, 88, 250] and has an impact on disease recurrence [250]. Altogether, these data point out a possible role of managing the microbiome to provide novel leverage on breast cancer. These strategic considerations encompass the use of prebiotics [36], probiotics [3745], diverse nutrition [4649], and the careful use antibiotics to reduce the risk for breast cancer incidence and recurrence [2233].
Microbiome changes with the stage, grade, or subtype of breast cancer. Hence, these stratifications should be assessed and patients in the study should be reported with all details. A good example of such a study design can be found in the paper of Plaza-Diaz and colleagues [273]. Along the same line, protocols for identifying external contaminations to the microbiome are very important [8, 69, 274], especially in samples with low biomass.
The microbiome possesses a large set of possible biomarkers; the different subtypes of breast cancer have different microbiomes that can be distinguished [70, 75]. Reports have identified possible markers both at the level of nucleic acids [69, 70, 72, 75] and at the protein level [81] in the breast and stool for the detection of the disease. Certain markers are quite promising (ROCs between 0.888 and 0.917). We would also like to point out a large number of review papers discussing diagnostic issues [275279]. As mentioned earlier, microbiome biomarkers can potentially be used to assess the microbiome-chemotherapy interactions and be a useful source of data in personalized medicine. Taken together, the microbiome-breast cancer interactions have wide applicability from the clinical perspective that warrants future studies and applied discoveries.

Declarations

Conflict of interest

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
4.
Zurück zum Zitat Miko, E., Kovacs, T., Sebo, E., Toth, J., Csonka, T., Ujlaki, G., et al. (2019). Microbiome-microbial metabolome-cancer cell interactions in breast cancer-familiar, but unexplored. Cells, 8(4), E293.PubMedCrossRef Miko, E., Kovacs, T., Sebo, E., Toth, J., Csonka, T., Ujlaki, G., et al. (2019). Microbiome-microbial metabolome-cancer cell interactions in breast cancer-familiar, but unexplored. Cells, 8(4), E293.PubMedCrossRef
10.
Zurück zum Zitat Miller, K. D., Nogueira, L., Mariotto, A. B., Rowland, J. H., Yabroff, K. R., Alfano, C. M., et al. (2019). Cancer treatment and survivorship statistics, 2019. CA: A Cancer Journal for Clinicians, 69(5), 363–385. Miller, K. D., Nogueira, L., Mariotto, A. B., Rowland, J. H., Yabroff, K. R., Alfano, C. M., et al. (2019). Cancer treatment and survivorship statistics, 2019. CA: A Cancer Journal for Clinicians, 69(5), 363–385.
14.
Zurück zum Zitat Senkus, E., Kyriakides, S., Ohno, S., Penault-Llorca, F., Poortmans, P., Rutgers, E., et al. (2015). Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology, 26(Suppl 5), v8-30.PubMedCrossRef Senkus, E., Kyriakides, S., Ohno, S., Penault-Llorca, F., Poortmans, P., Rutgers, E., et al. (2015). Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology, 26(Suppl 5), v8-30.PubMedCrossRef
15.
Zurück zum Zitat Minicozzi, P., Van Eycken, L., Molinie, F., Innos, K., Guevara, M., Marcos-Gragera, R., et al. (2019). Comorbidities, age and period of diagnosis influence treatment and outcomes in early breast cancer. International Journal of Cancer, 144(9), 2118–2127.PubMedCrossRef Minicozzi, P., Van Eycken, L., Molinie, F., Innos, K., Guevara, M., Marcos-Gragera, R., et al. (2019). Comorbidities, age and period of diagnosis influence treatment and outcomes in early breast cancer. International Journal of Cancer, 144(9), 2118–2127.PubMedCrossRef
17.
Zurück zum Zitat Cardoso, F., Kyriakides, S., Ohno, S., Penault-Llorca, F., Poortmans, P., Rubio, I. T., et al. (2019). Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology, 30(8), 1194–1220.PubMedCrossRef Cardoso, F., Kyriakides, S., Ohno, S., Penault-Llorca, F., Poortmans, P., Rubio, I. T., et al. (2019). Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology, 30(8), 1194–1220.PubMedCrossRef
20.
Zurück zum Zitat Badve, S. S., Beitsch, P. D., Bose, S., Byrd, D. R., Chen, V. W., Connolly, J. L., et al. (2018). Breast.In AJCC Cancer Staging Manual, Eighth Edition. Chicago, IL Badve, S. S., Beitsch, P. D., Bose, S., Byrd, D. R., Chen, V. W., Connolly, J. L., et al. (2018). Breast.In AJCC Cancer Staging Manual, Eighth Edition. Chicago, IL
23.
Zurück zum Zitat Kirkup, B., McKee, A., Makin, K., Paveley, J., Caim, S., Alcon-Giner, C., et al. (2019). Perturbation of the gut microbiota by antibiotics results in accelerated breast tumour growth and metabolic dysregulation. bioRxiv. https://doi.org/10.1101/553602CrossRef Kirkup, B., McKee, A., Makin, K., Paveley, J., Caim, S., Alcon-Giner, C., et al. (2019). Perturbation of the gut microbiota by antibiotics results in accelerated breast tumour growth and metabolic dysregulation. bioRxiv. https://​doi.​org/​10.​1101/​553602CrossRef
24.
Zurück zum Zitat Buchta Rosean, C., Bostic, R. R., Ferey, J. C. M., Feng, T. Y., Azar, F. N., Tung, K. S., et al. (2019). Preexisting commensal dysbiosis is a host-intrinsic regulator of tissue inflammation and tumor cell dissemination in hormone receptor-positive breast cancer. Cancer Research, 79(14), 3662–3675.PubMedCrossRef Buchta Rosean, C., Bostic, R. R., Ferey, J. C. M., Feng, T. Y., Azar, F. N., Tung, K. S., et al. (2019). Preexisting commensal dysbiosis is a host-intrinsic regulator of tissue inflammation and tumor cell dissemination in hormone receptor-positive breast cancer. Cancer Research, 79(14), 3662–3675.PubMedCrossRef
25.
Zurück zum Zitat Velicer, C. M., Heckbert, S. R., Lampe, J. W., Potter, J. D., Robertson, C. A., & Taplin, S. H. (2004). Antibiotic use in relation to the risk of breast cancer. JAMA, 291(7), 827–835.PubMedCrossRef Velicer, C. M., Heckbert, S. R., Lampe, J. W., Potter, J. D., Robertson, C. A., & Taplin, S. H. (2004). Antibiotic use in relation to the risk of breast cancer. JAMA, 291(7), 827–835.PubMedCrossRef
26.
Zurück zum Zitat Tamim, H. M., Hanley, J. A., Hajeer, A. H., Boivin, J. F., & Collet, J. P. (2008). Risk of breast cancer in relation to antibiotic use. Pharmacoepidemiology and Drug Safety, 17(2), 144–150.PubMedCrossRef Tamim, H. M., Hanley, J. A., Hajeer, A. H., Boivin, J. F., & Collet, J. P. (2008). Risk of breast cancer in relation to antibiotic use. Pharmacoepidemiology and Drug Safety, 17(2), 144–150.PubMedCrossRef
28.
Zurück zum Zitat Satram-Hoang, S., Moran, E. M., Anton-Culver, H., Burras, R. W., Heimann, T. M., Boggio, I., et al. (2010). A pilot study of male breast cancer in the Veterans Affairs healthcare system. Journal of Environmental Pathology, Toxicology and Oncology, 29(3), 235–244.PubMedCrossRef Satram-Hoang, S., Moran, E. M., Anton-Culver, H., Burras, R. W., Heimann, T. M., Boggio, I., et al. (2010). A pilot study of male breast cancer in the Veterans Affairs healthcare system. Journal of Environmental Pathology, Toxicology and Oncology, 29(3), 235–244.PubMedCrossRef
30.
Zurück zum Zitat Elkrief, A., Derosa, L., Kroemer, G., Zitvogel, L., & Routy, B. (2019). The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: A new independent prognostic factor? Annals of Oncology, 30(10), 1572–1579.PubMedCrossRef Elkrief, A., Derosa, L., Kroemer, G., Zitvogel, L., & Routy, B. (2019). The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: A new independent prognostic factor? Annals of Oncology, 30(10), 1572–1579.PubMedCrossRef
31.
Zurück zum Zitat Friedman, G. D., Oestreicher, N., Chan, J., Quesenberry, C. P., Jr., Udaltsova, N., & Habel, L. A. (2006). Antibiotics and risk of breast cancer: Up to 9 years of follow-up of 2.1 million women. Cancer Epidemiology, Biomarkers & Prevention, 15(11), 2102–2106.CrossRef Friedman, G. D., Oestreicher, N., Chan, J., Quesenberry, C. P., Jr., Udaltsova, N., & Habel, L. A. (2006). Antibiotics and risk of breast cancer: Up to 9 years of follow-up of 2.1 million women. Cancer Epidemiology, Biomarkers & Prevention, 15(11), 2102–2106.CrossRef
32.
Zurück zum Zitat Velicer, C. M., Heckbert, S. R., Rutter, C., Lampe, J. W., & Malone, K. (2006). Association between antibiotic use prior to breast cancer diagnosis and breast tumour characteristics (United States). Cancer Causes and Control, 17(3), 307–313.PubMedCrossRef Velicer, C. M., Heckbert, S. R., Rutter, C., Lampe, J. W., & Malone, K. (2006). Association between antibiotic use prior to breast cancer diagnosis and breast tumour characteristics (United States). Cancer Causes and Control, 17(3), 307–313.PubMedCrossRef
38.
Zurück zum Zitat Aragon, F., Carino, S., Perdigon, G., & de Moreno de LeBlanc, A. (2014). The administration of milk fermented by the probiotic Lactobacillus casei CRL 431 exerts an immunomodulatory effect against a breast tumour in a mouse model. Immunobiology, 219(6), 457–464.PubMedCrossRef Aragon, F., Carino, S., Perdigon, G., & de Moreno de LeBlanc, A. (2014). The administration of milk fermented by the probiotic Lactobacillus casei CRL 431 exerts an immunomodulatory effect against a breast tumour in a mouse model. Immunobiology, 219(6), 457–464.PubMedCrossRef
40.
Zurück zum Zitat Hassan, Z., Mustafa, S., Rahim, R. A., & Isa, N. M. (2016). Anti-breast cancer effects of live, heat-killed and cytoplasmic fractions of Enterococcus faecalis and Staphylococcus hominis isolated from human breast milk. In Vitro Cellular and Developmental Biology. Animal, 52(3), 337–348.PubMedCrossRef Hassan, Z., Mustafa, S., Rahim, R. A., & Isa, N. M. (2016). Anti-breast cancer effects of live, heat-killed and cytoplasmic fractions of Enterococcus faecalis and Staphylococcus hominis isolated from human breast milk. In Vitro Cellular and Developmental Biology. Animal, 52(3), 337–348.PubMedCrossRef
41.
Zurück zum Zitat Pourbaferani, M., Modiri, S., Norouzy, A., Maleki, H., Heidari, M., Alidoust, L., et al. (2021). A newly characterized potentially probiotic strain, Lactobacillus brevis MK05, and the Toxicity effects of its secretory proteins against MCF-7 breast cancer cells. Probiotics Antimicrob Proteins. https://doi.org/10.1007/s12602-021-09766-8CrossRefPubMed Pourbaferani, M., Modiri, S., Norouzy, A., Maleki, H., Heidari, M., Alidoust, L., et al. (2021). A newly characterized potentially probiotic strain, Lactobacillus brevis MK05, and the Toxicity effects of its secretory proteins against MCF-7 breast cancer cells. Probiotics Antimicrob Proteins. https://​doi.​org/​10.​1007/​s12602-021-09766-8CrossRefPubMed
42.
Zurück zum Zitat Méndez Utz, V. E., Pérez Visñuk, D., Perdigón, G., & de Moreno de LeBlanc, A. (2021). Milk fermented by Lactobacillus casei CRL431 administered as an immune adjuvant in models of breast cancer and metastasis under chemotherapy. Applied Microbiology and Biotechnology, 105(1), 327–340. https://doi.org/10.1007/s00253-020-11007-xCrossRefPubMed Méndez Utz, V. E., Pérez Visñuk, D., Perdigón, G., & de Moreno de LeBlanc, A. (2021). Milk fermented by Lactobacillus casei CRL431 administered as an immune adjuvant in models of breast cancer and metastasis under chemotherapy. Applied Microbiology and Biotechnology, 105(1), 327–340. https://​doi.​org/​10.​1007/​s00253-020-11007-xCrossRefPubMed
44.
Zurück zum Zitat Ranjbar, S., Seyednejad, S. A., Azimi, H., Rezaeizadeh, H., & Rahimi, R. (2019). Emerging roles of probiotics in prevention and treatment of breast cancer: A comprehensive review of their therapeutic potential. Nutrition and Cancer, 71(1), 1–12.PubMedCrossRef Ranjbar, S., Seyednejad, S. A., Azimi, H., Rezaeizadeh, H., & Rahimi, R. (2019). Emerging roles of probiotics in prevention and treatment of breast cancer: A comprehensive review of their therapeutic potential. Nutrition and Cancer, 71(1), 1–12.PubMedCrossRef
45.
Zurück zum Zitat Ranjbar, S., Seyednejad, S. A., Zakeri, S. E., Rezaeizadeh, H., & Rahimi, R. (2021). Probiotics for prophylaxis and management of breast cancer: Preclinical and clinical evidence. In P. K. Deol (Ed.), Probiotic Research in Therapeutics Applications in Cancers and Immunological Diseases (Vol. 1, pp. 159–189). Springer Singapore.CrossRef Ranjbar, S., Seyednejad, S. A., Zakeri, S. E., Rezaeizadeh, H., & Rahimi, R. (2021). Probiotics for prophylaxis and management of breast cancer: Preclinical and clinical evidence. In P. K. Deol (Ed.), Probiotic Research in Therapeutics Applications in Cancers and Immunological Diseases (Vol. 1, pp. 159–189). Springer Singapore.CrossRef
46.
Zurück zum Zitat Lecuyer, L., Dalle, C., Lefevre-Arbogast, S., Micheau, P., Lyan, B., Rossary, A., et al. (2019). Diet-related metabolomic signature of long-term breast cancer risk using penalized regression: An exploratory study in the SU.VI.MAX cohort. Cancer Epidemiology, Biomarkers & Prevention, 25, 1055–9965. Lecuyer, L., Dalle, C., Lefevre-Arbogast, S., Micheau, P., Lyan, B., Rossary, A., et al. (2019). Diet-related metabolomic signature of long-term breast cancer risk using penalized regression: An exploratory study in the SU.VI.MAX cohort. Cancer Epidemiology, Biomarkers & Prevention, 25, 1055–9965.
47.
Zurück zum Zitat Newman, T. M., Vitolins, M. Z., & Cook, K. L. (2019). From the table to the tumor: The role of mediterranean and western dietary patterns in shifting microbial-mediated signaling to impact breast cancer risk. Nutrients, 11(11), https://doi.org/10.3390/nu11112565 Newman, T. M., Vitolins, M. Z., & Cook, K. L. (2019). From the table to the tumor: The role of mediterranean and western dietary patterns in shifting microbial-mediated signaling to impact breast cancer risk. Nutrients, 11(11), https://​doi.​org/​10.​3390/​nu11112565
49.
Zurück zum Zitat Wu, Y., Huang, R., Wang, M., Bernstein, L., Bethea, T. N., Chen, C., et al. (2021). Dairy foods, calcium, and risk of breast cancer overall and for subtypes defined by estrogen receptor status: A pooled analysis of 21 cohort studies. American Journal of Clinical Nutrition, 114(2), 450–461. https://doi.org/10.1093/ajcn/nqab097CrossRef Wu, Y., Huang, R., Wang, M., Bernstein, L., Bethea, T. N., Chen, C., et al. (2021). Dairy foods, calcium, and risk of breast cancer overall and for subtypes defined by estrogen receptor status: A pooled analysis of 21 cohort studies. American Journal of Clinical Nutrition, 114(2), 450–461. https://​doi.​org/​10.​1093/​ajcn/​nqab097CrossRef
50.
Zurück zum Zitat Jones, G. S., Spencer Feigelson, H., Falk, R. T., Hua, X., Ravel, J., Yu, G., et al. (2019). Mammographic breast density and its association with urinary estrogens and the fecal microbiota in postmenopausal women. PLoS One., 14(5), e0216114.PubMedPubMedCentralCrossRef Jones, G. S., Spencer Feigelson, H., Falk, R. T., Hua, X., Ravel, J., Yu, G., et al. (2019). Mammographic breast density and its association with urinary estrogens and the fecal microbiota in postmenopausal women. PLoS One., 14(5), e0216114.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Fruge, A. D., Van der Pol, W., Rogers, L. Q., Morrow, C. D., Tsuruta, Y., & Demark-Wahnefried, W. (2018). 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. Journal of the Academy of Nutrition and Dietetics, 9(18), 164. Fruge, A. D., Van der Pol, W., Rogers, L. Q., Morrow, C. D., Tsuruta, Y., & Demark-Wahnefried, W. (2018). 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. Journal of the Academy of Nutrition and Dietetics, 9(18), 164.
54.
Zurück zum Zitat Wang, H., Altemus, J., Niazi, F., Green, H., Calhoun, B. C., Sturgis, C., et al. (2017). Breast tissue, oral and urinary microbiomes in breast cancer. Oncotarget, 8(50), 88122–88138.PubMedPubMedCentralCrossRef Wang, H., Altemus, J., Niazi, F., Green, H., Calhoun, B. C., Sturgis, C., et al. (2017). Breast tissue, oral and urinary microbiomes in breast cancer. Oncotarget, 8(50), 88122–88138.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Hieken, T. J., Chen, J., Hoskin, T. L., Walther-Antonio, M., Johnson, S., Ramaker, S., et al. (2016). The microbiome of aseptically collected human breast tissue in benign and malignant disease. Science and Reports, 6, 30751.CrossRef Hieken, T. J., Chen, J., Hoskin, T. L., Walther-Antonio, M., Johnson, S., Ramaker, S., et al. (2016). The microbiome of aseptically collected human breast tissue in benign and malignant disease. Science and Reports, 6, 30751.CrossRef
58.
Zurück zum Zitat Meng, S., Chen, B., Yang, J., Wang, J., Zhu, D., Meng, Q., et al. (2018). 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. Frontiers in Oncology, 8, 318.PubMedPubMedCentralCrossRef Meng, S., Chen, B., Yang, J., Wang, J., Zhu, D., Meng, Q., et al. (2018). 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. Frontiers in Oncology, 8, 318.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Smith, A., Pierre, J. F., Makowski, L., Tolley, E., Lyn-Cook, B., Lu, L., et al. (2019). Distinct microbial communities that differ by race, stage, or breast-tumor subtype in breast tissues of non-Hispanic Black and non-Hispanic White women. Science and Reports, 9(1), 11940.CrossRef Smith, A., Pierre, J. F., Makowski, L., Tolley, E., Lyn-Cook, B., Lu, L., et al. (2019). Distinct microbial communities that differ by race, stage, or breast-tumor subtype in breast tissues of non-Hispanic Black and non-Hispanic White women. Science and Reports, 9(1), 11940.CrossRef
61.
Zurück zum Zitat Costantini, L., Magno, S., Albanese, D., Donati, C., Molinari, R., Filippone, A., et al. (2018). Characterization of human breast tissue microbiota from core needle biopsies through the analysis of multi hypervariable 16S-rRNA gene regions. Science and Reports, 8(1), 16893.CrossRef Costantini, L., Magno, S., Albanese, D., Donati, C., Molinari, R., Filippone, A., et al. (2018). Characterization of human breast tissue microbiota from core needle biopsies through the analysis of multi hypervariable 16S-rRNA gene regions. Science and Reports, 8(1), 16893.CrossRef
64.
Zurück zum Zitat Banerjee, S., Wei, Z., Tan, F., Peck, K. N., Shih, N., Feldman, M., et al. (2015). Distinct microbiological signatures associated with triple negative breast cancer. Science and Reports, 5, 15162.CrossRef Banerjee, S., Wei, Z., Tan, F., Peck, K. N., Shih, N., Feldman, M., et al. (2015). Distinct microbiological signatures associated with triple negative breast cancer. Science and Reports, 5, 15162.CrossRef
65.
Zurück zum Zitat Banerjee, S., Tian, T., Wei, Z., Shih, N., Feldman, M. D., Peck, K. N., et al. (2018). Distinct microbial signatures associated with different breast cancer types. Frontiers in Microbiology, 9, 951.PubMedPubMedCentralCrossRef Banerjee, S., Tian, T., Wei, Z., Shih, N., Feldman, M. D., Peck, K. N., et al. (2018). Distinct microbial signatures associated with different breast cancer types. Frontiers in Microbiology, 9, 951.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Xuan, C., Shamonki, J. M., Chung, A., Dinome, M. L., Chung, M., Sieling, P. A., et al. (2014). Microbial dysbiosis is associated with human breast cancer. PLoS One, 9(1), e83744.PubMedPubMedCentralCrossRef Xuan, C., Shamonki, J. M., Chung, A., Dinome, M. L., Chung, M., Sieling, P. A., et al. (2014). Microbial dysbiosis is associated with human breast cancer. PLoS One, 9(1), e83744.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Thompson, K. J., Ingle, J. N., Tang, X., Chia, N., Jeraldo, P. R., Walther-Antonio, M. R., et al. (2017). A comprehensive analysis of breast cancer microbiota and host gene expression. PLoS One, 12(11), e0188873.PubMedPubMedCentralCrossRef Thompson, K. J., Ingle, J. N., Tang, X., Chia, N., Jeraldo, P. R., Walther-Antonio, M. R., et al. (2017). A comprehensive analysis of breast cancer microbiota and host gene expression. PLoS One, 12(11), e0188873.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Byrd, D. A., Vogtmann, E., Wu, Z., Han, Y., Wan, Y., Clegg-Lamptey, J.-N., et al. (2021). Associations of fecal microbial profiles with breast cancer and non-malignant breast disease in the Ghana Breast Health Study. International Journal of Cancer, n/a(n/a), https://doi.org/10.1002/ijc.33473 Byrd, D. A., Vogtmann, E., Wu, Z., Han, Y., Wan, Y., Clegg-Lamptey, J.-N., et al. (2021). Associations of fecal microbial profiles with breast cancer and non-malignant breast disease in the Ghana Breast Health Study. International Journal of Cancer, n/a(n/a), https://​doi.​org/​10.​1002/​ijc.​33473
75.
Zurück zum Zitat Zhu, J., Liao, M., Yao, Z., Liang, W., Li, Q., Liu, J., et al. (2018). Breast cancer in postmenopausal women is associated with an altered gut metagenome. Microbiome., 6(1), 136.PubMedPubMedCentralCrossRef Zhu, J., Liao, M., Yao, Z., Liang, W., Li, Q., Liu, J., et al. (2018). Breast cancer in postmenopausal women is associated with an altered gut metagenome. Microbiome., 6(1), 136.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Howe, C., Kim, S. J., Mitchell, J., Im, E., Kim, Y. S., Kim, Y. S., et al. (2018). Differential expression of tumor-associated genes and altered gut microbiome with decreased Akkermansia muciniphila confer a tumor-preventive microenvironment in intestinal epithelial Pten-deficient mice. Biochimica et Biophysica Acta, Molecular Basis of Disease, 1864(12), 3746–3758.PubMedCrossRef Howe, C., Kim, S. J., Mitchell, J., Im, E., Kim, Y. S., Kim, Y. S., et al. (2018). Differential expression of tumor-associated genes and altered gut microbiome with decreased Akkermansia muciniphila confer a tumor-preventive microenvironment in intestinal epithelial Pten-deficient mice. Biochimica et Biophysica Acta, Molecular Basis of Disease, 1864(12), 3746–3758.PubMedCrossRef
77.
Zurück zum Zitat Luu, T. H., Michel, C., Bard, J. M., Dravet, F., Nazih, H., & Bobin-Dubigeon, C. (2017). Intestinal proportion of Blautia sp. is associated with clinical stage and histoprognostic grade in patients with early-stage breast cancer. Nutrition and Cancer, 69(2), 267–275.PubMedCrossRef Luu, T. H., Michel, C., Bard, J. M., Dravet, F., Nazih, H., & Bobin-Dubigeon, C. (2017). Intestinal proportion of Blautia sp. is associated with clinical stage and histoprognostic grade in patients with early-stage breast cancer. Nutrition and Cancer, 69(2), 267–275.PubMedCrossRef
78.
Zurück zum Zitat Miko, E., Vida, A., Kovacs, T., Ujlaki, G., Trencsenyi, G., Marton, J., et al. (2018). Lithocholic acid, a bacterial metabolite reduces breast cancer cell proliferation and aggressiveness. Biochimica et Biophysica Acta, 1859(9), 958–974.PubMedCrossRef Miko, E., Vida, A., Kovacs, T., Ujlaki, G., Trencsenyi, G., Marton, J., et al. (2018). Lithocholic acid, a bacterial metabolite reduces breast cancer cell proliferation and aggressiveness. Biochimica et Biophysica Acta, 1859(9), 958–974.PubMedCrossRef
81.
Zurück zum Zitat Sári, Z., Mikó, E., Kovács, T., Jankó, L., Csonka, T., Sebő, E., et al. (2020). Indolepropionic acid, a metabolite of the microbiome, has cytostatic properties in breast cancer by activating AHR and PXR receptors and inducing oxidative stress. Cancers (Basel), 12(9), 2411. https://doi.org/10.3390/cancers12092411CrossRef Sári, Z., Mikó, E., Kovács, T., Jankó, L., Csonka, T., Sebő, E., et al. (2020). Indolepropionic acid, a metabolite of the microbiome, has cytostatic properties in breast cancer by activating AHR and PXR receptors and inducing oxidative stress. Cancers (Basel), 12(9), 2411. https://​doi.​org/​10.​3390/​cancers12092411CrossRef
83.
Zurück zum Zitat Goedert, J. J., Hua, X., Bielecka, A., Okayasu, I., Milne, G. L., Jones, G. S., et al. (2018). Postmenopausal breast cancer and oestrogen associations with the IgA-coated and IgA-noncoated faecal microbiota. British Journal of Cancer, 23(10), 435. Goedert, J. J., Hua, X., Bielecka, A., Okayasu, I., Milne, G. L., Jones, G. S., et al. (2018). Postmenopausal breast cancer and oestrogen associations with the IgA-coated and IgA-noncoated faecal microbiota. British Journal of Cancer, 23(10), 435.
84.
Zurück zum Zitat Bobin-Dubigeon, C., Luu, H. T., Leuillet, S., Lavergne, S. N., Carton, T., Le Vacon, F., et al. (2021). Faecal microbiota composition varies between patients with breast cancer and healthy women: A comparative case-control study. Nutrients, 13(8), https://doi.org/10.3390/nu13082705. Bobin-Dubigeon, C., Luu, H. T., Leuillet, S., Lavergne, S. N., Carton, T., Le Vacon, F., et al. (2021). Faecal microbiota composition varies between patients with breast cancer and healthy women: A comparative case-control study. Nutrients, 13(8), https://​doi.​org/​10.​3390/​nu13082705.
87.
90.
Zurück zum Zitat An, J., Kim, J. B., Yang, E. Y., Kim, H. O., Lee, W.-H., Yang, J., et al. (2021). Bacterial extracellular vesicles affect endocrine therapy in MCF7 cells. Medicine, 100(18). An, J., Kim, J. B., Yang, E. Y., Kim, H. O., Lee, W.-H., Yang, J., et al. (2021). Bacterial extracellular vesicles affect endocrine therapy in MCF7 cells. Medicine, 100(18).
92.
Zurück zum Zitat Zhang, N., Ma, Z. P., Wang, J., Bai, H. L., Li, Y. X., Sun, Q., et al. (2016). Human papillomavirus infection correlates with inflammatory Stat3 signaling activity and IL-17 expression in patients with breast cancer. American Journal of Translational Research, 8(7), 3214–3226. eCollection 2016.PubMedPubMedCentral Zhang, N., Ma, Z. P., Wang, J., Bai, H. L., Li, Y. X., Sun, Q., et al. (2016). Human papillomavirus infection correlates with inflammatory Stat3 signaling activity and IL-17 expression in patients with breast cancer. American Journal of Translational Research, 8(7), 3214–3226. eCollection 2016.PubMedPubMedCentral
94.
Zurück zum Zitat Miko, E., Vida, A., & Bai, P. (2016). Translational aspects of the microbiome-to be exploited. Cell Biology and Toxicology, 32(3), 153–156.PubMedCrossRef Miko, E., Vida, A., & Bai, P. (2016). Translational aspects of the microbiome-to be exploited. Cell Biology and Toxicology, 32(3), 153–156.PubMedCrossRef
95.
Zurück zum Zitat Kim, H. E., Kim, J., Maeng, S., Oh, B., Hwang, K. T., & Kim, B. S. (2021). Microbiota of breast tissue and its potential association with regional recurrence of breast cancer in Korean women. Journal of Microbiology and Biotechnology, 31(11), https://doi.org/10.4014/jmb.2106.06039. Kim, H. E., Kim, J., Maeng, S., Oh, B., Hwang, K. T., & Kim, B. S. (2021). Microbiota of breast tissue and its potential association with regional recurrence of breast cancer in Korean women. Journal of Microbiology and Biotechnology, 31(11), https://​doi.​org/​10.​4014/​jmb.​2106.​06039.
96.
Zurück zum Zitat Urbaniak, C., Cummins, J., Brackstone, M., Macklaim, J. M., Gloor, G. B., Baban, C. K., et al. (2014). Microbiota of human breast tissue. Applied and Environment Microbiology, 80(10), 3007–3014.CrossRef Urbaniak, C., Cummins, J., Brackstone, M., Macklaim, J. M., Gloor, G. B., Baban, C. K., et al. (2014). Microbiota of human breast tissue. Applied and Environment Microbiology, 80(10), 3007–3014.CrossRef
97.
Zurück zum Zitat Kovács, P., Csonka, T., Kovács, T., Sári, Z., Ujlaki, G., Sipos, A., et al. (2019). Lithocholic acid, a metabolite of the microbiome, increases oxidative stress in breast cancer. Cancers (Basel), 11(9), 1255.CrossRef Kovács, P., Csonka, T., Kovács, T., Sári, Z., Ujlaki, G., Sipos, A., et al. (2019). Lithocholic acid, a metabolite of the microbiome, increases oxidative stress in breast cancer. Cancers (Basel), 11(9), 1255.CrossRef
103.
Zurück zum Zitat Pavlides, S., Whitaker-Menezes, D., Castello-Cros, R., Flomenberg, N., Witkiewicz, A. K., Frank, P. G., et al. (2009). The reverse Warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle, 8(23), 3984–4001.PubMedCrossRef Pavlides, S., Whitaker-Menezes, D., Castello-Cros, R., Flomenberg, N., Witkiewicz, A. K., Frank, P. G., et al. (2009). The reverse Warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle, 8(23), 3984–4001.PubMedCrossRef
104.
Zurück zum Zitat Migneco, G., Whitaker-Menezes, D., Chiavarina, B., Castello-Cros, R., Pavlides, S., Pestell, R. G., et al. (2010). Glycolytic cancer associated fibroblasts promote breast cancer tumor growth, without a measurable increase in angiogenesis: Evidence for stromal-epithelial metabolic coupling. Cell Cycle, 9(12), 2412–2422.PubMedCrossRef Migneco, G., Whitaker-Menezes, D., Chiavarina, B., Castello-Cros, R., Pavlides, S., Pestell, R. G., et al. (2010). Glycolytic cancer associated fibroblasts promote breast cancer tumor growth, without a measurable increase in angiogenesis: Evidence for stromal-epithelial metabolic coupling. Cell Cycle, 9(12), 2412–2422.PubMedCrossRef
105.
Zurück zum Zitat Bonuccelli, G., Whitaker-Menezes, D., Castello-Cros, R., Pavlides, S., Pestell, R. G., Fatatis, A., et al. (2010). The reverse Warburg effect: Glycolysis inhibitors prevent the tumor promoting effects of caveolin-1 deficient cancer associated fibroblasts. Cell Cycle, 9(10), 1960–1971.PubMedCrossRef Bonuccelli, G., Whitaker-Menezes, D., Castello-Cros, R., Pavlides, S., Pestell, R. G., Fatatis, A., et al. (2010). The reverse Warburg effect: Glycolysis inhibitors prevent the tumor promoting effects of caveolin-1 deficient cancer associated fibroblasts. Cell Cycle, 9(10), 1960–1971.PubMedCrossRef
106.
Zurück zum Zitat Bonuccelli, G., Tsirigos, A., Whitaker-Menezes, D., Pavlides, S., Pestell, R. G., Chiavarina, B., et al. (2010). Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle, 9(17), 3506–3514.PubMedPubMedCentralCrossRef Bonuccelli, G., Tsirigos, A., Whitaker-Menezes, D., Pavlides, S., Pestell, R. G., Chiavarina, B., et al. (2010). Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle, 9(17), 3506–3514.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Kim, S., Kim, D. H., Jung, W. H., & Koo, J. S. (2013). Metabolic phenotypes in triple-negative breast cancer. Tumour Biology, 34(3), 1699–1712.PubMedCrossRef Kim, S., Kim, D. H., Jung, W. H., & Koo, J. S. (2013). Metabolic phenotypes in triple-negative breast cancer. Tumour Biology, 34(3), 1699–1712.PubMedCrossRef
108.
Zurück zum Zitat Choi, J., Kim, D. H., Jung, W. H., & Koo, J. S. (2013). Metabolic interaction between cancer cells and stromal cells according to breast cancer molecular subtype. Breast Cancer Research, 15(5), R78.PubMedPubMedCentralCrossRef Choi, J., Kim, D. H., Jung, W. H., & Koo, J. S. (2013). Metabolic interaction between cancer cells and stromal cells according to breast cancer molecular subtype. Breast Cancer Research, 15(5), R78.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Martinez-Outschoorn, U., Sotgia, F., & Lisanti, M. P. (2014). Tumor microenvironment and metabolic synergy in breast cancers: Critical importance of mitochondrial fuels and function. Seminars in Oncology, 41(2), 195–216.PubMedCrossRef Martinez-Outschoorn, U., Sotgia, F., & Lisanti, M. P. (2014). Tumor microenvironment and metabolic synergy in breast cancers: Critical importance of mitochondrial fuels and function. Seminars in Oncology, 41(2), 195–216.PubMedCrossRef
110.
Zurück zum Zitat Gang, B. P., Dilda, P. J., Hogg, P. J., & Blackburn, A. C. (2014). Targeting of two aspects of metabolism in breast cancer treatment. Cancer Biology and Therapy, 15(11), 1533–1541. 1510.4161/15384047.15382014.15955992.PubMedPubMedCentralCrossRef Gang, B. P., Dilda, P. J., Hogg, P. J., & Blackburn, A. C. (2014). Targeting of two aspects of metabolism in breast cancer treatment. Cancer Biology and Therapy, 15(11), 1533–1541. 1510.4161/15384047.15382014.15955992.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Elia, I., Schmieder, R., Christen, S., & Fendt, S. M. (2016). Organ-specific cancer metabolism and its potential for therapy. Handbook of Experimental Pharmacology, 233, 321–353.PubMedCrossRef Elia, I., Schmieder, R., Christen, S., & Fendt, S. M. (2016). Organ-specific cancer metabolism and its potential for therapy. Handbook of Experimental Pharmacology, 233, 321–353.PubMedCrossRef
113.
Zurück zum Zitat Martinez-Outschoorn, U. E., Lisanti, M. P., & Sotgia, F. (2014). Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Seminars in Cancer Biology, 25, 47–60.PubMedCrossRef Martinez-Outschoorn, U. E., Lisanti, M. P., & Sotgia, F. (2014). Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Seminars in Cancer Biology, 25, 47–60.PubMedCrossRef
117.
Zurück zum Zitat Dabek, M., McCrae, S. I., Stevens, V. J., Duncan, S. H., & Louis, P. (2008). Distribution of beta-glucosidase and beta-glucuronidase activity and of beta-glucuronidase gene gus in human colonic bacteria. FEMS Microbiology Ecology, 66(3), 487–495.PubMedCrossRef Dabek, M., McCrae, S. I., Stevens, V. J., Duncan, S. H., & Louis, P. (2008). Distribution of beta-glucosidase and beta-glucuronidase activity and of beta-glucuronidase gene gus in human colonic bacteria. FEMS Microbiology Ecology, 66(3), 487–495.PubMedCrossRef
118.
Zurück zum Zitat McIntosh, F. M., Maison, N., Holtrop, G., Young, P., Stevens, V. J., Ince, J., et al. (2012). Phylogenetic distribution of genes encoding beta-glucuronidase activity in human colonic bacteria and the impact of diet on faecal glycosidase activities. Environmental Microbiology, 14(8), 1876–1887.PubMedCrossRef McIntosh, F. M., Maison, N., Holtrop, G., Young, P., Stevens, V. J., Ince, J., et al. (2012). Phylogenetic distribution of genes encoding beta-glucuronidase activity in human colonic bacteria and the impact of diet on faecal glycosidase activities. Environmental Microbiology, 14(8), 1876–1887.PubMedCrossRef
119.
Zurück zum Zitat Gloux, K., Berteau, O., El Oumami, H., Beguet, F., Leclerc, M., & Dore, J. (2011). A metagenomic beta-glucuronidase uncovers a core adaptive function of the human intestinal microbiome. Proceedings of the National Academy of Sciences of the United States of America, 108(Suppl 1), 4539–4546.PubMedCrossRef Gloux, K., Berteau, O., El Oumami, H., Beguet, F., Leclerc, M., & Dore, J. (2011). A metagenomic beta-glucuronidase uncovers a core adaptive function of the human intestinal microbiome. Proceedings of the National Academy of Sciences of the United States of America, 108(Suppl 1), 4539–4546.PubMedCrossRef
120.
Zurück zum Zitat Kwa, M., Plottel, C. S., Blaser, M. J., & Adams, S. (2016). The Intestinal microbiome and estrogen receptor-positive female breast cancer. Journal of the National Cancer Institute, 108(8), djw029.PubMedCentral Kwa, M., Plottel, C. S., Blaser, M. J., & Adams, S. (2016). The Intestinal microbiome and estrogen receptor-positive female breast cancer. Journal of the National Cancer Institute, 108(8), djw029.PubMedCentral
123.
Zurück zum Zitat Ervin, S. M., Li, H., Lim, L., Roberts, L. R., Liang, X., Mani, S., et al. (2019). Gut microbial beta-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. Journal of Biological Chemistry, 294(49), 18586–18599.CrossRef Ervin, S. M., Li, H., Lim, L., Roberts, L. R., Liang, X., Mani, S., et al. (2019). Gut microbial beta-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. Journal of Biological Chemistry, 294(49), 18586–18599.CrossRef
125.
Zurück zum Zitat Sharon, G., Garg, N., Debelius, J., Knight, R., Dorrestein, P. C., & Mazmanian, S. K. (2014). Specialized metabolites from the microbiome in health and disease. Cell Metabolism, 20(5), 719–730.PubMedPubMedCentralCrossRef Sharon, G., Garg, N., Debelius, J., Knight, R., Dorrestein, P. C., & Mazmanian, S. K. (2014). Specialized metabolites from the microbiome in health and disease. Cell Metabolism, 20(5), 719–730.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Wilmanski, T., Rappaport, N., Earls, J. C., Magis, A. T., Manor, O., Lovejoy, J., et al. (2019). Blood metabolome predicts gut microbiome alpha-diversity in humans. Nature Biotechnology, 37(10), 1217–1228.PubMedCrossRef Wilmanski, T., Rappaport, N., Earls, J. C., Magis, A. T., Manor, O., Lovejoy, J., et al. (2019). Blood metabolome predicts gut microbiome alpha-diversity in humans. Nature Biotechnology, 37(10), 1217–1228.PubMedCrossRef
128.
Zurück zum Zitat Tenori, L., Oakman, C., Morris, P. G., Gralka, E., Turner, N., Cappadona, S., et al. (2015). Serum metabolomic profiles evaluated after surgery may identify patients with oestrogen receptor negative early breast cancer at increased risk of disease recurrence. Results from a retrospective study. Molecular Oncology, 9(1), 128–139. https://doi.org/10.1016/j.molonc.2014.07.012CrossRefPubMed Tenori, L., Oakman, C., Morris, P. G., Gralka, E., Turner, N., Cappadona, S., et al. (2015). Serum metabolomic profiles evaluated after surgery may identify patients with oestrogen receptor negative early breast cancer at increased risk of disease recurrence. Results from a retrospective study. Molecular Oncology, 9(1), 128–139. https://​doi.​org/​10.​1016/​j.​molonc.​2014.​07.​012CrossRefPubMed
132.
Zurück zum Zitat Harada-Shoji, N., Soga, T., Tada, H., Miyashita, M., Harada, M., Watanabe, G., et al. (2019). A metabolic profile of routine needle biopsies identified tumor type specific metabolic signatures for breast cancer stratification: A pilot study. Metabolomics, 15(11), 147.PubMedCrossRef Harada-Shoji, N., Soga, T., Tada, H., Miyashita, M., Harada, M., Watanabe, G., et al. (2019). A metabolic profile of routine needle biopsies identified tumor type specific metabolic signatures for breast cancer stratification: A pilot study. Metabolomics, 15(11), 147.PubMedCrossRef
137.
Zurück zum Zitat Soltysik, K., & Czekaj, P. (2013). Membrane estrogen receptors - is it an alternative way of estrogen action? Journal of Physiology and Pharmacology, 64(2), 129–142.PubMed Soltysik, K., & Czekaj, P. (2013). Membrane estrogen receptors - is it an alternative way of estrogen action? Journal of Physiology and Pharmacology, 64(2), 129–142.PubMed
138.
Zurück zum Zitat Radde, B. N., Ivanova, M. M., Mai, H. X., Salabei, J. K., Hill, B. G., & Klinge, C. M. (2015). Bioenergetic differences between MCF-7 and T47D breast cancer cells and their regulation by oestradiol and tamoxifen. The Biochemical Journal, 465(1), 49–61.PubMedCrossRef Radde, B. N., Ivanova, M. M., Mai, H. X., Salabei, J. K., Hill, B. G., & Klinge, C. M. (2015). Bioenergetic differences between MCF-7 and T47D breast cancer cells and their regulation by oestradiol and tamoxifen. The Biochemical Journal, 465(1), 49–61.PubMedCrossRef
139.
Zurück zum Zitat Radde, B. N., Ivanova, M. M., Mai, H. X., Alizadeh-Rad, N., Piell, K., Van Hoose, P., et al. (2016). Nuclear respiratory factor-1 and bioenergetics in tamoxifen-resistant breast cancer cells. Experimental Cell Research, 347(1), 222–231.PubMedPubMedCentralCrossRef Radde, B. N., Ivanova, M. M., Mai, H. X., Alizadeh-Rad, N., Piell, K., Van Hoose, P., et al. (2016). Nuclear respiratory factor-1 and bioenergetics in tamoxifen-resistant breast cancer cells. Experimental Cell Research, 347(1), 222–231.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Sotgia, F., & Lisanti, M. P. (2017). Mitochondrial mRNA transcripts predict overall survival, tumor recurrence and progression in serous ovarian cancer: Companion diagnostics for cancer therapy. Oncotarget, 8(40), 66925–66939.PubMedPubMedCentralCrossRef Sotgia, F., & Lisanti, M. P. (2017). Mitochondrial mRNA transcripts predict overall survival, tumor recurrence and progression in serous ovarian cancer: Companion diagnostics for cancer therapy. Oncotarget, 8(40), 66925–66939.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Zacksenhaus, E., Shrestha, M., Liu, J. C., Vorobieva, I., Chung, P. E. D., Ju, Y., et al. (2017). Mitochondrial OXPHOS induced by RB1 deficiency in breast cancer: Implications for anabolic metabolism, stemness, and metastasis. Trends in Cancer, 3(11), 768–779.PubMedCrossRef Zacksenhaus, E., Shrestha, M., Liu, J. C., Vorobieva, I., Chung, P. E. D., Ju, Y., et al. (2017). Mitochondrial OXPHOS induced by RB1 deficiency in breast cancer: Implications for anabolic metabolism, stemness, and metastasis. Trends in Cancer, 3(11), 768–779.PubMedCrossRef
143.
Zurück zum Zitat Maximov, P. Y., Abderrahman, B., Curpan, R. F., Hawsawi, Y. M., Fan, P., & Jordan, V. C. (2018). A unifying biology of sex steroid-induced apoptosis in prostate and breast cancers. Endocrine-Related Cancer, 25(2), R83–R113.PubMedCrossRef Maximov, P. Y., Abderrahman, B., Curpan, R. F., Hawsawi, Y. M., Fan, P., & Jordan, V. C. (2018). A unifying biology of sex steroid-induced apoptosis in prostate and breast cancers. Endocrine-Related Cancer, 25(2), R83–R113.PubMedCrossRef
144.
Zurück zum Zitat Al-Howail, H. A., Hakami, H. A., Al-Otaibi, B., Al-Mazrou, A., Daghestani, M. H., Al-Jammaz, I., et al. (2016). PAC down-regulates estrogen receptor alpha and suppresses epithelial-to-mesenchymal transition in breast cancer cells. BMC Cancer, 16, 540.PubMedPubMedCentralCrossRef Al-Howail, H. A., Hakami, H. A., Al-Otaibi, B., Al-Mazrou, A., Daghestani, M. H., Al-Jammaz, I., et al. (2016). PAC down-regulates estrogen receptor alpha and suppresses epithelial-to-mesenchymal transition in breast cancer cells. BMC Cancer, 16, 540.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Bouris, P., Skandalis, S. S., Piperigkou, Z., Afratis, N., Karamanou, K., Aletras, A. J., et al. (2015). Estrogen receptor alpha mediates epithelial to mesenchymal transition, expression of specific matrix effectors and functional properties of breast cancer cells. Matrix Biology, 43, 42–60.PubMedCrossRef Bouris, P., Skandalis, S. S., Piperigkou, Z., Afratis, N., Karamanou, K., Aletras, A. J., et al. (2015). Estrogen receptor alpha mediates epithelial to mesenchymal transition, expression of specific matrix effectors and functional properties of breast cancer cells. Matrix Biology, 43, 42–60.PubMedCrossRef
146.
Zurück zum Zitat Kulkoyluoglu-Cotul, E., Arca, A., & Madak-Erdogan, Z. (2019). Crosstalk between estrogen signaling and breast cancer metabolism. Trends in Endocrinology and Metabolism, 30(1), 25–38.PubMedCrossRef Kulkoyluoglu-Cotul, E., Arca, A., & Madak-Erdogan, Z. (2019). Crosstalk between estrogen signaling and breast cancer metabolism. Trends in Endocrinology and Metabolism, 30(1), 25–38.PubMedCrossRef
151.
152.
Zurück zum Zitat Shimazu, T., Hirschey, M. D., Newman, J., He, W., Shirakawa, K., Le Moan, N., et al. (2013). Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science, 339(6116), 211–214.PubMedCrossRef Shimazu, T., Hirschey, M. D., Newman, J., He, W., Shirakawa, K., Le Moan, N., et al. (2013). Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science, 339(6116), 211–214.PubMedCrossRef
154.
Zurück zum Zitat Arpaia, N., Campbell, C., Fan, X., Dikiy, S., van der Veeken, J., deRoos, P., et al. (2013). Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature, 504(7480), 451–455.PubMedPubMedCentralCrossRef Arpaia, N., Campbell, C., Fan, X., Dikiy, S., van der Veeken, J., deRoos, P., et al. (2013). Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature, 504(7480), 451–455.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Tan, J., McKenzie, C., Potamitis, M., Thorburn, A. N., Mackay, C. R., & Macia, L. (2014). The role of short-chain fatty acids in health and disease. Advances in Immunology, 121, 91–119.PubMedCrossRef Tan, J., McKenzie, C., Potamitis, M., Thorburn, A. N., Mackay, C. R., & Macia, L. (2014). The role of short-chain fatty acids in health and disease. Advances in Immunology, 121, 91–119.PubMedCrossRef
156.
Zurück zum Zitat Schulthess, J., Pandey, S., Capitani, M., Rue-Albrecht, K. C., Arnold, I., Franchini, F., et al. (2019). The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity, 50(2), 432–445.PubMedPubMedCentralCrossRef Schulthess, J., Pandey, S., Capitani, M., Rue-Albrecht, K. C., Arnold, I., Franchini, F., et al. (2019). The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity, 50(2), 432–445.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Salimi, V., Shahsavari, Z., Safizadeh, B., Hosseini, A., Khademian, N., & Tavakoli-Yaraki, M. (2017). Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids in Health and Disease, 16(1), 208.PubMedPubMedCentralCrossRef Salimi, V., Shahsavari, Z., Safizadeh, B., Hosseini, A., Khademian, N., & Tavakoli-Yaraki, M. (2017). Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids in Health and Disease, 16(1), 208.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Rodrigues, M. F., Carvalho, E., Pezzuto, P., Rumjanek, F. D., & Amoedo, N. D. (2015). Reciprocal modulation of histone deacetylase inhibitors sodium butyrate and trichostatin A on the energy metabolism of breast cancer cells. Journal of Cellular Biochemistry, 116(5), 797–808.PubMedCrossRef Rodrigues, M. F., Carvalho, E., Pezzuto, P., Rumjanek, F. D., & Amoedo, N. D. (2015). Reciprocal modulation of histone deacetylase inhibitors sodium butyrate and trichostatin A on the energy metabolism of breast cancer cells. Journal of Cellular Biochemistry, 116(5), 797–808.PubMedCrossRef
161.
Zurück zum Zitat Oh, H. K., Lee, J. Y., Lim, S. J., Kim, M. J., Kim, G. B., Kim, J. H., et al. (2008). Molecular cloning and characterization of a bile salt hydrolase from Lactobacillus acidophilus PF01. Journal of Microbiology and Biotechnology, 18(3), 449–456.PubMed Oh, H. K., Lee, J. Y., Lim, S. J., Kim, M. J., Kim, G. B., Kim, J. H., et al. (2008). Molecular cloning and characterization of a bile salt hydrolase from Lactobacillus acidophilus PF01. Journal of Microbiology and Biotechnology, 18(3), 449–456.PubMed
165.
Zurück zum Zitat Ridlon, J. M., Devendran, S., Alves, J. M., Doden, H., Wolf, P. G., Pereira, G. V., et al. (2020). The “in vivo lifestyle” of bile acid 7α-dehydroxylating bacteria: Comparative genomics, metatranscriptomic, and bile acid metabolomics analysis of a defined microbial community in gnotobiotic mice x. Gut Microbes, 11(3), 381–404. https://doi.org/10.1080/19490976.2019.1618173CrossRefPubMed Ridlon, J. M., Devendran, S., Alves, J. M., Doden, H., Wolf, P. G., Pereira, G. V., et al. (2020). The “in vivo lifestyle” of bile acid 7α-dehydroxylating bacteria: Comparative genomics, metatranscriptomic, and bile acid metabolomics analysis of a defined microbial community in gnotobiotic mice x. Gut Microbes, 11(3), 381–404. https://​doi.​org/​10.​1080/​19490976.​2019.​1618173CrossRefPubMed
167.
Zurück zum Zitat Ridlon, J. M., Harris, S. C., Bhowmik, S., Kang, D. J., & Hylemon, P. B. (2016). Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes, 7(1), 22–39.PubMedPubMedCentralCrossRef Ridlon, J. M., Harris, S. C., Bhowmik, S., Kang, D. J., & Hylemon, P. B. (2016). Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes, 7(1), 22–39.PubMedPubMedCentralCrossRef
171.
173.
Zurück zum Zitat de las Rivas, B., Marcobal, A., Carrascosa, A. V., & Munoz, R. (2006). PCR detection of foodborne bacteria producing the biogenic amines histamine, tyramine, putrescine, and cadaverine. Journal of Food Protection, 69(10), 2509–2514.PubMedCrossRef de las Rivas, B., Marcobal, A., Carrascosa, A. V., & Munoz, R. (2006). PCR detection of foodborne bacteria producing the biogenic amines histamine, tyramine, putrescine, and cadaverine. Journal of Food Protection, 69(10), 2509–2514.PubMedCrossRef
174.
Zurück zum Zitat Vattai, A., Akyol, E., Kuhn, C., Hofmann, S., Heidegger, H., von Koch, F., et al. (2017). Increased trace amine-associated receptor 1 (TAAR1) expression is associated with a positive survival rate in patients with breast cancer. Journal of cancer research and clinical oncology, 143(9), 1637–1647. https://doi.org/10.1007/s00432-017-2420-8CrossRefPubMed Vattai, A., Akyol, E., Kuhn, C., Hofmann, S., Heidegger, H., von Koch, F., et al. (2017). Increased trace amine-associated receptor 1 (TAAR1) expression is associated with a positive survival rate in patients with breast cancer. Journal of cancer research and clinical oncology, 143(9), 1637–1647. https://​doi.​org/​10.​1007/​s00432-017-2420-8CrossRefPubMed
175.
Zurück zum Zitat Sári, Z., Mikó, E., Kovács, T., Boratkó, A., Ujlaki, G., Jankó, L., et al. (2020). Indoxylsulfate, a metabolite of the microbiome, has cytostatic effects in breast cancer via activation of AHR and PXR receptors and induction of oxidative stress. Cancers (Basel), 12(10), 2915. https://doi.org/10.3390/cancers12102915CrossRef Sári, Z., Mikó, E., Kovács, T., Boratkó, A., Ujlaki, G., Jankó, L., et al. (2020). Indoxylsulfate, a metabolite of the microbiome, has cytostatic effects in breast cancer via activation of AHR and PXR receptors and induction of oxidative stress. Cancers (Basel), 12(10), 2915. https://​doi.​org/​10.​3390/​cancers12102915CrossRef
191.
Zurück zum Zitat Ridlon, J. M., Kang, D. J., & Hylemon, P. B. (2006). Bile salt biotransformations by human intestinal bacteria. Journal of Lipid Research, 47(2), 241–259.PubMedCrossRef Ridlon, J. M., Kang, D. J., & Hylemon, P. B. (2006). Bile salt biotransformations by human intestinal bacteria. Journal of Lipid Research, 47(2), 241–259.PubMedCrossRef
192.
Zurück zum Zitat Ridlon, J. M., Wolf, P. G., & Gaskins, H. R. (2016). Taurocholic acid metabolism by gut microbes and colon cancer. Gut Microbes, 7(3), 201–215.PubMedPubMedCentralCrossRef Ridlon, J. M., Wolf, P. G., & Gaskins, H. R. (2016). Taurocholic acid metabolism by gut microbes and colon cancer. Gut Microbes, 7(3), 201–215.PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat Tsuei, J., Chau, T., Mills, D., & Wan, Y. J. (2014). Bile acid dysregulation, gut dysbiosis, and gastrointestinal cancer. Experimental Biology and Medicine (Maywood, N.J.), 239(11), 1489–1504.CrossRef Tsuei, J., Chau, T., Mills, D., & Wan, Y. J. (2014). Bile acid dysregulation, gut dysbiosis, and gastrointestinal cancer. Experimental Biology and Medicine (Maywood, N.J.), 239(11), 1489–1504.CrossRef
194.
Zurück zum Zitat Merritt, M. E., & Donaldson, J. R. (2009). Effect of bile salts on the DNA and membrane integrity of enteric bacteria. Journal of Medical Microbiology, 58(Pt 12), 1533–1541.PubMedCrossRef Merritt, M. E., & Donaldson, J. R. (2009). Effect of bile salts on the DNA and membrane integrity of enteric bacteria. Journal of Medical Microbiology, 58(Pt 12), 1533–1541.PubMedCrossRef
195.
Zurück zum Zitat Garcia-Quintanilla, M., Prieto, A. I., Barnes, L., Ramos-Morales, F., & Casadesus, J. (2006). Bile-induced curing of the virulence plasmid in Salmonella enterica serovar Typhimurium. Journal of Bacteriology, 188(22), 7963–7965.PubMedPubMedCentralCrossRef Garcia-Quintanilla, M., Prieto, A. I., Barnes, L., Ramos-Morales, F., & Casadesus, J. (2006). Bile-induced curing of the virulence plasmid in Salmonella enterica serovar Typhimurium. Journal of Bacteriology, 188(22), 7963–7965.PubMedPubMedCentralCrossRef
196.
Zurück zum Zitat Prieto, A. I., Ramos-Morales, F., & Casadesus, J. (2006). Repair of DNA damage induced by bile salts in Salmonella enterica. Genetics, 174(2), 575–584.PubMedPubMedCentralCrossRef Prieto, A. I., Ramos-Morales, F., & Casadesus, J. (2006). Repair of DNA damage induced by bile salts in Salmonella enterica. Genetics, 174(2), 575–584.PubMedPubMedCentralCrossRef
197.
198.
Zurück zum Zitat Kandell, R. L., & Bernstein, C. (1991). Bile salt/acid induction of DNA damage in bacterial and mammalian cells: Implications for colon cancer. Nutrition and Cancer, 16(3–4), 227–238.PubMedCrossRef Kandell, R. L., & Bernstein, C. (1991). Bile salt/acid induction of DNA damage in bacterial and mammalian cells: Implications for colon cancer. Nutrition and Cancer, 16(3–4), 227–238.PubMedCrossRef
200.
Zurück zum Zitat Sorg, J. A., & Sonenshein, A. L. (2010). Inhibiting the initiation of Clostridium difficile spore germination using analogs of chenodeoxycholic acid, a bile acid. Journal of Bacteriology, 192(19), 4983–4990.PubMedPubMedCentralCrossRef Sorg, J. A., & Sonenshein, A. L. (2010). Inhibiting the initiation of Clostridium difficile spore germination using analogs of chenodeoxycholic acid, a bile acid. Journal of Bacteriology, 192(19), 4983–4990.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Slocum, M. M., Sittig, K. M., Specian, R. D., & Deitch, E. A. (1992). Absence of intestinal bile promotes bacterial translocation. American Surgeon, 58(5), 305–310. Slocum, M. M., Sittig, K. M., Specian, R. D., & Deitch, E. A. (1992). Absence of intestinal bile promotes bacterial translocation. American Surgeon, 58(5), 305–310.
203.
204.
Zurück zum Zitat Javitt, N. B., Budai, K., Miller, D. G., Cahan, A. C., Raju, U., & Levitz, M. (1994). Breast-gut connection: Origin of chenodeoxycholic acid in breast cyst fluid. Lancet, 343(8898), 633–635.PubMedCrossRef Javitt, N. B., Budai, K., Miller, D. G., Cahan, A. C., Raju, U., & Levitz, M. (1994). Breast-gut connection: Origin of chenodeoxycholic acid in breast cyst fluid. Lancet, 343(8898), 633–635.PubMedCrossRef
205.
Zurück zum Zitat Tang, W., Putluri, V., Ambati, C. R., Dorsey, T. H., Putluri, N., & Ambs, S. (2019). Liver- and microbiome-derived bile acids accumulate in human breast tumors and inhibit growth and improve patient survival. Clinical Cancer Research, 11, 1078–432. Tang, W., Putluri, V., Ambati, C. R., Dorsey, T. H., Putluri, N., & Ambs, S. (2019). Liver- and microbiome-derived bile acids accumulate in human breast tumors and inhibit growth and improve patient survival. Clinical Cancer Research, 11, 1078–432.
206.
Zurück zum Zitat Raju, U., Levitz, M., & Javitt, N. B. (1990). Bile acids in human breast cyst fluid: The identification of lithocholic acid. Journal of Clinical Endocrinology and Metabolism, 70(4), 1030–1034.PubMedCrossRef Raju, U., Levitz, M., & Javitt, N. B. (1990). Bile acids in human breast cyst fluid: The identification of lithocholic acid. Journal of Clinical Endocrinology and Metabolism, 70(4), 1030–1034.PubMedCrossRef
207.
Zurück zum Zitat Tang, X., Lin, C. C., Spasojevic, I., Iversen, E. S., Chi, J. T., & Marks, J. R. (2014). A joint analysis of metabolomics and genetics of breast cancer. Breast Cancer Research, 16(4), 415.PubMedPubMedCentralCrossRef Tang, X., Lin, C. C., Spasojevic, I., Iversen, E. S., Chi, J. T., & Marks, J. R. (2014). A joint analysis of metabolomics and genetics of breast cancer. Breast Cancer Research, 16(4), 415.PubMedPubMedCentralCrossRef
210.
Zurück zum Zitat Costarelli, V., & Sanders, T. A. (2002). Plasma bile acids and risk of breast cancer. IARC Scientific Publications, 156, 305–306.PubMed Costarelli, V., & Sanders, T. A. (2002). Plasma bile acids and risk of breast cancer. IARC Scientific Publications, 156, 305–306.PubMed
212.
Zurück zum Zitat Sreekanth, V., Bansal, S., Motiani, R. K., Kundu, S., Muppu, S. K., Majumdar, T. D., et al. (2013). Design, synthesis, and mechanistic investigations of bile acid-tamoxifen conjugates for breast cancer therapy. Bioconjugate Chemistry, 24(9), 1468–1484. https://doi.org/10.1021/bc300664k Epub 2013 Aug 21.CrossRefPubMed Sreekanth, V., Bansal, S., Motiani, R. K., Kundu, S., Muppu, S. K., Majumdar, T. D., et al. (2013). Design, synthesis, and mechanistic investigations of bile acid-tamoxifen conjugates for breast cancer therapy. Bioconjugate Chemistry, 24(9), 1468–1484. https://​doi.​org/​10.​1021/​bc300664k Epub 2013 Aug 21.CrossRefPubMed
213.
Zurück zum Zitat Yokoyama, M. T., & Carlson, J. R. (1979). Microbial metabolites of tryptophan in the intestinal tract with special reference to skatole. American Journal of Clinical Nutrition, 32(1), 173–178.CrossRef Yokoyama, M. T., & Carlson, J. R. (1979). Microbial metabolites of tryptophan in the intestinal tract with special reference to skatole. American Journal of Clinical Nutrition, 32(1), 173–178.CrossRef
214.
Zurück zum Zitat Danaceau, J. P., Anderson, G. M., McMahon, W. M., & Crouch, D. J. (2003). A liquid chromatographic-tandem mass spectrometric method for the analysis of serotonin and related indoles in human whole blood. Journal of Analytical Toxicology, 27(7), 440–444.PubMedCrossRef Danaceau, J. P., Anderson, G. M., McMahon, W. M., & Crouch, D. J. (2003). A liquid chromatographic-tandem mass spectrometric method for the analysis of serotonin and related indoles in human whole blood. Journal of Analytical Toxicology, 27(7), 440–444.PubMedCrossRef
215.
Zurück zum Zitat Rosas, H. D., Doros, G., Bhasin, S., Thomas, B., Gevorkian, S., Malarick, K., et al. (2015). A systems-level “misunderstanding”: The plasma metabolome in Huntington’s disease. Annals of Clinical Translational Neurology, 2(7), 756–768.PubMedPubMedCentralCrossRef Rosas, H. D., Doros, G., Bhasin, S., Thomas, B., Gevorkian, S., Malarick, K., et al. (2015). A systems-level “misunderstanding”: The plasma metabolome in Huntington’s disease. Annals of Clinical Translational Neurology, 2(7), 756–768.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Zelante, T., Iannitti, R. G., Cunha, C., De Luca, A., Giovannini, G., Pieraccini, G., et al. (2013). Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity, 39(2), 372–385.PubMedCrossRef Zelante, T., Iannitti, R. G., Cunha, C., De Luca, A., Giovannini, G., Pieraccini, G., et al. (2013). Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity, 39(2), 372–385.PubMedCrossRef
220.
Zurück zum Zitat Venkatesh, M., Mukherjee, S., Wang, H., Li, H., Sun, K., Benechet, A. P., et al. (2014). Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity, 41(2), 296–310.PubMedPubMedCentralCrossRef Venkatesh, M., Mukherjee, S., Wang, H., Li, H., Sun, K., Benechet, A. P., et al. (2014). Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity, 41(2), 296–310.PubMedPubMedCentralCrossRef
221.
Zurück zum Zitat Lamas, B., Richard, M. L., Leducq, V., Pham, H. P., Michel, M. L., Da Costa, G., et al. (2016). CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nature Medicine, 22(6), 598–605.PubMedPubMedCentralCrossRef Lamas, B., Richard, M. L., Leducq, V., Pham, H. P., Michel, M. L., Da Costa, G., et al. (2016). CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nature Medicine, 22(6), 598–605.PubMedPubMedCentralCrossRef
224.
Zurück zum Zitat Sonner, J. K., Keil, M., Falk-Paulsen, M., Mishra, N., Rehman, A., Kramer, M., et al. (2019). Dietary tryptophan links encephalogenicity of autoreactive T cells with gut microbial ecology. Nature Communications, 10(1), 4877.PubMedPubMedCentralCrossRef Sonner, J. K., Keil, M., Falk-Paulsen, M., Mishra, N., Rehman, A., Kramer, M., et al. (2019). Dietary tryptophan links encephalogenicity of autoreactive T cells with gut microbial ecology. Nature Communications, 10(1), 4877.PubMedPubMedCentralCrossRef
227.
Zurück zum Zitat Zhang, L., Nichols, R. G., & Patterson, A. D. (2017). The aryl hydrocarbon receptor as a moderator of host-microbiota communication. Current Opinion in Toxicology, 2, 30–35.PubMedPubMedCentralCrossRef Zhang, L., Nichols, R. G., & Patterson, A. D. (2017). The aryl hydrocarbon receptor as a moderator of host-microbiota communication. Current Opinion in Toxicology, 2, 30–35.PubMedPubMedCentralCrossRef
228.
Zurück zum Zitat Auslander, N., Yizhak, K., Weinstock, A., Budhu, A., Tang, W., Wang, X. W., et al. (2016). A joint analysis of transcriptomic and metabolomic data uncovers enhanced enzyme-metabolite coupling in breast cancer. Science and Reports, 6, 29662.CrossRef Auslander, N., Yizhak, K., Weinstock, A., Budhu, A., Tang, W., Wang, X. W., et al. (2016). A joint analysis of transcriptomic and metabolomic data uncovers enhanced enzyme-metabolite coupling in breast cancer. Science and Reports, 6, 29662.CrossRef
230.
Zurück zum Zitat Seiler, N., Bolkenius, F. N., & Rennert, O. M. (1981). Interconversion, catabolism and elimination of the polyamines. Medical Biology, 59(5–6), 334–346.PubMed Seiler, N., Bolkenius, F. N., & Rennert, O. M. (1981). Interconversion, catabolism and elimination of the polyamines. Medical Biology, 59(5–6), 334–346.PubMed
231.
Zurück zum Zitat Goodwin, A. C., Destefano Shields, C. E., Wu, S., Huso, D. L., Wu, X., Murray-Stewart, T. R., et al. (2011). Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America, 108(37), 15354–15359.PubMedPubMedCentralCrossRef Goodwin, A. C., Destefano Shields, C. E., Wu, S., Huso, D. L., Wu, X., Murray-Stewart, T. R., et al. (2011). Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America, 108(37), 15354–15359.PubMedPubMedCentralCrossRef
233.
Zurück zum Zitat Sittipo, P., Shim, J. W., & Lee, Y. K. (2019). Microbial metabolites determine host health and the status of some diseases. International Journal of Molecular Sciences, 20(21), E5296.PubMedCrossRef Sittipo, P., Shim, J. W., & Lee, Y. K. (2019). Microbial metabolites determine host health and the status of some diseases. International Journal of Molecular Sciences, 20(21), E5296.PubMedCrossRef
234.
Zurück zum Zitat Loser, C., Folsch, U. R., Paprotny, C., & Creutzfeldt, W. (1990). Polyamine concentrations in pancreatic tissue, serum, and urine of patients with pancreatic cancer. Pancreas, 5(2), 119–127.PubMedCrossRef Loser, C., Folsch, U. R., Paprotny, C., & Creutzfeldt, W. (1990). Polyamine concentrations in pancreatic tissue, serum, and urine of patients with pancreatic cancer. Pancreas, 5(2), 119–127.PubMedCrossRef
235.
Zurück zum Zitat Loser, C., Folsch, U. R., Paprotny, C., & Creutzfeldt, W. (1990). Polyamines in colorectal cancer. Evaluation of polyamine concentrations in the colon tissue, serum, and urine of 50 patients with colorectal cancer. Cancer, 65(4), 958–966.PubMedCrossRef Loser, C., Folsch, U. R., Paprotny, C., & Creutzfeldt, W. (1990). Polyamines in colorectal cancer. Evaluation of polyamine concentrations in the colon tissue, serum, and urine of 50 patients with colorectal cancer. Cancer, 65(4), 958–966.PubMedCrossRef
236.
Zurück zum Zitat Morrison, D. J., & Preston, T. (2016). Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes, 7(3), 189–200.PubMedPubMedCentralCrossRef Morrison, D. J., & Preston, T. (2016). Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes, 7(3), 189–200.PubMedPubMedCentralCrossRef
237.
Zurück zum Zitat Clausen, M. R., Mortensen, P. B., & Bendtsen, F. (1991). Serum levels of short-chain fatty acids in cirrhosis and hepatic coma. Hepatology, 14(6), 1040–1045.PubMedCrossRef Clausen, M. R., Mortensen, P. B., & Bendtsen, F. (1991). Serum levels of short-chain fatty acids in cirrhosis and hepatic coma. Hepatology, 14(6), 1040–1045.PubMedCrossRef
238.
Zurück zum Zitat Jakobsdottir, G., Bjerregaard, J. H., Skovbjerg, H., & Nyman, M. (2013). Fasting serum concentration of short-chain fatty acids in subjects with microscopic colitis and celiac disease: No difference compared with controls, but between genders. Scandinavian Journal of Gastroenterology, 48(6), 696–701.PubMedCrossRef Jakobsdottir, G., Bjerregaard, J. H., Skovbjerg, H., & Nyman, M. (2013). Fasting serum concentration of short-chain fatty acids in subjects with microscopic colitis and celiac disease: No difference compared with controls, but between genders. Scandinavian Journal of Gastroenterology, 48(6), 696–701.PubMedCrossRef
239.
Zurück zum Zitat Ktsoyan, Z. A., Mkrtchyan, M. S., Zakharyan, M. K., Mnatsakanyan, A. A., Arakelova, K. A., Gevorgyan, Z. U., et al. (2016). Systemic concentrations of short chain fatty acids are elevated in Salmonellosis and Exacerbation of Familial Mediterranean Fever. Frontiers in Microbiology, 7, 776.PubMedPubMedCentralCrossRef Ktsoyan, Z. A., Mkrtchyan, M. S., Zakharyan, M. K., Mnatsakanyan, A. A., Arakelova, K. A., Gevorgyan, Z. U., et al. (2016). Systemic concentrations of short chain fatty acids are elevated in Salmonellosis and Exacerbation of Familial Mediterranean Fever. Frontiers in Microbiology, 7, 776.PubMedPubMedCentralCrossRef
240.
Zurück zum Zitat Pryde, S. E., Duncan, S. H., Hold, G. L., Stewart, C. S., & Flint, H. J. (2002). The microbiology of butyrate formation in the human colon. FEMS Microbiology Letters, 217(2), 133–139.PubMedCrossRef Pryde, S. E., Duncan, S. H., Hold, G. L., Stewart, C. S., & Flint, H. J. (2002). The microbiology of butyrate formation in the human colon. FEMS Microbiology Letters, 217(2), 133–139.PubMedCrossRef
241.
Zurück zum Zitat Thirunavukkarasan, M., Wang, C., Rao, A., Hind, T., Teo, Y. R., Siddiquee, A. A., et al. (2017). Short-chain fatty acid receptors inhibit invasive phenotypes in breast cancer cells. PLoS One, 12(10), e0186334.PubMedPubMedCentralCrossRef Thirunavukkarasan, M., Wang, C., Rao, A., Hind, T., Teo, Y. R., Siddiquee, A. A., et al. (2017). Short-chain fatty acid receptors inhibit invasive phenotypes in breast cancer cells. PLoS One, 12(10), e0186334.PubMedPubMedCentralCrossRef
242.
Zurück zum Zitat Huang, C. K., Chang, P. H., Kuo, W. H., Chen, C. L., Jeng, Y. M., Chang, K. J., et al. (2017). Adipocytes promote malignant growth of breast tumours with monocarboxylate transporter 2 expression via beta-hydroxybutyrate. Nature Communications, 8, 14706.PubMedPubMedCentralCrossRef Huang, C. K., Chang, P. H., Kuo, W. H., Chen, C. L., Jeng, Y. M., Chang, K. J., et al. (2017). Adipocytes promote malignant growth of breast tumours with monocarboxylate transporter 2 expression via beta-hydroxybutyrate. Nature Communications, 8, 14706.PubMedPubMedCentralCrossRef
244.
Zurück zum Zitat Goldberg, A. A., Titorenko, V. I., Beach, A., & Sanderson, J. T. (2013). Bile acids induce apoptosis selectively in androgen-dependent and -independent prostate cancer cells. PeerJ, 1, e122.PubMedPubMedCentralCrossRef Goldberg, A. A., Titorenko, V. I., Beach, A., & Sanderson, J. T. (2013). Bile acids induce apoptosis selectively in androgen-dependent and -independent prostate cancer cells. PeerJ, 1, e122.PubMedPubMedCentralCrossRef
245.
Zurück zum Zitat Gafar, A. A., Draz, H. M., Goldberg, A. A., Bashandy, M. A., Bakry, S., Khalifa, M. A., et al. (2016). Lithocholic acid induces endoplasmic reticulum stress, autophagy and mitochondrial dysfunction in human prostate cancer cells. PeerJ, 4, e2445.PubMedPubMedCentralCrossRef Gafar, A. A., Draz, H. M., Goldberg, A. A., Bashandy, M. A., Bakry, S., Khalifa, M. A., et al. (2016). Lithocholic acid induces endoplasmic reticulum stress, autophagy and mitochondrial dysfunction in human prostate cancer cells. PeerJ, 4, e2445.PubMedPubMedCentralCrossRef
246.
Zurück zum Zitat Goldberg, A. A., Beach, A., Davies, G. F., Harkness, T. A., Leblanc, A., & Titorenko, V. I. (2011). Lithocholic bile acid selectively kills neuroblastoma cells, while sparing normal neuronal cells. Oncotarget, 2(10), 761–782.PubMedPubMedCentralCrossRef Goldberg, A. A., Beach, A., Davies, G. F., Harkness, T. A., Leblanc, A., & Titorenko, V. I. (2011). Lithocholic bile acid selectively kills neuroblastoma cells, while sparing normal neuronal cells. Oncotarget, 2(10), 761–782.PubMedPubMedCentralCrossRef
247.
Zurück zum Zitat Swales, K. E., Korbonits, M., Carpenter, R., Walsh, D. T., Warner, T. D., & Bishop-Bailey, D. (2006). The farnesoid X receptor is expressed in breast cancer and regulates apoptosis and aromatase expression. Cancer Research, 66(20), 10120–10126.PubMedCrossRef Swales, K. E., Korbonits, M., Carpenter, R., Walsh, D. T., Warner, T. D., & Bishop-Bailey, D. (2006). The farnesoid X receptor is expressed in breast cancer and regulates apoptosis and aromatase expression. Cancer Research, 66(20), 10120–10126.PubMedCrossRef
248.
Zurück zum Zitat Yager, J. D. (2015). Mechanisms of estrogen carcinogenesis: The role of E2/E1-quinone metabolites suggests new approaches to preventive intervention–a review. Steroids, 99(Pt A), 56–60.PubMedCrossRef Yager, J. D. (2015). Mechanisms of estrogen carcinogenesis: The role of E2/E1-quinone metabolites suggests new approaches to preventive intervention–a review. Steroids, 99(Pt A), 56–60.PubMedCrossRef
249.
Zurück zum Zitat Vergara, D., Simeone, P., Damato, M., Maffia, M., Lanuti, P., & Trerotola, M. (2019). The cancer microbiota: EMT and inflammation as shared molecular mechanisms associated with plasticity and progression. Journal of Oncology, 2019https://doi.org/10.1155/2019/1253727 Vergara, D., Simeone, P., Damato, M., Maffia, M., Lanuti, P., & Trerotola, M. (2019). The cancer microbiota: EMT and inflammation as shared molecular mechanisms associated with plasticity and progression. Journal of Oncology, 2019https://​doi.​org/​10.​1155/​2019/​1253727
250.
Zurück zum Zitat Chiba, A., Bawaneh, A., Velazquez, C., Clear, K. Y. J., Wilson, A. S., Howard-McNatt, M., et al. (2019). Neoadjuvant chemotherapy shifts breast tumor microbiota populations to regulate drug responsiveness and the development of metastasis. Molecular Cancer Research, 18, 1541–7786. Chiba, A., Bawaneh, A., Velazquez, C., Clear, K. Y. J., Wilson, A. S., Howard-McNatt, M., et al. (2019). Neoadjuvant chemotherapy shifts breast tumor microbiota populations to regulate drug responsiveness and the development of metastasis. Molecular Cancer Research, 18, 1541–7786.
251.
Zurück zum Zitat Ingman, W. V. (2019). The gut microbiome: A new player in breast cancer metastasis. Cancer Research, 79(14), 3539–3541.PubMedCrossRef Ingman, W. V. (2019). The gut microbiome: A new player in breast cancer metastasis. Cancer Research, 79(14), 3539–3541.PubMedCrossRef
252.
Zurück zum Zitat Menzies, K. J., Zhang, H., Katsyuba, E., & Auwerx, J. (2016). Protein acetylation in metabolism - metabolites and cofactors. Nature Reviews. Endocrinology, 12(1), 43–60.PubMedCrossRef Menzies, K. J., Zhang, H., Katsyuba, E., & Auwerx, J. (2016). Protein acetylation in metabolism - metabolites and cofactors. Nature Reviews. Endocrinology, 12(1), 43–60.PubMedCrossRef
254.
Zurück zum Zitat Yu, X., Shahir, A. M., Sha, J., Feng, Z., Eapen, B., Nithianantham, S., et al. (2014). Short chain fatty acids from periodontal pathogens suppress HDACs, EZH2, and SUV39H1 to promote Kaposi’s sarcoma-associated herpesvirus replication. Journal of Virology, 5, 5. Yu, X., Shahir, A. M., Sha, J., Feng, Z., Eapen, B., Nithianantham, S., et al. (2014). Short chain fatty acids from periodontal pathogens suppress HDACs, EZH2, and SUV39H1 to promote Kaposi’s sarcoma-associated herpesvirus replication. Journal of Virology, 5, 5.
255.
Zurück zum Zitat Haase, S., Haghikia, A., Wilck, N., Muller, D. N., & Linker, R. A. (2018). Impacts of microbiome metabolites on immune regulation and autoimmunity. Immunology, 154(2), 230–238.PubMedPubMedCentralCrossRef Haase, S., Haghikia, A., Wilck, N., Muller, D. N., & Linker, R. A. (2018). Impacts of microbiome metabolites on immune regulation and autoimmunity. Immunology, 154(2), 230–238.PubMedPubMedCentralCrossRef
256.
Zurück zum Zitat Ratajczak, W., Ryl, A., Mizerski, A., Walczakiewicz, K., Sipak, O., & Laszczynska, M. (2019). Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochimica Polonica, 66(1), 1–12.PubMed Ratajczak, W., Ryl, A., Mizerski, A., Walczakiewicz, K., Sipak, O., & Laszczynska, M. (2019). Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochimica Polonica, 66(1), 1–12.PubMed
257.
Zurück zum Zitat Fachi, J. L., Secca, C., Rodrigues, P. B., Mato, F. C. P., Di Luccia, B., Felipe, J. S., et al. (2020). Acetate coordinates neutrophil and ILC3 responses against C. difficile through FFAR2. Journal of Experimental Medicine, 217(3), (pii) 133544.CrossRef Fachi, J. L., Secca, C., Rodrigues, P. B., Mato, F. C. P., Di Luccia, B., Felipe, J. S., et al. (2020). Acetate coordinates neutrophil and ILC3 responses against C. difficile through FFAR2. Journal of Experimental Medicine, 217(3), (pii) 133544.CrossRef
259.
Zurück zum Zitat Routy, B., Le Chatelier, E., Derosa, L., Duong, C. P. M., Alou, M. T., Daillere, R., et al. (2018). Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science, 359(6371), 91–97.PubMedCrossRef Routy, B., Le Chatelier, E., Derosa, L., Duong, C. P. M., Alou, M. T., Daillere, R., et al. (2018). Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science, 359(6371), 91–97.PubMedCrossRef
260.
Zurück zum Zitat Viaud, S., Saccheri, F., Mignot, G., Yamazaki, T., Daillere, R., Hannani, D., et al. (2013). The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science, 342(6161), 971–976.PubMedPubMedCentralCrossRef Viaud, S., Saccheri, F., Mignot, G., Yamazaki, T., Daillere, R., Hannani, D., et al. (2013). The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science, 342(6161), 971–976.PubMedPubMedCentralCrossRef
261.
Zurück zum Zitat Xie, W., Huang, Y., Xie, W., Guo, A., & Wu, W. (2010). Bacteria peptidoglycan promoted breast cancer cell invasiveness and adhesiveness by targeting toll-like receptor 2 in the cancer cells. PLoS One, 5(5), e10850.PubMedPubMedCentralCrossRef Xie, W., Huang, Y., Xie, W., Guo, A., & Wu, W. (2010). Bacteria peptidoglycan promoted breast cancer cell invasiveness and adhesiveness by targeting toll-like receptor 2 in the cancer cells. PLoS One, 5(5), e10850.PubMedPubMedCentralCrossRef
267.
Zurück zum Zitat Luu, M., Weigand, K., Wedi, F., Breidenbend, C., Leister, H., Pautz, S., et al. (2018). Regulation of the effector function of CD8(+) T cells by gut microbiota-derived metabolite butyrate. Science and Reports, 8(1), 14430.CrossRef Luu, M., Weigand, K., Wedi, F., Breidenbend, C., Leister, H., Pautz, S., et al. (2018). Regulation of the effector function of CD8(+) T cells by gut microbiota-derived metabolite butyrate. Science and Reports, 8(1), 14430.CrossRef
274.
Zurück zum Zitat Armstrong, G., Martino, C., Rahman, G., Gonzalez, A., Vázquez-Baeza, Y., Mishne, G., et al. (2021). Uniform Manifold Approximation and Projection (UMAP) Reveals composite patterns and resolves visualization artifacts in microbiome data. mSystems, e0069121, https://doi.org/10.1128/mSystems.00691-21. Armstrong, G., Martino, C., Rahman, G., Gonzalez, A., Vázquez-Baeza, Y., Mishne, G., et al. (2021). Uniform Manifold Approximation and Projection (UMAP) Reveals composite patterns and resolves visualization artifacts in microbiome data. mSystems, e0069121, https://​doi.​org/​10.​1128/​mSystems.​00691-21.
275.
Zurück zum Zitat Dakubo, G. D. (2019). Body fluid microbiome as cancer biomarkers.In Cancer Biomarkers in Body Fluids. Cham: Springer Dakubo, G. D. (2019). Body fluid microbiome as cancer biomarkers.In Cancer Biomarkers in Body Fluids. Cham: Springer
279.
Zurück zum Zitat Parida, S., & Sharma, D. (2019). The power of small changes: Comprehensive analyses of microbial dysbiosis in breast cancer. Biochimica et Biophysica Acta - Reviews on Cancer, 11(19), 30042–30043. Parida, S., & Sharma, D. (2019). The power of small changes: Comprehensive analyses of microbial dysbiosis in breast cancer. Biochimica et Biophysica Acta - Reviews on Cancer, 11(19), 30042–30043.
Metadaten
Titel
The involvement of oncobiosis and bacterial metabolite signaling in metastasis formation in breast cancer
verfasst von
Tünde Kovács
Edit Mikó
Gyula Ujlaki
Heba Yousef
Viktória Csontos
Karen Uray
Peter Bai
Publikationsdatum
30.12.2021
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 4/2021
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-021-10013-3

Weitere Artikel der Ausgabe 4/2021

Cancer and Metastasis Reviews 4/2021 Zur Ausgabe

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.