throbber
Biochemical and Biophysical Research Communications 477 (2016) 1005e1010
`
`Contents lists available at ScienceDirect
`
`Biochemical and Biophysical Research Communications
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / y b b r c
`
`Specificity of anti-prostate cancer CYP17A1 inhibitors on androgen
`biosynthesis
`
`Sameer S. Udhane a, b, Bernhard Dick b, c, Qingzhong Hu d, 1, Rolf W. Hartmann d, e,
`Amit V. Pandey a, b, *
`
`a Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, University Children’s Hospital Bern, 3010 Bern, Switzerland
`b Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
`c Department of Nephrology, Hypertension and Clinical Pharmacology, University Hospital of Bern, Bern, Switzerland
`d Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, Saarbrücken, Germany
`e Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E 8.1, 66123 Saarbrücken, Germany
`
`a r t i c l e i n f o
`
`a b s t r a c t
`
`Article history:
`Received 26 June 2016
`Accepted 4 July 2016
`Available online 6 July 2016
`
`Keywords:
`CYP17A1
`Orteronel
`Abiraterone
`CYP21A2
`PCOS
`Premature adrenarche
`Castration resistant prostate cancer
`
`The orteronel, abiraterone and galeterone, which were developed to treat castration resistant prostate
`cancer, inhibit 17,20 lyase activity but little is known about their effects on adrenal androgen biosyn-
`thesis. We studied the effect of several inhibitors and found that orteronel was selective towards 17,20
`lyase activity than abiraterone and galeterone. Gene expression analysis showed that galeterone altered
`the expression of HSD3B2 but orteronel did not change the expression of HSD3B2, CYP17A1 and AKR1C3.
`The CYP19A1 activity was not inhibited except by compound IV which lowered activity by 23%. Sur-
`prisingly abiraterone caused complete blockade of CYP21A2 activity. Analysis of steroid metabolome by
`gas chromatography e mass spectrometry revealed changes in steroid levels caused by different in-
`hibitors. We can conclude that orteronel is a highly specific inhibitor of 17,20 lyase activity. The discovery
`of these specific drug actions on steroidogenic enzyme activities would be valuable for understanding
`the regulation of androgens.
`
`© 2016 Elsevier Inc. All rights reserved.
`
`1. Introduction
`
`Androgens are essential for sexual differentiation and repro-
`duction in both male and female. Androgen production in humans
`occurs mainly in the testis, ovaries and the adrenal glands. The
`initial steps of steroidogenesis in both the adrenal and the gonads
`are same and use similar enzymes for steroid biosynthesis path-
`ways [1]. Both the adrenal and the ovary produce dehydroepian-
`drosterone (DHEA) as the key precursor of androgens and
`estrogens. Premature adrenarche and polycystic ovary syndrome
`(PCOS) are common hyperandrogenic disorders. During adrenarche
`(functional activation of adrenal zona reticularis) production of
`adrenal androgens (DHEA and DHEA sulfate) is at a higher level [1].
`
`* Corresponding author. Pediatric Endocrinology, Diabetology & Metabolism,
`KIKL C837, University Children’s Hospital Bern, Freiburgstrasse 15, CH-3010 Bern,
`Switzerland.
`E-mail address: amit@pandeylab.org (A.V. Pandey).
`1 Current address: Department of Chemistry, University of Cambridge, Lensfield
`Road, Cambridge, CB2 1EW, U.K.
`
`http://dx.doi.org/10.1016/j.bbrc.2016.07.019
`0006-291X/© 2016 Elsevier Inc. All rights reserved.
`
`In case of premature adrenarche, the concentration of circulating
`adrenal androgens increases at an earlier age. Several studies link
`premature adrenarche with post pubertal adrenal and ovarian
`hyperandrogenism [2,3]. Premature adrenarche is also linked to the
`hyperandrogenism in PCOS [4]. In hyperandrogenic women with
`PCOS, androgen production from both the ovaries and the adrenals
`is higher [5]. Changes in cytochrome P450 17A1 (P450c17,
`CYP17A1) activities could explain the increase in androgen secre-
`tion that causes hyperandrogenic disorders [1].
`The CYP17A1 plays a vital role in regulating adrenal androgen
`production (Suppl Fig. 1). CYP17A1 is localized in the endoplasmic
`reticulum and responsible for 17a-hydroxylase and 17,20 lyase re-
`actions. The 17,20 lyase activity regulates androgen production and
`is dependent on cytochrome P450 oxidoreductase (POR) [6], cyto-
`chrome b5 (CYB5) [7] and phosphorylation [7,8]. Treatment of
`androgen dependent prostate cancer is by androgen deprivation
`therapy, androgen receptor (AR) antagonists or anti androgen
`agents and CYP17A1 inhibitors that block androgen production [9].
`Hormonal therapy such as using a gonadotrophin hormonal
`analogue for treatment of hyperandrogenic disorders may not be
`
`Amerigen Exhibit 1107
`Amerigen v. Janssen IPR2016-00286
`
`

`
`1006
`
`S.S. Udhane et al. / Biochemical and Biophysical Research Communications 477 (2016) 1005e1010
`
`specific enough to control the extra-gonadal and the gonadal
`androgen production. Drugs targeting androgen production are
`being used for treatment of prostate cancer and to decrease the
`androgen levels in hyperandrogenic women [10,11]. But anti
`androgen treatment can have side effects on sexuality of women
`with hyperandrogenism [12]. One example is flutamide, a non-
`steroidal anti androgen drug used to treat hyperandrogenic
`women [13e16]. Flutamide has many side effects like diarrhea,
`gynecomastia, muscle cramps, hematological alterations, and
`hepatotoxicity [17,18].
`CYP17A1 inhibitors are designed for the treatment of the
`androgen dependent prostate cancer patients (Suppl Fig. 2). Abir-
`aterone prolonged the survival of castration-resistant prostate
`cancer (CRPC) patients in phase III clinical trials [19]. Abiraterone
`also has modest affinity towards the androgen receptor [20]. In the
`hyperandrogenic disorders like premature or exaggerated adre-
`narche and PCOS, use of total CYP17A1 inhibitors will also block the
`17a-hydroxylase activity. New inhibitors of CYP17A1, orteronel and
`galeterone were also designed for treatment of castration-resistant
`prostate cancer [21,22]. Effect of abiraterone, orteronel and gale-
`terone on adrenal androgen production is not clear.
`Here we are reporting the effects of CYP17A1 inhibitors on the
`adrenal androgen production. We compared CYP17A1 inhibitors
`orteronel, galeterone, abiraterone and recently reported com-
`pounds I and IV and studied the specificity of their action. We found
`that orteronel is a potent inhibitor of 17,20 lyase activity with no
`impact on other cytochrome P450 enzymes involved in steroid
`hormone biosynthesis. We also analyzed the impact of these in-
`hibitors on the steroid metabolome, focusing on C19 steroids. Use
`of specific 17,20 lyase inhibitors can be a treatment option for
`hyperandrogenic disorders for the control of androgen production.
`
`2. Materials and methods
`
`2.1. Cell culture and treatment
`
`Human adrenocortical NCI-H295R cells obtained from American
`Type Culture Collection (ATCC; CRL-2128) were cultivated in
`DMEM/Ham’s F-12 medium with additives as described in
`supplementary material. Orteronel, galeterone and abiraterone
`were purchased from the Selleckchem (Houston, TX, USA).
`CYP17A1 inhibitors compound I and IV were described earlier [23].
`
`2.2. Steroid profiling
`
`Steroid profiling was performed with the cells grown on 6 well
`plates by adding 100,000 cpm [3H] pregnenolone (preg) to the
`culture medium for 90 min. To study specific CYP17A1 enzyme
`activities, cells were first treated with 1 mM or 2 mM CYP17A1 in-
`hibitors. After 24 h of inhibitor treatment, cells were treated with
`1 mM trilostane (a specific blocker of HSD3B) for 90 min before
`adding [3H] preg. For CYP21A2 activity, cells were treated with
`1 mM of different inhibitors for 24 h and then labeled with [3H] 17a-
`hydroxyprogesterone (17-OH Prog) for 60 min to monitor the
`conversion of 11-deoxycortisol. Steroids were extracted from cell
`supernatants and separated by thin layer chromatography (TLC) as
`previously described [24]. Steroid conversion was calculated as
`percentage of
`total
`radioactivity incorporated into specific
`products.
`
`2.3. Quantitative real time PCR (qRT-PCR)
`
`2.4. CYP19A1 enzyme activity using titrated water release assay
`
`Human Placental JEG3 cells were grown on 12 well plates and
`treated with CYP17A1 inhibitors for 24 h before aromatase activity
`was assayed. Aromatase activity was assayed with 3H labeled an-
`drostenedione ([1b-3H(N)]-androstene-3,17-dione; ~100,000 cpm/
`well). Androstenedione was added to the treated cells for 6 h.
`Aromatase activity was then assessed by the titrated water release
`assay as previously described [25,26].
`
`2.5. GC-MS measurement of steroid metabolites
`
`The measurement of steroid hormone metabolites in human
`adrenal H295R cells was performed by gas chromatography e mass
`spectrometry (GC-MS) as previously described [27]. The cells were
`grown in 10 cm plates in normal growth medium for 24 h, then
`medium was replaced, and cells were treated with 1 mM of CYP17A1
`inhibitors in medium without NU-I serum for 24 h. After 12 h of
`treatment, 1 mM of pregnenolone was added. At the end, superna-
`tant was collected and concentrated samples were used for steroid
`analysis by GCeMS. All measurements were performed in the
`steroid laboratory of the Department of Nephrology, Hypertension
`and Clinical Pharmacology at the University Hospital of Bern,
`Switzerland.
`
`3. Results and discussion
`
`3.1. CYP17A1 inhibitors modulate the adrenal androgen production
`
`CYP17A1 is required for androgen production in humans and
`control of its enzyme activity is essential for treatment of hyper-
`androgenic disorders. Abiraterone, galeterone and orteronel are
`inhibitors of CYP17A1 (Suppl Fig. 2). Abiraterone is an effective
`active site-directed inhibitor, designed for the treatment of CRPC
`patients to suppress the androgen production by inhibition of
`CYP17A1 activities [28,29]. The galeterone and orteronel were
`designed to selectively inhibit 17,20 lyase activity of CYP17A1 for
`the treatment of CRPC [21,22]. There have been suggestions that
`galeterone is more specific towards the 17,20 lyase activity of
`CYP17A1 than abiraterone but this effect is not clear [9,30]. Func-
`tional studies on the effects of orteronel in vivo (cynomolgus
`monkey) and in vitro (human adrenal tumor cells) showed that
`orteronel is a potent inhibitor of 17,20 lyase activity [31]. In addition
`to these well-known inhibitors, Hartmann group synthesized
`inhibitors of steroid biosynthesis [23]. They observed
`several
`compound IV was a more potent inhibitor of 17,20 lyase activity
`compared to its effects on 17a-hydroxylase activity [23]. However,
`the effect of these drugs on adrenal androgen production remains
`unknown.
`We used the steroidogenic human adrenal cortex H295R cells to
`study the effect of CYP17A1 inhibitors (orteronel, galeterone,
`compound I and IV and abiraterone). We found that orteronel and
`galeterone drastically decreased the DHEA and androstenedione
`production (Fig. 1A). Compounds I and IV decreased DHEA,
`androstendione and 11-DOC/11-deoxycortisol production. In the
`cells treated with abiraterone, we only observed the conversion of
`preg to prog and other products in the pathway were completely
`blocked (Fig. 1B). This suggests that abiraterone inhibits CYP17A1
`and possibly other enzymes that are involved in steroidogenesis.
`
`3.2. Orteronel is a specific inhibitor of the CYP17A1 17,20 lyase
`activity
`
`The H295R cells were treated with inhibitors for 72 h. After the
`treatment, total RNA was isolated and qRT-PCR was done as pre-
`viously described [24]. Details are in supplementary material.
`
`We studied the effect of CYP17A1 inhibitors in more detail for
`the specificity of the inhibition of CYP17A1 enzyme activities. Cells
`
`

`
`S.S. Udhane et al. / Biochemical and Biophysical Research Communications 477 (2016) 1005e1010
`
`1007
`
`Fig. 1. Effect of the CYP17A1 inhibitors on adrenal steroid profile. For the steroid profiling H295R cells were treated with 1 mm orteronel, galeterone, compound I and IV and
`abiraterone for 24 h. Steroid production was labeled with [3H] preg for 90 min. Steroids were extracted and resolved by TLC. A) Steroid profiling of adrenal H295R cells treated with
`DMSO and 1 mM of orteronel or galeterone B) Steroid profiling of adrenal H295R cells treated with 1 mM of compound I, compound IV and abiraterone. Representative TLCs are
`depicted on the left, summaries and quantifications of the results are given on the right. Prog, progesterone; D4A, androstenedione; Preg, pregnenolone; DHEA, dehydroepian-
`drosterone; 17-OH Preg, 17a-hydroxypregnenolone; 17-OH Prog, 17a-hydroxy progesterone; Data are presented as mean ± SD of three independent experiments. *p < 0.05,
`**p < 0.01.
`
`Fig. 2. Effects of CYP17A1 inhibitors on NCI-H295 cells. A and B . Effect of inhibitors on CYP17A1 enzyme activity. The enzyme activities of CYP17A1 were checked in H295R
`cells treated with control (DMSO) and 1 mM of orteronel, galeterone, compound I, compound IV and abiraterone. CYP17A1 activities were assessed by the conversion of [3H] preg to
`17-OH preg for 17a-hydroxylase activity and 17-OH preg to DHEA for 17,20 lyase activity. Representative TLCs are depicted on the left, summaries and quantifications of the results
`are given on the right. Data are given as mean ± SD of three independent experiments. Preg, pregnenolone; DHEA, dehydroepiandrosterone; 17-OH Preg, 17a-hydroxypregnenolone.
`*p < 0.05,**p < 0.01. C. Impact of CYP17A1 inhibitors on genes involved in androgen biosynthesis. H295R cells were treated with control (DMSO) or 1 mM orteronel and
`galeterone for 72 h and then RNA was isolated and transcribed to cDNA. Results of qRT-PCR validation of HSD3B2, CYP17A1 and AKR1C3 genes relative to the housekeeping gene
`cyclophilin A. Expression of the genes was analyzed by SYBR Green real-time PCR. Analysis of relative gene expression was determined by the 2DDCt method. *p < 0.05.
`
`

`
`1008
`
`S.S. Udhane et al. / Biochemical and Biophysical Research Communications 477 (2016) 1005e1010
`
`were treated with inhibitors and then with 1 mM trilostane (a
`specific blocker of HSD3B) for 90 min before labeling with [3H]
`preg. We found that the orteronel decreased only DHEA production,
`suggesting selectivity of inhibition towards the 17,20 lyase activity
`of CYP17A1 (Fig. 2A). Treatments of galeterone, abiraterone, com-
`pound I and IV showed a decrease in the conversion from preg to
`17-OH preg and to DHEA which suggests these compounds
`inhibited both the hydroxylase as well as lyase activity of CYP17A1
`(Fig. 2AeB). These data indicate that orteronel is a specific inhibitor
`of 17,20 lyase activity, as observed in previous studies [31,32].
`
`3.3. Effect of CYP17A1 inhibitors on genes involved in androgen
`biosynthesis
`
`Abiraterone and galeterone are steroidal structures that also act
`as androgen receptor (AR) antagonist. Orteronel is a non-steroidal
`CYP17A1 inhibitor. Abiraterone has been reported to alter the
`androgen regulated gene expression in prostate cancer cells
`through the AR-mediated signaling [33]. Since, both the orteronel
`and galeterone have shown potential as potent CYP17A1 inhibitors,
`we conducted further experiments to find out if these inhibitors
`have any impact on gene transcription. The H295R cells were
`treated with control (DMSO) or 1 mM orteronel and galeterone for
`72 h. Gene expressions were studied by relative quantification PCR
`(qRT-PCR). We studied the expression of three key genes CYP17A1,
`HSD3B2 and AKR1C3 that are involved in androgen biosynthesis.
`Interestingly, we found a slight increase in HSD3B2 gene expression
`upon treatment with galeterone (Fig. 2C). In presence of orteronel
`no significant changes in gene expression were observed (Fig. 2C).
`
`3.4.
`
`Influence of CYP17A1 inhibitors on aromatase activity
`
`To further study the specificity of the CYP17A1 inhibitors, we
`determined their impact on other related steroidogenic enzyme
`activities. First, we studied the effect of inhibitors on aromatase
`(CYP19A1) activity. The activity and expression of aromatase differs
`between different cell types depending on the need of cells for
`estrogens. Aromatase is localized in endoplasmic reticulum and
`catalyzes the last step of estrogen biosynthesis from androgens
`(converts androstenedione to estrone and testosterone to estra-
`diol). The orteronel, galeterone, compound I and abiraterone
`showed no effect of on aromatase activity (Fig. 3A). Compound IV
`treatment resulted in a decrease of aromatase activity by 23%
`(Fig. 3A), which was in accordance with its reported modest inhi-
`bition of aromatase in vitro (IC50 ¼ 228 nM) [34].
`
`3.5. Abiraterone severely inhibits 21-hydroxylase (CYP21A2)
`activity
`
`We then studied the effect of these inhibitors on the 21-
`hydroxylase (CYP21A2) enzyme activity. The 21-hydroxylase plays
`a role in producing glucocorticoids (cortisol) and mineralocorticoid
`(aldosterone). Cortisol helps maintain blood sugar levels, protects
`the body from stress and suppresses inflammation. Maintaining the
`cortisol level is required for the gonadal function, as disruption in
`cortisol production may affect
`fertility by the regulation of
`hypothalamic-pituitary-adrenal (HPA) and gonadal (HPG) axis [35].
`CYP21A2 activities were assessed by checking the conversion of
`[3H] 17-OH Prog to 11-deoxycortisol. Orteronel, galeterone, com-
`pound I and IV treatment did not change 11-deoxycortisol pro-
`duction, suggesting no effect on CYP21A2 activity. Surprisingly,
`abiraterone was found to completely inhibit the CYP21A2 activity
`(Fig. 3B). This suggests that abiraterone is not a specific inhibitor of
`CYP17A1 and can also inhibit CYP21A2 activity. Protein sequence
`comparison of CYP17A1 and CYP21A2 shows an identity of around
`
`Fig. 3. Influence of CYP17A1 inhibitors on other steroid metabolizing cytochrome
`P450 activity. The effect of
`inhibitors on P450 aromatase (CYP19A1) and 21-
`hydroxylase (CYP21A2). A) Effect of inhibitors on aromatase. Effect of inhibitors
`was studied on aromatase activity in human placental JEG-3 cells. Cells were treated
`with control (DMSO) and 1 mM of orteronel, galeterone, abiraterone, compound I and
`IV for 24 h before aromatase activity was tested. Aromatase activity was assayed with
`androstenedione using 2 mM cold substrate and 3H labeled androstenedione ([1b-
`3H(N)]-androstene-3,17-dione; ~100,000 cpm/well) as radioactive tracer. Andro-
`stenedione was added to the treated cells for 6 h. Aromatase activity was measured by
`observing the conversion of 3H-androstenedione to estrone using the titrated water
`release assay. Data are presented as mean ± SEM of two independent experiments. B)
`Effect on CYP21A1 activity. The 21-hydroxylase activity was checked in H295R cells
`treated with control (DMSO) and 1 mM of orteronel, galeterone, abiraterone, compound
`I and compound IV for 24 h. CYP21A2 activities were measured by observing the
`conversion of [3H] 17-OH prog to 11-deoxycortisol. Representative TLCs are depicted in
`the upper panel, summaries and quantifications of the results are given in the lower
`panel. Data are presented as mean ± SD of three independent experiments. **p < 0.01.
`
`28% but structures of CYP17A1 and CYP21A2 have many similarities
`and may retain similar binding sites. The inhibition of CYP21A2
`could be a serious problem since using this drug inhibits whole
`adrenal androgen biosynthesis. Use of abiraterone for androgen
`lowering effects, where inhibition of CYP21A2 is not desired, will
`create additional complications.
`
`Impact of CYP17A1 inhibitors on the metabolomics of C19
`3.6.
`steroids by GC-MS
`
`The CYP17A1 inhibitors orteronel, galeterone and abiraterone
`showed significant decrease in potent androgens like testosterone
`and dihydrotestosterone (Table 1). Galeterone lowered all classical
`and backdoor pathway metabolites of androgens in human adrenal
`cells. However, orteronel and abiraterone treatment lead to an in-
`crease of androgen metabolite androsterone, which is involved in
`
`

`
`S.S. Udhane et al. / Biochemical and Biophysical Research Communications 477 (2016) 1005e1010
`
`1009
`
`Table 1
`The table shows the GC-MS measurement of secreted steroid metabolites in the human adrenal H295R cells in the untreated vs the treated condition. The change in me-
`tabolites are indicated based on untreated levels which are set as hundred percent compared to CYP17A1 inhibitor treatments. *nd: Not determined; þ: Increased metabolite.
`
`Metabolites
`
`Untreated
`
`Orteronel
`
`Galeterone
`
`Abiraterone
`
`Androgen metabolites
`Androsterone
`Etiocholanolone
`Androstenediol
`11b-Hydroxy-Androsterone
`11b-Hydroxy-Etiocholanolone
`Dehydroepiandrosterone
`5a androstendiol
`16a-Hydroxy-DHEA
`5a androstenetriol
`5-Pregnenetriol
`Testosterone
`5a-Dihydrotestosterone
`Estrogen metabolites
`Estriol
`17-b Estradiol
`Progesterone metabolites
`17a-OH-Pregnanolone
`Pregnanetriol
`Corticosterone metabolites
`Tetrahydrocorticosterone
`5a-Tetrahydrocorticosterone
`Cortisol-metabolites
`Cortisone
`Tetrahydrocortisone
`a-Cortolone
`b-Cortolone
`20a-Dihydrocortisone
`20b-Dihydrocortisone
`Cortisol
`TH-Cortisol
`5a-Tetrahydrocortisol
`a-Cortol
`b-Cortol
`20a-Dihydrocortisol
`
`100
`100
`100
`100
`nd
`100
`100
`100
`100
`100
`100
`100
`
`100
`nd
`
`100
`100
`
`nd
`nd
`
`nd
`100
`100
`nd
`nd
`nd
`100
`100
`100
`nd
`nd
`nd
`
`448
`3915
`328
`104
`

`48
`80
`74
`108
`36
`50
`nd
`
`103
`

`
`4162
`802
`
`nd
`nd
`
`nd
`90
`873
`

`nd
`nd
`22
`nd
`nd
`

`nd
`120
`
`30
`51
`nd
`94
`nd
`30
`66
`84
`39
`71
`29
`nd
`
`58
`

`
`56
`49
`

`nd
`
`nd
`96
`374
`

`nd
`nd
`22
`83
`83
`

`

`162
`
`1593
`3030
`152
`1834
`

`10
`63
`81
`133
`9
`26
`nd
`
`55
`

`
`4408
`2011
`

`

`

`6372
`45,600
`

`

`

`40
`2617
`2617
`

`

`161
`
`the backdoor pathway of dihydrotestosterone biosynthesis. One
`possible explanation is that abiraterone can influence backdoor
`pathway by elevating CYP17A1 expression and induce progesterone
`accumulation [36]. The progesterone accumulation can compete
`against abiraterone for binding to CYP17A1 leading to induction of
`androgen metabolites involved in backdoor pathway [36]. Orter-
`onel seems to be more specific for inhibiting the 17,20 lyase activity
`in the classical androgen biosynthesis pathway and might not in-
`fluence the androgen metabolism via backdoor pathway. Addi-
`tionally, we found that orteronel and abiraterone treatment also
`lead to an increase in other androgen metabolites like etiochola-
`nolone and 11b-hydroxy-etiocholanolone. For the first time the
`data here show the effect of CYP17A1 inhibitors on the human
`steroid metabolome especially on androgen metabolites, in adrenal
`H295R cells. These data indicate that orteronel, galeterone and
`abiraterone can influence or alter different steroid metabolites in
`human adrenal cells apart from blocking the CYP17A1 activity.
`Based on our results, we can conclude that orteronel is a more
`specific inhibitor for CYP17A1 17,20 lyase activity compared to other
`inhibitors tested here. Orteronel had no effect on other tested en-
`zymes that are involved in steroid hormone biosynthesis. Hence, it
`can be considered as a treatment option for hyperandrogenic dis-
`orders like polycystic ovary syndrome. Using abiraterone for
`lowering androgen production in hyperandrogenic disorders may
`create additional undesired complications in patients because of its
`inhibitory effect on CYP21A2 activities. For the first time we
`showed the effect of CYP17A1 inhibitors abiraterone, orteronel and
`galeterone on steroid metabolome in human adrenal H295R cells.
`Our data provide evidence for the significantly altered steroids
`
`metabolites by CYP17A1 inhibitors. The steroid metabolome model
`may be used as an efficient diagnostic tool for further studies on
`urine or serum samples of hyperandrogenic patients treated with
`CYP17A1 inhibitors. Our observations that abiraterone is also a very
`potent inhibitor of CYP21A2, suggests that abiraterone should not
`be used to treat hyperandrogenic disorders when inhibition of
`CYP21A2 is an undesirable side effect. Orteronel may be tried for a
`more specific inhibition of CYP17A1 activities without affecting
`other steroid metabolizing enzymes.
`
`Acknowledgments
`
`This work was supported by grants to AVP from the Swiss Na-
`tional Science Foundation (31003A-134926). We thank Dr. Nasser
`Dhayat from the Department of Nephrology, Hypertension and
`Clinical Pharmacology, University Hospital of Bern, Bern,
`Switzerland for help with the steroid pathway figure.
`
`Appendix A. Supplementary data
`
`Supplementary data related to this article can be found at http://
`dx.doi.org/10.1016/j.bbrc.2016.07.019.
`
`Transparency document
`
`Transparency document related to this article can be found
`online at http://dx.doi.org/10.1016/j.bbrc.2016.07.019.
`
`

`
`1010
`
`S.S. Udhane et al. / Biochemical and Biophysical Research Communications 477 (2016) 1005e1010
`
`References
`
`[1] W.L. Miller, Androgen synthesis in adrenarche, Rev. Endocr. Metab. Disord. 10
`(2009) 3e17.
`[2] L. Ibanez, N. Potau, R. Virdis, M. Zampolli, C. Terzi, M. Gussinye, A. Carrascosa,
`E. Vicens-Calvet, Postpubertal outcome in girls diagnosed of premature
`pubarche during childhood:
`increased frequency of
`functional ovarian
`hyperandrogenism, J. Clin. Endocrinol. Metab. 76 (1993) 1599e1603.
`[3] L. Ibanez, N. Potau, M.V. Marcos, F. De Zegher, Adrenal hyperandrogenism in
`adolescent girls with a history of low birthweight and precocious pubarche,
`Clin. Endocrinol. (Oxf) 53 (2000) 523e527.
`[4] R.L. Rosenfield, Clinical review: identifying children at risk for polycystic ovary
`syndrome, J. Clin. Endocrinol. Metab. 92 (2007) 787e796.
`[5] E. Carmina, T. Koyama, L. Chang, F.Z. Stanczyk, R.A. Lobo, Does ethnicity in-
`fluence the prevalence of adrenal hyperandrogenism and insulin resistance in
`polycystic-ovary-syndrome, Am. J. Obstet. Gynecol. 167 (1992) 1807e1812.
`[6] A.V. Pandey, C.E. Flück, NADPH P450 oxidoreductase: structure, function, and
`pathology of diseases, Pharmacol. Ther. 138 (2013) 229e254.
`[7] A.V. Pandey, W.L. Miller, Regulation of 17,20 lyase activity by cytochrome b5
`and by serine phosphorylation of P450c17,
`J. Biol. Chem. 280 (2005)
`13265e13271.
`[8] L.H. Zhang, H. Rodriguez, S. Ohno, W.L. Miller, Serine phosphorylation of
`human P450c17 increases 17,20-lyase activity: implications for adrenarche
`and the polycystic ovary syndrome, Proc. Natl. Acad. Sci. U. S. A. 92 (1995)
`10619e10623.
`[9] M.N. Stein, N. Patel, A. Bershadskiy, A. Sokoloff, E.A. Singer, Androgen syn-
`thesis inhibitors in the treatment of castration-resistant prostate cancer, Asian
`J. Androl. 16 (2014) 387e400.
`[10] L. Falsetti, A. Gambera, C. Platto, L. Legrenzi, Management of hirsutism, Der-
`matol. 1 (2000) 89e99.
`[11] D. Gillatt, Antiandrogen treatments in locally advanced prostate cancer: are
`they all the same? J. Cancer Res. Clin. Oncol. 132 (Suppl. 1) (2006) S17eS26.
`[12] H. Appelt, B. Strauss, Effects of antiandrogen treatment on the sexuality of
`women with hyperandrogenism, Psychother. Psychosom. 42 (1984) 177e181.
`[13] L. Cusan, A. Dupont, J.L. Gomez, R.R. Tremblay, F. Labrie, Comparison of flu-
`tamide and spironolactone in the treatment of hirsutism: a randomized
`controlled trial, Fertil. Steril. 61 (1994) 281e287.
`[14] E. Diamanti-Kandarakis, A. Mitrakou, M.M. Hennes, D. Platanissiotis, N. Kaklas,
`J. Spina, E. Georgiadou, R.G. Hoffmann, A.H. Kissebah, S. Raptis,
`Insulin
`sensitivity and antiandrogenic therapy in women with polycystic ovary syn-
`drome, Metabolism 44 (1995) 525e531.
`[15] E. Diamanti-Kandarakis, A. Mitrakou, S. Raptis, G. Tolis, A.J. Duleba, The effect
`of a pure antiandrogen receptor blocker, flutamide, on the lipid profile in the
`polycystic ovary syndrome, J. Clin. Endocrinol. Metab. 83 (1998) 2699e2705.
`[16] L. Ibanez, F. de Zegher, Low-dose flutamide-metformin therapy for hyper-
`insulinemic hyperandrogenism in non-obese adolescents and women, Hum.
`Reprod. Update 12 (2006) 243e252.
`[17] V. Bruni, E. Peruzzi, M. Dei, S. Nannini, V. Seravalli, G. Sisti, M. Fambrini,
`Hepatotoxicity with low- and ultralow-dose flutamide: a surveillance study
`on 203 hyperandrogenic young females, Fertil. Steril. 98 (2012) 1047e1052.
`[18] J. Brahm, M. Brahm, R. Segovia, R. Latorre, R. Zapata, J. Poniachik, E. Buckel,
`L. Contreras, Acute and fulminant hepatitis induced by flutamide: case series
`report and review of the literature, Ann. Hepatol. 10 (2011) 93e98.
`[19] B.A. Gartrell, F. Saad, Abiraterone in the management of castration-resistant
`prostate cancer prior to chemotherapy, Ther. Adv. Urol. 7 (2015) 194e202.
`[20] J. Richards, A.C. Lim, C.W. Hay, A.E. Taylor, A. Wingate, K. Nowakowska,
`C. Pezaro, S. Carreira, J. Goodall, W. Arlt, I.J. McEwan, J.S. de Bono, G. Attard,
`Interactions of abiraterone, eplerenone, and prednisolone with wild-type and
`mutant androgen receptor: a rationale for increasing abiraterone exposure or
`combining with MDV3100, Cancer Res. 72 (2012) 2176e2182.
`[21] T. Kaku, T. Hitaka, A. Ojida, N. Matsunaga, M. Adachi, T. Tanaka, T. Hara,
`
`M. Yamaoka, M. Kusaka, T. Okuda, S. Asahi, S. Furuya, A. Tasaka, Discovery of
`orteronel (TAK-700), a naphthylmethylimidazole derivative, as a highly se-
`lective 17,20-lyase inhibitor with potential utility in the treatment of prostate
`cancer, Bioorg. Med. Chem. 19 (2011) 6383e6399.
`[22] B. Montgomery, M.A. Eisenberger, M.B. Rettig, F. Chu, R. Pili, J.J. Stephenson,
`N.J. Vogelzang, A.J. Koletsky, L.T. Nordquist, W.J. Edenfield, K. Mamlouk,
`K.J. Ferrante, M.E. Taplin, Androgen receptor modulation optimized for
`response (ARMOR) phase I and II studies: galeterone for the treatment of
`castration-resistant prostate Cancer, Clin. Cancer Res. 22 (2016) 1356e1363,
`http://dx.doi.org/10.1158/1078-0432.CCR-15-1432.
`PubMed
`PMID:
`26527750.
`[23] S.J. Krug, Q. Hu, R.W. Hartmann, Hits identified in library screening demon-
`strate selective CYP17A1 lyase inhibition, J. Steroid Biochem. Mol. Biol. 134
`(2013) 75e79.
`[24] S.S. Udhane, A.V. Pandey, G. Hofer, P.E. Mullis, C.E. Fluck, Retinoic acid re-
`ceptor beta and angiopoietin-like protein 1 are involved in the regulation of
`human androgen biosynthesis, Sci. Rep. 5 (2015) 10132.
`[25] E.D. Lephart, E.R. Simpson, Assay of aromatase activity, Methods Enzymol. 206
`(1991) 477e483.
`[26] A.V. Pandey, P. Kempna, G. Hofer, P.E. Mullis, C.E. Flück, Modulation of human
`CYP19A1 activity by mutant NADPH P450 oxidoreductase, Mol. Endocrinol. 21
`(2007) 2579e2595.
`[27] N.A. Dhayat, A.C. Frey, B.M. Frey, C.H. d’Uscio, B. Vogt, V. Rousson, B. Dick,
`C.E. Fluck, Estimation of reference curves for the urinary steroid metabolome
`in the first year of life in healthy children: tracing the complexity of human
`postnatal steroidogenesis, J. Steroid Biochem. Mol. Biol. 154 (2015) 226e236.
`[28] G.A. Potter, S.E. Barrie, M. Jarman, M.G. Rowlands, Novel steroidal inhibitors of
`human cytochrome P45017 alpha (17 alpha-hydroxylase-C17,20-lyase): po-
`tential agents for the treatment of prostatic cancer, J. Med. Chem. 38 (1995)
`2463e2471.
`[29] G. Attard, A.H. Reid, T.A. Yap, F. Raynaud, M. Dowsett, S. Settatree, M. Barrett,
`C. Parker, V. Martins, E. Folkerd, J. Clark, C.S. Cooper, S.B. Kaye, D. Dearnaley,
`G. Lee, J.S. de Bono, 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. 26 (2008) 4563e4571.
`[30] L. Yin, Q. Hu, CYP17 inhibitorseabiraterone, C17,20-lyase inhibitors and
`multi-targeting agents, Nat. Rev. Urol. 11 (2014) 32e42.
`[31] M. Yamaoka, T. Hara, T. Hitaka, T. Kaku, T. Takeuchi, J. Takahashi, S. Asahi,
`H. Miki, A. Tasaka, M. Kusaka, Orteronel (TAK-700), a novel non-steroidal
`17,20-lyase inhibitor: effects on steroid synthesis in human and monkey
`adrenal cells and serum steroid levels in cynomolgus monkeys, J. Steroid
`Biochem. Mol. Biol. 129 (2012) 115e128.
`[32] T. Hara, J. Kouno, T. Kaku, T. Takeuchi, M. Kusaka, A. Tasaka, M. Yamaoka,
`Effect of a novel 17,20-lyase inhibitor, orteronel (TAK-700), on androgen
`synthesis in male rats, J. Steroid Biochem. Mol. Biol. 134 (2013) 80e91.
`[33] E.A. Mostaghel, B.T. Marck, S.R. Plymate, R.L. Vessella, S. Balk, A.M. Matsumoto,
`P.S. Nelson, R.B. Montgomery, Resistance to CYP17A1 inhibition with abir-
`aterone in castration-resistant prostate cancer: induction of steroidogenesis
`and androgen receptor splice variants, Clin. Cancer Res. 17 (2011) 5913e5925.
`[34] L. Yin, S. Lucas, F. Maurer, U. Kazmaier, Q. Hu, R.W. Hartmann, Novel imidazol-
`1-ylmethyl substituted 1,2,5,6-Tetrahydropyrrolo[3,2,1-ij]quinolin-4-ones as
`potent and selective CYP11B1 inhibitors for the treatment of Cushing’s syn-
`drome, J. Med. Chem. 55 (2012) 6629e6633.
`[35] S. Whirledge, J.A. Cidlowski, Glucocorticoids, stress, and fertility, Minerva
`Endocrinol. 35 (2010) 109e125.
`[36] C. Cai, S. Chen, P. Ng, G.J. Bubley, P.S. Nelson, E.A. Mostaghel, B. Marck,
`A.M. Matsumoto, N.I. Simon, H. Wang, S.P. Balk, Intratumoral de novo steroid
`synthesis activates androgen receptor in castration-resistant prostate cancer
`and is upregulate

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