throbber
NIH Public Access
`Author Manuscript
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`Published in final edited form as:
`Cell. 2013 December 5; 155(6): 1309–1322. doi:10.1016/j.cell.2013.11.012.
`
`Glucocorticoid Receptor Confers Resistance to Anti-Androgens
`
`by Bypassing Androgen Receptor Blockade
`
`Vivek K. Arora1,2, Emily Schenkein1, Rajmohan Murali1,3, Sumit K. Subudhi2, John
`Wongvipat1, Minna D. Balbas1,4, Neel Shah1,4, Ling Cai1, Eleni Efstathiou5, Chris
`Logothetis5, Deyou Zheng6, and Charles L. Sawyers1,7
`1Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New
`York, NY 10065.
`
`2Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
`
`3Department of Pathology Memorial Sloan-Kettering Cancer Center, New York, NY 10065
`
`4Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan-Kettering
`Cancer Center, New York, NY 10065.
`
`5Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer
`Center Houston, TX 77030
`
`6Departments of Neurology, Genetics and Neuroscience, Albert Einstein College of Medicine,
`Bronx, NY 10461
`
`7Howard Hughes Medical Institute, Chevy Chase, MD 20815
`
`Summary
`
`The treatment of advanced prostate cancer has been transformed by novel antiandrogen therapies
`such as enzalutamide. Here we identify induction of glucocorticoid receptor (GR) expression as a
`common feature of drug resistant tumors in a credentialed preclinical model, a finding also
`confirmed in patient samples. GR substituted for the androgen receptor (AR) to activate a similar
`but distinguishable set of target genes and was necessary for maintenance of the resistant
`phenotype. The GR agonist dexamethasone was sufficient to confer enzalutamide resistance
`whereas a GR antagonist restored sensitivity. Acute AR inhibition resulted in GR upregulation in a
`subset of prostate cancer cells due to relief of AR-mediated feedback repression of GR expression.
`These findings establish a novel mechanism of escape from AR blockade through expansion of
`cells primed to drive AR target genes via an alternative nuclear receptor upon drug exposure.
`
`Introduction
`
`Recently approved drugs that target androgen receptor (AR) signaling such as abiraterone
`and enzalutamide have rapidly become standard therapies for advanced stage prostate cancer
`(Scher et al., 2012b) (de Bono et al., 2011). Despite their success, sustained response with
`these agents is limited by acquired resistance which typically develops within ~6-12 months.
`Clinical success of kinase inhibitors in other tumors such as melanoma, lung cancer,
`
`© 2013 Elsevier Inc. All rights reserved.
`
`Correspondence: sawyersc@mskcc.org.
`
`Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
`customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
`the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
`discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Amerigen Exhibit 1137
`Amerigen v. Janssen IPR2016-00286
`
`

`
`Arora et al.
`
`Page 2
`
`leukemia and sarcoma is similarly transient (Sawyers et al., 2002) (Chapman et al., 2011)
`(Demetri et al., 2002) (Maemondo et al., 2010), resulting in numerous efforts to define
`mechanisms of acquired resistance. One strategy that has proven particularly useful is
`prolonged treatment of drug-sensitive preclinical models to derive drug-resistant sublines,
`followed by genome-wide profiling studies to ascertain differences that may play a causal
`role in conferring drug resistance. A common mechanism that has emerged from these
`kinase inhibitor studies is reactivation of the signaling pathway targeted by the drug, directly
`by mutation of the kinase target or indirectly by bypassing pathway inhibitor blockade
`through amplification of an alternative kinase (Glickman and Sawyers, 2012). Both
`scenarios have been validated in clinical specimens and are guiding efforts to discover next
`generation inhibitors and to develop rational drug combinations.
`
`Clinically relevant mechanisms of resistance to hormone therapy in prostate cancer have
`also been elucidated using preclinical models. Hormone therapy, through the use of drugs
`that lower serum testosterone or competitively block the binding of androgens to AR, has
`been the mainstay of treatment for metastatic prostate cancer for decades but is not curative.
`The late stage of disease, which is refractory to hormone therapy, is termed castration
`resistant prostate cancer (CRPC). We previously examined the molecular basis of
`progression to CRPC in mouse models and discovered that increased AR expression was the
`primary mechanism (Chen et al., 2004). We then used this observation to screen for novel
`anti-androgens that restore AR inhibition in the setting of increased AR levels. These efforts
`yielded three second-generation anti-androgens: enzalutamide, ARN-509, and RD162 (Tran
`et al., 2009) (Clegg et al., 2012). Enzalutamide and ARN-509 were further developed for
`clinical use, culminating in FDA approval of enzalutamide in 2012 based on increased
`survival (Scher et al., 2012b).
`
`Now with widespread use, resistance to enzalutamide is a major clinical problem. We and
`others have recently identified an AR point mutation as one resistance mechanism by
`derivation of drug-resistant sublines following prolonged exposure to enzalutamide or
`ARN-509 (Balbas et al., 2013) (Joseph et al., 2013) (Korpal et al., 2013). This AR mutation
`has also been recovered from patients with resistance to ARN-509 but only in a minority of
`cases (Joseph et al., 2013). Here we define a novel and potentially more prevalent
`mechanism of resistance by which tumors bypass AR blockade through upregulation of the
`glucocorticoid receptor (GR).
`
`Results
`
`GR is expressed in antiandrogen-resistant tumors
`
`We previously showed that LNCaP/AR xenograft tumors regress during the first 28 days of
`treatment with ARN-509 (Clegg et al., 2012), enzalutamide or RD162 (Tran et al., 2009). In
`a pilot study to explore mechanisms of acquired resistance to these drugs, we treated mice
`continually and harvested tumors after progression (mean 163 days, Supplemental Table
`1A). Tissue from fifteen resistant tumors obtained from long term antiandrogen treated mice
`(n=6 ARN-509, n=9 RD162) and from three control tumors from vehicle treated mice were
`analyzed by expression array. Aggregated data from resistant and control tumors in this pilot
`cohort were compared to identify expression changes commonly associated with resistance
`(Figure 1A). Among the most up-regulated genes in the resistant tumors was the
`glucocorticoid receptor (GR, gene symbol NR3C1) which shares overlapping target
`specificity with AR (Mangelsdorf et al., 1995). Of note, several of the most differentially
`expressed genes were known androgen regulated genes (confirmed by transcriptome
`analysis of short term DHT treated LnCaP/AR cells, in vitro (Supplemental Table 1B)), but
`they were altered in directions that did not reflect restored AR signaling. On the one hand,
`SGK1 (Serum Glucocorticoid Induced Kinase 1), a known AR and GR-induced target gene,
`
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Arora et al.
`
`Page 3
`
`was among the most up-regulated genes, but several other androgen-induced genes
`(PMEPA1, SNAI2, KCNN2, LONRF1, SPOCK1) were among the most repressed.
`Conversely, several androgen-repressed genes (UGT2B15, PMP22, CAMK2N1, UGT2B17)
`were among the most up-regulated (Figure 1A). These findings indicated that resistance in
`this model system is unlikely to be mediated by simple restoration of AR activity and raised
`the possibility that GR may play a role.
`
`To explore this question further, we generated an independent set of drug-resistant tumors
`(the validation cohort), focusing on the two second generation antiandrogens in clinical use,
`enzalutamide and ARN-509 (Figure 1B). GR mRNA levels in 10 control, 8 short term
`treated (4 day) and 16 resistant tumors were substantially higher in resistant tissues
`compared to control (median 26.9-fold increase) or 4 day treated tumors (Figure 1C). Of the
`tissues analyzed by RT-qPCR, most were also analyzed for GR expression by western blot,
`based on availability of protein lysates (control n=6, 4 day n=5, resistant n=13). No GR was
`detected in control samples, minimal expression was noted in 4 day treated samples, and
`substantial expression was found in most resistant tumors in a pattern that tended to
`correlate with GR mRNA levels (Figure 1D). There was no correlation between GR
`expression and the specific antiandrogen treatment used (Supplemental Table 1C). In
`contrast to GR, AR RNA or proteins levels were not consistently different across the
`treatment groups (Figure 1C,1D).
`
`To explore AR and GR signaling in more detail, we established cells lines from control and
`drug-resistant tumors by adaptation to growth in vitro. LREX’ (LnCaP/AR Resistant to
`Enzalutamide Xenograft derived) was derived from an enzalutamide-resistant tumor with
`high GR expression, and CS1 was derived from a vehicle treated tumor. We also developed
`a flow cytometry-based assay to measure GR expression on a cell-by-cell basis. In both
`LNCaP/AR and CS1, most cells showed no evidence of GR expression, with the exception
`of a small subpopulation (black arrow, discussed later) (Figure 1E). In contrast, essentially
`all LREX’ cells expressed GR. Intracellular AR staining confirmed that AR levels in LREX’
`did not notably differ from control cells (Figure S1A).
`
`LREX’ tumors are dependent on GR for enzalutamide-resistant growth
`
`Having established the LREX’ model as representative of high GR expression, we next
`confirmed that these cells maintain a resistant phenotype in vivo. LREX’ or control cells
`were injected into castrated mice that were then immediately initiated on antiandrogen
`treatment. LREX’ showed robust growth whereas LNCaP/AR or CS1 lines were unable to
`establish tumors in the presence of antiandrogen (Figure 2A,2B). Strong expression of GR
`was confirmed in multiple LREX’ xenograft tumors by western blot and by IHC (Figure
`S1B, 2C). As expected, untreated LNCaP/AR tumors were negative for GR expression with
`the exception of rare GR-positive cells (Figure 2C). Although many of these GR-positive
`cells had morphologic features of stromal or endothelial cells (blue arrows), some appeared
`epithelial (black arrow), consistent the with flow cytometry analysis (Figure 1E, black
`arrows).
`
`To determine whether GR expression is required to maintain the drug-resistant phenotype,
`LREX’ cells were infected with a shRNA targeting GR (shGR) and stable knockdown of
`GR protein was confirmed (Figure 2F). Tumor growth of shGR infected LREX’ cells was
`significantly delayed relative to shNT (non targeted)-infected cells in castrated mice treated
`with enzalutamide (Figure 2D). In contrast, shGR had no impact on the growth of GR-
`negative CS1 xenografts, diminishing the possibility of an off-target effect (Figure 2E). Of
`note, shGR LREX’ xenografts harvested on day 49 showed decreased GR protein
`knockdown compared to the pre-implantation levels, indicative of selective pressure against
`
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Arora et al.
`
`Page 4
`
`GR silencing in the setting of enzalutamide treatment (Figure 2F). These findings provide
`direct evidence that GR drives enzalutamide resistance in vivo.
`
`GR expression is associated with clinical resistance to enzalutamide
`
`To determine whether GR expression is a feature of clinical antiandrogen resistance, we
`evaluated GR expression in bone metastases from patients receiving enzalutamide. Bone
`marrow samples were obtained prior to enzalutamide treatment (baseline) and again after 8
`weeks of treatment, as previously reported in a cohort of abiraterone-treated patients
`(Efstathiou et al., 2012). Using a GR IHC assay optimized for use in bone marrow samples,
`we quantified the percentage of GR-positive tumor cells and dichotomized the data based on
`clinical response. Patients who continued to benefit from therapy for greater than 6 months
`were defined as good responders, while those in whom therapy was discontinued earlier than
`6 months due to a lack of clinical benefit were classified as poor responders (Figure 3A).
`Consistent with the designation of good versus poor clinical response based on treatment
`status at 6 months, 11 of 13 good responders but only 1 of 14 poor responders had a
`maximal PSA decline greater than 50% (Figure 3C). Akin to the findings in the preclinical
`model, GR positivity at baseline was low: 3% of tumor cells in good responders and 8% in
`poor responders. Of note, 3 of 22 tumors had evidence of high GR expression at baseline (≥
`20% of tumor cells) and all three had a poor clinical response (Figure 3C,D). At 8 weeks,
`the mean percentage of GR positive cells was higher than baseline levels in both response
`groups but was more significantly elevated in poor responders (29% vs 8%, p=.009). In
`addition, the percentage of GR-positive cells at 8 weeks was significantly higher in poor
`compared to good responders (29% versus 10%, p=.02) (Figure 3C,D), and similar results
`were obtained when the analysis was limited to patients from whom matched baseline and 8
`week samples were available for analysis (Figure 3E). Furthermore, when GR IHC data was
`dichotomized based on PSA decline instead of clinical response, GR induction was also
`associated with a limited PSA decline (Figure S2). These findings establish a correlation
`between GR expression and clinical response to enzalutamide and raise the possibility that
`AR inhibition may induce GR expression in some patients. The fact that PSA levels also
`correlate with GR expression raises the question of whether transcriptional regulation of a
`canonical AR target gene may be regulated by GR.
`
`GR expressing drug-resistant tumors show uneven restoration of AR target genes
`
`Having implicated GR as a potential mediator of antiandrogen resistance, we next asked if
`restored AR pathway activity also plays a role by comparing the mRNA transcript levels of
`74 direct AR target genes in control, 4 day, and resistant tumors from the validation cohort
`(Figures S3) as well as eight LREX’ tumors (Figure 4A) (see experimental procedures and
`Supplementary Table 2 for details on gene selection). Consistent with the data generated in
`the pilot cohort (Figure 1A), some AR target genes in resistant tissues showed elevated
`levels relative to control (SGK1, STK39) while other genes (NDRG1, TIPARP, PMEPA1)
`showed no evidence of restored expression.
`
`To examine restoration of AR signaling across the entire set of 74 target genes, we
`calculated a fractional restoration value using log 2 transformed expression values and the
`equation (Resistant – 4 day) / (Control – 4 day). With this approach, a gene whose
`expression in resistant tissue equals the expression in control tumors calculates as 1, while a
`gene whose expression in resistance equals its expression after 4 days of antiandrogen
`treatment equals 0. (Values greater than one indicate hyper-restoration in resistance relative
`to control and values below zero suggest further inhibition as compared to acute treatment.)
`These data confirmed that the pattern of restoration varied gene by gene, but this pattern was
`consistent in LREX’ xenografts and in the validation cohort tumors (Pearson r .64, p = 7.54
`× 10−10, Figure 4B). This finding is most consistent with a model in which AR remains
`
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Arora et al.
`
`Page 5
`
`inhibited in drug-resistant tumors but expression of certain AR target genes is restored by an
`alternative transcription factor, possibly GR. The fact that AR restoration values were
`somewhat higher in the LREX’ analysis correlates with higher GR expression in these
`tumors (Figure 4C).
`
`GR drives expression of AR target genes in resistant tissues
`
`To determine if GR can drive expression of this subset of AR target genes, we compared, in
`vitro, DHT-induced (AR) and dexamethasone (Dex)-induced (GR) expression of 7 AR
`targets that represent the spectrum of restoration noted in the in vivo analysis, as well as PSA
`(Figure 4D). All 8 genes were regulated by DHT as expected, and this regulation was
`blocked by enzalutamide. Thus, AR signaling remains intact and can be inhibited by
`antiandrogens in these drug-resistant cells, making an AR-dependent mechanism of drug
`resistance less likely.
`
`In contrast to DHT, the effect of Dex on these same target genes was variable but closely
`matched the pattern observed in drug resistant xenografts. For example, Dex strongly
`induced SGK1 and STK39 but did not induce TIPARP, NDRG1, and PMEPA1. Of note,
`KLK3 (PSA) was comparably induced by either DHT or Dex, providing evidence that
`persistent PSA expression in patients responding poorly to enzalutamide could be driven by
`GR. As expected, enzalutamide did not notably affect Dex activity. To confirm that this
`pattern of GR-dependent gene expression is not unique to LREX’ cells, we introduced a GR
`expressing retrovirus into parental LNCaP/AR cells and observed a similar pattern of DHT-
`versus Dex-induced gene expression (Figure S4A, S4B). To be sure that the effects of Dex
`in these models are mediated through GR, we co-treated cells with a previously described
`competitive GR antagonist that lacks AR binding called compound 15 (Wang et al., 2006).
`Compound 15 significantly decreased expression of Dex-induced genes, confirming that
`Dex activity in the LREX’ model is GR-dependent (Figure S4C). Lastly, siRNA
`experiments targeting AR confirmed that AR is not necessary for Dex-mediated gene
`activation (Figure S4D). Collectively these experiments demonstrate that GR is able to drive
`expression of certain AR target genes independent of AR.
`
`AR and GR have overlapping transcriptomes and cistromes
`
`To explore AR and GR transcriptomes in an unbiased fashion, we performed expression
`profiling after short-term treatment of LREX’ cells with DHT or Dex in the presence or
`absence of enzalutamide. AR and GR signatures were respectively defined as all genes with
`absolute expression change greater than 1.6 fold (FDR<.05) after 1 nM DHT or 100 nM Dex
`treatment (Supplementary Table 3). Of the 105 AR signature genes and 121 GR signature
`genes, 52 were common to both lists (Figure 5A). An even larger proportion of AR or GR
`signature genes (>80%) showed evidence of regulation by the reciprocal receptor using
`different thresholds for expression differences (Supplementary Table 3). Heatmap analysis
`of these genes confirmed significant overlap in DHT- versus Dex-induced gene expression
`and showed that Dex-induced gene expression is not impacted by enzalutamide treatment
`(Figure 5B). These findings support the hypothesis that GR activity can bypass
`enzalutamide-mediated AR inhibition by regulating a distinct but significantly overlapping
`transcriptome.
`
`We next addressed the question of whether transcriptomes of enzalutamide-resistant tumors
`are more likely to be explained by AR- or GR-driven gene expression using gene set
`enrichment analysis (GSEA). To define gene sets that distinguish AR and GR activity,
`expression of AR and GR signature genes was first evaluated by GSEA in the DHT- and
`Dex-treated samples from which they were derived. As expected, GR signature genes were
`enriched in the Dex-treated samples and AR signature genes were enriched with DHT
`
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Arora et al.
`
`Page 6
`
`treatment (Figure 5C). Because several of the genes did not distinguish AR and GR status
`due to their overlapping transcriptional activities, we refined the lists into AR selective
`genes (defined as the AR induced signature genes that were also more highly expressed in
`DHT treated samples relative to Dex treated samples, n=39) and GR selective genes (defined
`as the converse, n=67) (Supplementary Table 3). GSEA analysis of these selective gene lists
`revealed that GR selective genes were strongly enriched in the enzalutamide-resistant
`LREX’ tumors whereas AR selective genes were strongly enriched in the control tumors
`(Figure 5D). These data provide compelling, unbiased evidence that drug resistance is
`associated with a transition from AR- to GR-driven transcriptional activity.
`
`One prediction of this model is that GR should occupy a substantial portion of AR binding
`sites in drug resistant cells. To address this question, we conducted ChIP-seq experiments to
`define AR and GR DNA binding sites in LREX’ cells after DHT and Dex treatment
`respectively. Of note, 52% of the AR binding sites identified after DHT treatment were
`bound by GR after Dex treatment (Figure 5E). We examined the remaining 48% of AR
`peaks more closely to be sure that these peaks were not scored as GR negative simply
`because they fell just below the threshold set by our peak calling parameters. When we
`plotted the average AR and GR signal as a measure of the relative strength of AR and GR
`peaks, we found little evidence of GR binding at the AR unique sites (Figure S5A),
`confirming that these peaks were indeed unique to AR. Next we conducted motif analysis to
`explore potential differences between AR/GR overlap versus AR unique sites. The core
`ARE/GRE consensus sequence was present in both groups (66% and 68% of peaks) but AR/
`GR overlap peaks were relatively enriched for the FoxA1 motif (64% versus 45% of peaks,
`p=2.2×10-16) (Figure 5E). Similar analysis of the GR cistrome defined GR unique and AR/
`GR overlap peaks and revealed that a higher proportion of GR binding sites were unique to
`GR. Interestingly, GR unique peaks were highly enriched for the FoxA motif (Figure 5F),
`while the classic ARE/GRE was not reported by the motif discovery algorithm (MEME) and
`was found only 25% of the time.
`
`Although these cistrome studies provide evidence of substantial overlap between AR and
`GR binding sites in enzaluamide-resistant cells, several lines of evidence indicate that the
`transcriptional differences in DHT- versus Dex-induced gene expression cannot be
`explained solely by DNA binding. For example, ChIP RT-qPCR experiments showed
`significant AR and GR DNA binding at genes induced by both receptors (SGK1, FKBP5,
`PSA) but also at genes such as NDRG1 that are transcriptionally activated by DHT but not
`Dex (Figure S5B). Integrative ChIP-seq and transcriptome analysis provided further
`evidence that DNA binding is not sufficient to determine transcriptional competence. Of the
`56 AR signature genes found to have an AR binding peak, 49 showed at least some
`transcriptional regulation by GR (1.2 fold expression change, p<.05). 38 of these 49 GR
`regulated genes (78%) had an overlapping AR/GR binding peak, confirming substantial
`overlap at co-regulated genes. But GR peaks were also found in 3 of the 7 AR targets genes
`(43%) with no apparent GR transcriptional regulation (Figure S4C). Others have reported
`evidence of allosteric regulation of hormone receptor complexes by specific DNA sequences
`independent of binding affinity (Meijsing et al., 2009), a phenomenon that may also be
`relevant here.
`
`Activation of GR by dexamethasone is sufficient to confer enzalutamide resistance
`
`Whereas LNCaP/AR cells acquire GR expression after prolonged exposure to enzalutamide,
`some prostate cancer cell lines derived from CRPC patients (DU145, PC3, VCaP) express
`endogenous GR (Figure 6A). DU145 and PC3 cells are AR-negative and hence resistant to
`enzalutamide but VCaP cells are enzalutamide-sensitive in vitro (Tran et al., 2009). IHC
`analysis showed diffuse, primarily cytoplasmic GR expression under standard culture
`
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Arora et al.
`
`Page 7
`
`conditions that lack glucocorticoid supplementation (Figure S6A). To test if GR activation
`by addition of glucocorticoids impacts antiandrogen sensitivity, we treated VCaP cells with
`enzalutamide in the presence or absence of Dex. Enzalutamide inhibited growth as expected,
`but co-treatment with Dex reversed this growth inhibition (Figure 6B). Additional studies
`with the GR antagonist, compound 15, or with GR shRNA restored enzalutamide sensitivity,
`provided pharmacologic and genetic evidence that GR confers resistance (Figure 6C, 6D,
`6E). Of note, GR knockdown (which inhibits GR more completely than compound 15,
`which has mixed agonist/antagonist properties(Wang et al., 2006)) augmented the activity of
`enzalutamide even in the absence of Dex (Figure 6D,F), suggesting that even the weak basal
`GR activity seen under our standard cultures conditions can confer relative resistance to
`enzalutamide. This result also suggests that a pure GR antagonist could enhance the activity
`of enzalutamide in prostate cancers co-expressing GR and AR.
`
`To determine if Dex activates a subset of AR target genes in VCaP (as we observed in the
`LREX’ model), we derived a list of AR target genes in VCaP cells exposed to DHT and
`asked whether Dex could modulate these same AR target genes in the presence of
`enzalutamide. Dex restored expression of some targets (KLK2, FKBP5, HOMER2,
`SLC45A3) but not others (DHCR24, SLC2A3, TRPM8, TMEM79), analogous to the uneven
`restoration we observed in the LNCaP/AR model (Figure 6G). Dex also induced expression
`of the clinical biomarker PSA in these cells, further supporting the hypothesis that GR can
`drive PSA progression in enzalutamide-resistant patients (Figures S6B, C). To confirm that
`Dex activated genes via the glucocorticoid receptor, we evaluated the effect of compound 15
`on Dex induced transcriptional activity. As expected, compound 15 reduced Dex induction
`of the GR targets KLK2 and FKBP5 (Figure 6H). Similarly, GR knock-down prevented
`Dex-mediated induction of target genes (Figure S6C). As in the LREX’ system
`(Supplementary Table 3), the vast majority of genes robustly regulated by GR activation in
`VCaP cells were also regulated by AR activation with DHT (Supplementary Table 4).
`Others have recently shown substantial overlap in the AR and GR cistromes in VCAP as
`well(Sahu et al., 2013). These findings extend our hypothesis that GR promotes
`enzalutamide resistance largely by replacing AR activity at a subset of genes to a second
`model system.
`
`A subset of prostate cancer are primed for GR induction in the setting of AR inhibition
`
`In considering potential mechanisms for increased GR expression in drug-resistant tumors,
`we noted several observations that suggested two distinct models. First, flow cytometry
`analysis of LNCaP/AR and CS1 cells revealed GR expression in a rare subset of cells (Fig
`1E), raising the possibility that these cells clonally expand under the selective pressure of
`antiandrogen therapy. Consistent with this model, we observed rare GR-positive cells in a
`tissue microarray analysis of 59 untreated primary prostate cancers (Supplementary Table
`5). However, we also observed a modest (~2 fold) but significant increase in GR mRNA
`levels in LNCaP/AR xenografts after only 4 days of antiandrogen treatment, reminiscent of
`an older report of increased GR expression in normal ventral rat prostate after castration
`(Davies and Rushmere, 1990). These findings suggest a second model of adaptive resistance
`whereby AR inhibition causes an increase in GR levels due to loss of AR-mediated negative
`feedback.
`
`To investigate the relationship between AR activity and GR expression, we first asked if the
`high level of GR expression in LREX’ tumors is maintained after discontinuation of
`enzalutamide. Remarkably, GR mRNA levels dropped by ~5 fold 8 days after treatment
`discontinuation (Figure 7A). Because enzalutamide has a prolonged half-life in mice (Tran
`et al., 2009), it is difficult to make definitive conclusions about negative feedback loops
`using in vivo models. Therefore, we conducted similar enzalutamide withdrawal
`
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Arora et al.
`
`Page 8
`
`experiments in LREX’ cells cultured in vitro. GR mRNA levels dropped as early as 1 day
`after discontinuation and continued to decline throughout the 23 days of the experiment
`(Figure 7B). Additional experiments with LREX’ cells using earlier timepoints in charcoal
`stripped media showed reduced GR mRNA levels after only 8 hours DHT exposure and this
`reduction was reversed by co-treatment with enzalutamide (Figure 7C). This reduction
`correlated precisely with the recruitment of an AR binding peak in an intronic enhancer of
`GR identified by ChIP, suggesting AR directly represses GR expression in these cells
`(Figure 7D).
`
`To determine if the loss of GR expression upon enzalutamide withdrawal occurs across the
`entire cell population or is restricted to a subset of cells, we conducted flow cytometry
`experiments, where a shift in median signal intensity can be used to identify expression
`changes in the bulk cell population. (Expression changes limited to a minority sub-
`population would not affect the median and would instead be identified as a tail population
`by histogram plot.) We observed an exponential decay in median GR protein signal (half-
`life 7.6 days) (Figure 7E,top row, 7F), confirming that the loss in GR expression occurs
`across the entire LREX’ cell population. Extension of this experiment to later time points
`(17 weeks) revealed a plateau in loss of GR expression by 7 weeks (Figure S7A).
`
`Next we conducted the reciprocal experiment of re-exposure of LREX’ cells to
`enzalutamide following GR downregulation after prolonged enzalutamide withdrawal
`(LREX’off). GR expression was regained with induction kinetics essentially reciprocating
`the rate of decay previously seen with removal of drug (doubling time 6.8 days),
`establishing that the resistant line remained poised for GR induction in the setting of AR
`inhibition (Figure 7E,F). Consistent with the time scale, continued drug exposure for 7
`weeks was associated with a clear shift in GR expression in essentially all cells (Figure
`S7A).
`
`We next determined if AR inhibition is sufficient to induce GR expression in LNCaP/AR or
`CS1 cells that had not previously been exposed to enzalutamide. In contrast to LREX’, there
`was no change in median expression intensity in CS1 or LnCaP/AR over the 4 week
`experiment, indicating that most cells do not turn on GR expression simply as a consequence
`of AR inhibition (Figures 7E, 7F, S7C). However, the area under the GR staining population
`did increase. Given the weak antiproliferative effect of enzalutamide in vitro (Figure S7B),
`we conclude that this increase in GR expression is most likely explained by loss of AR-
`mediated negative feedback rather than by clonal expansion. Together, these findings
`support a model in which a subset of prostate cancer cells are “primed” for GR induction in
`the context of AR inhibition through an adaptive resistance mechanism (via AR-mediated
`negative feedback). We postulate that these cells then clonally expand under the selective
`pressure of AR blockade, eventually emerging as drug-resistant tumors whose expression
`profiles may resemble those of AR-driven tumors but are driven by GR (Figure 7G).
`
`Discussion
`
`Following the recent approvals of the next generation AR pathway inhibitors abiraterone
`and enzalutamide, the treatment of metastatic prostate cancer has evolved to a two-stage
`process. Initially patients receive conventional androgen deprivation therapy, typically with
`a gonadotropin-releasing hormone agonist that lowers testosterone (castration), often in
`conjunction with an anti-androgen such as bicalutamide. Preclinical and clinical studies have
`conclusively demonstrated that acquired resistance to conventional androgen deprivation
`therapy is caused by restoration of AR pathway activation, primarily due to increased AR
`expression. These discoveries provided the rationale for the development of next generation
`AR therapies.
`
`Cell. Author manuscript; available in PMC 2014 June 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Arora et al.
`
`Page 9
`
`Here

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket