throbber
Priority Report
`
`Cancer
`Research
`
`Interactions of Abiraterone, Eplerenone, and Prednisolone
`with Wild-type and Mutant Androgen Receptor: A Rationale
`for Increasing Abiraterone Exposure or Combining
`with MDV3100
`
`Juliet Richards1, Ai Chiin Lim1, Colin W. Hay3, Angela E. Taylor4, Anna Wingate1, Karolina Nowakowska1,
`Carmel Pezaro1,2, Suzanne Carreira1, Jane Goodall1, Wiebke Arlt4, Iain J. McEwan3,
`Johann S. de Bono1,2, and Gerhardt Attard1,2
`
`Abstract
`
`Prostate cancer progression can be associated with androgen receptor (AR) mutations acquired following
`treatment with castration and/or an antiandrogen. Abiraterone, a rationally designed inhibitor of CYP17A1
`recently approved for the treatment of docetaxel-treated castration-resistant prostate cancer (CRPC), is often
`effective, but requires coadministration with glucocorticoids to curtail side effects. Here, we hypothesized that
`progressive disease on abiraterone may occur secondary to glucocorticoid-induced activation of mutated AR. We
`found that prednisolone plasma levels in patients with CRPC were sufficiently high to activate mutant AR.
`Mineralocorticoid receptor antagonists, such as spironolactone and eplerenone that are used to treat side effects
`related to mineralocorticoid excess, can also bind to and activate signaling through wild-type or mutant AR.
`Abiraterone inhibited in vitro proliferation and AR-regulated gene expression of AR-positive prostate cancer cells,
`which could be explained by AR antagonism in addition to inhibition of steroidogenesis. In fact, activation of
`mutant AR by eplerenone was inhibited by MDV3100, bicalutamide, or greater concentrations of abiraterone.
`Therefore, an increase in abiraterone exposure could reverse resistance secondary to activation of AR by residual
`ligands or coadministered drugs. Together, our findings provide a strong rationale for clinical evaluation of
`combined CYP17A1 inhibition and AR antagonism. Cancer Res; 72(9); 2176–82. Ó2012 AACR.
`
`Introduction
`
`The small-molecule CYP17A1 inhibitor, abiraterone acetate
`(Zytiga, Janssen), was recently approved for the treatment of
`men with castration-resistant prostate cancer (CRPC) progres-
`sing after docetaxel chemotherapy. Despite a significant sur-
`vival advantage with 1,000 mg abiraterone daily and objective
`tumor responses in up to 60% of patients with CRPC, progres-
`sive disease on treatment invariably develops (1, 2). MDV3100
`is a novel antiandrogen (3, 4) that has also recently been
`
`Authors' Affiliations: 1Section of Medicine, The Institute of Cancer
`Research; 2The Royal Marsden NHS Foundation Trust, Sutton, Surrey;
`3School of Medical Sciences, University of Aberdeen, Foresterhill, Aberd-
`een; and 4Centre for Endocrinology, Diabetes and Metabolism, School of
`Clinical and Experimental Medicine, University of Birmingham, Birming-
`ham, United Kingdom
`
`Note: Supplementary data for this article are available at Cancer Research
`Online (http://cancerres.aacrjournals.org/).
`
`J.S. de Bono and G. Attard are joint senior authors.
`
`Corresponding Author: Gerhardt Attard, Section of Medicine, The Insti-
`tute of Cancer Research and the Royal Marsden NHS Foundation Trust,
`Downs Road, Sutton, Surrey SM2 5PT, United Kingdom. Phone: 0044-
`7793077493; Fax: 0044-2086427979; E-mail: Gerhardt.attard@icr.ac.uk
`
`doi: 10.1158/0008-5472.CAN-11-3980
`
`Ó2012 American Association for Cancer Research.
`
`reported to confer a survival advantage in patients with CRPC
`progressing after docetaxel (5). As prostate-specific antigen
`(PSA) level often increases at progression on both these agents,
`we have hypothesized that resistance occurs secondary to
`reactivation of androgen receptor (AR) signaling. Inhibition
`of CYP17A1 results in significant suppression of androgens and
`estrogens but also of cortisol that is associated with a com-
`pensatory increase in adrenocorticotropic hormone level (2).
`Abiraterone acetate has therefore been developed in combi-
`nation with exogenous glucocorticoids. However, up to 40% of
`patients on prednisone/prednisolone alone and 55% of
`patients on abiraterone acetate and prednisone/prednisolone
`develop a syndrome of secondary mineralocorticoid excess
`characterized by hypokalemia, hypertension, and fluid over-
`load that can be controlled by increasing the dose of predni-
`sone or adding a mineralocorticoid receptor antagonist (MRA)
`such as eplerenone (1). Eplerenone is currently recommended
`in preference to spironolactone as previous studies showed
`that eplerenone did not bind and activate wild-type (WT)-AR
`(2, 6). However, as eplerenone is not invariably available,
`spironolactone is also being used.
`Point mutations of the AR, which appear to cluster in the
`ligand-binding domain, are rare in therapy naive patients but
`occur in 15% to 45% of castration-resistant disease and can
`increase AR affinity for a wide range of steroids (7, 8). Over 100
`
`2176
`
`Cancer Res; 72(9) May 1, 2012
`
`Amerigen Exhibit 1136
`Amerigen v. Janssen IPR2016-00286
`
`

`
`mutations have been described and many have been shown to
`give a functional advantage to maintain AR signaling. We
`hypothesized that progressive disease on abiraterone acetate
`could occur secondary to activation of mutated "promiscuous"
`AR by steroidal agents administered to patients to prevent or
`treat side effects of mineralocorticoid excess.
`
`Materials and Methods
`
`Materials
`FBS and charcoal-stripped serum (CSS) were purchased
`from Gibco. Bicalutamide, dexamethasone, prednisone, and
`titrated [3H]-
`dihydrotestosterone (DHT; Sigma-Aldrich),
`R1881 (Perkin-Elmer), R1881 (Steraloids), eplerenone and spir-
`onolactone (Tocris-Bioscience) were obtained from commer-
`cial sources. Abiraterone and MDV3100 were synthesized using
`the publicly available chemical structures and checked by mass
`spectrometry. Drugs were dissolved in dimethyl sulfoxide
`(DMSO) and then diluted to a maximum DMSO concentration
`of 0.2%. LNCaP, VCaP, PC-3, DU145, and COS-7 cells were
`obtained from American Type Culture Collection (ATCC; LGC
`Standards), grown according to ATCC recommendations, used
`less than 6 months from receipt and freeze down and con-
`firmed mycoplasma free.
`
`Luciferase reporter assays
`We constructed a PSA-ARE3-luc luciferase reporter plasmid
`that was cotransfected with a human AR expression plasmid,
`F527-AR [wild-type or mutant as stated; mutations confirmed
`by sequencing (Beckman Coulter Genomics)] into PC-3 cells.
`These were seeded in white opaque 384-well plates and grown
`in 10% CSS-supplemented phenol red–free RPMI-1640 for 30
`hours. Cells were then treated with the indicated concentra-
`tion of compound and R1881 for 16 hours. Luciferase activity
`was determined by adding ONE Glo (Promega) and measuring
`luminescence on a TopCount plate reader (Perkin-Elmer).
`Transfection efficiency and protein expression are shown in
`Supplementary Fig. S1.
`
`Cell viability
`LNCaP and VCaP cells were seeded in 96-well plates and
`grown in CSS-supplemented phenol red–free or FBS-supple-
`mented media for 7 days. Cells were treated with compound at
`24 and 96 hours after plating and cell viability was determined
`on day 7 by adding CellTiter Glo (Promega) and measuring
`luminescence.
`
`Ligand-binding assay
`PC-3 cells transfected with wild-type or T877A mutant AR or
`LNCaP cells were seeded in 24-well plates and grown in CSS-
`supplemented phenol red–free media for 24 hours. To deter-
`mine the kinetics of [3H]-R1881 binding to the wild-type and
`T877A AR, cells were treated with 0.25 to 25 nmol/L [3H]-R1881
`for 2 hours, then washed, lysed, and radioactivity was measured
`(1900CA analyzer, Perkin-Elmer). The Kd and Bmax were deter-
`mined by nonlinear regression using GraphPad Prism soft-
`ware. When the concentration of [3H]-R1881 required to
`almost saturate AR in both wild-type and T877A AR mutant
`
`Interaction of AR with Abiraterone and Eplerenone
`
`transfections was established (5 nmol/l), displacement of [3H]-
`R1881 by test compound was determined. The concentration
`at which 50% of [3H]-R1881 was displaced (EC50) was estab-
`lished using nonlinear regression (GraphPad Prism).
`
`Quantitative real time-PCR
`LNCaP and VCaP cells were seeded in 6-well plates and
`grown in CSS-supplemented phenol red–free media for 24
`hours and then treated for 5 hours as indicated. Following
`RNA extraction and cDNA synthesis, quantitative PCR
`(qPCR) was carried out on the Mx3000P QPCR System
`(Agilent) using the RT2 SYBR Green ROX qPCR Mastermix
`(SABiosciences). Every sample was run in duplicate and each
`reaction contained 50 ng of cDNA in a total volume of 20 mL.
`DCt for each gene was determined after normalization to
`actin
`and
`glyceraldehyde-3-phosphate
`dehydrogenase
`(GAPDH), and DDCt was calculated relative to the designat-
`ed reference sample. Gene expression values were set equal
`to 2DDCt (Applied Biosystems). Primers were purchased
`from SABiosciences.
`
`Measurement of plasma prednisolone
`Plasma was collected from patients with CRPC after 48 days
`of continuous daily abiraterone acetate and prednisolone. All
`patients provided written, informed consent to blood with-
`drawal for research purposes, and this study was approved by
`the Royal Marsden Hospital ethics review committees. Pred-
`nisolone was quantified by comparison to a calibration series
`ranging from 5 to 500 ng/mL prepared in 50:50 methanol:water.
`A Waters Xevo mass spectrometer with Acquity uPLC system
`was used, fitted with a HSS T3, 1.8 mm, 1.2  50 mm2 column
`(Waters). The column temperature was maintained at 60C
`and the settings used were an electrospray source in positive
`ionization mode; capillary voltage 4.0 kV; source temperature,
`150C; and desolvation temperature, 500C.
`
`Results
`
`The selective mineralocorticoid receptor antagonist,
`eplerenone, activates mutant AR
`We first cotransfected PC-3 AR-negative prostate cancer
`cells with PSA-ARE2-luc and either wild-type (WT)-AR or 3
`mutations previously described in CRPC (T877A-AR, D879G-
`AR, and W741C-AR). The T877A mutation has been identified
`in several studies in patients treated with flutamide (8, 9) and
`has been extensively studied as it is found in the LNCaP
`prostate cancer cell line (Supplementary Table S1). D879G
`and W741C mutations have been identified in patients previ-
`ously treated with bicalutamide (8, 9). We then compared
`activation of wild-type or mutant AR by synthetic androgen
`(R1881) to activation by the MRAs, eplerenone, and spirono-
`lactone. In keeping with previous reports, spironolactone
`activates WT-AR (7) and also T877A-AR, D879G-AR, and
`W741C-AR only 2-log less potently than R1881 does (Fig. 1A
`and B and Supplementary Fig. S2). Eplerenone does not
`activate WT-AR, D879G-AR, or W741C-AR but importantly
`can activate T877A-AR with a dose-proportional response
`and an EC50 value of 5.2 mmol/L [95% confidence interval (CI),
`
`www.aacrjournals.org
`
`Cancer Res; 72(9) May 1, 2012
`
`2177
`
`

`
`Richards et al.
`
`Figure 1. Eplerenone activates T877A-AR and spironolactone activates both T877A-AR and wild-type (WT)-AR. Sigmoidal dose–response curves show
`activation of WT-AR by R1881 and spironolactone (A) and T877A-AR by R1881, spironolactone, eplerenone, prednisolone, dexamethasone. (B) Fold
`change from the DMSO control was plotted and EC50 values calculated using nonlinear regression (GraphPad). EC50 values and 95% CIs are given. C, LNCaP
`and VCaP prostate cancer cells in CSS were treated with eplerenone or spironolactone alone or in combination with 0.1, 1, or 5 mmol/L abiraterone, 10 mmol/L
`bicalutamide, or 10 mmol/L MDV3100 for 7 days and then analyzed for cell viability. Fold change from the DMSO control was then calculated and plotted.
`Significance is shown for stimulation by eplerenone or spironolactone compared with DMSO control ( , horizontal) and for inhibition by bicalutamide,
`MDV3100, or abiraterone when compared with stimulated levels ( , vertical). D, LNCaP and VCaP cells were treated with 0.1 nmol/L R1881 or 0.1 to 10 mmol/L
`eplerenone for 5 hours. RNA was extracted and cDNA synthesized for analysis by qPCR to determine relative levels of PSA and TMPRSS2 mRNA expression.
`Significance compared with DMSO controls is shown. Data shown for all experiments are the mean (error bars, SEM) of 3 independent experiments of 16
`replicates (A and B) or in duplicate (C and D). , P < 0.05; , P < 0.01; , P < 0.001, one-way ANOVA with the Bonferroni correction.
`
`2.89–9.37 mmol/L; Fig. 1A and B and Supplementary Fig. S2).
`Pharmacokinetic studies with eplerenone report a Cmax of 1.72
` 0.28 mg/mL (equivalent to 4.2  0.7 mmol/L) and a half-life of
`3 hours with 100 mg eplerenone (6); doses of eplerenone
`between 50 and 200 mg are used to treat toxicities secondary
`to mineralocorticoid excess from abiraterone in patients with
`CRPC (Supplementary Table S2). We proceeded to confirm
`that both spironolactone and eplerenone (1 and 10 mmol/L)
`increased proliferation of hormone-stripped LNCaP (T877A-
`AR) but only spironolactone increased the proliferation of
`VCaP (WT-AR; Fig. 1C). The increase in proliferation was
`inhibited by AR antagonism, suggesting this effect was sec-
`ondary to binding to and activation of the AR (Fig. 1C).
`Similarly, eplerenone significantly increased expression of the
`androgen-regulated and clinically important genes PSA and
`TMPRSS2 in LNCaP but not in VCaP (Fig. 1D).
`
`Exogenous glucocorticoids can activate mutant AR at
`clinically relevant doses observed in CRPC patients
`treated with abiraterone acetate
`Prednisolone or its precursor prednisone are commonly
`administered in combination with abiraterone acetate
`although 2 phase II studies combined abiraterone acetate with
`dexamethasone (2, 10). Prednisone and dexamethasone do not
`activate WT-AR but activate T877A-AR with EC50 values of 25.1
`mmol/L (95% CI, 12.64–36.83 mmol/L) and 21.6 mmol/L (95% CI,
`12.53–50.26 mmol/L), respectively (Fig. 1A and B). Previous
`reports have shown that other AR mutations such as T877A in
`combination with L701H are highly sensitive to glucocorticoids
`with activation by concentrations as low as 10 nmol/L (11). We
`therefore proceeded to measure plasma levels of prednisolone
`in 15 patients with CRPC on continuous daily treatment with
`1,000 mg abiraterone acetate and 10 mg prednisolone.
`
`2178
`
`Cancer Res; 72(9) May 1, 2012
`
`Cancer Research
`
`

`
`Interaction of AR with Abiraterone and Eplerenone
`
`to confirm downregulation by qPCR of PSA and TMPRSS2 in
`LNCaP cells treated with abiraterone (Fig. 3D).
`
`Binding of abiraterone or eplerenone to the AR is
`confirmed by competitive displacement of [3H]-R1881
`To confirm that AR antagonism by abiraterone and agon-
`ism by eplerenone (both previously undescribed) occurred
`secondary to binding to the AR ligand–binding domain, we
`used a competitive radiolabeled assay to show displacement
`of R1881 from PC-3 cells transfected with either WT-AR or
`T877A-AR. The EC50 value of eplerenone for WT-AR was
`6-fold higher than T877A-AR (EC50, 2.4 mmol/l; 95% CI, 2.0–
`2.9 mmol/L; Fig. 4A and B). In keeping with the inhibitory
`activity of abiraterone observed in our reporter luciferase
`studies, abiraterone displaced ligand from both WT-AR
`(EC50, 13.4 mmol/L; 95% CI, 10.3–17.4 mmol/L) and T877A
`(EC50, 7.9 mmol/L; 95% CI, 6.7–9.3 mmol/L; Fig. 4A and B). We
`also confirmed displacement of radiolabeled R1881 from
`LNCaP with abiraterone (EC50, 2.6 mmol/L; 95% CI, 1.0–6.8
`mmol/L) and eplerenone (EC50, 4.3 mmol/L; 95% CI, 2.4–7.8
`mmol/L; Supplementary Fig. S5).
`
`Mutant AR activation by eplerenone can be inhibited by
`abiraterone or bicalutamide but most effectively by
`MDV3100
`We observed dose-proportional growth inhibition with
`abiraterone of LNCaP cells stimulated by eplerenone and of
`LNCaP and VCaP cells stimulated by spironolactone (Fig. 1C).
`Similar levels of inhibition were observed with bicalutamide,
`with more profound inhibition by MDV3100 (Fig. 1C). Abir-
`aterone, MDV3100, and bicalutamide achieved similar levels
`of
`inhibition of upregulation of PSA by eplerenone but
`MDV3100 inhibited induction of TMPRSS2 expression more
`significantly than bicalutamide or abiraterone (Fig. 3D). Sim-
`ilarly, MDV3100 showed more significant inhibition of spir-
`onolactone-stimulated PSA and TMPRSS2 expressions than
`abiraterone or bicalutamide (Supplementary Fig. S6). Also,
`abiraterone (5 mmol/L) significantly inhibited activation of
`T877A-AR (in transfected PC-3) by 1 mmol/L eplerenone but
`not by 10 mmol/L eplerenone; stimulation by 10 mmol/L
`eplerenone was significantly inhibited by both bicalutamide
`and MDV3100 (Fig. 4C).
`
`Increased hormone levels reduce AR inhibition by
`MDV3100
`Recent studies have suggested that intratumoral testoster-
`one levels increase in patients treated with MDV3100 (12). We
`found that 1mmol/L and 10 mmol/L MDV3100 significantly
`inhibited WT-AR luciferase activity stimulated by 0.1 nmol/L
`R1881 or 1 nmol/L DHT, respectively, but 50mmol/L
`MDV3100 was required to significantly inhibit AR stimulated
`by 1 nmol/L R1881 (Fig. 4D) or 10 nmol/L DHT (Supplementary
`Fig. S7).
`
`Discussion
`
`Abiraterone was developed as a specific CYP17A1 inhib-
`itor (13). Previous studies have failed to identify binding of
`
`Figure 2. Plasma concentrations (nmol/L) of prednisolone in 15 patients
`with CRPC treated with abiraterone acetate measured with liquid
`chromatography/tandem mass spectrometry. The median concentration
`of 152 nmol/L (SD, 100 nmol/L) is marked by the solid line. The 10 nmol/L
`limit above which activation of T877A-L701H-AR has been previously
`reported to occur is shown by the dashed line.
`
`Prednisolone levels were less than 4 nmol/L in 2 patients but
`more than 30 nmol/L in the other 13 patients. The median
`concentration was 153 nmol/L (range, <4–305 nmol/L; Fig. 2
`and Supplementary Table S2).
`
`Abiraterone binds and inhibits wild-type and mutant AR
`Following the observation of activation of T877A-AR
`by eplerenone, we proceeded to evaluate the effect of abirater-
`one on wild-type and mutant AR (T877A, D879G, R629Q,
`W741C, and M749L). We did not observe an increase in
`reporter luciferase activity with doses of abiraterone up to
`25 mmol/L with WT-AR or any mutation tested (Supplemen-
`tary Fig. S3) but observed dose-proportional inhibition of
`stimulated wild-type and mutant AR activity (Fig. 3A) with
`significant inhibition observed at doses 10 mmol/L. Inhibi-
`tion was however not as potent as for same concentrations of
`MDV3100. We then proceeded to confirm our findings by
`comparing inhibition of AR activation using abiraterone or
`MDV3100 in a different model system (COS-7 cells cotrans-
`fected with AR and a GRE2-TATA-luc reporter gene and
`activated by 10 nmol/L DHT for 24 hours). Similarly we
`observed dose-proportional inhibition of WT-AR, T877A-AR,
`G142V-AR, P533S-AR, T575A-AR, and H874Y-AR by abirater-
`one (Fig. 3B). Higher concentrations of abiraterone were
`required for inhibition of R629Q-AR in this system than was
`observed in PC-3 cells transfected with an ARE3-luc assay (Fig.
`3A). We also confirmed significant inhibition of proliferation of
`the AR-positive prostate cancer cell lines LNCaP and VCaP
`with doses of abiraterone 1 mmol/L (Fig. 3C). No inhibitory
`effect was observed with the AR-negative prostate cancer cell
`lines, PC-3, and DU145 (Supplementary Fig. S4). We proceeded
`
`www.aacrjournals.org
`
`Cancer Res; 72(9) May 1, 2012
`
`2179
`
`

`
`Richards et al.
`
`Inhibition of wild-type and mutant stimulated AR activity by abiraterone, bicalutamide, and MDV3100. A, PC-3 cells were cotransfected with ARE3-luc
`Figure 3.
`and wild-type or mutant AR (T877A, D879G, W741C, M749L, and R629Q). Cells were treated with 0.1 to 25 mmol/L abiraterone, 10 mmol/L bicalutamide, or 10
`mmol/L MDV3100 in CSS medium containing 0.1 nmol/L R1881 for 16 hours and then analyzed for luciferase activity. Fold change from the DMSO control was
`calculated and then percentage change relative to the R1881-stimulated DMSO control was determined. Data shown are representative of 3 independent
`experiments and represent mean and SEM of 8 replicates. B, COS-7 cells were cotransfected with GRE2-TATA-luc and the wild-type or mutant human
`expression plasmid pSVARo (T877A, G142V, P533S, T575A, H874Y, R629Q). Cells were treated with 0.1 to 5 mmol/L abiraterone or MDV3100 in CSS medium
`containing 10 nmol/L DHT for 24 hours. The luciferase activities were assayed in duplicate and normalized for the amounts of expressed AR determined
`immunologically by dot blot analysis and normalized for protein concentration. The change in normalized luciferase activity relative to cells incubated
`without any compound for each AR variant was determined. Data shown represent 2 or 3 independent experiments carried out in quadruplicate.
`C, dose-proportional inhibition of proliferation of LNCaP and VCaP cells by abiraterone, MDV3100, and bicalutamide. LNCaP and VCaP prostate cancer cells
`in FBS were treated with 0.1, 1, 5 or 10 mmol/L abiraterone, 0.1 or 10 mmol/L bicalutamide, or 0.1 or 10 mmol/L MDV3100 for 7 days and then analyzed
`for cell viability. Fold change from the DMSO control was then calculated and plotted. Data shown are the mean (error bars, SEM) of 3 independent experiments
`in quadruplicate. D, LNCaP cells were treated with 0.1 nmol/L R1881 or 10 mmol/L eplerenone in combination with DMSO, 10 mmol/L bicalutamide, 10 mmol/L
`MDV3100, or 5 mmol/L abiraterone for 5 hours. RNA was extracted and cDNA synthesized for analysis by qPCR to determine relative levels of PSA and
`TMPRSS2 mRNA expression. Data shown are the mean and SEM of 3 independent experiments in duplicate. Significance is shown for , P < 0.05;
` , P < 0.01; , P < 0.001; , P < 0.0001 relative to DMSO control (one-way ANOVA with the Bonferroni correction).
`
`abiraterone to the AR (14). However, in this study we used
`both reporter luciferase and competitive radiolabeled assays
`to show that abiraterone binds and inhibits WT-AR. Another
`study published while our article was under review reported
`supporting evidence that abiraterone binds the AR and
`produces a dose-dependent decrease in AR levels (15). This
`study failed to identify the EC50 value with wild-type
`or mutant AR but predicted it as over 3 mmol/L. We
`also tested 8 AR mutations selected from a screen of 42
`mutations for causing a differential response to various
`hormones. We included mutations in the amino terminal
`(G142V, P533S), DNA-binding (T575A), and ligand-binding
`(W741C, M749L, T877A, D879G, and H874Y) domains and
`
`the hinge region (R629Q; Supplementary Table S1). As
`previously
`described,
`bicalutamide
`activated W741C
`(4, 16) but no agonistic activity was observed with any
`mutation and abiraterone. Similarly MDV3100 potently
`inhibited WT-AR and all mutant AR tested. However, these
`mutations were mostly identified in patients progressing on
`bicalutamide or flutamide and different, new mutations may
`develop in patients progressing on abiraterone or MDV3100.
`Abiraterone is an active treatment for CRPC due to CYP17A1
`inhibition and significant suppression of hormones (2). How-
`ever, we observed up to 32% AR inhibition with 1 mmol/L
`abiraterone, with significantly greater inhibition at 5 and 10
`mmol/L. Pharmacokinetic studies have reported maximum
`
`2180
`
`Cancer Res; 72(9) May 1, 2012
`
`Cancer Research
`
`

`
`Interaction of AR with Abiraterone and Eplerenone
`
`Figure 4. Displacement of [3H] R1881 by eplerenone and abiraterone in PC-3 cells transfected with wild-type or T877A mutant AR. PC-3 cells were transfected
`with WT-AR (A) or T877A mutant AR (B) and then treated with CSS media containing 5 nmol/L of [3H] R1881 in combination with cold R1881, DHT,
`eplerenone, abiraterone, or bicalutamide at the concentrations shown for 2 hours. Abiraterone was insoluble in cell media at concentrations greater than 25
`mmol/L. Cell-associated radioactivity was measured and the data analyzed by nonlinear regression to determine the EC50 value for each test compound
`(GraphPad Prism). Data shown are the mean and SEM of 3 independent experiments in triplicate for percentage [3H]-R1881 bound versus log10 of
`concentration (mmol/L) of cold competitor. EC50 and 95% CI values are given. C, inhibition of eplerenone-stimulated AR activation by bicalutamide, MDV3100,
`and abiraterone. PC-3 cells were cotransfected with ARE3-luc and T877A mutant AR. Cells were treated with DMSO (control) or eplerenone in combination
`with DMSO, 10 mmol/L bicalutamide, 10 mmol/L MDV3100, or 5 mmol/L abiraterone for 16 hours and then analyzed for luciferase activity. Fold change
`from the DMSO control was calculated. Data shown are from 3 independent experiments and represent mean and SEM of 13 replicates. D, increased
`hormone levels reduce AR inhibition by MDV3100. PC-3 cells were cotransfected with ARE3-luc and WT-AR. Cells were treated with R1881 in combination
`with DMSO or MDV3100 at the concentrations indicated for 16 hours and then analyzed for luciferase activity. Fold change from the DMSO control
`was calculated. Data shown are from 3 independent experiments and represent mean and SEM of 24 replicates. , P < 0.01 relative to R1881
`or DHT control with DMSO (one-way ANOVA with the Bonferroni correction).
`
`plasma levels after a single 1,000 mg dose of abiraterone acetate
`in fasting patients of 1.2 to 5 mmol/L, confirming AR antag-
`onism could occur at clinically achievable doses (Supplemen-
`tary Table S2; refs. 2, 17). Higher doses of abiraterone up to at
`least 2,000 mg daily are safely tolerated (2) and greater activity
`could be observed with increased drug exposure despite
`complete CYP17A1 inhibition at lower doses. This could be
`achieved by administration with food (2, 17). Moreover, several
`studies of abiraterone have now reported preclinical in vitro
`and in vivo antitumor activity and inhibition of AR nuclear
`localization and AR-regulated transcription that was attribut-
`ed entirely to inhibition of steroidogenesis (18, 19) but could in
`fact be partly explained by AR antagonism. Similarly, in vitro
`inhibition of LNCaP and VCaP cells in our study could also be
`explained by abiraterone's effect on steroidogenesis.
`Significant activation of both wild-type and mutant AR is
`observed with spironolactone that should be avoided in all
`patients with CRPC. We also show activation of T877A-AR by
`
`eplerenone that was developed as a novel, non–AR-binding
`MRA (6). This could underlie clinical resistance in a proportion
`of patients. Similarly, exogenous glucocorticoids that are
`currently administered in combination with abiraterone
`reach levels in patients that have been previously shown to
`activate mutant L701H T877A AR (11). Activation of "promis-
`cuous" AR by coadministered drugs or residual hormones (as
`we reported recently ref. 20) could be inhibited by increasing
`the dose of abiraterone or possibly more effectively, combining
`with a potent antiandrogen such as MDV3100. Because of
`toxicity the dose of MDV3100 selected for phase III develop-
`ment was 160 mg daily that achieves median plasma concen-
`trations up to approximately 35 mmol/L (3, 4). AR inhibition at
`these concentrations could be overcome by an increase in
`hormones that would be prevented by combination with
`abiraterone acetate. Overall these observations provide a
`strong rationale for clinical evaluation of combined CYP17A1
`inhibition and AR antagonism.
`
`www.aacrjournals.org
`
`Cancer Res; 72(9) May 1, 2012
`
`2181
`
`

`
`Richards et al.
`
`Disclosure of Potential Conflicts of Interest
`Abiraterone acetate was developed at The Institute of Cancer Research, which
`therefore has a commercial interest in the development of this agent. J.S. de Bono
`has received consulting fees from Ortho Biotech Oncology Research and Devel-
`opment (a unit of Cougar Biotechnology), consulting fees and travel support
`from Amgen, Astellas, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb,
`Dendreon, Enzon, Exelixis, Genentech, GlaxoSmithKline, Medivation, Merck,
`Novartis, Pfizer, Roche, Sanofi-Aventis, Supergen, and Takeda, and grant support
`from AstraZeneca. G. Attard has received consulting fees from Janssen-Cilag,
`Veridex and Millennium Pharmaceuticals, lecture fees from Janssen-Cilag, Ipsen
`and Sanofi-Aventis, and grant support from AstraZeneca. G. Attard is on The ICR
`rewards to inventors list of abiraterone acetate. No potential conflicts of interest
`were disclosed by the other authors.
`
`Acknowledgments
`The authors thank Elaine Barrie in the Centre for Cancer Therapeutics, The
`Institute of Cancer Research, Sutton, Surrey for helpful discussions on exper-
`imental design; Elizabeth Folkerd and Mitch Dowsett in the Royal Marsden
`Hospital Academic Biochemistry Department, Fulham Road, London for assis-
`tance with radiolabeled ligand-binding studies; and Penny Flohr, Cancer Bio-
`
`markers, The Institute of Cancer Research, Sutton, Surrey for assistance with
`processing clinical samples.
`
`Grant Support
`The Institute of Cancer Research authors are employees of the Section of
`Medicine that is supported by a Cancer Research UK programme grant and
`an Experimental Cancer Medical Centre (ECMC) grant from Cancer Research
`UK and the Department of Health (ref: C51/A7401). G. Attard is also
`supported by an NIHR clinical lectureship, a Welcome Trust Starter Grant
`for Clinical Lecturers and a young investigator award from the Prostate
`Cancer Foundation, Santa Monica, CA. J. Richards and A. Wingate were
`supported by Prostate Action, London, UK. W. Arlt is in receipt of a Medical
`Research Council (MRC) UK Strategic Biomarker Grant (G0801473). C.W. Hay
`was supported by the Chief Scientist's Office, Scottish Government. The
`authors also acknowledge NHS funding to the RMH NIHR Biomedical
`Research Centre.
`
`Received December 7, 2011; revised January 30, 2012; accepted February 22,
`2012; published OnlineFirst March 12, 2012.
`
`References
`
`4.
`
`1. de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al.
`Abiraterone and increased survival in metastatic prostate cancer. N
`Engl J Med 2011;364:1995–2005.
`2. Attard G, Reid AH, Yap TA, Raynaud F, Dowsett M, Settatree S, et al.
`Phase I clinical trial of a selective inhibitor of CYP17, abiraterone
`acetate, confirms that castration-resistant prostate cancer commonly
`remains hormone driven. J Clin Oncol 2008;26:4563–71.
`3. Scher HI, Beer TM, Higano CS, Anand A, Taplin M-E, Efstathiou E, et al.
`Antitumour activity of MDV3100 in castration-resistant prostate can-
`cer: a phase 1–2 study. Lancet 2010;375:1437–46.
`Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, et al. Devel-
`opment of a second-generation antiandrogen for
`treatment of
`advanced prostate cancer. Science 2009;324:787–90.
`5. Scher H, Fizazi K, Saad F, Taplin ME, Sternberg C, Miller K, et al. Effect
`of MDV3100, an androgen receptor signaling inhibitor (ARSI), on
`overall survival in patients with prostate cancer postdocetaxel: results
`from the phase III AFFIRM study. J Clin Oncol 30, 2012 (suppl 5; abstr
`LBA1).
`6. Ravis WR, Reid S, Sica DA, Tolbert DS. Pharmacokinetics of epler-
`enone after single and multiple dosing in subjects with and without
`renal impairment. J Clin Pharmacol 2005;45:810–21.
`Luthy IA, Begin DJ, Labrie F. Androgenic activity of synthetic proges-
`tins and spironolactone in androgen-sensitive mouse mammary car-
`cinoma (Shionogi) cells in culture. J Steroid Biochem 1988;31:845–52.
`Taplin ME, Bubley GJ, Shuster TD, Frantz ME, Spooner AE, Ogata GK,
`et al. Mutation of the androgen-receptor gene in metastatic androgen-
`independent prostate cancer. N Engl J Med 1995;332:1393–8.
`Taplin M-E, Rajeshkumar B, Halabi S, Werner CP, Woda BA, Picus J,
`et al. Androgen receptor mutations in androgen-independent prostate
`cancer: Cancer and Leukemia Group B Study 9663. J Clin Oncol
`2003;21:2673–8.
`10. Reid AH, Attard G, Danila DC, Oommen NB, Olmos D, Fong PC, et al.
`Significant and sustained antitumor activity in post-docetaxel, cas-
`tration-resistant prostate cancer with the CYP17 inhibitor abiraterone
`acetate. J Clin Oncol 2010;28:1489–95.
`11. Zhao XY, Malloy PJ, Krishnan AV, Swami S, Navone NM, Peehl DM,
`et al. Glucocorticoids can promote androgen-independent growth of
`prostate cancer cells through a mutated androgen receptor. Nat Med
`2000;6:703–6.
`
`12. Efstathiou E, Titus MA, Tsavachidou A, Hoang A, Karlou M, Wen S,
`et al. MDV3100 effects on androgen receptor (AR) signaling and bone
`marrow testosterone concentration modulation: a preliminary report. J
`Clin Oncol 29: 2011 (suppl; abstr 4501).
`13. Potter GA, Barrie SE, Jarman M, Rowlands MG. Novel steroidal
`inhibitors of human cytochrome P45017 alpha (17 alpha-hydroxy-
`lase-C17,20-lyase): potential agents for the treatment of prostatic
`cancer. J Med Chem 1995;38:2463–71.
`14. Handratta VD, Vasaitis TS, Njar VCO, Gediya LK, Kataria R, Chopra P,
`et al. Novel C-17-heteroaryl steroidal CYP17 inhibitors/antiandrogens:
`synthesis, in vitro biological activity, pharmacokinetics, and antitumor
`activity in the LAPC4 human prostate cancer xenograft model. J Med
`Chem 2005;48:29

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