Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
2.
Oncogene ; 40(5): 997-1011, 2021 02.
Article in English | MEDLINE | ID: mdl-33323970

ABSTRACT

Estrogen receptor alpha gene (ESR1) mutations occur frequently in ER-positive metastatic breast cancer, and confer clinical resistance to aromatase inhibitors. Expression of the ESR1 Y537S mutation induced an epithelial-mesenchymal transition (EMT) with cells exhibiting enhanced migration and invasion potential in vitro. When small subpopulations of Y537S ESR1 mutant cells were injected along with WT parental cells, tumor growth was enhanced with mutant cells becoming the predominant population in distant metastases. Y537S mutant primary xenograft tumors were resistant to the antiestrogen tamoxifen (Tam) as well as to estradiol (E2) withdrawal. Y537S ESR1 mutant primary tumors metastasized efficiently in the absence of E2; however, Tam treatment significantly inhibited metastasis to distant sites. We identified a nine-gene expression signature, which predicted clinical outcomes of ER-positive breast cancer patients, as well as breast cancer metastasis to the lung. Androgen receptor (AR) protein levels were increased in mutant models, and the AR agonist dihydrotestosterone significantly inhibited estrogen-regulated gene expression, EMT, and distant metastasis in vivo, suggesting that AR may play a role in distant metastatic progression of ESR1 mutant tumors.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Receptor alpha/genetics , Receptors, Androgen/genetics , Tamoxifen/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Estradiol/metabolism , Estrogens/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mutation/genetics , Neoplasm Metastasis , Receptors, Androgen/drug effects , Xenograft Model Antitumor Assays
3.
Br J Cancer ; 124(1): 191-206, 2021 01.
Article in English | MEDLINE | ID: mdl-33257837

ABSTRACT

BACKGROUND: Oestrogen Receptor 1 (ESR1) mutations are frequently acquired in oestrogen receptor (ER)-positive metastatic breast cancer (MBC) patients who were treated with aromatase inhibitors (AI) in the metastatic setting. Acquired ESR1 mutations are associated with poor prognosis and there is a lack of effective therapies that selectively target these cancers. METHODS: We performed a proteomic kinome analysis in ESR1 Y537S mutant cells to identify hyperactivated kinases in ESR1 mutant cells. We validated Recepteur d'Origine Nantais (RON) and PI3K hyperactivity through phospho-immunoblot analysis, organoid growth assays, and in an in vivo patient-derived xenograft (PDX) metastatic model. RESULTS: We demonstrated that RON was hyperactivated in ESR1 mutant models, and in acquired palbociclib-resistant (PalbR) models. RON and insulin-like growth factor 1 receptor (IGF-1R) interacted as shown through pharmacological and genetic inhibition and were regulated by the mutant ER as demonstrated by reduced phospho-protein expression with endocrine therapies (ET). We show that ET in combination with a RON inhibitor (RONi) decreased ex vivo organoid growth of ESR1 mutant models, and as a monotherapy in PalbR models, demonstrating its therapeutic efficacy. Significantly, ET in combination with the RONi reduced metastasis of an ESR1 Y537S mutant PDX model. CONCLUSIONS: Our results demonstrate that RON/PI3K pathway inhibition may be an effective treatment strategy in ESR1 mutant and PalbR MBC patients. Clinically our data predict that ET resistance mechanisms can also contribute to CDK4/6 inhibitor resistance.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Female , Humans , Mice , Mutation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Xenograft Model Antitumor Assays
4.
World Allergy Organ J ; 13(2): 100100, 2020 02.
Article in English | MEDLINE | ID: mdl-32099590

ABSTRACT

Background: Although oral food challenge (OFC) is an important clinical procedure for diagnosing food allergy, there is a paucity of literature on the outcome of the procedure and specifically the patients on whom the procedure is performed from the aspects of their age, sex, race/ethnicity, health insurance status, and serum specific IgE to the food tested. Objective: We aimed to review results of OFC and determine the impact of patient age, sex, race/ethnicity, insurance status, private or public, and food specific serum IgE on the outcome of OFC. Methods: A retrospective chart review was performed of patients undergoing OFCs at a children's hospital outpatient allergy clinic over a two-year period. The outcome of OFC was allergic or non-allergic based on determination and documentation by the treating physician. A logistic regression model was built to determine the association between the OFC outcomes, age, and symptoms at the time of OFC. A Chi-square analysis was performed to check for any significant relationship between the OFC outcome and age when stratified by insurance status. Results: Five hundred and eight children underwent 641 OFCs. Twenty nine percent of OFCs had an allergic outcome with the most commonly challenged foods being peanuts, eggs, and milk. Patient age and gender, when stratified by insurance status, did not have a significant effect on OFC outcomes. Serum IgE to peanuts and egg was significantly different between allergic OFC and non-allergic outcome. Vomiting and urticaria/angioedema correlated with an allergic OFC outcome. Conclusion: OFCs confirm the food allergy diagnosis in about one-third of patients tested, and they should continue to be used when possible for an accurate diagnosis. Age, sex, and insurance status do not have a significant association with the outcome of OFC and cannot be added as predictive factors.

6.
Breast Cancer Res Treat ; 157(2): 253-265, 2016 06.
Article in English | MEDLINE | ID: mdl-27178332

ABSTRACT

The purpose of this study was to address the role of ESR1 hormone-binding mutations in breast cancer. Soft agar anchorage-independent growth assay, Western blot, ERE reporter transactivation assay, proximity ligation assay (PLA), coimmunoprecipitation assay, silencing assay, digital droplet PCR (ddPCR), Kaplan-Meier analysis, and statistical analysis. It is now generally accepted that estrogen receptor (ESR1) mutations occur frequently in metastatic breast cancers; however, we do not yet know how to best treat these patients. We have modeled the three most frequent hormone-binding ESR1 (HBD-ESR1) mutations (Y537N, Y537S, and D538G) using stable lentiviral transduction in human breast cancer cell lines. Effects on growth were examined in response to hormonal and targeted agents, and mutation-specific changes were studied using microarray and Western blot analysis. We determined that the HBD-ESR1 mutations alter anti-proliferative effects to tamoxifen (Tam), due to cell-intrinsic changes in activation of the insulin-like growth factor receptor (IGF1R) signaling pathway and levels of PIK3R1/PIK3R3. The selective estrogen receptor degrader, fulvestrant, significantly reduced the anchorage-independent growth of ESR1 mutant-expressing cells, while combination treatments with the mTOR inhibitor everolimus, or an inhibitor blocking IGF1R, and the insulin receptor significantly enhanced anti-proliferative responses. Using digital drop (dd) PCR, we identified mutations at high frequencies ranging from 12 % for Y537N, 5 % for Y537S, and 2 % for D538G in archived primary breast tumors from women treated with adjuvant mono-tamoxifen therapy. The HBD-ESR1 mutations were not associated with recurrence-free or overall survival in response in this patient cohort and suggest that knowledge of other cell-intrinsic factors in combination with ESR1 mutation status will be needed determine anti-proliferative responses to Tam.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Mutation , Receptors, Somatomedin/genetics , Tamoxifen/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Models, Genetic , Receptor, IGF Type 1 , Receptors, Estrogen/metabolism , Receptors, Somatomedin/metabolism , Signal Transduction , Tamoxifen/therapeutic use
8.
Breast Cancer Res Treat ; 154(2): 225-37, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26487496

ABSTRACT

Tamoxifen (Tam) resistance represents a significant clinical problem in estrogen receptor (ER) α-positive breast cancer. We previously showed that decreased expression of Rho guanine nucleotide dissociation inhibitor (Rho GDI) α, a negative regulator of the Rho GTPase pathway, is associated with Tam resistance. We now discover that androgen receptor (AR) is overexpressed in cells with decreased Rho GDIα and seek to determine AR's contribution to resistance. We engineered ERα-positive cell lines with stable knockdown (KD) of Rho GDIα (KD cells). Resistance mechanisms were examined using microarray profiling, protein-interaction studies, growth and reporter gene assays, and Western blot analysis combined with a specific AR antagonist and other signaling inhibitors. Tam-resistant tumors and cell lines with low Rho GDIα levels exhibited upregulated AR expression. Microarray of Rho GDIα KD cells indicated that activation of EGFR and ERα was associated with Tam treatment. When AR levels were elevated, interaction between AR and EGFR was detected. Constitutive and Tam-induced phosphorylation of EGFR and ERK1/2 was blocked by the AR antagonist Enzalutamide, suggesting that AR-mediated EGFR activation was a mechanism of resistance in these cells. Constitutive ERα phosphorylation and transcriptional activity was inhibited by Enzalutamide and the EGFR inhibitor gefitinib, demonstrating that AR-mediated EGFR signaling activated ERα. Tam exhibited agonist activity in AR overexpressing cells, stimulating ERα transcriptional activity and proliferation, which was blocked by Enzalutamide and gefitinib. We describe a novel model of AR-mediated Tam resistance through activation of EGFR signaling leading to ER activation in ERα-positive cells with low expression of Rho GDIα.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , ErbB Receptors/genetics , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Receptors, Androgen/metabolism , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Female , Gene Expression , Gene Expression Profiling , Humans , MAP Kinase Signaling System/drug effects , Protein Binding , Receptors, Androgen/genetics , Tamoxifen/therapeutic use , Transcriptional Activation , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism
9.
Breast Cancer Res Treat ; 150(3): 535-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25820519

ABSTRACT

The purpose of this study was to discover novel nuclear receptor targets in triple-negative breast cancer. Expression microarray, Western blot, qRT-PCR analyses, MTT growth assay, soft agar anchorage-independent growth assay, TRE reporter transactivation assay, and statistical analysis were performed in this study. We performed microarray analysis using 227 triple-negative breast tumors, and clustered the tumors into five groups according to their nuclear receptor expression. Thyroid hormone receptor beta (TRß) was one of the most differentially expressed nuclear receptors in group 5 compared to other groups. TRß low expressing patients were associated with poor outcome. We evaluated the role of TRß in triple-negative breast cancer cell lines representing group 5 tumors. Knockdown of TRß increased soft agar colony and reduced sensitivity to docetaxel and doxorubicin treatment. Docetaxel or doxorubicin long-term cultured cell lines also expressed decreased TRß protein. Microarray analysis revealed cAMP/PKA signaling was the only KEGG pathways upregulated in TRß knockdown cells. Inhibitors of cAMP or PKA, in combination with doxorubicin further enhanced cell apoptosis and restored sensitivity to chemotherapy. TRß-specific agonists enhanced TRß expression, and further sensitized cells to both docetaxel and doxorubicin. Sensitization was mediated by increased apoptosis with elevated cleaved PARP and caspase 3. TRß represents a novel nuclear receptor target in triple-negative breast cancer; low TRß levels were associated with enhanced resistance to both docetaxel and doxorubicin treatment. TRß-specific agonists enhance chemosensitivity to these two agents. Mechanistically enhanced cAMP/PKA signaling was associated with TRß's effects on response to chemotherapy.


Subject(s)
Drug Resistance, Neoplasm , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Docetaxel , Doxorubicin/pharmacology , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , Prognosis , Signal Transduction/drug effects , Taxoids/pharmacology , Triple Negative Breast Neoplasms/drug therapy
10.
Clin Cancer Res ; 17(20): 6510-21, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21878538

ABSTRACT

PURPOSE: Tamoxifen (Tam) is the most prescribed hormonal agent for treatment of estrogen receptor α (ERα)-positive breast cancer patients. Using microarray analysis, we observed that metastatic breast tumors resistant to Tam therapy had elevated levels of Dicer. EXPERIMENTAL DESIGN: We overexpressed Dicer in ERα-positive MCF-7 human breast cancer cells and observed a concomitant increase in expression of the breast cancer resistance protein (BCRP). We thus hypothesized that Tam resistance associated with Dicer overexpression in ERα-positive breast cancer cells may involve BCRP. We analyzed BCRP function in Dicer-overexpressing cells using growth in soft agar and mammosphere formation and evaluated intracellular Tam efflux. RESULTS: In the presence of Tam, Dicer-overexpressing cells formed resistant colonies in soft agar, and treatment with BCRP inhibitors restored Tam sensitivity. Tumor xenograft studies confirmed that Dicer-overexpressing cells were resistant to Tam in vivo. Tumors and distant metastases could be initiated with as few as five mammosphere cells from both vector and Dicer-overexpressing cells, indicating that the mammosphere assay selected for cells with enhanced tumor-initiating and metastatic capacity. Dicer-overexpressing cells with elevated levels of BCRP effluxed Tam more efficiently than control cells, and BCRP inhibitors were able to inhibit efflux. CONCLUSION: Dicer-overexpressing breast cancer cells enriched for cells with enhanced BCRP function. We hypothesize that it is this population which may be involved in the emergence of Tam-resistant growth. BCRP may be a novel clinical target to restore Tam sensitivity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , DEAD-box RNA Helicases/pharmacology , Drug Resistance, Neoplasm/genetics , Estrogen Antagonists/therapeutic use , Neoplasm Proteins/metabolism , Ribonuclease III/pharmacology , Tamoxifen/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Estrogen Receptor alpha/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/genetics , Tamoxifen/pharmacology , Up-Regulation
11.
Breast Cancer Res Treat ; 129(3): 819-27, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21792626

ABSTRACT

Endocrine therapy in patients with breast cancer can be limited by the problem of resistance. Preclinical studies suggest that complete blockade of the estrogen receptor (ER) combined with inhibition of the epidermal growth factor receptor can overcome endocrine resistance. We tested this hypothesis in a phase II neoadjuvant trial of anastrozole and fulvestrant combined with gefitinib in postmenopausal women with newly diagnosed ER-positive breast cancer. After a baseline tumor core biopsy, patients were randomized to receive anastrozole and fulvestrant or anastrozole, fulvestrant, and gefitinib (AFG) for 3 weeks. After a second biopsy at 3 weeks, all patients received AFG for 4 months and surgery was done if the tumor was operable. The primary endpoint was best clinical response by RECIST criteria and secondary endpoints were toxicity and change in biomarkers. The study closed after 15 patients were enrolled because of slow accrual. Median patient age was 67 years and median clinical tumor size was 7 cm. Four patients had metastatic disease present. Three patients withdrew before response was assessed. In the remaining 12 patients, there were two complete clinical responses (17%), three partial responses (25%), five had stable disease (41%), and two (17%) had progressive disease. Most common adverse events were rash in four patients, diarrhea in four, joint symptoms in three, and abnormal liver function tests in three. There were no grade 4 toxicities and all toxicities were reversible. At 3 weeks, cell proliferation as measured by Ki-67 was significantly reduced in the AFG group (P value = 0.01), with a parallel reduction in the expression of the Cyclin D1 (P value = 0.02). RNA microarray data showed a corresponding decrease in the expression of cell cycle genes. These results suggest that AFG was an effective neoadjuvant therapy and consistently reduced proliferation in ER-positive tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Anastrozole , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/therapy , Breast Neoplasms, Male/drug therapy , Cell Cycle/genetics , Cyclin D1/metabolism , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Female , Fulvestrant , Gefitinib , Humans , Ki-67 Antigen/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Neoadjuvant Therapy , Nitriles/administration & dosage , Oncogene Protein v-akt/metabolism , Proto-Oncogene Proteins/metabolism , Quinazolines/administration & dosage , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Triazoles/administration & dosage
12.
J Natl Cancer Inst ; 103(7): 538-52, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21447808

ABSTRACT

BACKGROUND: Estrogen receptor (ER) α is a successful therapeutic target in breast cancer, but patients eventually develop resistance to antiestrogens such as tamoxifen. METHODS: To identify genes whose expression was associated with the development of tamoxifen resistance and metastasis, we used microarrays to compare gene expression in four primary tumors from tamoxifen-treated patients whose breast cancers did not recur vs five metastatic tumors from patients whose cancers progressed during adjuvant tamoxifen treatment. Because Rho guanine dissociation inhibitor (GDI) α was underexpressed in the tamoxifen-resistant group, we stably transfected ERα-positive MCF-7 breast cancer cells with a plasmid encoding a short hairpin (sh) RNA to silence Rho GDIα expression. We used immunoblots and transcription assays to examine the role of Rho GDIα in ER-related signaling and growth of cells in vitro and as xenografts in treated nude mice (n = 8-9 per group) to examine the effects of Rho GDIα blockade on hormone responsiveness and metastatic behavior. The time to tumor tripling as the time in weeks from randomization to a threefold increase in total tumor volume over baseline was examined in treated mice. The associations of Rho GDIα and MTA2 levels with tamoxifen resistance were examined in microarray data from patients. All statistical tests were two-sided. RESULTS: Rho GDIα was expressed at lower levels in ERα-positive tumors that recurred during tamoxifen treatment than in ERα-positive tamoxifen-sensitive primary tumors. MCF-7 breast cancer cells in which Rho GDIα expression had been silenced were tamoxifen-resistant, had increased Rho GTPase and p21-activated kinase 1 activity, increased phosphorylation of ERα at serine 305, and enhanced tamoxifen-induced ERα transcriptional activity compared with control cells. MCF-7 cells in which Rho GDIα expression was silenced metastasized with high frequency when grown as tumor xenografts. When mice were treated with estrogen or estrogen withdrawal, tripling times for xenografts from cells with Rho GDIα silencing were similar to those from vector-containing control cells; however, tripling times were statistically significantly faster than control when mice were treated with tamoxifen (median tripling time for tumors with Rho GDIα small interfering RNA = 2.34 weeks; for control tumors = not reached, hazard ratio = 4.13, 95% confidence interval = 1.07 to 15.96, P = .040 [adjusted for multiple comparisons, P = .119]). Levels of the metastasis-associated protein MTA2 were also increased upon Rho GDIα silencing, and combined Rho GDIα and MTA2 levels were associated with recurrence in 250 tamoxifen-treated patients. CONCLUSION: Loss of Rho GDIα enhances metastasis and resistance to tamoxifen via effects on both ERα and MTA2 in models of ERα-positive breast cancer and in tumors of tamoxifen-treated patients.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Drug Resistance, Neoplasm , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Histone Deacetylases/metabolism , Neoplasm Recurrence, Local/prevention & control , Repressor Proteins/metabolism , Signal Transduction , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation , Estrogen Antagonists/therapeutic use , Estrogen Receptor alpha/drug effects , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Genome-Wide Association Study , Guanine Nucleotide Dissociation Inhibitors/genetics , Histone Deacetylases/genetics , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Nude , Neoplasm Recurrence, Local/metabolism , Odds Ratio , Phenotype , Plasmids , Protein Array Analysis , RNA, Small Interfering/metabolism , Random Allocation , Repressor Proteins/genetics , Retrospective Studies , Secondary Prevention/methods , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Tamoxifen/therapeutic use , Time Factors , Transcriptional Activation , Transplantation, Heterologous , Tumor Stem Cell Assay , rho GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors
13.
Breast Cancer Res Treat ; 121(1): 1-11, 2010 May.
Article in English | MEDLINE | ID: mdl-19533338

ABSTRACT

Although the androgen receptor (AR) is a known clinical target in prostate cancer, little is known about its possible role in breast cancer. We have investigated the role of AR expression in human breast cancer in response to treatment with the antiestrogen tamoxifen. Resistance to tamoxifen is a major problem in treating women with breast cancer. By gene expression profiling, we found elevated AR and reduced estrogen receptor (ER) alpha mRNA in tamoxifen-resistant tumors. Exogenous overexpression of AR rendered ERalpha-positive MCF-7 breast cancer cells resistant to the growth-inhibitory effects of tamoxifen in anchorage-independent growth assays and in xenograft studies in athymic nude mice. AR-overexpressing cells remained sensitive to growth stimulation with dihydrotestosterone. Treatment with the AR antagonist Casodex (bicalutamide) reversed this resistance, demonstrating the involvement of AR signaling in tamoxifen resistance. In AR-overexpressing cells, tamoxifen induced transcriptional activation by ERalpha that could be blocked by Casodex, suggesting that AR overexpression enhances tamoxifen's agonistic properties. Our data suggest a role for AR overexpression as a novel mechanism of hormone resistance, so that AR may offer a new clinical therapeutic target in human breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Receptors, Androgen/biosynthesis , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Western , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
14.
Cancer Res ; 69(11): 4724-32, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487288

ABSTRACT

Aromatase inhibitors (AI) are rapidly becoming the first choice for hormonal treatment of estrogen receptor-alpha (ERalpha)-positive breast cancer in postmenopausal women. However, de novo and acquired resistance frequently occurs. We have previously identified a lysine to arginine transition at residue 303 (K303R) in ERalpha in premalignant breast lesions and invasive breast cancers, which confers estrogen hypersensitivity and resistance to tamoxifen treatment. Thus, we questioned whether resistance to AIs could arise in breast cancer cells expressing the ERalpha mutation. As preclinical models to directly test this possibility, we generated K303R-overexpressing MCF-7 cells stably transfected with an aromatase expression vector. Cells were stimulated with the aromatase substrate, androstenedione, with or without the AI anastrozole (Ana). We found that Ana decreased androstenedione-stimulated growth of wild-type cells, whereas K303R-expressing cells were resistant to the inhibitory effect of Ana on growth. We propose that a mechanism of resistance involves an increased binding between the mutant receptor and the p85alpha regulatory subunit of phosphatidylinositol-3-OH kinase (PI3K), leading to increased PI3K activity and activation of protein kinase B/Akt survival pathways. Inhibition of the selective "addiction" to the PI3K/Akt pathway reversed AI resistance associated with expression of the mutant receptor. Our findings suggest that the K303R ERalpha mutation might be a new predictive marker of response to AIs in mutation-positive breast tumors, and that targeting the PI3K/Akt pathway may be a useful strategy for treating patients with tumors resistant to hormone therapy.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/genetics , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Amino Acid Substitution/genetics , Animals , Arginine/genetics , Aromatase Inhibitors/pharmacology , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , CHO Cells , Cricetinae , Cricetulus , Female , Humans , Lysine/genetics , Mice , Mice, Nude , Mutation, Missense/physiology , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Clin Cancer Res ; 13(11): 3235-43, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545528

ABSTRACT

PURPOSE: Estrogen receptor alpha (ERalpha) predicts the natural history of breast cancer without intervening therapy. Here, we have optimized the detection of a somatic mutation, an A908G transition of ERalpha, and examined its association with clinical and biological features of invasive breast cancer. EXPERIMENTAL DESIGN: We compared two methods of sequencing to detect the A908G ERalpha mutation. We then used primer extension sequencing with genomic DNA isolated from invasive breast tumors to determine whether the mutation was associated with clinical outcome in 267 axillary node-negative and axillary node-positive breast tumors. The presence of the mutation and clinical variables were analyzed for association with recurrence-free survival and overall survival by Cox proportional hazards regression models. RESULTS: We determined that dye-labeled terminator sequencing was not adequate for detection of the A908G ERalpha mutation. The mutation was detected at a high frequency (50%) in invasive breast tumors using primer extension sequencing, and was found to be associated with clinical measures of poor outcome, including larger tumor size and axillary lymph node positivity. Although the mutation was associated with recurrence-free survival in univariate analysis, it was not an independent predictor of outcomes in multivariate analysis. CONCLUSIONS: Consistent with our previous finding of this somatic ERalpha mutation in breast ductal hyperplasias, we now present evidence that the A908G mutation is present in invasive breast tumors using an optimized sequencing method. We find that the mutation is significantly associated with aggressive biological tumor features, and with an unfavorable prognosis, but was not an independent prognostic marker in untreated patients.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Gene Expression Regulation, Neoplastic , Mutation , Adult , DNA Primers/chemistry , DNA, Neoplasm/chemistry , Humans , Lymphatic Metastasis , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Prognosis , Proportional Hazards Models , Treatment Outcome
16.
Mol Cancer Ther ; 5(12): 3023-31, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172405

ABSTRACT

It has long been appreciated that estrogenic signaling contributes to breast cancer progression. c-Src is also required for a number of processes involved in tumor progression and metastasis. We have previously identified the K303R mutant estrogen receptor alpha (ERalpha) that confers hypersensitivity to low levels of estrogen. Because ERalpha and c-Src have been shown to interact in a number of different systems, we wanted to evaluate the role of c-Src kinase in estrogen-stimulated growth and survival of ERalpha-positive breast cancer cells. MCF-7 cells stably expressing the mutant receptor showed increased c-Src kinase activity and c-Src tyrosine phosphorylation when compared with wild-type ERalpha-expressing cells. A c-Src inhibitor, AZD0530, was used to analyze the biological effects of pharmacologically inhibiting c-Src kinase activity. MCF-7 cells showed an anchorage-dependent growth IC50 of 0.47 micromol/L, which was increased 4-fold in the presence of estrogen. In contrast, cells stably expressing the mutant ERalpha had an elevated IC50 that was only increased 1.4-fold by estrogen stimulation. The c-Src inhibitor effectively inhibited the anchorage-independent growth of both of these cells, and estrogen was able to reverse these effects. When cells were treated with suboptimal concentrations of c-Src inhibitor and tamoxifen, synergistic inhibition was observed, suggesting a cooperative interaction between c-Src and ERalpha. These data clearly show an important role for ERalpha and estrogen signaling in c-Src-mediated breast cancer cell growth and survival. Here, we show that c-Src inhibition is blocked by estrogen signaling; thus, the therapeutic use of c-Src inhibitors may require inhibition of ERalpha in estrogen-dependent breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzodioxoles/pharmacology , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/biosynthesis , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Tamoxifen/pharmacology , Benzodioxoles/administration & dosage , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Humans , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/metabolism , Quinazolines/administration & dosage , Tamoxifen/administration & dosage , src-Family Kinases
17.
J Androl ; 24(2): 192-200, 2003.
Article in English | MEDLINE | ID: mdl-12634305

ABSTRACT

Members of the transforming growth factor beta type (TGFbeta) superfamily and their receptors are expressed in the testis, and are believed to play important paracrine and autocrine roles during testicular development and spermatogenesis. The Smad proteins are downstream mediators for the family of TGFbeta growth factors. Smad2 and Smad3 are associated with both TGFbeta and activin signaling. However, very little is known about the expression and regulation of the Smad signaling proteins in the testis. In the present study, we have determined that Smad2 and Smad3 proteins are expressed in the postnatal testes of rats from 5 days to 60 days of age. Expression levels for both proteins are higher in young rats than in sexually mature rats. Smad2 and Smad3 messenger RNA levels parallel protein expression. Smad2 and Smad3 proteins are mainly localized in the cytoplasm of meiotic germ cells, Sertoli cells, and Leydig cells. Smad3 protein is localized to the nucleus of preleptotene to zygotene primary spermatocytes in young rats. Both proteins are expressed throughout all stages of the cycle of seminiferous tubules but are expressed at their lowest levels at stages VII-VIII in the seminiferous epithelium of adult rats. The presence of these downstream mediators in these cell types supports a role for TGFbeta and activin during spermatogenesis. The difference between the expression of Smad2 and Smad3 suggests that they may have different functions within the testis.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Testis/growth & development , Testis/physiology , Trans-Activators/genetics , Age Factors , Animals , Male , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/growth & development , Seminiferous Epithelium/physiology , Smad2 Protein , Smad3 Protein , Spermatocytes/physiology , Testis/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...