throbber
Original Article
`
`BLOCKADE OF ANDROGENIC STEROID SYNTHESIS FOR TREATING PROSTATE CANCER
` ATTARD
` et al.
`
`The use of C17,20-lyase inhibitors
`in prostate cancer is described by
`authors from Los Angeles. These
`agents suppress the generation of
`testosterone and potentially active
`androgenic precursors, perhaps
`reversing castration resistance.
`Abiraterone is an orally
`bioavailable lyase inhibitor
`structurally related to
`pregnenolone, and is currently
`under clinical assessment.
`
`Another paper is presented by the
`same group of authors in Los
`Angeles addressing some of the
`controversies about the continual
`need for the traditional radical
`open surgical management of RCC,
`and evaluates the oncological
`principles which ensure the
`persistent need for this approach.
`
`Selective blockade of androgenic
`steroid synthesis by novel lyase
`inhibitors as a therapeutic strategy for
`treating metastatic prostate cancer
`
`GERHARDT ATTARD, ARIE S. BELLDEGRUN* and JOHANN S. DE BONO
`Institute for Cancer Research/The Royal Marsden NHS Foundation Trust, Centre for Cancer
`Therapeutics, Sutton, Surrey, UK, and *UCLA David Geffen School of Medicine, Department of
`Urology, Los Angeles, California, USA
`Accepted for publication 21 July 2005
`
`KEYWORDS
`
`castration refractory prostate cancer,

`17
`-hydroxylase, C
`-lyase, CYP450c17,
`17,20
`abiraterone, CB7630
`
`THE NEED FOR NOVEL AGENTS TO TREAT
`PROSTATE CANCER
`
`Prostate cancer is the most common
`malignancy in Western societies and the
`second most common cause of male cancer-
`related death in the UK and USA, accounting
`≈
`for
`12% of all male cancer-related deaths
`[1,2]. When confined to the prostate gland,
`the disease is curable with local therapy
`(radical prostatectomy, external beam
`radiotherapy, brachytherapy or cryotherapy).
`However, despite the use of PSA for screening,
`in 15–33% of men local therapy fails and they
`develop incurable metastatic disease [3,4].
`Activation of the androgen receptor (AR) by
`androgenic steroids including testosterone

`and its more potent 5
`-reduced metabolite,

`5
`-dihydrotestosterone (DHT), regulates the
`transcription of a diverse range of target
`genes involved in prostate cell proliferation,
`differentiation, and apoptosis [5]. Androgen
`deprivation by medical or surgical castration
`remains the mainstay of treatment and
`>
`90% of men with metastatic prostate
`
`cancer initially respond rapidly and
`often dramatically to castration, with
`improvements in bone pain, regression of soft
`tissue metastasis, and steep declines in PSA
`level. However, the duration of response is
`frequently short (12–33 months) and in
`almost all patients is followed by the
`emergence of castration-resistant prostate
`cancer (CRPC) that, untreated, is invariably
`fatal within 9–12 months [3]. Systemic
`chemotherapy with docetaxel for patients
`with CRPC confers a modest survival
`advantage (2–3 months) and is effective
`for palliating symptoms [6], but the
`duration and rate of response is limited.
`Other chemotherapy, e.g. mitoxantrone, has
`not been shown to improve survival but might
`help with symptom control [6]. There is an
`urgent need for new agents that provide
`palliation and improve survival.
`
`Continued androgen-dependent activation of
`a ‘hypersensitive’ AR in castrated patients
`secondary to AR gene amplification [7],
`mutations in the AR gene [5], increased AR
`expression [8], or alterations in AR co-
`repressor/co-activator function [9] appear to
`be important mechanisms of castration
`resistance. These could be reversed by
`suppressing circulating androgens, inhibiting
`the binding of biologically active androgenic
`steroids to the ARs or disrupting AR
`

`
`
`
`2 0 0 5 B J U I N T E R N A T I O N A L | 9 6 , 1 2 4 1 – 1 2 4 6 | doi:10.1111/j.1464-410X.2005.05821.x
`
`1 2 4 1
`
`<T>1,16<END1>1<END2>14<END3>(577,-14)<E4>22</E4>0<E5>1<E6>18<E7>11<E8>12/1/2015 12:00:00 AM15:00:05.4497467<E9></T>
`
`MYLAN PHARMS. INC. EXHIBIT 1023 PAGE 1
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`A T TA R D
`
`E T A L .
`
`generation with drugs such as HSP90
`inhibitors. This review discusses the rationale
`behind using selective lyase inhibitors (e.g.
`abiraterone acetate, also known as CB7630),
`to suppress circulating androgens.
`
`TARGETING THE AR
`
`The benefit of androgen deprivation therapy
`(ADT) was first reported by Huggins and
`Hodges in 1941 [10] when surgical castration
`was found to give symptomatic benefit and in
`some cases, complete responses in metastatic
`prostate cancer. The role of ADT by castration
`has since expanded to the neoadjuvant and,
`more commonly, the adjuvant settings, and to
`patients with a high PSA level but no clinical
`or radiological evidence of metastatic disease
`[3]. Medical castration with LHRH agonists
`(e.g. goserelin and leuprorelin) is often a more
`popular alternative to orchidectomy. These act
`by continuous stimulation of the anterior
`pituitary gland, resulting in inhibition of LH
`secretion and suppression of testicular
`androgen synthesis. Antiandrogens (either
`steroidal, e.g. cyproterone acetate, or
`nonsteroidal, e.g. bicalutamide or flutamide)
`prevent androgen binding to the AR. These
`can be used in combination with LHRH
`analogues to inhibit binding of residual
`androgens to the AR to attempt ‘maximum
`androgen blockade’ [3]. However, this
`combined strategy has not significantly
`prolonged the survival of patients with
`advanced prostate cancer [3]. In addition, up
`to 30% of patients show a decrease in PSA
`level after stopping AR antagonists [11]. This
`can partly be explained by the development
`of AR gene mutations or increased AR
`expression that might cause AR antagonists
`to behave as weak agonists [7,8]. For many
`years, oestrogens, including diethylstilbestrol,
`were the primary medical treatment for
`metastatic prostate cancer, until they were
`superseded by LHRH agonists [3]. Now they
`have a role in treating castrated patients in
`whom castration and antiandrogen therapy
`have failed, but although PSA response rates
`in a series of phase II trials were 21–80%, the
`<
`median response duration is
`6 months
`and the concomitant use of prophylactic
`warfarin has not eliminated the risk of
`thromboembolic events [12].
`
`The presence of AR protein expression and
`significant AR mRNA levels in tumour samples
`from patients with CRPC strongly suggest
`that prostate tumours evolve mechanisms to
`
`1 2 4 2
`
`reactivate AR signalling and AR-responsive
`pathways after ADT [7,13]. This is supported
`by preclinical models, which have suggested
`that, as prostate cancer cells become
`castration resistant (otherwise, but less
`appropriately known as hormone- or
`androgen-resistant), they acquire the ability
`to grow in the presence of low levels of
`androgens (equivalent to castrate), by
`the up-regulation of AR expression [8]. It
`was also reported that, despite castration,
`intraprostatic levels of testosterone and DHT
`in CRPC might remain sufficient to maintain
`tumour growth [14]. The source of these
`androgens is unclear, but altered regulation of
`enzymes involved in the synthesis and
`inactivation of androgens might be one cause
`of their accumulation. In support of this,
`increased expression of enzymes involved in
`androgen synthesis were reported in prostate
`cancer cells acquired from biopsies of CRPC
`[13,15]. These data suggest the possibility of
`endogenous production of steroids by CRPC.
`Overall, these reports indicate that the
`currently used ADTs fail to reverse AR
`signalling in CRPC and support the use of
`novel drugs that target the AR directly or
`indirectly by suppressing the generation of
`ligands.
`
`Plasma testosterone is not completely
`suppressed by castration, in part because of
`peripheral conversion of adrenal androgenic
`steroids to testosterone by 17-ketoreductase
`[3]. Adrenal androgen synthesis can be
`inhibited by targeting the hypothalamo-
`pituitary-adrenal axis or by inhibiting key
`enzymes involved in adrenal steroid
`biosynthesis. Suppression of the
`hypothalamo-pituitary-adrenal axis and
`consequently the generation of adrenal
`androgens by low-dose steroids has not been
`unequivocally shown to occur in patients with
`CRPC, but it could be one explanation for their
`anti-neoplastic activity [16,17]. However,
`ketoconazole, an imidazole antifungal agent
`that weakly and non-selectively inhibits
`several cytochrome P450 enzymes involved in
`adrenal steroid synthesis, induces a transient
`PSA response in 20–30% of patients with
`CRPC [11]. Interestingly, patients who respond
`to ketoconazole and subsequently progress
`show a significant decrease, associated with
`an increase with disease progression of
`adrenal androgens (dehydroepiandrosterone
`(DHEA) and androstenedione) suggesting that
`ketoconazole resistance is caused by adrenal
`androgens [11]. These clinical results lend
`credence to the inhibition of the adrenal
`
`steroid synthesis pathway as a therapeutic
`strategy.
`
`THE ROLE OF CYTOCHROME P450c17 IN
`ANDROGEN BIOSYNTHESIS
`
`The two sites thought to produce most of the
`androgenic steroids in humans are the testis
`and the adrenal cortex. The principal
`enzymatic reaction in steroid biosynthesis
`involves cleavage of a six-carbon group from
`cholesterol by CYP450ssc (desmolase) [18]. A
`series of subsequent reactions catalysed by
`members of the cytochrome P450 family then
`produce the glucocorticoid and
`mineralocorticoid 21-carbon steroid
`hormones vital to human survival (Fig. 1).
`CYP450c17 is a single microsomal enzyme,
`encoded for by the single human CYP17 gene,
`that catalyses the two independently-

`regulated steroids 17
`-hydroxylase and
`C
`-lyase activities needed to produce
`17,20
`the 19-carbon precursors of the sex
`steroids in both the adrenal cortex and the

`testis [18]. The 17
`-hydroxylase activity

`typically converts pregnenolone to 17
`-
`hydroxypregnenolone and progesterone

`to 17
`-hydroxyprogesterone, and the

`C
`-lyase activity converts 17
`-
`17,20

`-
`hydroxypregnenolone to DHEA and 17
`hydroxyprogesterone to androstenedione
`(Fig. 1). The C
`-lyase activity is roughly
`17,20
`50 times more efficient for converting

`17
`-hydroxypregnenolone to DHEA than

`for converting 17
`-hydroxyprogesterone to
`androstenedione [18,19]. The products of
`steroid biosynthesis, androstenedione and
`DHEA, are weak as androgens but in the testis
`can be converted to testosterone by the

`enzyme 17
`-hydroxysteroid dehydrogenase
`[20]. Castration blocks testosterone from this
`source but does not prevent the synthesis of
`adrenal androgens. These androgens might be
`a clinically important source of androgenic
`steroids for activating the AR on prostate
`cancer cells. Steroidogenesis in the human
`adrenal is divided into three morphologically
`and functionally distinct zones. The zona
`glomerulosa, located just below the adrenal
`capsule, does not express CYP450c17 and
`produces the 17-hydroxy 21-carbon steroid
`aldosterone, the principal mineralocorticoid,
`under the regulation of angiotensin II [21].
`The middle layer, the zona fasciculata,

`expresses CYP450c17 and has abundant 17
`-
`hydroxylase but very little C
`-lyase activity,
`17,20
`and produces the 17-hydroxy 21-carbon
`steroid cortisol, the principal glucocorticoid,
`

`
` 2 0 0 5 B J U I N T E R N A T I O N A L
`
`MYLAN PHARMS. INC. EXHIBIT 1023 PAGE 2
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`B L O C K A D E O F A N D R O G E N I C S T E R O I D S Y N T H E S I S F O R T R E A T I N G P R O S T A T E C A N C E R
`

`FIG. 1. The impact of abiraterone on the adrenal steroid synthesis pathway. Abiraterone inhibits CYP450c17 17
`
`-lyase activity (crossed out in
`-hydroxylase and C
`17,20
`red) and suppresses androstenedione, DHEA and their androgenic precursors (blue arrows). Suppression of cortisol and its precursors causing a compensatory rise in
`ACTH and excess synthesis of aldosterone and its precursors is predicted (blue arrows).
`
`Cholesterol
`
`desmolase
`
`Adrenal Cortex
`
`Pregnenolone
`
`Progesterone
`
`DOC
`
`Corticosterone
`
`18OH-
`Corticosterone
`
`Aldosterone
`
`3-BHSD-I
`
`17a-hydroxylase
`
`21-hydroxylase
`
`11b-hydroxylase
`
`18-hydroxylase
`
`18-oxidase
`
`17OH-pregnenolone
`
`17OH-progesterone
`
`11-DOC
`
`Cortisol
`
`3-BHSD-I
`
`C17-20 lyase
`
`Peripheral Tissues
`
`
`
`DHEA
`
`Androstenedione
`
`Testosterone
`
`DHT
`
`3-BHSD-I
`
`17-keto-reductase
`
`5a-reductase
`
`under the regulation of adrenocorticotropic
`hormone (ACTH) corticotrophin [21]. The inner
`zona reticularis, which does not become
`morphologically identifiable until the onset of
`adrenarche, expresses CYP450c17 and has

`both 17
`-hydroxylase and C
`-lyase
`17,20
`activities, and thus produces 17-hydroxy
`19-carbon precursors of sex steroids [18,21].
`The C
`-lyase activity of human CYP450c17
`17,20
`is enhanced by serine phosphorylation
`of CYP450c17 and by the presence of
`cytochrome b5. The expression of cytochrome
`b5 increases in the adrenal zona reticularis at
`the onset of adrenarche [18].
`
`Congenital deficiencies in CYP450c17 are a
`rare form of congenital adrenal hyperplasia in
`which not only adrenal but also gonadal
`steroidogenesis is impaired [19]. This results in
`abrogation of gonadal sex steroid production
`and adrenal biosynthesis of cortisol and
`androgens. However, corticosterone synthesis
`is not impaired, and as corticosterone has
`glucocorticoid properties, patients rarely
`manifest symptoms of adrenal insufficiency.
`In fact, rodents lack CYP450c17 and
`use corticosterone as their principal
`glucocorticoid [19]. However, because
`corticosterone is a weaker glucocorticoid than
`

`
` 2 0 0 5 B J U I N T E R N A T I O N A L
`
`cortisol, abnormally high corticosterone
`production is necessary before feedback
`inhibition of pituitary ACTH secretion occurs,
`establishing a new steady state. To produce
`enough corticosterone to compensate for
`the absence of cortisol, more intermediate
`steroids might be generated. These include
`progesterone, 11-deoxycorticosterone (DOC),
`18-hydroxycorticosterone and 19-nor-DOC.
`This ACTH-driven overproduction of
`mineralocorticoids often leads to
`hypertension, a characteristic presenting
`feature of this disease, usually in early
`adulthood [19]. At diagnosis, sexual
`infantilism in 46 XX females and ambiguous
`genitalia in 46 XY males is usually manifest
`and laboratory investigations will often find
`hypokalaemia [19]. An extremely rare disorder
`is isolated C
`-lyase deficiency caused
`17,20
`-
`by mutations that destroy most C
`17,20

`lyase activity but preserve most 17
`-
`hydroxylase activity. Patients do not have
`mineralocorticoid excess but show the
`consequences of absent sex steroids [19],
`so CYP450c17 is a logical target for the
`development of new drugs to treat CRPC.
`The most potent and selective inhibitor of
`CYP450c17 currently in clinical studies is
`abiraterone acetate.
`
`PRECLINICAL DEVELOPMENT OF
`ABIRATERONE ACETATE
`
`The serendipitous discovery that some esters
`of 4-pyridylacetic acid are effective inhibitors
`of the hydroxylase-lyase enzyme in rat testis
`[22] led to the study of a variety of esters of 3-

`and 4-pyridylacetic acid and their
`-alkylated

`derivatives on human testicular 17
`-
`hydroxylase/C
`-lyase [23,24]. The most
`17,20

`-
`potent inhibitors of human testicular 17
`hydroxylase/C
`-lyase had, as a common
`17,20
`structural feature, the 17-(-3-pyridyl)
`substituent that contains nitrogen capable of
`forming a co-ordinate bond with the haem
`iron of the enzyme (Fig. 2) [23,24]. The 3-
`pyridyl substituent results in a several orders
`more potent inhibition of CYP450c17 than
`the 2-pyridyl and 4-pyridyl substituents [23].
`The double bond in the 16,17-position of
`the steroidal skeleton is essential for the
`irreversible inhibition of CYP450c17 [25]. Two
`compounds, CB7598 (abiraterone), which is
`closely related structurally to the natural
`substrate pregnenolone, and CB7627, were
`identified as the most potent, and were
`selected for further development at the
`Institute of Cancer Research in London. With
`<
`a Ki
` of
`1 n
`, both compounds were
`M
`app
`
`1 2 4 3
`
`MYLAN PHARMS. INC. EXHIBIT 1023 PAGE 3
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`A T TA R D
`
`E T A L .
`
`TABLE 1
`
`Summary of phase I evaluation of abiraterone acetate
`
`Variable
`Number of patients
`Dose levels, mg
`Testosterone level at
`inclusion, nmol/L
`Effect on testosterone (T)
`
`Effect on androstenedione
`Effect on LH
`
`Phase I study patient population
`Single dose in castrate men
`Single dose in intact men
`16
`4
`10, 30, 100, 200, 500
`200, 500
`<
`>
`2
`9
`
`Continuous dosing (12 days) in intact men
`6
`500, 800
`>
`9
`
`Target suppression at
`500 mg
`Same as for T
`Suppressed at inclusion
`
`Suppression with nadir on second
`day, recovery 6–9 days.
`Same as for T
`Maximal rise on third day, recovery
`by 10th day
`
`Suppression with nadir on first to third day and
`sustained for up to 9 days
`Same as for T
`Maximal rise on third day
`
`several times more potent inhibitors of rat
`hydroxylase/lyase activity than ketoconazole
`[23,24]. Unlike CB7627, abiraterone was a
`highly selective inhibitor of CYP450c17 and so
`was chosen as the main candidate for further
`development [24]. However, due to poor
`bioavailability and a susceptibility to
`hydrolysis by esterases, a prodrug for
`abiraterone was sought [26,27]. The amide
`was a several orders less potent inhibitor of

`
`-O-acetate
`hydroxylase/lyase [24] but the 3
`form (abiraterone acetate, CB7630) (Fig. 2)
`
`in vivo was
`was resistant to esterases and
`rapidly deacetylated to abiraterone resulting
`in potent inhibition of CYP450c17 [26,27].
`
`When adult mice given abiraterone acetate or
`ketoconazole i.p. daily for 14 days were
`compared to a control group dosed with
`vehicle alone, abiraterone significantly
`reduced plasma testosterone concentrations
`for at least 24 h despite a four-fold increase
`in LH concentrations [27]. This was associated
`with reduced weights of the ventral prostate
`(48%) and seminal vesicles (87%), compared
`to the controls. The kidneys and testis were
`also reduced in weight by 37% and 62%,
`respectively [27]. There was no weight loss in
`the androgen-dependent organs of the group
`given ketoconazole [27]. There was a marked
`increase in the weight of the adrenal glands in
`the group administered ketoconazole, but no
`change in the control group or in the animals
`dosed with abiraterone, indicating no
`inhibition of corticosterone production by
`abiraterone. Animal toxicology studies found
`no effects on haematological, biochemical or
`renal function variables and only mild
`toxicities. Abiraterone acetate was therefore
`considered safe enough to be studied in
`humans [27].
`
`1 2 4 4
`
`FIG. 2.
`The structure of abiraterone and abiraterone acetate (centre). The effect variations in the orientation

`of the pyridyl substituent have on the potency of 17
`-hydroxylase/C
`-lyase inhibition.
`
`17,20
`
`N
`
`N
`
`N
`
`H O
`
`R O
`
`H O
`
`2-pyridyl
`
`IC50 (nm)
`lyase 76
`hydroxylase 270
`
`3-pyridyl
`
`IC50 (nm)
`lyase 2.9
`hydroxylase 4
`
`4-pyridyl
`
`IC50 (nm)
`lyase 1000
`hydroxylase 4000
`
`
`
`R = H: Abiraterone (CB7598); 17-(3-pyridyl)androsta-5,16-dien-3b -ol
`R = Ac: Abiraterone acetate (CB7630)
`
`CLINICAL EVALUATION OF
`ABIRATERONE ACETATE
`
`A series of three phase I studies were
`performed in men with histologically
`confirmed prostate cancer who had shown a
`biochemical (PSA) relapse after two lines of
`hormone treatment but for whom, at the time
`of participation, no alternative treatment for
`symptomatic or progressive disease was
`considered necessary (Table 1). The first in-
`human study investigated the effect of a
`single dose of abiraterone acetate in castrate
`<
`patients (testosterone confirmed
`2 nmol/L,
`58 ng/mL) with prostate cancer in whom
`treatment with antiandrogens had failed.
`
`Nine patients were treated in groups of three
`at doses of 10, 30 and 100 mg. At these doses,
`the plasma level of abiraterone was below the
`level of detection and there was no consistent
`effect on testosterone [28]. One patient was
`then given 200 mg and six of them 500 mg.
`There were detectable plasma levels of
`>
`200 mg [28].
`abiraterone at doses of
`Abiraterone at 500 mg suppressed
`<
`0.14 nmol/L
`testosterone concentrations, to
`(4 ng/mL), or by 75% when baseline
`>
`0.6 nmol/L (20 ng/
`testosterone levels were
`mL), and androstenedione concentrations.
`Suppression was sustained from the second
`to the fifth day after abiraterone acetate.
`There was no corresponding suppression of
`

`
` 2 0 0 5 B J U I N T E R N A T I O N A L
`
`MYLAN PHARMS. INC. EXHIBIT 1023 PAGE 4
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`B L O C K A D E O F A N D R O G E N I C S T E R O I D S Y N T H E S I S F O R T R E A T I N G P R O S T A T E C A N C E R
`

`-hydroxyprogesterone. This first study
`17
`was followed by a second investigating the
`effect of a single dose of abiraterone acetate
`in intact men with prostate cancer who had
`previously been treated with an antiandrogen
`and an LHRH agonist. One patient was given
`200 mg, but the testosterone level was not
`suppressed. Three patients were then given
`500 mg, and in all three there was a reduction
`in testosterone level of more than half from
`baseline. The testosterone nadir was on the
`second day after therapy, with recovery to
`pretreatment levels 6–9 days later. There was
`a corresponding compensatory surge in LH
`levels, maximal on the third day, with recovery
`to pretreatment levels by the 10th day. There
`was no change in cortisol levels in either
`study [28]. The third and last in this series of
`phase I evaluations investigated 12-day
`continuous dosing in intact men with
`prostate cancer who had previously received
`antiandrogens and LHRH agonists. The
`duration of drug treatment was limited by a
`lack of availability of the drug. Three patients
`were given 500 mg daily and another three
`were given 800 mg daily. The testosterone
`level was suppressed after the first day of
`dosing and was sustained for 3 days, and
`then reversed by a rise in LH. The patients
`given 800 mg appeared to show greater
`suppression of testosterone but as in the
`500 mg cohort, the effect of abiraterone was
`insufficient to offset the rise in LH. All six
`patients had a reduced cortisol response to
`ACTH stimulation on the 11th day after
`dosing, suggesting reduced adrenocortical
`reserve. There was a mild reduction in evening
`cortisol levels also in the three patients
`given 800 mg but there were no clinical
`manifestations of adrenocortical insufficiency
`[28]. Pharmacokinetic studies suggested good
`>
`oral bioavailability at doses of
`200 mg.
`The mean (
`) T
` for abiraterone was 2.70
`SD
`max
`(2.71) h with an elimination half-life of 27.6
`(20.17) h, supporting once-daily oral dosing.
`There was an inter-patient 10-fold variation
`in the area under the curve for a given
`dose, making analysis of dose-dependent
`pharmacokinetic relationships difficult.
`Several factors could have caused this wide
`level of variation. As with all oral drugs,
`absorption might have been altered by
`residual food in the stomach despite a 2-h
`fast, by intrinsic inter-individual differences in
`upper gastrointestinal pH, by concomitant
`medication(s) effecting gastric pH, or by
`variable first-pass hepatic metabolism.
`Abiraterone acetate was well tolerated, and
`no serious adverse events or haematological
`

`
` 2 0 0 5 B J U I N T E R N A T I O N A L
`
`or biochemical changes were reported. Mild
`mood variations, flushing attacks, testicular
`discomfort, and headaches were described in
`individual patients [28].
`
`FUTURE DEVELOPMENT OF NOVEL
`LYASE INHIBITORS
`
`Several other derivatives of naturally
`occurring steroidal substrates, including
`pregnane and androstane, are potent

`inhibitors of 17
`-hydroxylase/C
`-lyase
`17,20
`[29,30]. Sa40 (17-(5-pyrimidyl)androsta-5,16-

`diene-3
`-ol) and its 3-acetyl derivative, Sa41,
`
`in vitro
`are three times more potent for
`inhibition of human CYP450c17 than
`abiraterone and abiraterone acetate,
`respectively, but have a poorer
`in vivo rodent
`
`pharmacokinetic profile in
`experiments [29]. Clinical assessment in
`patients is required. L-2 (20-hydroximinio-
`′
`4,16-pregnadien-3-one), L-36 (17-(3
`-
`pyrazolyl)androsta-4,16-dien-3-one) and L-
`′
`39 (17-(5
`-isoxazolyl)androsta-4,16-dien-3-
`one) are steroidal compounds that in addition

`to inhibiting 17
`-hydroxylase/C
`-lyase,
`17,20

`also inhibit 5
`-reductase with a potency
`similar to finasteride [30]. They also interact
`with wild-type AR and the mutated AR on
`cells from the LNCaP cell line [30]. L-36 shows
`an agonistic interaction whereas L-39 is
`
`in vitro LNCaP cell
`antagonistic and inhibits
`growth [30]. L-39 has therefore been
`proposed as a candidate for further

`development. However, the benefit of 5
`-
`reductase inhibitors for treating metastatic
`prostate cancer is unclear [3]. Although
`conversion of testosterone to its more active
`metabolite, DHT, is inhibited, testosterone
`consequently accumulates and can activate
`the AR. Also, interaction with the AR might
`not be desirable as changes in the AR can lead
`to antagonists behaving agonistically [7,8].
`Other nonsteroidal compounds, including TX-
`977 and its two diastereoisomers, reported
`
`in vivo inhibitors of
`to be more potent
`testosterone synthesis, have been associated
`with an increase in weight of the adrenal
`glands in rodent models, suggesting lower
`specificity of lyase inhibition vs glucocorticoid
`synthesis [31]. Abiraterone acetate’s
`selectivity for CYP450c17 and its failure to
`interact with the AR might be preferable for
`treating patients with CRPC. However, at
`present, while the available data are
`encouraging with respect to side-effects
`and endocrine effectiveness with short-term
`dosing, there is no evidence of clinical
`efficacy. A safety and efficacy evaluation of
`
`abiraterone acetate administered daily and
`continuously to castrate men with advanced
`prostate cancer progressing despite hormone
`treatment is planned. Concomitant castration
`is expected to prevent a compensatory LH rise,
`and sustained, profound suppression of
`serum testosterone and androgenic precursor
`levels is predicted. Patients will be closely
`monitored for the development of
`glucocorticoid insufficiency or hypertension.
`Synthesis of corticosterone, a precursor of
`aldosterone and the primary glucocorticoid in
`rodents, will not be inhibited by abiraterone
`and its continued synthesis might prevent
`clinical manifestations of glucocorticoid
`insufficiency. In fact, hypertension due to
`increased ACTH and not glucocorticoid
`insufficiency is described in congenital
`CYP45017c deficiency [19]. The results of
`these studies are keenly awaited.
`
`CONFLICT OF INTEREST
`
`A. Belldegrun: Vice Chairman, Board of
`Directors and Chairman, Scientific Advisory
`Board, Cougar Biotechnology.
`
`REFERENCES
`
`1
`
`2
`
`3
`
`4
`
`5
`
`6
`
`Cancer Research UK. Mortality
`
`Statistics. Available at http://
`www.cancerresearchuk.org/aboutcancer/
`=
`statistics/mortality?version
`1. Accessed
`July 2005
`Jemal A, Murray T, Samuels A, Ghafoor
`A, Ward E, Thun MJ.
`Cancer statistics,
`
`53: 5–26
`
`CA Cancer J Clin 2003;
`2003.
`Hellerstedt BA, Pienta KJ.
`The current
`state of hormonal therapy for prostate
`
`52:
`
`CA Cancer J Clin 2002;
`cancer.
`154–79
`Han M, Partin AW, Zahurak M,
`Piantadosi S, Epstein JI, Walsh PC.
`Biochemical (prostate specific antigen)
`recurrence probability following radical
`prostatectomy for clinically localized
`
`
`J Urol 2003; 169: 517–23
`
`prostate cancer.
`Buchanan G, Greenberg NM, Scher HI,
`Harris JM, Marshall VR, Tilley WD.
`Collocation of androgen receptor gene
`Clin Cancer
`mutations in prostate cancer.
`
`7: 1273–81
`Res
` 2001;
`Tannock IF, de Wit R, Berry WR
` Docetaxel plus prednisone or
`et al.
`mitoxantrone plus prednisone for
`
`advanced prostate cancer.
`
`351: 1502–12
`2004;
`
`N Engl J Med
`
`1 2 4 5
`
`MYLAN PHARMS. INC. EXHIBIT 1023 PAGE 5
`
`

`
`A T TA R D
`
`E T A L .
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`7
`
`8
`
`9
`
`10
`
`11
`
`Edwards J, Krishna NS, Grigor KM,
`Bartlett JM.
`Androgen receptor gene
`amplification and protein expression in
`Br J
`hormone refractory prostate cancer.
`
`89: 552–6
`
`Cancer 2003;
`Chen CD, Welsbie DS, Tran C
`
`et al.
`Molecular determinants of resistance to
`
`
`antiandrogen therapy. Nat Med 2004; 10:
`
`
`33–9
`Li P, Yu X, Ge K, Melamed J, Roeder RG,
`Wang Z.
`Heterogeneous expression
`and functions of androgen receptor
`co-factors in primary prostate cancer.
`
`161: 1467–74
`
`Am J Pathol 2002;
`Huggins C, Hodges CV.
`Studies on
`prostatic cancer. I. The effect of
`castration, estrogen and androgen
`injection on serum phosphatases in
`metastatic carcinoma of the prostate.
`
`1: 293–7
`
`Cancer Res 1941;
`Small EJ, Halabi S, Dawson NA
`
`et al.
`Antiandrogen withdrawal alone or in
`combination with ketoconazole in
`androgen-independent prostate cancer
`patients: a phase III trial (CALGB 9583).
`
`22: 1025–33
`
`J Clin Oncol 2004;
`Oh WK.
`The evolving role of estrogen
`Clin Prostate
`therapy in prostate cancer.
`
`1: 81–9
`
`Cancer 2002;
`Holzbeierlein J, Lal P, LaTulippe E
`
`et al.
`Gene expression analysis of human
`prostate carcinoma during hormonal
`therapy identifies androgen-responsive
`genes and mechanisms of therapy
`
`164: 217–
`
`Am J Pathol 2004;
`resistance.
`27
`Mohler JL, Gregory CW, Ford OH 3rd
` The androgen axis in recurrent
`et al.
`
`
`
`440–8
`Castagnetta LA, Carruba G, Traina
`A
`et al. Expression of different 17beta-
`hydroxysteroid dehydrogenase types
`and their activities in human prostate
`cancer cells. Endocrinology 1997; 138:
`4876–82
`16 Khandwala HM, Vassilopoulou-Sellin R,
`Logethetis CJ, Friend KE. Corticosteroid-
`induced inhibition of adrenal androgen
`production in selected patients with
`prostate cancer. Endocr Pract 2001; 7: 11–
`5
`
`12
`
`13
`
`14
`
`15
`
`
`
`prostate cancer. Clin Cancer Res 2004; 10:
`
`17 Akakura K, Suzuki H, Ueda T et al.
`Possible mechanism of dexamethasone
`therapy for prostate cancer: suppression
`of circulating level of interleukin-6.
`Prostate 2003; 56: 106–9
`18 Miller WL, Auchus RJ, Geller DH. The
`regulation of 17,20 lyase activity. Steroids
`1997; 62: 133–42
`19 Auchus RJ. The genetics,
`pathophysiology, and management of
`human deficiencies of P450c17.
`Endocrinol Metab Clin North Am 2001;
`30: 101–19
`20 O’Shaughnessy PJ, Baker PJ, Heikkila
`M, Vainio S, McMahon AP. Localization
`of 17beta-hydroxysteroid dehydrogenase/
`17-ketosteroid reductase isoform
`expression in the developing mouse testis
`– androstenedione is the major androgen
`secreted by fetal/neonatal leydig cells.
`Endocrinology 2000; 141: 2631–7
`21 Dickerman Z, Grant DR, Faiman C,
`Winter JS. Intraadrenal steroid
`concentrations in man: zonal differences
`and developmental changes. J Clin
`Endocrinol Metab 1984; 59: 1031–6
`22 McCague R, Rowlands MG, Barrie SE,
`Houghton J. Inhibition of enzymes of
`estrogen and androgen biosynthesis by
`esters of 4-pyridylacetic acid. J Med Chem
`1990; 33: 3050–5
`23 Rowlands MG, Barrie SE, Chan F et al.
`Esters of 3-pyridylacetic acid that
`combine potent inhibition of 17 alpha-
`hydroxylase/C17,20-lyase (cytochrome
`P45017 alpha) with resistance to
`esterase hydrolysis. J Med Chem 1995;
`38: 4191–7
`24 Potter GA, Barrie SE, Jarman M,
`Rowlands MG. Novel steroidal inhibitors
`of human cytochrome P45017 alpha
`(17 alpha-hydroxylase-C17,20-lyase):
`potential agents for the treatment of
`prostatic cancer. J Med Chem 1995; 38:
`2463–71
`25 Jarman M, Barrie SE, Llera JM. The
`16,17-double bond is needed for
`irreversible inhibition of human
`cytochrome p45017alpha by abiraterone
`(17-(3-pyridyl) androsta-5, 16-dien-
`3beta-ol) and related steroidal inhibitors.
`J Med Chem 1998; 41: 5375–81
`
`26 Chan FC, Potter GA, Barrie SE
`et al. 3- and 4-pyridylalkyl
`adamantanecarboxylates: inhibitors of
`human cytochrome, P450 (17alpha)
`(17alpha-hydroxylase/C17,20-lyase).
`Potential nonsteroidal agents for the
`treatment of prostatic cancer. J Med
`Chem 1996; 39: 3319–23
`27 Barrie SE, Potter GA, Goddard PM,
`Haynes BP, Dowsett M, Jarman M.
`Pharmacology of novel steroidal
`inhibitors of cytochrome P450(17)
`alpha (17 alpha-hydroxylase/C17–20
`lyase). J Steroid Biochem Mol Biol, 1994;
`50: 267–73
`28 O’Donnell A, Judson I, Dowsett M
`et al. Hormonal impact of the 17alpha-
`hydroxylase/C(17,20)–lyase inhibitor
`abiraterone acetate (CB

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