Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 12(15)2023 07 27.
Article in English | MEDLINE | ID: mdl-37566024

ABSTRACT

Emerging evidence suggests a profound association between the microbiota composition in the gastrointestinal tract and breast cancer progression. The gut microbiota plays a crucial role in modulating the immune response, releasing metabolites, and modulating estrogen levels, all of which have implications for breast cancer growth. However, recent research has unveiled a novel aspect of the relationship between the microbiota and breast cancer, focusing on microbes residing within the mammary tissue, which was once considered sterile. These localized microbial communities have been found to change in the presence of a tumor as compared to healthy mammary tissue, unraveling their potential contribution to tumor progression. Studies have identified specific bacterial species that are enriched within breast tumors and have highlighted the mechanisms by which even these microbes influence cancer progression through immune modulation, direct carcinogenic activity, and effects on cellular pathways involved in cell proliferation or apoptosis. This review aims to provide an overview of the current knowledge on the mechanisms of crosstalk between the gut/mammary microbiota and breast cancer. Understanding this intricate interplay holds promise for developing innovative therapeutic approaches.


Subject(s)
Breast Neoplasms , Breast , Gastrointestinal Microbiome , Animals , Humans , Breast/microbiology , Breast Neoplasms/microbiology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Immunity , Symbiosis , Host Microbial Interactions
2.
Cancer Lett ; 555: 216041, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36565918

ABSTRACT

The mammary gland hosts a microbiota, which differs between malignant versus normal tissue. We found that aerosolized antibiotics decrease murine mammary tumor growth and strongly limit lung metastasis. Oral absorbable antibiotics also reduced mammary tumors. In ampicillin-treated nodules, the immune microenvironment consisted of an M1 profile and improved T cell/macrophage infiltration. In these tumors, we noted an under-representation of microbial recognition and complement pathways, supported by TLR2/TLR7 protein and C3-fragment deposition reduction. By 16S rRNA gene profiling, we observed increased Staphylococcus levels in untreated tumors, among which we isolated Staphylococcus epidermidis, which had potent inflammatory activity and increased Tregs. Conversely, oral ampicillin lowered Staphylococcus epidermidis in mammary tumors and expanded bacteria promoting an M1 phenotype and reducing MDSCs and tumor growth. Ampicillin/paclitaxel combination improved the chemotherapeutic efficacy. Notably, an Amp-like signature, based on genes differentially expressed in murine tumors, identified breast cancer patients with better prognosis and high immune infiltration that correlated with a bacteria response signature. This study highlights the significant influence of mammary tumor microbiota on local immune status and the relevance of its treatment with antibiotics, in combination with breast cancer therapies.


Subject(s)
Mammary Neoplasms, Animal , Staphylococcus epidermidis , Mice , Animals , RNA, Ribosomal, 16S/genetics , Ampicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Tumor Microenvironment
3.
Cancers (Basel) ; 14(22)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36428742

ABSTRACT

While the results thus far demonstrate the clinical benefit of trastuzumab in breast cancer (BC), some patients do not respond to this drug. HER2 mRNA, alone or combined with other genes/biomarkers, has been proven to be a powerful predictive marker in several studies. Here, we provide evidence of the association between HER2 mRNA levels and the response to anti-HER2 treatment in HER2-positive BC patients treated with adjuvant trastuzumab and show that this association is independent of estrogen receptor (ER) tumor positivity. While HER2 mRNA expression was significantly correlated with HER2 protein levels in ER-negative tumors, no correlation was found in ER-positive tumors, and HER2 protein expression was not associated with relapse risk. Correlation analyses in the ER-positive subset identified ER activity as the pathway inversely associated with HER2 mRNA. Associations between HER2 levels and oncogene addiction, as well as between HER2 activation and trastuzumab sensitivity, were also observed in vitro in HER2-positive BC cell lines. In ER-positive but not ER-negative BC cells, HER2 transcription was increased by reducing ligand-dependent ER activity or inducing ER degradation. Accordingly, HER2 mRNA levels in patients were found to be inversely correlated with blood levels of estradiol, the natural ligand of ER that induces ER activation. Moreover, low estradiol levels were associated with a lower risk of relapse in HER2-positive BC patients treated with adjuvant trastuzumab. Overall, we found that HER2 mRNA levels, but not protein levels, indicate the HER2 dependency of tumor cells and low estrogen-dependent ER activity in HER2-positive tumors.

4.
Front Oncol ; 12: 947188, 2022.
Article in English | MEDLINE | ID: mdl-35912227

ABSTRACT

The microbiota is emerging as a key player in cancer due to its involvement in several host physiological functions, including digestion, development of the immune system, and modulation of endocrine function. Moreover, its participation in the efficacy of anticancer treatments has been well described. For instance, the involvement of the breast microbiota in breast cancer (BC) development and progression has gained ground in the past several years. In this review, we report and discuss new findings on the impact of the gut and breast microbiota on BC, focusing on the HER2+ BC subtype, and the possibility of defining microbial signatures that are associated with disease aggressiveness, treatment response, and therapy toxicity. We also discuss novel insights into the mechanisms through which microorganism-host interactions occur and the possibility of microbiota editing in the prevention and treatment optimization of BC.

5.
Cell ; 184(21): 5338-5356.e21, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34624222

ABSTRACT

The tumor microenvironment (TME) influences cancer progression and therapy response. Therefore, understanding what regulates the TME immune compartment is vital. Here we show that microbiota signals program mononuclear phagocytes in the TME toward immunostimulatory monocytes and dendritic cells (DCs). Single-cell RNA sequencing revealed that absence of microbiota skews the TME toward pro-tumorigenic macrophages. Mechanistically, we show that microbiota-derived stimulator of interferon genes (STING) agonists induce type I interferon (IFN-I) production by intratumoral monocytes to regulate macrophage polarization and natural killer (NK) cell-DC crosstalk. Microbiota modulation with a high-fiber diet triggered the intratumoral IFN-I-NK cell-DC axis and improved the efficacy of immune checkpoint blockade (ICB). We validated our findings in individuals with melanoma treated with ICB and showed that the predicted intratumoral IFN-I and immune compositional differences between responder and non-responder individuals can be transferred by fecal microbiota transplantation. Our study uncovers a mechanistic link between the microbiota and the innate TME that can be harnessed to improve cancer therapies.


Subject(s)
Interferon Type I/metabolism , Membrane Proteins/metabolism , Microbiota , Monocytes/metabolism , Tumor Microenvironment , Akkermansia/drug effects , Akkermansia/physiology , Animals , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dietary Fiber/pharmacology , Dinucleoside Phosphates/administration & dosage , Dinucleoside Phosphates/pharmacology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunomodulation/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Macrophages/drug effects , Macrophages/metabolism , Melanoma/immunology , Melanoma/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbiota/drug effects , Monocytes/drug effects , Phagocytes/drug effects , Phagocytes/metabolism , Transcription, Genetic/drug effects , Tumor Microenvironment/drug effects
6.
iScience ; 24(9): 103012, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34522855

ABSTRACT

The gut microbiota's function in regulating health has seen it linked to disease progression in several cancers. However, there is limited research detailing its influence in breast cancer (BrCa). This study found that antibiotic-induced perturbation of the gut microbiota significantly increases tumor progression in multiple BrCa mouse models. Metagenomics highlights the common loss of several bacterial species following antibiotic administration. One such bacteria, Faecalibaculum rodentium, rescued this increased tumor growth. Single-cell transcriptomics identified an increased number of cells with a stromal signature in tumors, and subsequent histology revealed an increased abundance of mast cells in the tumor stromal regions. We show that administration of a mast cell stabilizer, cromolyn, rescues increased tumor growth in antibiotic treated animals but has no influence on tumors from control cohorts. These findings highlight that BrCa-microbiota interactions are different from other cancers studied to date and suggest new research avenues for therapy development.

7.
Cancer Res ; 81(8): 2195-2206, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33483370

ABSTRACT

Emerging evidence indicates that gut microbiota affect the response to anticancer therapies by modulating the host immune system. In this study, we investigated the impact of gut microbiota on immune-mediated trastuzumab antitumor efficacy in preclinical models of HER2-positive breast cancer and in 24 patients with primary HER2-positive breast cancer undergoing trastuzumab-containing neoadjuvant treatment. In mice, the antitumor activity of trastuzumab was impaired by antibiotic administration or fecal microbiota transplantation from antibiotic-treated donors. Modulation of the intestinal microbiota was reflected in tumors by impaired recruitment of CD4+ T cells and granzyme B-positive cells after trastuzumab treatment. Antibiotics caused reductions in dendritic cell (DC) activation and the release of IL12p70 upon trastuzumab treatment, a mechanism that was necessary for trastuzumab effectiveness in our model. In patients, lower α-diversity and lower abundance of Lachnospiraceae, Turicibacteraceae, Bifidobacteriaceae, and Prevotellaceae characterized nonresponsive patients (NR) compared with those who achieved pathologic complete response (R), similar to antibiotic-treated mice. The transfer of fecal microbiota from R and NR into mice bearing HER2-positive breast cancer recapitulated the response to trastuzumab observed in patients. Fecal microbiota ß-diversity segregated patients according to response and positively correlated with immune signature related to interferon (IFN) and NO2-IL12 as well as activated CD4+ T cells and activated DCs in tumors. Overall, our data reveal the direct involvement of the gut microbiota in trastuzumab efficacy, suggesting that manipulation of the gut microbiota is an optimal future strategy to achieve a therapeutic effect or to exploit its potential as a biomarker for treatment response. SIGNIFICANCE: Evidence of gut microbiota involvement in trastuzumab efficacy represents the foundation for new therapeutic strategies aimed at manipulating commensal bacteria to improve response in trastuzumab-resistant patients.See related commentary by Sharma, p. 1937 GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2195/F1.large.jpg.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Gastrointestinal Microbiome/physiology , Receptor, ErbB-2 , Trastuzumab/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Breast Neoplasms/immunology , Bridged-Ring Compounds/therapeutic use , CD4-Positive T-Lymphocytes , Cyclophosphamide/therapeutic use , Cytokines/blood , Dendritic Cells/drug effects , Doxorubicin/therapeutic use , Fecal Microbiota Transplantation , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Granzymes , Humans , Immune System , Immunity, Mucosal , Interferons/metabolism , Interleukin-12/metabolism , Mice , Neoadjuvant Therapy , Nitric Oxide/metabolism , Streptomycin/pharmacology , Taxoids/therapeutic use , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vancomycin/pharmacology
8.
Oncoimmunology ; 8(1): e1512942, 2019.
Article in English | MEDLINE | ID: mdl-30546951

ABSTRACT

Through whole-transcriptome profiling of HER2+ breast carcinomas (BCs), we previously showed that those sensitive to trastuzumab are addicted to this oncoprotein and are enriched in immune pathways, raising the hypothesis that HER2 itself regulates immune cell recruitment. In the present study we investigated the relationship between HER2 activity and the pro-trastuzumab tumor immune milieu. Gene expression profiling and immunohistochemistry analysis of 53 HER2+ BCs showed that trastuzumab-sensitive tumors expressed significantly higher levels of chemokines involved in immune cell recruitment, with higher infiltration of T cells and monocytes, and higher levels of PD-1 ligands than tumors that do not benefit from trastuzumab. In vitro analysis in HER2+ BC cells revealed that CCL2 production was induced by HER2 stimulation with EGF/HRG via the PI3K-NF-kB axis, and down-modulated by HER2 inhibition with trastuzumab. CCL2 expression was higher in HER2+/ER- than HER2+/ER+ BC cell lines, and degradation of ER by fulvestrant induced an enhancement in NF-κB transcriptional activity and consequent CCL2 expression. Trastuzumab efficacy relied on CCL2 levels and monocytes present in the tumor microenvironment in FVB mice bearing HER2+ mammary carcinoma cells. HER2 signals were also found to sustain the expression of PD-1 ligands in tumor cells via the MEK pathway. Overall, our results support the concept that the activated HER2 oncogene regulates recruitment and activation of tumor infiltrating immune cells and trastuzumab activity by inducing CCL2 and PD-1 ligands and that ER activity negatively controls the HER2-driven pro-trastuzumab tumor microenvironment.

9.
Br J Cancer ; 119(12): 1487-1494, 2018 12.
Article in English | MEDLINE | ID: mdl-30478407

ABSTRACT

BACKGROUND: Optimising the selection of HER2-targeted regimens by identifying subsets of HER2-positive breast cancer (BC) patients who need more or less therapy remains challenging. We analysed BC samples before and after treatment with 1 cycle of trastuzumab according to the response to trastuzumab. METHODS: Gene expression profiles of pre- and post-treatment tumour samples from 17 HER2-positive BC patients were analysed on the Illumina platform. Tumour-associated immune pathways and blood counts were analysed with regard to the response to trastuzumab. HER2-positive murine models with differential responses to trastuzumab were used to reproduce and better characterise these data. RESULTS: Patients who responded to single-agent trastuzumab had basal tumour biopsies that were enriched in immune pathways, particularly the MHC-II metagene. One cycle of trastuzumab modulated the expression levels of MHC-II genes, which increased in patients who had a complete response on treatment with trastuzumab and chemotherapy. Trastuzumab increased the MHC-II-positive cell population, primarily macrophages, only in the tumour microenvironment of responsive mice. In patients who benefited from complete trastuzumab therapy and in mice that harboured responsive tumours circulating neutrophil levels declined, but this cell subset rose in nonresponsive tumours. CONCLUSIONS: Short treatment with trastuzumab induces local and systemic immunomodulation that is associated with clinical outcomes.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Immunologic Factors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/pharmacology , Animals , Breast Neoplasms/immunology , Female , Genes, MHC Class II , Humans , Ki-67 Antigen/analysis , Mice , Receptor, ErbB-2/analysis , Transcriptome , Trastuzumab/therapeutic use , Tumor Microenvironment
10.
Cancer Lett ; 384: 94-100, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27693459

ABSTRACT

Exosomes-secreted microRNAs play an important role in metastatic spread. During this process breast cancer cells acquire the ability to transmigrate through blood vessels by inducing changes in the endothelial barrier. We focused on miR-939 that is predicted to target VE-cadherin, a component of adherens junction involved in vessel permeability. By in silico analysis miR-939 was found highly expressed in the basal-like tumor subtypes and in our cohort of 63 triple-negative breast cancers (TNBCs) its expression significantly interacted with lymph node status in predicting disease-free survival probability. We demonstrated, in vitro, that miR-939 directly targets VE-cadherin leading to an increase in HUVECs monolayer permeability. MDA-MB-231 cells transfected with a miR-939 mimic, released miR-939 in exosomes that, once internalized in endothelial cells, favored trans-endothelial migration of MDA-MB-231-GFP cells by the disruption of the endothelial barrier. Notably, when up taken in endothelial cells exosomes caused VE-cadherin down-regulation specifically through miR-939 as we demonstrated by inhibiting miR-939 expression in exosomes-releasing TNBC cells. Together, our data indentify an extracellular pro-tumorigenic role for tumor-derived, exosome-associated miR-939 that can explain its association with worse prognosis in TNBCs.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/metabolism , Transendothelial and Transepithelial Migration , Triple Negative Breast Neoplasms/metabolism , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Disease-Free Survival , Down-Regulation , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/pathology , Humans , MicroRNAs/genetics , Neoplasm Metastasis , Paracrine Communication , Permeability , Proportional Hazards Models , Signal Transduction , Time Factors , Transfection , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
11.
Dis Markers ; 2017: 7849108, 2017.
Article in English | MEDLINE | ID: mdl-29403144

ABSTRACT

HER2 is overexpressed in 20% of invasive breast cancers (BCs) and correlates with a more aggressive disease. Until the advent of targeted agents, HER2 was associated with worse outcomes. Rationally designed HER2-targeted agents have been developed and introduced into clinical practice for women with HER2-amplified BC, improving disease-free and overall survival for primary and metastatic tumors. Trastuzumab, a recombinant humanized anti-HER2 monoclonal antibody, combined with chemotherapy, remains the standard of care for patients with HER2-positive BCs. However, many patients do not respond to this agent, whereas newer drugs have proven to be efficacious in clinical trials. The identification of biomarkers that select sensitive tumors and patients who will benefit from these new agents would help the incorporation of these therapies, limiting the risk of side effects and overtreatment and improving the outcomes of all patients with early-stage HER2-positive BC. We review the mechanisms of action of HER2-targeting agents, focusing on the involvement of the immune system and related predictive biomarkers.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/blood , Breast Neoplasms/blood , Female , Humans , Protein Kinase Inhibitors/administration & dosage , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
12.
Oncotarget ; 7(1): 255-65, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26595802

ABSTRACT

Recent clinical data indicate a synergistic therapeutic effect between trastuzumab and taxanes in neoadjuvantly treated HER2-positive breast cancer (BC) patients. In HER2+ BC experimental models and patients, we investigated whether this synergy depends on the ability of drug-induced stress to improve NK cell effectiveness and thus trastuzumab-mediated ADCC. HER2+ BC cell lines BT474 and MDAMB361 treated with docetaxel showed up-modulation of NK activator ligands both in vitro and in vivo, accompanied by a 15-40% increase in in vitro trastuzumab-mediated ADCC; antibodies blocking the NKG2D receptor significantly reduced this enhancement. NKG2D receptor expression was increased by docetaxel treatment in circulating and splenic NK cells from mice xenografted with tumor cells, an increase related to expansion of the CD11b+Ly6G+ cell population. Accordingly, NK cells derived from HER2+ BC patients after treatment with taxane-containing therapy expressed higher levels of NKG2D receptor than before treatment. Moreover, plasma obtained from these patients recapitulated the modulation of NKG2D on healthy donors' NK cells, improving their trastuzumab-mediated activity in vitro. This enhancement occurred mainly using plasma from patients with low NKG2D basal expression. Our results indicate that taxanes increase tumor susceptibility to ADCC by acting on tumor and NK cells, and suggest that taxanes concomitantly administered with trastuzumab could maximize the antibody effect, especially in patients with low basal immune effector cytotoxic activity.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Killer Cells, Natural/drug effects , NK Cell Lectin-Like Receptor Subfamily K/immunology , Animals , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/pharmacology , Cell Line, Tumor , Cells, Cultured , Docetaxel , Drug Synergism , Female , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, SCID , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Taxoids/administration & dosage , Taxoids/pharmacology , Trastuzumab/administration & dosage , Trastuzumab/pharmacology , Xenograft Model Antitumor Assays
13.
Int J Cancer ; 134(12): 2789-97, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24242003

ABSTRACT

Altered degradation and deposition of extracellular matrix are hallmarks of tumor progression and response to therapy. From a microarray supervised analysis on a dataset of chemotherapy-treated breast carcinoma patients, maspin, a member of the serpin protease inhibitor family, has been the foremost variable identified in non-responsive versus responsive tumors. Accordingly, in a series of 52 human breast carcinomas, we detected high maspin expression in tumors that progressed under doxorubicin (DXR)-based chemotherapy. Our analysis of the role of maspin in response to chemotherapy in human MCF7 and MDAMB231 breast and SKOV3 ovarian carcinoma cells transfected to overexpress maspin and injected into mice showed that maspin overexpression led to DXR resistance through the maspin-induced collagen-enriched microenvironment and that an anti-maspin neutralizing monoclonal antibody reversed the collagen-dependent DXR resistance. Impaired diffusion and decreased DXR activity were also found in tumors derived from Matrigel-embedded cells, where abundant collagen fibers characterize the tumor matrix. Conversely, liposome-based DXR reached maspin-overexpressing tumor cells despite the abundant extracellular matrix and was more efficient in reducing tumor growth. Our results identify maspin-induced accumulation of collagen fibers as a cause of disease progression under DXR chemotherapy for breast cancer. Use of a more hydrophilic DXR formulation or of a maspin inhibitor in combination with chemotherapy holds the promise of more consistent responses to maspin-overexpressing tumors and dense-matrix tumors in general.


Subject(s)
Breast Neoplasms/metabolism , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Serpins/metabolism , Tumor Microenvironment/drug effects , Animals , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/immunology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen/metabolism , Disease Progression , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Serpins/biosynthesis , Serpins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...