Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 35
Filtrer
1.
NPJ Breast Cancer ; 9(1): 41, 2023 May 20.
Article de Anglais | MEDLINE | ID: mdl-37210417

RÉSUMÉ

This clinical trial combined fulvestrant with the anti-androgen enzalutamide in women with metastatic ER+/HER2- breast cancer (BC). Eligible patients were women with ECOG 0-2, ER+/HER2- measurable or evaluable metastatic BC. Prior fulvestrant was allowed. Fulvestrant was administered at 500 mg IM on days 1, 15, 29, and every 4 weeks thereafter. Enzalutamide was given at 160 mg po daily. Fresh tumor biopsies were required at study entry and after 4 weeks of treatment. The primary efficacy endpoint of the trial was the clinical benefit rate at 24 weeks (CBR24). The median age was 61 years (46-87); PS 1 (0-1); median of 4 prior non-hormonal and 3 prior hormonal therapies for metastatic disease. Twelve had prior fulvestrant, and 91% had visceral disease. CBR24 was 25% (7/28 evaluable). Median progression-free survival (PFS) was 8 weeks (95% CI: 2-52). Adverse events were as expected for hormonal therapy. Significant (p < 0.1) univariate relationships existed between PFS and ER%, AR%, and PIK3CA and/or PTEN mutations. Baseline levels of phospho-proteins in the mTOR pathway were more highly expressed in biopsies of patients with shorter PFS. Fulvestrant plus enzalutamide had manageable side effects. The primary endpoint of CBR24 was 25% in heavily pretreated metastatic ER+/HER2- BC. Short PFS was associated with activation of the mTOR pathway, and PIK3CA and/or PTEN mutations were associated with an increased hazard of progression. Thus, a combination of fulvestrant or other SERD plus AKT/PI3K/mTOR inhibitor with or without AR inhibition warrants investigation in second-line endocrine therapy of metastatic ER+ BC.

2.
Cells ; 12(3)2023 01 29.
Article de Anglais | MEDLINE | ID: mdl-36766786

RÉSUMÉ

Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype for which no effective targeted therapies are available. Growing evidence suggests that chemotherapy-resistant cancer cells with stem-like properties (CSC) may repopulate the tumor. The androgen receptor (AR) is expressed in up to 50% of TNBCs, and AR inhibition decreases CSC and tumor initiation. Runt-related transcription factor 1 (RUNX1) correlates with poor prognosis in TNBC and is regulated by the AR in prostate cancer. Our group has shown that RUNX1 promotes TNBC cell migration and regulates tumor gene expression. We hypothesized that RUNX1 is regulated by the AR and that both may work together in TNBC CSC to promote disease recurrence following chemotherapy. Chromatin immunoprecipitation sequencing (ChIP-seq) experiments in MDA-MB-453 revealed AR binding to RUNX1 regulatory regions. RUNX1 expression is upregulated by dihydrotestosterone (DHT) in MDA-MB-453 and in an AR+-TNBC HCI-009 patient-derived xenograft (PDX) tumors (p < 0.05). RUNX1 is increased in a CSC-like experimental model in MDA-MB-453 and SUM-159PT cells (p < 0.05). Inhibition of RUNX1 transcriptional activity reduced the expression of CSC markers. Interestingly, RUNX1 inhibition reduced cell viability and enhanced paclitaxel and enzalutamide sensitivity. Targeting RUNX1 may be an attractive strategy to potentiate the anti-tumor effects of AR inhibition, specifically in the slow-growing CSC-like populations that resist chemotherapy which lead to metastatic disease.


Sujet(s)
Tumeurs du sein triple-négatives , Humains , Lignée cellulaire tumorale , Sous-unité alpha 2 du facteur CBF/génétique , Récidive tumorale locale , Récepteurs aux androgènes/métabolisme , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Femelle
3.
Cancers (Basel) ; 14(14)2022 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-35884604

RÉSUMÉ

Triple-negative breast cancer (TNBC) often undergoes at least partial epithelial-to-mesenchymal transition (EMT) to facilitate metastasis. Identifying EMT-associated characteristics can reveal novel dependencies that may serve as therapeutic vulnerabilities in this aggressive breast cancer subtype. We found that NPC1, which encodes the lysosomal cholesterol transporter Niemann-Pick type C1 is highly expressed in TNBC as compared to estrogen receptor-positive (ER+) breast cancer, and is significantly elevated in high-grade disease. We demonstrated that NPC1 is directly targeted by microRNA-200c (miR-200c), a potent suppressor of EMT, providing a mechanism for its differential expression in breast cancer subtypes. The silencing of NPC1 in TNBC causes an accumulation of cholesterol-filled lysosomes, and drives decreased growth in soft agar and invasive capacity. Conversely, overexpression of NPC1 in an ER+ cell line increases invasion and growth in soft agar. We further identified TNBC cell lines as cholesterol auxotrophs, however, they do not solely depend on NPC1 for adequate cholesterol supply. The silencing of NPC1 in TNBC cell lines led to altered mitochondrial function and morphology, suppression of mTOR signaling, and accumulation of autophagosomes. A small molecule inhibitor of NPC1, U18666A, decreased TNBC proliferation and synergized with the chemotherapeutic drug, paclitaxel. This work suggests that NPC1 promotes aggressive characteristics in TNBC, and identifies NPC1 as a potential therapeutic target.

4.
NPJ Breast Cancer ; 8(1): 62, 2022 May 10.
Article de Anglais | MEDLINE | ID: mdl-35538119

RÉSUMÉ

Both TP53 and ESR1 mutations occur frequently in estrogen receptor positive (ER+) metastatic breast cancers (MBC) and their distinct roles in breast cancer tumorigenesis and progression are well appreciated. Recent clinical studies discovered mutual exclusivity between TP53 and ESR1 mutations in metastatic breast cancers; however, mechanisms underlying this intriguing clinical observation remain largely understudied and unknown. Here, we explored the interplay between TP53 and ESR1 mutations using publicly available clinical and experimental data sets. We first confirmed the robust mutational exclusivity using six independent cohorts with 1,056 ER+ MBC samples and found that the exclusivity broadly applies to all ER+ breast tumors regardless of their clinical and distinct mutational features. ESR1 mutant tumors do not exhibit differential p53 pathway activity, whereas we identified attenuated ER activity and expression in TP53 mutant tumors, driven by a p53-associated E2 response gene signature. Further, 81% of these p53-associated E2 response genes are either direct targets of wild-type (WT) p53-regulated transactivation or are mutant p53-associated microRNAs, representing bimodal mechanisms of ER suppression. Lastly, we analyzed the very rare cases with co-occurrences of TP53 and ESR1 mutations and found that their simultaneous presence was also associated with reduced ER activity. In addition, tumors with dual mutations showed higher levels of total and PD-L1 positive macrophages. In summary, our study utilized multiple publicly available sources to explore the mechanism underlying the mutual exclusivity between ESR1 and TP53 mutations, providing further insights and testable hypotheses of the molecular interplay between these two pivotal genes in ER+ MBC.

5.
Breast Cancer Res ; 23(1): 102, 2021 11 04.
Article de Anglais | MEDLINE | ID: mdl-34736512

RÉSUMÉ

PURPOSE: Accumulating evidence has attracted attention to the androgen receptor (AR) as a biomarker and therapeutic target in breast cancer. We hypothesized that AR activity within the tumor has clinical implications and investigated whether androgen responsive serum factors might serve as a minimally invasive indicator of tumor AR activity. METHODS: Based on a comprehensive gene expression analysis of an AR-positive, triple negative breast cancer patient-derived xenograft (PDX) model, 163 dihydrotestosterone (DHT)-responsive genes were defined as an androgen responsive gene set. Among them, we focused on genes that were DHT-responsive that encode secreted proteins, namely KLK3, AZGP1 and PIP, that encode the secreted factors prostate specific antigen (PSA), zinc-alpha-2-glycoprotein (ZAG) and prolactin induced protein (PIP), respectively. Using AR-positive breast cancer cell lines representing all breast cancer subtypes, expression of candidate factors was assessed in response to agonist DHT and antagonist enzalutamide. Gene set enrichment analysis (GSEA) was performed on publically available gene expression datasets from breast cancer patients to analyze the relationship between genes encoding the secreted factors and other androgen responsive gene sets in each breast cancer subtype. RESULTS: Anti-androgen treatment decreased proliferation in all cell lines tested representing various tumor subtypes. Expression of the secreted factors was regulated by AR activation in the majority of breast cancer cell lines. In GSEA, the candidate genes were positively correlated with an androgen responsive gene set across breast cancer subtypes. CONCLUSION: KLK3, AZGP1 and PIP are AR regulated and reflect tumor AR activity. Further investigations are needed to examine the potential efficacy of these factors as serum biomarkers.


Sujet(s)
Adipokines/métabolisme , Tumeurs du sein/métabolisme , Kallicréines/métabolisme , Protéines de transport membranaire/métabolisme , Antigène spécifique de la prostate/métabolisme , Récepteurs aux androgènes/métabolisme , Adipokines/génétique , Antagonistes du récepteur des androgènes/pharmacologie , Androgènes/pharmacologie , Animaux , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Kallicréines/génétique , Mâle , Protéines de transport membranaire/génétique , Souris , Antigène spécifique de la prostate/génétique , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
6.
NPJ Breast Cancer ; 7(1): 64, 2021 May 27.
Article de Anglais | MEDLINE | ID: mdl-34045467

RÉSUMÉ

Many immune suppressive mechanisms utilized by triple negative breast cancer (TNBC) are regulated by oncogenic epithelial-to-mesenchymal transition (EMT). How TNBC EMT impacts innate immune cells is not fully understood. To determine how TNBC suppresses antitumor macrophages, we used microRNA-200c (miR-200c), a powerful repressor of EMT, to drive mesenchymal-like mouse mammary carcinoma and human TNBC cells toward a more epithelial state. MiR-200c restoration significantly decreased growth of mouse mammary carcinoma Met-1 cells in culture and in vivo. Cytokine profiling of Met-1 and human BT549 cells revealed that miR-200c upregulated cytokines, such as granulocyte-macrophage colony-stimulating factor (GM-CSF), promoted M1 antitumor macrophage polarization. Cytokines upregulated by miR-200c correlated with an epithelial gene signature and M1 macrophage polarization in BC patients and predicted a more favorable overall survival for TNBC patients. Our findings demonstrate that immunogenic cytokines (e.g., GM-CSF) are suppressed in aggressive TNBC, warranting further investigation of cytokine-based therapies to limit disease recurrence.

7.
Mol Cancer Ther ; 20(6): 1062-1071, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33722849

RÉSUMÉ

Triple-negative breast cancer (TNBC) is an aggressive subtype, with a peak recurrence rate within the first few years after diagnosis. Few targeted therapies are available to treat this breast cancer subtype, defined by the lack of estrogen receptor (ER) and progesterone receptor and without amplification of human epidermal growth factor receptor 2 (HER2). Although cell cycle cyclin-dependent kinase (CDK) 4/6 inhibitors are approved for treatment of ER-positive (ER+) breast cancer, they have not proven effective as monotherapy in patients with TNBC. The androgen receptor (AR) has emerged as a therapeutic target in a subset of TNBCs and with significant clinical benefit observed in multiple trials. The purpose of this study was to investigate the preclinical activity of the CDK4/6 inhibitor, abemaciclib, in combination with an agent that targets both androgen biosynthesis and AR activity, seviteronel, using TNBC cell lines expressing high AR, cell line xenografts, and an AR-positive (AR+), androgen-responsive TNBC patient-derived xenograft (PDX). Single-cell RNA sequencing demonstrated heterogeneity in AR levels, even in a highly AR+ cell line, and identified cell cycle pathway activation in ARHigh- versus ARLow-expressing cells. Combination treatment with the cell cycle CDK4/6 inhibitor, abemaciclib, and seviteronel showed synergy in an AR+ TNBC model compared with each drug alone. Although cell cycle inhibitors are FDA approved for use in ER+ breast cancer, our studies suggest that they may also be effective in AR+ TNBC, perhaps combined with AR-targeted agents.


Sujet(s)
Antagonistes du récepteur des androgènes/usage thérapeutique , Cycle cellulaire/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/traitement médicamenteux , Antagonistes du récepteur des androgènes/pharmacologie , Animaux , Modèles animaux de maladie humaine , Humains , Souris
8.
Endocrinology ; 162(2)2021 02 01.
Article de Anglais | MEDLINE | ID: mdl-33294922

RÉSUMÉ

Triple-negative breast cancer (TNBC) is an aggressive subtype with peak recurrence as metastatic disease within the first few years of diagnosis. Androgen receptor (AR) expression is increased in anchorage-independent cells in TNBC preclinical models. Both AR knockdown and inhibition lead to reduced TNBC invasion in vitro, reduced tumorigenicity, and less recurrence in vivo in preclinical models. Transforming growth factor ß (TGFß) pathway gene signatures also increased during anchorage-independent survival both in vitro and in vivo in preclinical models and in circulating tumor cells (CTCs) from patients during emergence of chemo resistant disease. We hypothesized that a positive loop between AR and TGFß signaling facilitates TNBC anchorage-independent survival. We find that multiple components of the TGFß pathway, including TGFß1 and 3, as well as pathway activity measured by nuclear localization and transcriptional activity of phosphorylated Smad3, are enhanced in anchorage-independent conditions. Further, exogenous TGFß increased AR protein while TGFß inhibition decreased AR and TNBC viability, particularly under anchorage-independent culture conditions. ChIP-seq experiments revealed AR binding to TGFB1 and SMAD3 regulatory regions in MDA-MB-453 cells. In clinical datasets, TGFB3 and AR positively correlate and high expression of both genes together corresponded to significantly worse recurrence-free and overall survival in both ER-negative and basal-like breast cancer. Finally, inhibiting both AR and TGFß decreased cell survival, particularly under anchorage-independent conditions. These findings warrant further investigations into whether combined inhibition of AR and TGFß pathways might decrease metastatic recurrence rates and mortality from TNBC.


Sujet(s)
Anoïkis , Récidive tumorale locale/étiologie , Récepteurs aux androgènes/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Tumeurs du sein triple-négatives/métabolisme , Lignée cellulaire tumorale , Rétrocontrôle physiologique , Humains , Tumeurs du sein triple-négatives/mortalité
9.
Cancer Res ; 81(3): 732-746, 2021 02 01.
Article de Anglais | MEDLINE | ID: mdl-33184106

RÉSUMÉ

Mutations in ESR1 that confer constitutive estrogen receptor alpha (ER) activity in the absence of ligand are acquired by ≥40% of metastatic breast cancers (MBC) resistant to adjuvant aromatase inhibitor (AI) therapy. To identify targetable vulnerabilities in MBC, we examined steroid hormone receptors and tumor-infiltrating immune cells in metastatic lesions with or without ER mutations. ER and progesterone receptor (PR) were significantly lower in metastases with wild-type (WT) ER compared with those with mutant ER, suggesting that metastases that evade AI therapy by mechanism(s) other than acquiring ER mutations lose dependency on ER and PR. Metastases with mutant ER had significantly higher T regulatory and Th cells, total macrophages, and programmed death ligand-1 (PD-L1)-positive immune-suppressive macrophages than those with WT ER. Breast cancer cells with CRISPR-Cas9-edited ER (D538G, Y537S, or WT) and patient-derived xenografts harboring mutant or WT ER revealed genes and proteins elevated in mutant ER cells, including androgen receptor (AR), chitinase-3-like protein 1 (CHI3L1), and IFN-stimulated genes (ISG). Targeting these proteins blunted the selective advantage of ER-mutant tumor cells to survive estrogen deprivation, anchorage independence, and invasion. Thus, patients with mutant ER MBC might respond to standard-of-care fulvestrant or other selective ER degraders when combined with AR or CHI3L1 inhibition, perhaps with the addition of immunotherapy. SIGNIFICANCE: Targetable alterations in MBC, including AR, CHI3L1, and ISG, arise following estrogen-deprivation, and ER-mutant metastases may respond to immunotherapies due to elevated PD-L1+ macrophages.See related article by Arnesen et al., p. 539.


Sujet(s)
Tumeurs du sein , Récepteur alpha des oestrogènes , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Récepteur alpha des oestrogènes/génétique , Fulvestrant/pharmacologie , Expression des gènes , Humains , Mutation
10.
Cancer Res ; 77(13): 3455-3466, 2017 07 01.
Article de Anglais | MEDLINE | ID: mdl-28512248

RÉSUMÉ

Preclinical and early clinical trials indicate that up to 50% of triple-negative breast cancers (TNBC) express androgen receptor (AR) and are potentially responsive to antiandrogens. However, the function of AR in TNBC and the mechanisms by which AR-targeted therapy reduces tumor burden are largely unknown. We hypothesized that AR maintains a cancer stem cell (CSC)-like tumor-initiating population and serves as an antiapoptotic factor, facilitating anchorage independence and metastasis. AR levels increased in TNBC cells grown in forced suspension culture compared with those in attached conditions, and cells that expressed AR resisted detachment-induced apoptosis. Culturing TNBC cells in suspension increased the CSC-like population, an effect reversed by AR inhibition. Pretreatment with enzalutamide (Enza) decreased the tumor-initiating capacity of TNBC cells and reduced tumor volume and viability when administered simultaneously or subsequent to the chemotherapeutic paclitaxel; simultaneous treatment more effectively suppressed tumor recurrence. Overall, our findings suggest that AR-targeted therapies may enhance the efficacy of chemotherapy even in TNBCs with low AR expression by targeting a CSC-like cell population with anchorage independence and invasive potential. Cancer Res; 77(13); 3455-66. ©2017 AACR.


Sujet(s)
Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Récepteurs aux androgènes/métabolisme , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Antagonistes du récepteur des androgènes/pharmacologie , Animaux , Benzamides , Lignée cellulaire tumorale , Femelle , Techniques de knock-down de gènes , Hétérogreffes , Humains , Souris nude , Nitriles , Paclitaxel/pharmacologie , 3-Phényl-2-thiohydantoïne/analogues et dérivés , 3-Phényl-2-thiohydantoïne/pharmacologie , Récepteurs aux androgènes/génétique , Activation de la transcription , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/prévention et contrôle , Régulation positive
11.
Horm Cancer ; 8(2): 69-77, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28194662

RÉSUMÉ

Triple-negative breast cancer (TNBC) has a faster rate of metastasis compared to other breast cancer subtypes, and no effective targeted therapies are currently FDA-approved. Recent data indicate that the androgen receptor (AR) promotes tumor survival and may serve as a potential therapeutic target in TNBC. Studies of AR in disease progression and the systemic effects of anti-androgens have been hindered by the lack of an AR-positive (AR+) immunocompetent preclinical model. In this study, we identified the transgenic MMTV-PyMT (mouse mammary tumor virus-polyoma middle tumor-antigen) mouse mammary gland carcinoma model of breast cancer and Met-1 cells derived from this model as tools to study the role of AR in breast cancer progression. AR protein expression was examined in late-stage primary tumors and lung metastases from MMTV-PyMT mice as well as in Met-1 cells by immunohistochemistry (IHC). Sensitivity of Met-1 cells to the AR agonist dihydrotestosterone (DHT) and anti-androgen therapy was examined using cell viability, migration/invasion, and anchorage-independent growth assays. Late-stage primary tumors and lung metastases from MMTV-PyMT mice and Met-1 cells expressed abundant nuclear AR protein, while negative for estrogen and progesterone receptors. Met-1 sensitivity to DHT and AR antagonists demonstrated a reliance on AR for survival, and AR antagonists inhibited invasion and anchorage-independent growth. These data suggest that the MMTV-PyMT model and Met-1 cells may serve as valuable tools for mechanistic studies of the role of AR in disease progression and how anti-androgens affect the tumor microenvironment.


Sujet(s)
Tumeurs du poumon/anatomopathologie , Tumeurs expérimentales de la mamelle/métabolisme , Récepteurs aux androgènes/métabolisme , Tumeurs du sein triple-négatives/métabolisme , Antagonistes des androgènes/administration et posologie , Antagonistes des androgènes/pharmacologie , Animaux , Lignée cellulaire tumorale , Noyau de la cellule/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , 5alpha-Dihydrotestostérone/administration et posologie , 5alpha-Dihydrotestostérone/pharmacologie , Évolution de la maladie , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Tumeurs du poumon/secondaire , Virus de la tumeur mammaire de la souris/physiologie , Souris , Souris transgéniques
12.
Development ; 143(22): 4236-4248, 2016 11 15.
Article de Anglais | MEDLINE | ID: mdl-27729410

RÉSUMÉ

Profiling of RNA from mouse mammary epithelial cells (MECs) isolated on pregnancy day (P)14 and lactation day (L)2 revealed that the majority of differentially expressed microRNA declined precipitously between late pregnancy and lactation. The decline in miR-150, which exhibited the greatest fold-decrease, was verified quantitatively and qualitatively. To test the hypothesis that the decline in miR-150 is crucial for lactation, MEC-specific constitutive miR-150 was achieved by crossing ROSA26-lox-STOP-lox-miR-150 mice with WAP-driven Cre recombinase mice. Both biological and foster pups nursed by bitransgenic dams exhibited a dramatic decrease in survival compared with offspring nursed by littermate control dams. Protein products of predicted miR-150 targets Fasn, Olah, Acaca, and Stat5B were significantly suppressed in MECs of bitransgenic mice with constitutive miR-150 expression as compared with control mice at L2. Lipid profiling revealed a significant reduction in fatty acids synthesized by the de novo pathway in L2 MECs of bitransgenic versus control mice. Collectively, these data support the hypothesis that a synchronized decrease in miRNAs, such as miR-150, at late pregnancy serves to allow translation of targets crucial for lactation.


Sujet(s)
Lactation/génétique , Lipogenèse/génétique , Glandes mammaires animales/métabolisme , microARN/génétique , Animaux , Cellules cultivées , Régulation négative/génétique , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Lactation/métabolisme , Souris , Souris de lignée C57BL , Souris transgéniques , microARN/métabolisme , Analyse sur microréseau , Grossesse/génétique , Grossesse/métabolisme
13.
Mol Cancer Res ; 14(11): 1054-1067, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27565181

RÉSUMÉ

Androgen receptor (AR) is expressed in 90% of estrogen receptor alpha-positive (ER+) breast tumors, but its role in tumor growth and progression remains controversial. Use of two anti-androgens that inhibit AR nuclear localization, enzalutamide and MJC13, revealed that AR is required for maximum ER genomic binding. Here, a novel global examination of AR chromatin binding found that estradiol induced AR binding at unique sites compared with dihydrotestosterone (DHT). Estradiol-induced AR-binding sites were enriched for estrogen response elements and had significant overlap with ER-binding sites. Furthermore, AR inhibition reduced baseline and estradiol-mediated proliferation in multiple ER+/AR+ breast cancer cell lines, and synergized with tamoxifen and fulvestrant. In vivo, enzalutamide significantly reduced viability of tamoxifen-resistant MCF7 xenograft tumors and an ER+/AR+ patient-derived model. Enzalutamide also reduced metastatic burden following cardiac injection. Finally, in a comparison of ER+/AR+ primary tumors versus patient-matched local recurrences or distant metastases, AR expression was often maintained even when ER was reduced or absent. These data provide preclinical evidence that anti-androgens that inhibit AR nuclear localization affect both AR and ER, and are effective in combination with current breast cancer therapies. In addition, single-agent efficacy may be possible in tumors resistant to traditional endocrine therapy, as clinical specimens of recurrent disease demonstrate AR expression in tumors with absent or refractory ER. IMPLICATIONS: This study suggests that AR plays a previously unrecognized role in supporting E2-mediated ER activity in ER+/AR+ breast cancer cells, and that enzalutamide may be an effective therapeutic in ER+/AR+ breast cancers. Mol Cancer Res; 14(11); 1054-67. ©2016 AACR.


Sujet(s)
Tumeurs du sein/génétique , Chromatine/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , 3-Phényl-2-thiohydantoïne/analogues et dérivés , Récepteurs aux androgènes/métabolisme , Récepteurs des oestrogènes/génétique , Tamoxifène/administration et posologie , Anilides/pharmacologie , Benzamides , Sites de fixation , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Noyau de la cellule/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cyclohexanes/pharmacologie , Évolution de la maladie , Oestradiol , Femelle , Humains , Cellules MCF-7 , Nitriles , 3-Phényl-2-thiohydantoïne/administration et posologie , 3-Phényl-2-thiohydantoïne/pharmacologie , Récepteurs des oestrogènes/métabolisme , Tamoxifène/pharmacologie
14.
Hum Pathol ; 56: 40-51, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27260947

RÉSUMÉ

Dicer is an RNase III enzyme responsible for cleaving double-stranded RNAs into small interfering RNAs and microRNAs, which either target messenger RNA transcripts for degradation or inhibit translation. Dicer protein levels have been examined in breast cancer with contradictory results. Our goal was to resolve whether Dicer levels differ in breast cancer versus normal breast epithelium and between estrogen receptor-α-positive (ER+) or estrogen receptor-α-negative (ER-) primary breast cancers. We compared 3 different Dicer antibodies: Abcam 4A6, Abcam ab5818, and Sigma HPA000694, using immunohistochemistry and Western blot analyses. All 3 Dicer antibodies detected higher levels of Dicer in ER+ breast cancer cell lines versus ER-, and all 3 recognized exogenous overexpressed Dicer. In clinical specimens, all 3 antibodies detected higher Dicer in ER+ breast cancers versus triple-negative breast cancer (TNBC) but had very different staining patterns by immunohistochemistry on the same tumor samples. Using the optimal antibody, ab5818, selected for its sensitivity and specificity, Dicer protein expression was significantly higher in ER+ versus TNBC clinical specimens of primary tumor (P<.0001, unpaired t test). Dicer was also significantly higher in adjacent normal breast epithelium versus TNBC (P<.0001, paired t test; n=18 pairs). Differences in antibody performance may explain contrasting results observed in the literature regarding Dicer protein in breast cancer. If Dicer becomes more clinically relevant as a prognostic indicator, further antibody optimization and standardization will be critical.


Sujet(s)
Anticorps/immunologie , Spécificité des anticorps , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/enzymologie , DEAD-box RNA helicases/métabolisme , Immunohistochimie , Récepteurs des oestrogènes/analyse , Ribonuclease III/métabolisme , Tumeurs du sein triple-négatives/enzymologie , Adulte , Sujet âgé , Animaux , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/immunologie , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , DEAD-box RNA helicases/génétique , DEAD-box RNA helicases/immunologie , Femelle , Cellules HEK293 , Humains , Souris , Adulte d'âge moyen , Valeur prédictive des tests , Récepteur ErbB-2/analyse , Récepteurs à la progestérone/analyse , Reproductibilité des résultats , Ribonuclease III/génétique , Ribonuclease III/immunologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/anatomopathologie , Régulation positive , Jeune adulte
15.
Cancer Res ; 75(21): 4651-64, 2015 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-26363006

RÉSUMÉ

The ability of a cancer cell to develop resistance to anoikis, a programmed cell death process triggered by substratum detachment, is a critical step in the metastatic cascade. Triple-negative breast cancers (TNBC) exhibit higher rates of metastasis after diagnosis, relative to estrogen-positive breast cancers, but while TNBC cells are relatively more resistant to anoikis, the mechanisms involved are unclear. Through gene expression and metabolomic profiling of TNBC cells in forced suspension culture, we identified a molecular pathway critical for anchorage-independent cell survival. TNBC cells in suspension upregulated multiple genes in the kynurenine pathway of tryptophan catabolism, including the enzyme tryptophan 2,3-dioxygenase (TDO2), in an NF-κB-dependent manner. Kynurenine production mediated by TDO2 in TNBC cells was sufficient to activate aryl hydrocarbon receptor (AhR), an endogenous kynurenine receptor. Notably, pharmacologic inhibition or genetic attenuation of TDO2 or AhR increased cellular sensitivity to anoikis, and also reduced proliferation, migration, and invasion of TNBC cells. In vivo, TDO2 inhibitor-treated TNBC cells inhibited colonization of the lung, suggesting that TDO2 enhanced metastatic capacity. In clinical specimens of TNBC, elevated expression of TDO2 was associated with increased disease grade, estrogen receptor-negative status, and shorter overall survival. Our results define an NF-κB-regulated signaling axis that promotes anoikis resistance, suggest functional connections with inflammatory modulation by the kynurenine pathway, and highlight TDO2 as an attractive target for treatment of this aggressive breast cancer subtype.


Sujet(s)
Anoïkis/physiologie , Récepteurs à hydrocarbure aromatique/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tryptophane 2,3-dioxygenase/métabolisme , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Cynurénine/métabolisme , Souris , Souris de lignée NOD , Souris SCID , Facteur de transcription NF-kappa B/métabolisme , Invasion tumorale/génétique , Métastase tumorale , Interférence par ARN , Petit ARN interférent , Récepteurs à hydrocarbure aromatique/antagonistes et inhibiteurs , Récepteurs à hydrocarbure aromatique/génétique , Transduction du signal , Tryptophane 2,3-dioxygenase/antagonistes et inhibiteurs , Tryptophane 2,3-dioxygenase/génétique
16.
J Invest Dermatol ; 135(8): 2068-2076, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-25822579

RÉSUMÉ

In vitiligo, the autoimmune destruction of epidermal melanocytes produces white spots that can be repigmented by melanocyte precursors from the hair follicles, following stimulation with UV light. We examined by immunofluorescence the distribution of melanocyte markers (C-KIT, DCT, PAX3, and TYR) coupled with markers of proliferation (KI-67) and migration (MCAM) in precursors and mature melanocytes from the hair follicle and the epidermis of untreated and narrow band UVB (NBUVB)-treated human vitiligo skin. NBUVB was associated with a significant increase in the number of melanocytes in the infundibulum and with restoration of the normal melanocyte population in the epidermis, which was lacking in the untreated vitiligo. We identified several precursor populations (melanocyte stem cells, melanoblasts, and other immature phenotypes), and progressively differentiating melanocytes, some with putative migratory and/or proliferative abilities. The primary melanocyte germ was present in the untreated and treated hair follicle bulge, whereas a possible secondary melanocyte germ composed of C-KIT+ melanocytes was found in the infundibulum and interfollicular epidermis of UV-treated vitiligo. This is an exceptional model for studying the mobilization of melanocyte stem cells in human skin. Improved understanding of this process is essential for designing better treatments for vitiligo, ultimately based on melanocyte stem cell activation and mobilization.


Sujet(s)
Mélanocytes/anatomopathologie , Cellules souches/anatomopathologie , Rayons ultraviolets , Traitement par ultraviolets , Vitiligo/anatomopathologie , Vitiligo/radiothérapie , Différenciation cellulaire/effets des radiations , Mouvement cellulaire/effets des radiations , Prolifération cellulaire/effets des radiations , Épiderme/métabolisme , Épiderme/anatomopathologie , Épiderme/effets des radiations , Follicule pileux/métabolisme , Follicule pileux/anatomopathologie , Follicule pileux/effets des radiations , Humains , Intramolecular oxidoreductases/métabolisme , Mélanocytes/métabolisme , Mélanocytes/effets des radiations , Facteur de transcription PAX3 , Facteurs de transcription PAX/métabolisme , Phénotype , Protéines proto-oncogènes c-kit/métabolisme , Cellules souches/métabolisme , Cellules souches/effets des radiations , Vitiligo/métabolisme
17.
Mol Cancer Ther ; 14(3): 769-78, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25713333

RÉSUMÉ

Triple-negative breast cancer (TNBC) has the lowest 5-year survival rate of invasive breast carcinomas, and currently there are no approved targeted therapies for this aggressive form of the disease. The androgen receptor (AR) is expressed in up to one third of TNBC and we find that all AR(+) TNBC primary tumors tested display nuclear localization of AR, indicative of transcriptionally active receptors. While AR is most abundant in the "luminal AR (LAR)" molecular subtype of TNBC, here, for the first time, we use both the new-generation anti-androgen enzalutamide and AR knockdown to demonstrate that the other non-LAR molecular subtypes of TNBC are critically dependent on AR protein. Indeed, AR inhibition significantly reduces baseline proliferation, anchorage-independent growth, migration, and invasion and increases apoptosis in four TNBC lines (SUM159PT, HCC1806, BT549, and MDA-MB-231), representing three non-LAR TNBC molecular subtypes (mesenchymal-like, mesenchymal stem-like, and basal-like 2). In vivo, enzalutamide significantly decreases viability of SUM159PT and HCC1806 xenografts. Furthermore, mechanistic analysis reveals that AR activation upregulates secretion of the EGFR ligand amphiregulin (AREG), an effect abrogated by enzalutamide in vitro and in vivo. Exogenous AREG partially rescues the effects of AR knockdown on proliferation, migration, and invasion, demonstrating that upregulation of AREG is one mechanism by which AR influences tumorigenicity. Together, our findings indicate that non-LAR subtypes of TNBC are AR dependent and, moreover, that enzalutamide is a promising targeted therapy for multiple molecular subtypes of AR(+) TNBC.


Sujet(s)
3-Phényl-2-thiohydantoïne/analogues et dérivés , Récepteurs aux androgènes/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Antagonistes des androgènes/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Benzamides , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/génétique , Humains , Souris , Souris nude , Nitriles , 3-Phényl-2-thiohydantoïne/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique
18.
PLoS One ; 10(1): e0116895, 2015.
Article de Anglais | MEDLINE | ID: mdl-25603314

RÉSUMÉ

Ewing Sarcoma is an aggressive malignancy of bone and soft tissue affecting children and young adults. Ewing Sarcoma is driven by EWS/Ets fusion oncoproteins, which cause widespread alterations in gene expression in the cell. Dysregulation of receptor tyrosine kinase signaling, particularly involving IGF-1R, also plays an important role in Ewing Sarcoma pathogenesis. However, the basis of this dysregulation, including the relative contribution of EWS/Ets-dependent and independent mechanisms, is not well understood. In the present study, we identify variable expression of two modifiers of PI3K signaling activity, PIK3R3 and PTEN, in Ewing Sarcoma, and examine the consequences of this on PI3K pathway regulation and oncogenic phenotypes. Our findings indicate that PIK3R3 plays a growth-promotional role in Ewing Sarcoma, but suggest that this role is not strictly dependent on regulation of PI3K pathway activity. We further show that expression of PTEN, a well-established, potent tumor suppressor, is lost in a subset of Ewing Sarcomas, and that this loss strongly correlates with high baseline PI3K pathway activity in cell lines. In support of functional importance of PTEN loss in Ewing Sarcoma, we show that re-introduction of PTEN into two different PTEN-negative Ewing Sarcoma cell lines results in downregulation of PI3K pathway activity, and sensitization to the IGF-1R small molecule inhibitor OSI-906. Our findings also suggest that PTEN levels may contribute to sensitivity of Ewing Sarcoma cells to the microtubule inhibitor vincristine, a relevant chemotherapeutic agent in this cancer. Our studies thus identify PIK3R3 and PTEN as modifiers of oncogenic phenotypes in Ewing Sarcoma, with potential clinical implications.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Phosphohydrolase PTEN/génétique , Phosphatidylinositol 3-kinases/génétique , Sarcome d'Ewing/métabolisme , Sarcome d'Ewing/anatomopathologie , Lignée cellulaire tumorale , Humains , Phosphohydrolase PTEN/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéine proto-oncogène c-fli-1/génétique , Protéine proto-oncogène c-fli-1/métabolisme , Sarcome d'Ewing/génétique
19.
Horm Cancer ; 5(6): 374-89, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25213330

RÉSUMÉ

The anti-diabetic drug metformin (1,1-dimethylbiguanide hydrochloride) reduces both the incidence and mortality of several types of cancer. Metformin has been shown to selectively kill cancer stem cells, and triple-negative breast cancer (TNBC) cell lines are more sensitive to the effects of metformin as compared to luminal breast cancer. However, the mechanism underlying the enhanced susceptibility of TNBC to metformin has not been elucidated. Expression profiling of metformin-treated TNBC lines revealed fatty acid synthase (FASN) as one of the genes most significantly downregulated following 24 h of treatment, and a decrease in FASN protein was also observed. Since FASN is critical for de novo fatty acid synthesis and is important for the survival of TNBC, we hypothesized that FASN downregulation facilitates metformin-induced apoptosis. Profiling studies also exposed a rapid metformin-induced increase in miR-193 family members, and miR-193b directly targets the FASN 3'UTR. Addition of exogenous miR-193b mimic to untreated TNBC cells decreased FASN protein expression and increased apoptosis of TNBC cells, while spontaneously immortalized, non-transformed breast epithelial cells remained unaffected. Conversely, antagonizing miR-193 activity impaired the ability of metformin to decrease FASN and cause cell death. Further, the metformin-stimulated increase in miR-193 resulted in reduced mammosphere formation by TNBC lines. These studies provide mechanistic insight into metformin-induced killing of TNBC.


Sujet(s)
Fatty acid synthase type I/métabolisme , Metformine/usage thérapeutique , microARN/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/traitement médicamenteux , Régions 3' non traduites/génétique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Fatty acid synthase type I/génétique , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Glandes mammaires humaines/effets des médicaments et des substances chimiques , microARN/génétique , Cellules souches tumorales/anatomopathologie , Tumeurs du sein triple-négatives/anatomopathologie
20.
Breast Cancer Res ; 16(1): R7, 2014 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-24451109

RÉSUMÉ

INTRODUCTION: The androgen receptor (AR) is widely expressed in breast cancers and has been proposed as a therapeutic target in estrogen receptor alpha (ER) negative breast cancers that retain AR. However, controversy exists regarding the role of AR, particularly in ER + tumors. Enzalutamide, an AR inhibitor that impairs nuclear localization of AR, was used to elucidate the role of AR in preclinical models of ER positive and negative breast cancer. METHODS: We examined nuclear AR to ER protein ratios in primary breast cancers in relation to response to endocrine therapy. The effects of AR inhibition with enzalutamide were examined in vitro and in preclinical models of ER positive and negative breast cancer that express AR. RESULTS: In a cohort of 192 women with ER + breast cancers, a high ratio of AR:ER (≥2.0) indicated an over four fold increased risk for failure while on tamoxifen (HR = 4.43). The AR:ER ratio had an independent effect on risk for failure above ER % staining alone. AR:ER ratio is also an independent predictor of disease-free survival (HR = 4.04, 95% CI: 1.68, 9.69; p = 0.002) and disease specific survival (HR = 2.75, 95% CI: 1.11, 6.86; p = 0.03). Both enzalutamide and bicalutamide inhibited 5-alpha-dihydrotestosterone (DHT)-mediated proliferation of breast cancer lines in vitro; however, enzalutamide uniquely inhibited estradiol (E2)-mediated proliferation of ER+/AR + breast cancer cells. In MCF7 xenografts (ER+/AR+) enzalutamide inhibited E2-driven tumor growth as effectively as tamoxifen by decreasing proliferation. Enzalutamide also inhibited DHT- driven tumor growth in both ER positive (MCF7) and negative (MDA-MB-453) xenografts, but did so by increasing apoptosis. CONCLUSIONS: AR to ER ratio may influence breast cancer response to traditional endocrine therapy. Enzalutamide elicits different effects on E2-mediated breast cancer cell proliferation than bicalutamide. This preclinical study supports the initiation of clinical studies evaluating enzalutamide for treatment of AR+ tumors regardless of ER status, since it blocks both androgen- and estrogen- mediated tumor growth.


Sujet(s)
Antagonistes des androgènes/usage thérapeutique , Antagonistes du récepteur des androgènes/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Récepteur alpha des oestrogènes/métabolisme , 3-Phényl-2-thiohydantoïne/analogues et dérivés , Anilides/usage thérapeutique , Animaux , Antinéoplasiques hormonaux/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Benzamides , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie sans rechute , Femelle , Humains , Cellules MCF-7 , Souris , Adulte d'âge moyen , Transplantation tumorale , Nitriles/usage thérapeutique , 3-Phényl-2-thiohydantoïne/usage thérapeutique , Récepteurs aux androgènes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tamoxifène/usage thérapeutique , Composés tosyliques/usage thérapeutique , Transplantation hétérologue
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...