<<

Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

Clinical Cancer Therapy: Preclinical Research

Glucocorticoid Receptor Antagonism as a Novel Therapy for Triple-Negative Breast Cancer

Maxwell N. Skor1, Erin L. Wonder1, Masha Kocherginsky2, Anju Goyal1, Ben A. Hall1, Yi Cai3, and Suzanne D. Conzen1,4

Abstract Purpose: Triple-negative breast cancer (TNBC) accounts for 10% to 20% of newly diagnosed invasive breast cancer. Finding effective targets for -resistant TNBC has proven difficult in part because of TNBC’s molecular heterogeneity. We have previously reported that likely because of the antiapoptotic activity of receptor (GR) in receptor (ER)-negative breast epithelial and cancer cells, high GR expression/activity in early-stage TNBC significantly correlates with chemotherapy resistance and increased recurrence. We hypothesized that pretreatment with , a GR antagonist, would potentiate the efficacy of chemotherapy in GRþ TNBCs by inhibiting the antiapoptotic signaling pathways of GR and increasing the cytotoxic efficiency of chemotherapy. Experimental Design: TNBC cell apoptosis was examined in the context of physiologic glucocorticoid concentrations, chemotherapy, and/or pharmacologic concentrations of mifepristone. We used high- throughput live microscopy with continuous recording to measure apoptotic cells stained with a fluorescent dye and Western blot analysis to detect caspase-3 and PARP cleavage. The effect of mifepristone on GR- mediated gene expression was also measured. TNBC xenograft studies were performed in female severe combined immunodeficient (SCID) mice and tumors were measured following treatment with vehicle, , or mifepristone/paclitaxel. Results: We found that although mifepristone treatment alone had no significant effect on TNBC cell viability or clonogenicity in the absence of chemotherapy, the addition of mifepristone to dexamethasone/ paclitaxel treatment significantly increased cytotoxicity and caspase-3/PARP cleavage. Mifepristone also antagonized GR-induced SGK1 and MKP1/DUSP1 gene expression while significantly augmenting pacli- taxel-induced GRþ MDA-MB-231 xenograft tumor shrinkage in vivo. Conclusions: These results suggest that mifepristone pretreatment could be a useful strategy for increasing tumor cell apoptosis in chemotherapy-resistant GRþ TNBC. Clin Cancer Res; 19(22); 6163–72. 2013 AACR.

Introduction gene expression, thereby changing the complement of pro- (GCs) are secreted from the adrenal teins regulating key signaling pathways (4, 5). In its ligand- gland in response to exposure to emotional and physiologic bound state, GR initiates or represses gene expression in a stressors and are responsible for modulating essential met- cell-type–specific manner (1). For example, GR activation abolic, cardiovascular, immune, and behavioral functions can induce apoptosis in lymphocytes (6, 7), whereas its (1–3). The (GR) belongs to a family activation results in inhibition of apoptosis in breast epi- of nuclear hormone receptors that are ligand-dependent thelial cells (8). Furthermore, in a TNBC xenograft model, transcription factors involved in activating and repressing the activation of tumor GR by dexamethasone (dex), a synthetic GC, diminished chemotherapy effectiveness in vivo (9). Until now, the use of a GR antagonist in an in vivo model of GRþ triple-negative breast cancer (TNBC) has not Authors' Affiliations: Departments of 1Medicine, 2Health Studies, and 3Surgery, 4Ben May Department for Cancer Research, The University of been reported. Chicago, Chicago, Illinois It was previously shown by our group and others that Note: Supplementary data for this article are available at Clinical Cancer GR activation initiates potent antiapoptotic signaling Research Online (http://clincancerres.aacrjournals.org/). pathways in breast epithelial cells, at least in part, via Corresponding Author: Suzanne D. Conzen, Department of , The transcriptional regulation of genes encoding cell survival University of Chicago, 900 East 59th Street, Chicago, IL 60637. Phone: 773- pathway proteins (5, 10–12). For example, genes encod- 834-2604; Fax: 773-702-9268; E-mail: [email protected] ing the antiapoptotic proteins serum and glucocorticoid- doi: 10.1158/1078-0432.CCR-12-3826 inducible protein kinase-1 (SGK1) and mitogen-activated 2013 American Association for Cancer Research. protein kinase phosphatase-1 (MKP1/DUSP1)areboth

www.aacrjournals.org 6163

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

Skor et al.

atmosphere with 5% CO . Before treatment with glucocor- Translational Relevance 2 ticoids, mifepristone, and/or chemotherapy, cells were Triple-negative breast cancer (TNBC) lacks effective grown in DMEM or RPMI-1640 supplemented with 2.5% targeted therapies. Approximately 25% of invasive charcoal-stripped FCS and 1% penicillin-streptomycin. TNBCs are glucocorticoid receptor (GR)-positive [>10% of tumor cells strongly GR-positive by immunohis- Cell death assay tochemistry (IHC) and/or significantly increased tumor TNBC cell lines (MDA-MB-231 at 2 103 cells/well, MDA- NR3C1 (GR) mRNA levels compared with median TNBC MB-468 at 5 103 cells/well, and BT-20 at 3.5 103 cells/ NR3C1 expression]. High tumor GR expression signifi- well) were plated in 96-well plates and allowed to adhere cantly correlates with earlier relapse in early-stage TNBC. overnight in DMEM or RPMI supplemented with 10% FCS. Mifepristone is a potent GR and receptor Media were then changed to 2.5% charcoal-stripped serum (PR) modulator. Here, we report that in GRþ TNBC (CS-FCS) for 48 hours. Cells were treated with vehicle (EtOH (which lacks expression of the PR), pretreatment with 0.1% v/v), dexamethasone (100 nmol/L) or mifepristone mifepristone potentiates paclitaxel-induced cytotoxicity, (100 nmol/L) alone or dexamethasone/mifepristone (100 presumably by antagonizing GR-activated TNBC cell nmol/L) 1 hour before paclitaxel (100 nmol/L) treatment for survival pathways that otherwise contribute to chemo- 72 hours. A cyanine dimer nucleic acid dye, YOYO-1 (Life therapy resistance. Technologies, Y3601) that causes green fluorescence if the cellular membrane is compromised was used to detect dead cells. Two images (1.90 1.52 mm) in separate regions of each well were captured with a 10 objective at upregulated following GR activation (13–15). Moreover, 4-hour intervals using the IncuCyte FLR HD real-time in short-hairpin RNAs (shRNA) expression targeting either vitro micro-imaging system (Essen Instruments). Cell SGK1 MKP1/DUSP1 or demonstrated a requirement for death (detected as YOYO-1-positive) and total cell counts the induction of these proteins to induce GR-mediated (phase contrast) were measured computationally by Ima- cell survival (13). Furthermore, mifepristone, a dual GR geJ Version 1.46r (16) using investigator-coded software and PR modulator, significantly antagonizes the induc- for analysis (Supplementary Method S1). The "cytotoxic SGK1 MKP1/DUSP1 tion of and expression in ER-negative index" represents the number of dead cells/total cells for breast cell lines treated with glucocorticoids (13). each image. In this study, we tested the hypothesis that GR modulation Images collected between 12 and 72 hours were used in (using mifepristone) could potentiate chemotherapy-in- the analysis. The cytotoxic index was log-transformed to duced cytotoxicity in TNBC models where GR (but neither satisfy the normality assumption. Data were analyzed using ER or PR) is expressed. Our data suggest that mifepristone repeated measures analysis of variance models. A separate blocks GR-mediated tumor cell survival by antagonizing model was fitted for each cell line. The fixed effects included associated gene expression and inhibiting apoptotic path- were treatment, time, time2, time3, and all corresponding ways that are usually activated by endogenous glucocorti- interactions between treatment and time terms. Random coids, thereby augmenting chemotherapy-induced cell death effects included random intercept terms for biologic and in vivo and decreasing TNBC tumor growth. technical replicates and a random slope for the biologic replicate. Correlation between serial measurements was Experimental Procedures modeled using AR (1) covariance structure. A generalized Materials F-test was used to test the composite hypothesis of no Paclitaxel (Sigma Cat. No.T7402) and dexamethasone difference between treatment, trt time, trt time2, and (Sigma Cat. No. D4902) were purchased from Sigma- trt time3, effectively comparing the entire curves over Aldrich. Initially, mifepristone was purchased from Enzo time. Analyses were performed in SAS 9.2. Life Sciences (Cat. No. BML-S510-0025) and later experi- ments were repeated with pharmaceutical-grade mifepris- Clonogenic assay tone provided by Corcept Therapeutics. For in vivo experi- MDA-MB-231 cells (10,000 cells per 10 cm dish) were ments, pharmaceutical-grade paclitaxel liquid suspension allowed to adhere overnight in DMEM supplemented with was purchased from Bedford Laboratories. 10% FCS. Media was then changed to 2.5% CS-FCS for 48 hours. Cells were treated with vehicle (EtOH 0.1% V/V), Cell culture dexamethasone (100 nmol/L) or mifepristone (100 nmol/L) MDA-MB-231, BT-20, and MDA-MB-468 cell lines were alone or dex/mif (100 nmol/L) 1 hour before paclitaxel (100 purchased from American Type Culture Collection. MDA- nmol/L) treatment for 4 days. The colonies were fixed with MB-231 and BT-20 cells were cultured in Dulbecco’s mod- 3.7% paraformaldehyde and stained with 0.05% crystal ified Eagle Medium (DMEM; Lonza) and MDA-MB-468 violet in PBS. Five random 1 cm diameter circles were then cells in RPMI-1640 (Thermo Fisher Scientific), both sup- drawn with a black marker, and the colonies within these 5 plemented with 10% fetal calf serum (FCS; Gemini Bio- regions were counted under a stereoscope at a magnification Products) and (1% penicillin-streptomycin, of 13. A minimum of 5 cells per colony were required to Lonza). All cell lines were cultured at 37C in a humidified meet the criterion for counting as a "colony."

6164 Clin Cancer Res; 19(22) November 15, 2013 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

GR Antagonism in TNBC

A mixed-effects analysis of variance model was fitted, with containing phosphatase and protease inhibitor cocktails colony count as the responsible variable, treatment group as (Roche). Protein concentrations were measured using the the fixed effect, and biologic replicate and technical replicate BCA Assay Kit (Thermo Scientific) and 2 Laemmli buffer nested within the biologic replicates as the random effects. supplemented with 5% 2-mercaptoethanol was added to an Dexamethasone/paclitaxel versus dexamethasone/mifep- equivalent volume of protein lysate. Total proteins (60 mg ristone/paclitaxel groups were compared on the basis of per lane) were resolved by SDS-PAGE and then transferred this fitted model, and the reported P values are unadjusted to polyvinylidene difluoride membranes (Bio-Rad). After for multiple comparisons. the membranes were washed 3 times, they were incubated with 5% bovine serum albumin (BSA; Fisher Scientific) in Quantitative real-time PCR 0.1% Tween20 in TBS (TBS/T) for 1 hour at room temper- MDA-MB-231, MDA-MB-468, and BT-20 cells were seed- ature, followed by overnight incubation at 4C with pri- ed at approximately 50% confluence and allowed to adhere mary antibody against human GR-alpha (GR-XP Cell Sig- overnight in DMEM with 10% FCS, then cultured in 2.5% naling), caspase-3 (Cell Signaling Technology), PARP (Cell CS-FCS for an additional 48 hours. Media were removed Signaling Technology), or human b-Actin (Sigma-Aldrich). and equal volumes of either vehicle (ethanol), dexameth- After additional washing, membranes were incubated for 1 asone (100 nmol/L) or dexamethasone/mifepristone (100 hour at room temperature with either Alexa Fluor 680 goat nmol/L FC) diluted in DMEM or RPMI supplemented with anti-rabbit (Invitrogen) or 800 goat anti-mouse (LI-COR) 2.5% CS-FCS was then added. After 4 hours of treatment, secondary antibody, rinsed and scanned using the Odyssey 100 mL of RNA-Solv Reagent (EaZy Nucleic Acid Isolation infrared imaging system (LI-COR) at a wavelength of 700 or Kit) supplemented with 2% 2-mercaptoethanol was added 800 nm. to each well to harvest RNA. Total RNA was extracted using the Qiagen All-Prep DNA/RNA Mini Kit. cDNA was then Mammary fat pad xenograft studies in female SCID reverse transcribed from 0.5 mg of total RNA with Quanta mice reverse transcription reagents (Quanta Biosciences) using All experiments were carried out in accordance with the the GeneAmp PCR System 9700 (Applied BioSystems) U.S. Public Health Service Policy on Humane Care and Use per manufacturer’s instruction. The cDNA was diluted in of Laboratory Animals and approved by the University of PerfeCTa SYBR Green FastMix (Quanta Biosciences), and Chicago Institutional Animals Care and Use Committee. quantitative real-time PCR (qRT-PCR) was carried out in Suspensions of MDA-MB-231 cells (1 107)in50mL of PBS a BioRad PCR System MyIQ (BioRad Life Sciences). were injected subcutaneously into the right pectoral mam- The following primers were used: SGK1,50-AGGCCCA- mary fat pad of a 5- to 6-week-old female SCID mouse TCCTTCTCTGTTT-30 (forward) and 50-TTCACTGCTCCC- (Taconic). Tumors were allowed to reach 200 mm3 and CTCAGTCT-30 (reverse); MKP1/DUSP1,50-CCTGACAG- then mice were treated via intraperitoneal (i.p.) injection CGCGGAATCT-30 (forward) and 50-GATTTCCACCGGGC- with paclitaxel (10 mg/kg) suspended in castor oil (1:10 CAC-30 (reverse); NR3C1/GR,50-TCTGAACTTCCCTGGTC- v/v); vehicle-treated animals received two injections and GAA-30 (forward) and 50- GTGGTCCTGTTGTTGCTGTT-30 paclitaxel-treated animals received an additional injection (reverse); Actin-B 50-CAGCGGAACCGCTCATTGCCAATGG- of mifepristone (15 mg/kg/d) or vehicle (ethanol and 30 (forward) and 50-TCACCCCCTGTGCCCATCTACGA-30 sesame seed oil 1:10 v/v) 1 hour before the paclitaxel (reverse). The samples were loaded in duplicate. Relative (17). The longest (L) and shortest (S) diameters of the quantification of gene expression was calculated according tumors were measured 3 times a week with electronic to the standard curve method, as described by Applied calipers, and tumor volume was calculated using the for- Biosystems User Bulletin 2, October 2001, based on the mula for an ellipsoid sphere: S2 L (0.52; ref. 9). Tumors DDCt approach (15). A ratio of GR target gene expression to were dissected and cut lengthwise into mirror image sec- Actin-B expression was calculated. tions. One section was minced in lysis buffer and frozen (for For SGK1 and MKP1/DUSP1 qRT-PCR analysis, a mixed- protein analysis), and the other was fixed in 10% neutral- effects ANOVA model was fitted with Ct as the response buffered formalin [for IHC and immunofluorescence (IF)]. variable: treatment, gene type (target or reference), and Tumor growth data were analyzed using repeated measures treatment gene interaction as the fixed effects and repli- analysis of variance models as previously described (9). cate as the random effect. A linear contrast was then con- structed to estimate DDCt and its confidence interval, and Histopathological examination the results were exponentiated to obtain the estimate of For anti-GR IF analysis of tumor xenografts, samples were DDC 2 t and its confidence intervals; 68% confidence inter- formalin-fixed for 24 hours and embedded in paraffin vals, corresponding to SEM under the normality assump- immediately after necropsy. Sections (5 mm thick) were tion. Analyses were performed in SAS 9.2. adhered to positively charged slides, dewaxed in xylene, and hydrated using graded ethanol washes. Heat-induced Antibodies and Western blotting antigen retrieval was performed using Tris-EDTA Buffer (10 Cells were allowed to adhere overnight in media contain- mmol/L Tris base, 1 mmol/L EDTA solution, pH 9.0) ing 10% FCS. The following day, media were changed to incubation in a pressure cooker for 3 minutes. After 30 2.5% CS-FCS and cultured for 48 hours, then lysed in buffer minutes of blocking in 10% normal goat serum in PBS,

www.aacrjournals.org Clin Cancer Res; 19(22) November 15, 2013 6165

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

Skor et al.

slides were incubated with either a 1:100 dilution of anti-GR qRT-PCR (Fig. 1C) and total GR protein expression by rabbit polyclonal antibody (Santa Cruz Biotechnology, H- Western blot analysis (Fig. 1D). Q-RT-PCR confirmed 300 sc-8992) followed by a secondary Alexa Fluor goat anti- expression of GR (NR3C1) mRNA levels in all 3 cells lines. rabbit IgG (Cell Signaling Technology). The highest total GR mRNA and protein levels were found in Entire scans of each tumor section were captured using MDA-MB-231 cells; interestingly, several translational iso- the CRi Panoramic Whole Slide Scanner (PerkinElmer Life forms (GR-A, GR-B, GR-C, GR-D) were also observed in all Sciences). Twenty random individual images of each scan three cell lines (20, 21). were then analyzed from different locations of the slide. The proportion of GRþ staining cells over the total cell Mifepristone induces caspase-3–associated cell death count for each image was calculated (details in Supplemen- The molecular mechanisms underlying GR-mediated cell tary Method S2) and log-transformed to satisfy the normal- survival in the context of chemotherapy-induced apoptosis ity assumption. A mixed-effects model was fitted, with are not well understood (12). We therefore characterized treatment as the fixed effect, and tumor as the random caspase-3 and PARP cleavage in association with paclitaxel- effect, to account for the correlation between multiple induced apoptosis. We found that chemotherapy-induced images of the same tumor section. apoptosis and accompanying caspase-3 and PARP cleavage increased following the addition of mifepristone (Fig. 2). The increased cleavage of caspase-3 and PARP is seen after Results 24 hours of treatment but enhanced after 48 hours of treat- Mifepristone enhances paclitaxel-induced TNBC cell ment. A second biologic replicate of caspase-3 cleavage at 24 death and 48 hours is reported in Supplementary Fig. S4. These We have previously showed that high GR-expressing ER- data suggest that mifepristone reverses GR-mediated cell sur- negative breast cancer cells exposed to physiologic stress vival, at least in part, through blocking an apoptotic mecha- dose concentrations of glucocorticoids (1 mmol/L) are rel- nism involving increased cleavage of caspase-3 and PARP. atively resistant to chemotherapy-induced cell death (13). Here, we examined whether or not pharmacologically rel- Mifepristone antagonizes GR-mediated gene evant concentrations of mifepristone (100 nmol/L) could expression in TNBC cell lines reverse the cytoprotective effects of physiological glucocor- SGK1 and MKP1/DUSP1 genes are directly upregulated ticoid concentrations (100 mmol/L) in MDA-MB-231, by GR transactivation in mammary epithelial cells (2). To MDA-MB-468, and BT-20 TNBC cell lines (18). We used examine whether mifepristone antagonizes this GR-medi- the IncuCyte system, a real-time microscopic-imaging sys- ated gene expression, we treated MDA-MB-231, BT-20, and tem (16), to determine the percentage of cell death contin- MDA-MB-468 cells with dex (100 nmol/L) mifepristone uously over several days using software outlined in Supple- (100 nmol/L) for 4 hours. In MDA-MB-231 cells, Q-RT-PCR mentary Method S1. Cells were treated with vehicle (EtOH showed an average dex-associated 2.13-fold increase in 0.1% V/V), dexamethasone (100 nmol/L), mifepristone SGK1 and 6.62-fold increase in MKP1/DUSP1 mRNA levels (100 nmol/L), or dexamethasone/mifepristone (100 over vehicle alone; the dex-mediated increase in both GR nmol/L) 1 hour before paclitaxel (100 nmol/L) before being target genes was significantly reversed with the addition of placed in the IncuCyte assay system. Figure 1A shows the mifepristone (Fig. 3A). In BT-20 cells, SGK1 (2.10-fold percentage of cell death measured over several days. The increase) and MKP1/DUSP1 (1.70-fold increase) mRNA addition of mifepristone to MDA-MB-231 (Fig. 1A, top) and levels were both inhibited by mifepristone (Fig. 3B). In BT-20 (Fig. 1A, middle) cells significantly reversed the MDA-MB-468 cells, the dex-associated increase in SGK1 protective effect of physiologic glucocorticoid concentra- (2.85-fold increase) was significantly inhibited by mifep- tions (dexamethasone, 100 nmol/L). The addition of ristone, whereas MKP1/DUSP1 (1.52-fold increase) expres- mifepristone to MDA-MB-468 cells also increased cell death sion was reversed although it did not meet statistical sig- from dexamethasone/paclitaxel, although not significantly nificance (Fig. 3C). These data suggest the degree of GR (P ¼ 0.68; Fig. 1A, bottom). Interestingly, treating MDA- inhibition with mifepristone is variable depending on cell MB-231, BT-20, and MDA-MB-468 cells with mifepristone type and the particular gene studied. alone (no chemotherapy) had no significant effect on either apoptosis (Fig. 1) or cell proliferation (Supplementary Fig. Mifepristone treatment potentiates paclitaxel S1). A representative image of cells before counting is shown effectiveness in an MDA-MB-231 TNBC xenograft model in Supplementary Fig. S2. Figure 1B shows images from the In cell lines with robust mifepristone-mediated antago- IncuCyte detection system where green cells reflect apopto- nism of GR target gene expression, we hypothesized that tic cells reported in Fig. 1A (19). Trypan blue exclusion mifepristone administered in vivo might reverse the endog- assays (Supplementary Fig. S3) further supported the con- enous GR activity of TNBC and increase chemotherapy sen- clusion that mifepristone treatment partially reversed GR- sitivity. Therefore, SCID mice bearing MDA-MB-231 GRþ mediated protection from chemotherapy-induced apopto- TNBC mammary fat pad tumor xenografts (200 mm3) sis in TNBC. were treated with either vehicle, paclitaxel (10 mg/kg/d), We next measured GR (NR3C1) mRNA transcript expres- or mifepristone (15 mg/kg/d) 1 hour before paclitaxel sion in MDA-MB-231, MDA-MB-468, and BT-20 cells by (10 mg/kg/d) for 5 consecutive days (17). Tumors were

6166 Clin Cancer Res; 19(22) November 15, 2013 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

GR Antagonism in TNBC

Figure 1. In vitro analysis of paclitaxel-induced cell death mifepristone in MBA-MB-231, BT-20, and MDA-MB-468 cells. A, TNBC cell lines were treated with vehicle (EtOH 0.01%), mifepristone (mif; 100 nmol/L), paclitaxel (pac; 100 nmol/L), dexamethasone (dex; 100 nmol/L)/paclitaxel (100 nmol/L), or dexamethasone (100 nmol/L)/mifepristone (100 nmol/L)/paclitaxel (100 nmol/L). Error bars represent SEM. , two treatments compared to derive the P value shown. B, representative phase-contrast and cytotoxic fluorescent dye images of 3 TNBC cell lines using automated IncuCyte imaging. C, total GR (NR3C1) mRNA expression was first normalized to b-actin mRNA expression and then compared (fold difference) to one technical replicate associated with MDA-MB-231 NR3C1 levels (error bars reflect SEM of 3 independent experiments). D, Western blot analysis of GR and b-actin. GRa translational isoforms are labeled. then measured 3 times per week for 35 days (Fig. 4A) except (SEM ¼ 13) on day 35, whereas the average tumor volumes for vehicle-treated animals, which were sacrificed once the for the mifepristone/paclitaxel cohort were 300 mm3 (SEM tumor had reached approximately 3,000 mm3. Analysis of ¼ 12) on day 18 and 800 mm3 (SEM ¼ 12) on day 35. the mean tumor volumes SEMs for each treatment group Finally, the generalized F-test showed a significant differ- are shown in Fig. 4A. Repeated measures ANOVA model ence between mifepristone/paclitaxel- and vehicle/paclitax- found a significant treatment day2 interaction (P < 0.001) el-treated tumors over time (P ¼ 0.0417). Thus, the addition indicating significant differences in the pattern of tumor of mifepristone treatment 1 hour before chemotherapy growth over time. Differences at each time point in pacli- significantly improved the efficacy of paclitaxel in MDA- taxel versus mifepristone/paclitaxel treatment groups MB-231 xenografts. were evaluated using post hoc testing. Significant differences To explore how the addition of mifepristone treat- between the paclitaxel and mifepristone/paclitaxel treat- ment increases chemotherapy-induced TNBC tumor ment groups appeared at day 18 (P ¼ 0.02) and remained shrinkage, paraffin-embedded tumor xenograft sections significantly different throughout the rest of the experi- were examined with a green fluorescence–labeled anti-GR ment. The average tumor volumes for the paclitaxel cohort antibody and stained with DAPI (representative image were 500 mm3 (SEM ¼ 14) on day 18 and 2,200 mm3 Supplementary Fig. S5). As shown in Fig. 4B, tumors from

www.aacrjournals.org Clin Cancer Res; 19(22) November 15, 2013 6167

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

Skor et al.

Figure 2. Caspase-3 and PARP cleavage following treatment with mifepristone. MDA-MB-231 cells were treated with vehicle (EtOH 0.01%), dexamethasone (dex; 100 nmol/L), mifepristone (mif; 100 nmol/L), paclitaxel (pac; 100 nmol/L), dexamethasone/ paclitaxel, or dexamethasone/ mifepristone/paclitaxel for 24 and 48 hours. The addition of mifepristone resulted in increased cleaved caspase-3 and PARP compared with dexamethasone/ paclitaxel treatment alone.

mice (n ¼ 6) treated with paclitaxel alone had a trend sequencing of TNBC samples also strongly suggests that this toward a higher percentage of strongly GRþ residual tumor type of breast cancer can be further classified into several cells compared to tumors from mice (n ¼ 5) treated with molecular subtypes (24). Defining the major driver path- mifepristone and paclitaxel (19% vs. 8%, P ¼ 0.11). Soft- ways dividing TNBC into various phenotypic subtypes is a ware code is described in Supplementary Method S2. This critical question to be answered to improve TNBC outcome. difference in GR expression was further supported by West- Over a decade ago, we discovered that GR activation by ern blot analysis of total GR protein expression in the physiologic concentrations of glucocorticoids mediates residual tumors (Supplementary Fig. S6). These data suggest potent antiapoptotic signaling in the context of either growth that the addition of mifepristone treatment results in factor deprivation (8, 14) or chemotherapy-induced apo- improved cytotoxicity of highly GR-expressing MDA-MB- ptosis (13) in GRþ, ER-negative premalignant breast epi- 231 cells, ultimately resulting in a smaller population of thelial and TNBC cell lines. Furthermore, mifepristone, a high GRþ tumor cells and greater tumor shrinkage. GR antagonist, was observed to reverse the cell survival The effect of mifepristone in combination with glucocor- effects of GR activation in c-Myc–overexpressing ER-negative ticoids and paclitaxel on colony formation in vitro was MCF10A cells, an in vitro model for preinvasive TNBC (8). further evaluated with a clonogenic assay (Fig. 4C). MDA- Here we hypothesized that increased sensitivity to chemo- MB-231 cells were treated with vehicle (EtOH 0.1% V/V), dex therapy-induced apoptosis would result from concomitant (100 nmol/L), mifepristone (100 nmol/L), or dexametha- GR antagonism with mifepristone of TNBCs. Indeed, we sone/mifepristone (100 nmol/L) 1 hour before paclitaxel found that cotreatment with the GR-antagonist mifepristone (100 nmol/L) in 2.5% CS-FCS. Colonies were counted 96 both reverses GR-mediated gene expression in 3 TNBC cell hours after treatment. The addition of mifepristone to lines and augments chemotherapy-induced apoptosis. dex/pac significantly decreased the number of colonies The mechanism by which GR activation inhibits apopto- formed in comparison to dex/pac alone (P ¼ 0.0047). A sis appears to require GR-mediated transcriptional induc- representative image of each treatment at 3 different magni- tion of both SGK1 and MKP1/DUSP1 (13). SGK1 over- fications (15,24,and38) is reported in Supplementary expression was previously found to inhibit PARP-depen- Fig. S7. These data suggest that mifepristone significantly dent apoptosis in a variety of cell lines (8), in part, through reduces tumor viability in the setting of chemotherapy inde- phosphorylation and inactivation of the Forkhead tran- pendently of an effect on cell proliferation. scription factors (8). Here, we find that GR activation blunts paclitaxel (and deprivation)-induced cleavage of caspase-3 and PARP. The addition of mifepristone to Discussion dexamethasone reversed the reduced caspase-3 and PARP The limited success of targeted treatments for patients cleavage resulting from growth factor deprivation as well as with TNBC highlights the complex heterogeneity of ER/ paclitaxel therapy, suggesting that GR activity is an impor- PR/HER2 breast cancer (22, 23). Next-generation tant mediator of caspase-3 and PARP-dependent apoptosis,

6168 Clin Cancer Res; 19(22) November 15, 2013 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

GR Antagonism in TNBC

regardless of initiating cellular insult (8). We are currently characterizing additional antiapoptotic GR target genes and networks modified by GR activity to better understand the transcriptional regulatory mechanisms through which glucocorticoids and mifepristone modify chemotherapy- induced cytotoxicity. Although most cell lines used in TNBC research express GR-alpha, levels vary (14). In a cohort of more than 300 patients with early-stage ER-negative cancer, we reported that the highest quartile of GR-expressing primary tumors (by Affymetrix gene arrays) had a significantly worse long- term prognosis compared to patients with tumors expres- sing the bottom quartile of GR expression (2). In these studies, increased SGK1 and MKP1/DUSP1 tumor expres- sion also correlated with high GR expression, suggesting that these GR target genes as well as others are likely reflecting increased GR activity (2). On the basis of the results reported here, we anticipate that GR antagonism will be a particularly useful treatment for patients with GRþ tumors that are likely chemotherapy-resistant TNBC. Recently, next-generation sequencing of more than 100 primary TNBCs identified the NR3C1 (GR-encoding) gene as among the top genes undergoing somatic mutation in association with significant changes in expression of shared GR network genes (24, 25). These findings support the hypothesis that GR activity is a significant driver in the biology of a subset of TNBC. A previous study by Pietenpol and colleagues (26) char- acterized somatic mutations in a panel of TNBC cell lines including the MDA-MB-231, BT-20, and MDA-MB-468 lines studied here. MDA-MB-468 cells, the least sensitive to the addition of mifepristone in our assay, were classified as "heavily enriched for mutations relating to cell cycle and cell division pathways" including mutations in both p53 and RB (ref. 26; see Table 1). Interestingly, this cell line was most sensitive to paclitaxel monotherapy (approximately twice the percentage cell death was observed in MDA-MB- 468 cells compared to either MDA-MB-231 or BT-20 cells). In contrast, MDA-MB-231 and BT-20 cells predominately harbor somatic mutations in genes encoding proteins relat- ed to cell survival and growth factor signaling (respectively PDGFRA and PIK3CA) and were the least sensitive to paclitaxel monotherapy (ref. 26; Table 1). This suggests that TNBCs with mutations in survival signaling pathways (rather than cell-cycle pathways) could benefit the most from GR antagonism because GR antagonism blocks cell survival but has little effect on proliferation in ER cells (Supplementary Fig. S1). Further clinical/translational studies could test the hypothesis that the "basal" type TNBC harboring p53 and RB mutations (both affecting cell cycle) Figure 3. SGK1 and MKP1/DUSP1 steady-state mRNA analysis following will benefit less from GR antagonism; these tumors also 4 hours of dex versus dex/mif treatment. MDA-MB-231, MDA-MB-468, or BT-20 cells were treated with vehicle (EtOH 0.01%), dex (100 nmol/L), or dex/mif (100 nmol/L) for 4 hours. SGK1 and MKP1/DUSP1 mRNA MKP1/DUSP1 P ¼ expression was normalized to b-actin mRNA levels. Normalized SGK1 and ( 0.005) mRNA expression with the addition of SGK1 and MKP1/DUSP1 fold change relative to vehicle treatment is shown mifepristone. C, in MDA-MB-468 cells, dexamethasone-induced P ¼ fi (n ¼ 3 experiments SEM). A, in MDA-MB-231 cells, dex-induced SGK1 ( 0.001) steady-state mRNA levels were signi cantly higher compared (P ¼ 0.02) and MKP1/DUSP1 (P ¼ 0.008) mRNA expression was with dexamethasone/mifepristone-treated cells, although the difference MKP1/DUSP1 fi P < significantly reversed following concomitant mifepristone treatment. B, in mRNA levels was not signi cant (NS). , 0.05; P < P < BT-20 cells showed significantly less induction of both SGK1 (P ¼ 0.004) , 0.01; , 0.001.

www.aacrjournals.org Clin Cancer Res; 19(22) November 15, 2013 6169

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

Skor et al.

Table 1. Subtypes and associated somatic mutations in TNBC cell lines (26)

TNBC subtype Cell line Mutated genes MDA-MB-231 Mesenchymal-like TP53, CDKN2A, PDGFRA, BRAF, KRAS, NF2 BT-20 Unclassified TP53, CDKN2A, PIK3CA MDA-MB-468 Basal-like TP53, RB1, PTEN, SMAD4

NOTE: The underlined somatically mutated genes encode proteins having primary roles in cell cycle regulation, while those without underlining have primary roles in malignant transformation including epithelial/mesenchymal transition (EMT). BT-20 and MDA-MB-231 are predicted to have mainly cell cycle gene mutations, while MDA-MB-231 cell have many mutations involving genes regulating cell cycle and transformation.

The residual MDA-MB-231 xenograft tumors we excised and examined approximately one month following treat- ment with paclitaxel mifepristone suggest that residual tumor GR expression is relatively low following dual pac- litaxel and mifepristone treatment compared to tumor remaining after paclitaxel monotherapy. Interestingly, not all individual MDA-MB-231 cells had the same intensity of immunofluorescent GR expression (Fig. 4B). This implies that mifepristone may preferentially target high GR-expres- sing MDA-MB-231 cells for chemotherapy-induced cell death. Another possibility is that mifepristone results in increased non-metabolized paclitaxel concentrations based on the ability of GR ligands (such as mifepristone and dexamethasone) to inhibit the CYP3A4 enzyme, there- Figure 4. Paclitaxel mifepristone treatment of MDA-MB-231 tumor by increasing active paclitaxel levels (27). However, we have xenografts. A, mice bearing MDA-MB-231 human TNBC xenograft tumors previously measured plasma paclitaxel levels from SCID in the thoracic mammary fat pad were treated (arrows) with vehicle (closed mice treated with either vehicle/paclitaxel or dex/paclitaxel squares, n ¼ 5), paclitaxel (open circles, n ¼ 7) mifepristone (closed triangles, n ¼ 6) via i.p. injection for 5 consecutive days. Tumors were and found no significant differences in active paclitaxel measured 3 times a week for 35 days. Daily pretreatment with mifepristone concentrations (9). Therefore, the lower percentage of significantly improved tumor response compared to treatment with GR-expressing cells following mifepristone treatment is paclitaxel alone (P ¼ 0.04). To determine the significance of treatment more likely due to greater sensitivity of high GR-expressing effects on tumor growth, repeated measures ANOVA was performed. Error cells to chemotherapy-induced apoptosis with the addition bars represent SEM. B, tumors were removed at the endpoint of the experiment, fixed, paraffin-embedded, and sectioned followed by of mifepristone. fluorescent immunohistochemistry. A higher percentage of GRþ cells The binding affinity of mouse to remained in tumors following treatment with paclitaxel alone (n ¼ 6animals) human GR is 1.5 to 3 times lower that (28). While compared to tumors from SCID mice treated with mif/paclitaxel (n ¼ 5mice; we observed a statistically significant difference between the P ¼ 0.11). C, MDA-MB-231 cells were treated with vehicle (EtOH 0.01%), in vivo mifepristone (mif; 100 nmol/L), paclitaxel (pac; 100 nmol/L), dexamethasone pac and mif/pac treatment , the decreased binding (dex; 100 nmol/L)/paclitaxel (100 nmol/L), or dexamethasone (100 nmol/L)/ affinity of mouse corticosterone to the human MDA-MB- mifepristone (100 nmol/L)/paclitaxel (100 nmol/L), and colonies were 231 xenografted cells could dampen the antagonistic effect counted after 96 hours of treatment. Error bars represent SEM. P,two of mifepristone. However, we also conducted in vitro cell P treatments compared to derive the value shown. viability assays with dexamethasone at 100 nmol/L, which is similar to plasma cortisol concentrations in patients (29). tend to be more sensitive to initial chemotherapy treat- In these experiments, we also observed an increase in tumor ment compared to the "mesenchymal" TNBC subtypes that cell death with the addition of mifepristone to dex/pac. include MDA-MB-231 (26). Furthermore, clonogenicity of MDA-MB-231 cells was

6170 Clin Cancer Res; 19(22) November 15, 2013 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

GR Antagonism in TNBC

significantly decreased with dex/mif/pac compared to dex/ PR-negative, HER2-negative but GR-positive breast cancer pac. Taken together, these data support the hypothesis that will be determined. mifepristone co-treatment targets high GR-expressing tumor cells that would otherwise be resistant to chemo- Disclosure of Potential Conflicts of Interest therapy alone. The University of Chicago, S.D. Conzen and M. Kocherginsky were Mifepristone has been studied extensively since the 1980s assigned a patent for the use of GR antagonists in ER-negative breast cancer while this manuscript was under review. No other conflicts of interest were for its ability to antagonize progesterone and glucocorticoid disclosed by the other authors. receptors in various human tissues (30). However, previous studies using mifepristone in breast cancer have only con- sidered the use of mifepristone to target the PR in ERþ/PRþ Authors' Contributions Conception and design: M.N. Skor, S.D. Conzen tumors, never to target the GR in TNBC. For example, Development of methodology: M.N. Skor, M. Kocherginsky, B.A. Hall, several ERþ/PRþ breast tumor xenograft studies have sug- S.D. Conzen Acquisition of data (provided animals, acquired and managed patients, gested a decrease in volume following treatment with provided facilities, etc.): M.N. Skor, E.L. Wonder, A. Goyal, B.A. Hall, S.D. mifepristone alone (subcutaneous administration, doses Conzen ranging from 25 to 50 mg/kg) or in combination with Analysis and interpretation of data (e.g., statistical analysis, biosta- tistics, computational analysis): M.N. Skor, M. Kocherginsky, Y. Cai, S.D. anti-estrogen therapy. In addition, a phase II Conzen with 28 patients found that daily single-agent oral mifep- Writing, review, and/or revision of the manuscript: M.N. Skor, E.L. ristone treatment (200 mg) for recurrent ERþ/PRþ breast Wonder, M. Kocherginsky, S.D. Conzen Administrative, technical, or material support (i.e., reporting or orga- cancer resulted in 3 partial responses for an overall response nizing data, constructing databases): M.N. Skor, E.L. Wonder, S.D. rate of 10.7% and only mild-to-moderate side effects (nau- Conzen sea, lethargy, anorexia, and hot flashes were noted; ref. 31). Study supervision: M.N. Skor, S.D. Conzen In our studies, an in vitro mifepristone concentration of 100 nmol/L was used to target the GR in PR-nonexpressing Acknowledgments breast tumors based on previous human pharmacokinetic The authors thank Robert Roe MD (Corcept Therapeutics) for supplying pharmaceutical-grade mifepristone and for helpful discussions on pharma- studies where subjects received between 100 and 800 mg/d cology, Christine Labno PhD of the University of Chicago’s Integrated of drug. The concentration of mifepristone in the serum Microscopy Core Facility and Russell Szmulewitz MD for expert help with immunofluorescence, and Paul Volden and other members of the Conzen 24 hours after administration was found to be approxi- and Matthew Brady laboratories for valuable feedback and expertise. mately 2.0 mmol/L irrespective of dose (32). In addition, chorionic villi tissue levels in women receiving a single dose of 200 mg of mifepristone found cytosolic concentra- Grant Support The study was supported by NIH R01 CA089208, The University of tions on average of 238 nmol/L (18). These human data Chicago Comprehensive Cancer Center NIH P30 CA014599, and Susan suggest that 100 nmol/L is a physiologically achievable G. Komen for the Cure IIR12223772. Corcept Therapeutics provided phar- mifepristone concentration in tumor tissue. macological-grade mifepristone only. The costs of publication of this article were defrayed in part by the An ongoing phase I clinical trial is evaluating the safety payment of page charges. This article must therefore be hereby marked of mifepristone (300 mg/d) for 2 days followed imme- advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate diately on day 2 by a weekly dose of nab-paclitaxel. In this this fact. study, the safety and tolerability of mifepristone in com- Received December 14, 2012; revised August 8, 2013; accepted August 27, bination with chemotherapy for advanced ER-negative, 2013; published OnlineFirst September 9, 2013.

References 1. Bamberger CM, Schulte HM, Chrousos GP. Molecular determinants of 7. Wyllie AH. Glucocorticoid-induced thymocyte apoptosis is associ- glucocorticoid receptor function and tissue sensitivity to glucocorti- ated with endogenous endonuclease activation. Nature 1980;284: coids. Endocr Rev 1996;17:245–61. 555–6. 2. Pan D, Kocherginsky M, Conzen SD. Activation of the glucocorticoid 8. Moran TJ, Gray S, Mikosz CA, Cozen SD. The glucocorticoid receptor receptor is associated with poor prognosis in estrogen receptor- mediates a survival signal in human mammary epithelial cells. Cancer negative breast cancer. Cancer Res 2011;71:6360–70. Res 2000;60:867–72. 3. Armaiz-Pena GN, Lutgendorf SK, Cole SW, Sood AK. Neuroendo- 9. Pang D, Kocherginsky M, Krausz T, Kim SY, Conzen SD. Dexa- crine modulation of cancer progression. Brain Behav Immun 2009; decreases xenograft response to Paclitaxel through 23:10–5. inhibition of tumor cell apoptosis. Cancer Biol Ther 2006;5: 4. McKenna NJ, O'Malley BW. Combinatorial control of gene ex- 933–40. pression by nuclear receptors and coregulators. Cell 2002;108: 10. Brickley DR, Mikosz CA, Hagan CR, Conzen SD. Ubiquitin modification 465–74. of serum and glucocorticoid-induced protein kinase-1 (SGK-1). J Biol 5. Lewis-Tuffin LJ, Cidlowski JA. The physiology of human glucocorti- Chem 2002;277:43064–70. coid receptor beta (hGRbeta) and glucocorticoid resistance. Ann N Y 11. Huang Y, Johnson KR, Norris JS, Fan W. Nuclear factor-kappaB/ Acad Sci 2006;1069:1–9. IkappaB signaling pathway may contribute to the mediation of pac- 6. Gibson S, Tu S, Oyer R, Anderson SM, Johnson GL. Epidermal litaxel-induced apoptosis in solid tumor cells. Cancer Res 2000;60: growth factor protects epithelial cells against Fas-induced apo- 4426–32. ptosis. Requirement for Akt activation. J Biol Chem 1999;274: 12. Zhang C, Wenger T, Mattern J, Ilea S, Frey C, Gutwein P, et al. Clinical 17612–8. and mechanistic aspects of glucocorticoid-induced chemotherapy

www.aacrjournals.org Clin Cancer Res; 19(22) November 15, 2013 6171

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

Skor et al.

resistance in the majority of solid tumors. Cancer Biol Ther 2007;6: 22. O'Shaughnessy J, Osborne C, Pippen JE, Yoffe M, Patt D, Rocha C, 278–87. et al. Iniparib plus chemotherapy in metastatic triple-negative breast 13. Wu W, Chaudhuri S, Brickley DR, Pang D, Karrison T, Conzen SD. cancer. N Engl J Med 2011;364:205–14. Microarray analysis reveals glucocorticoid-regulated survival genes 23. Arnedos M, Bihan C, Delaloge S, Andre F. Triple-negative breast cancer: that are associated with inhibition of apoptosis in breast epithelial cells. are we making headway at least? Ther Adv Med Oncol 2012;4:195–210. Cancer Res 2004;64:1757–64. 24. Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, et al. The clonal and 14. Mikosz CA, Brickley DR, Sharkey MS, Moran TW, Conzen SD. Glu- mutational evolution spectrum of primary triple-negative breast can- cocorticoid receptor-mediated protection from apoptosis is associ- cers. Nature 2012;486:395–9. ated with induction of the serine/threonine survival kinase gene, sgk-1. 25. Koboldt DC, Fulton RS, McLellan MD, Schmidt H, Kalicki-Veizer J, J Biol Chem 2001;276:16649–54. McMichael JF, et al. Comprehensive molecular portraits of human 15. Melhem A, Yamada SD, Fleming GF, Delgado B, Brickley DR, Wu W, breast tumours. Nature 2012;490:61–70. et al. Administration of glucocorticoids to ovarian cancer patients is 26. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr associated with expression of the anti-apoptotic genes SGK1 and Y, et al. Identification of human triple-negative breast cancer subtypes MKP1/DUSP1 in ovarian tissues. Clin Cancer Res 2009;15:3196–204. and preclinical models for selection of targeted therapies. J Clin 16. Thon JN, Devine MT, Begonja AJ, Tibbitts J, Italiano JE Jr. High- Investig 2011;121:2750–67. content live-cell imaging assay used to establish mechanism of tras- 27. He K, Woolf TF, Hollenberg PF. Mechanism-based inactivation of tuzumab emtansine (T-DM1)-mediated inhibition of produc- cytochrome P-450-3A4 by mifepristone (RU486). J Pharmacol Exp tion. Blood 2012;120:1975–84. Ther 1999;288:791–7. 17. Masson MJ, Collins LA, Carpenter LD, Graf ML, Ryan PM, Bourdi M, 28. Giannopoulos G, Keichline D. Species-related differences in steroid- et al. Pathologic role of stressed-induced glucocorticoids in drug- binding specificity of glucocorticoid receptors in lung. Endocrinology induced liver injury in mice. Biochem Biophys Res Commun 2010;397: 1981;108:1414–9. 453–8. 29. Weitzman ED, Fukushim D, Nogeire C, Roffwarg H, Gallaghe Tf, 18. Wang JD, Shi WL, Zhang GQ, Bai XM. Tissue and serum levels of Hellman L. 24 hour pattern of episodic secretion of cortisol in normal steroid-hormones and RU-486 after administration of mifepristone. subjects. J Clin Endocrinol Metab 1971;40:850–5. Contraception 1994;49:245–53. 30. Spitz IM, Bardin CW. Drug-therapy – Mifepristone (RU-486) – a mod- 19. Abraham VC, Towne DL, Waring JE, Warrior U, Burns DJ. Application ulator of progestin and glucocorticoid action. N Engl J Med 1993; of a high-content multiparameter cytotoxicity assay to prioritize com- 329:404–12. pounds based on toxicity potential in humans. J Biomol Screening 31. Perrault D, Eisenhauer EA, Pritchard KI, Panasci L, Norris B, Vanden- 2008;13:527–37. berg T, et al. Phase II study of the progesterone antagonist mifepris- 20. Lu NZ, Cidlowski JA. Translational regulatory mechanisms generate N- tone in patients with untreated metastatic breast carcinoma: a National terminal glucocorticoid receptor isoforms with unique transcriptional Cancer Institute of Canada Clinical Trials Group study. J Clin Oncol target genes. Mol Cell 2005;18:331–42. 1996;14:2709–12. 21. Gross KL, Oakley RH, Scoltock AB, Jewell CM, Cidlowski JA. Gluco- 32. Heikinheimo O, Kekkonen R, Lahteenmaki P. The pharmacokinet- corticoid receptor alpha isoform-selective regulation of antiapoptotic ics of mifepristone in humans reveal insights into differential genes in osteosarcoma cells: a new mechanism for glucocorticoid mechanisms of antiprogestin action. Contraception 2003;68: resistance. Mol Endocrinol 2011;25:1087–99. 421–6.

6172 Clin Cancer Res; 19(22) November 15, 2013 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst September 9, 2013; DOI: 10.1158/1078-0432.CCR-12-3826

Glucocorticoid Receptor Antagonism as a Novel Therapy for Triple-Negative Breast Cancer

Maxwell N. Skor, Erin L. Wonder, Masha Kocherginsky, et al.

Clin Cancer Res 2013;19:6163-6172. Published OnlineFirst September 9, 2013.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-12-3826

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2013/09/09/1078-0432.CCR-12-3826.DC1

Cited articles This article cites 32 articles, 11 of which you can access for free at: http://clincancerres.aacrjournals.org/content/19/22/6163.full#ref-list-1

Citing articles This article has been cited by 19 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/19/22/6163.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/19/22/6163. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research.