throbber
Abiraterone for the Treatment
`of mCRPC
`
`9
`
`Zafeiris Zafeiriou, Niven Mehra and Johann S. de Bono
`
`Introduction
`
`(PCa) was a
`identified that prostate cancer
`Huggins and Hodges first
`hormone-driven disease and established as standard treatment androgen deprivation
`by surgical castration or medical castration (administration of estrogens) [1]. Vir-
`tually all metastatic PCa respond to castration but disease invariably progresses
`after a median of 18–24 months. This state was initially characterized as “hormone
`insensitive” or “hormone resistant” PCa but these terms have now been abandoned
`for the term “castration-resistant PCa (CRPC)”.
`It was hypothesized early that the effect of castration could be augmented by
`suppression of adrenal androgen biosynthesis as after castration androgens of
`extragonadal origin were still existent at about 10 % of the pre-castration level. For
`that purpose adrenalectomy [2] and hypophysectomy [3] were attempted with poor
`
`Z. Zafeiriou N. Mehra
`Prostate Cancer Targeted Therapy Group, Institute of Cancer Research/the Royal
`Marsden NHS Foundation Trust, Sycamore House, Downs Road,
`Sutton SM2 5PT, Surrey, UK
`e-mail: Zafeiris.Zafeiriou@icr.ac.uk
`
`N. Mehra
`e-mail: niven.mehra@icr.ac.uk
`
`J.S. de Bono (&)
`Department of Drug Development Unit, Institute of Cancer Research/the Royal
`Marsden NHS Foundation Trust, Sutton, Surrey, UK
`e-mail: johann.de-bono@icr.ac.uk
`
`© Springer International Publishing Switzerland 2016
`K.C. Balaji (ed.), Managing Metastatic Prostate Cancer In Your
`Urological Oncology Practice, DOI 10.1007/978-3-319-31341-2_9
`
`125
`Amerigen Exhibit 1134
`Amerigen v. Janssen
`IPR2016-00286
`
`

`
`126
`
`Z. Zafeiriou et al.
`
`outcome but provided evidence of activity and support for the rationale of sup-
`pression of extra-testicular androgens. Later this was pursued with administration of
`corticosteroids to suppress the hypothalamic ACTH axis [4]. Ketoconazole, an
`imidazole antifungal, was subsequently used [5] which inhibits enzymes involved
`in adrenal steroid synthesis, including CYP17, albeit non-selectively and incom-
`pletely [6], with PSA responses in approximately 60 % of patients [7], but also
`grade 3–4 toxicities in approximately 20 % of them [8].
`
`Early Development of Abiraterone
`
`Ketoconazole’s efficacy in CRPC sparkled the development of more potent and
`selective inhibitors of CYP17A1 [9, 10]. Pioneering work at the ICR in Sutton, UK,
`identified in the early 1990s that both steroidal [11] and nonsteroidal pyridyl esters
`[12, 13] exhibited selectivity for CYP17A1 inhibition.
`The steroidal inhibitors are derived from the natural substrates of CYP17A1,
`pregnenolone or progesterone [14], bind irreversibly to CYP17A1 and have
`increased inhibitory properties over many nonsteroidal agents. The 3-pyridyl ana-
`logues (Figs. 9.1 and 9.2) result in more potent inhibition of CYP17 relative to the
`2-pyridyl and 4-pyridyl analogues [12] and the 16,17-double bond adjacent to the
`pyridyl residue is necessary for the irreversible binding [15].
`One such steroidal compound, CB7630, later known as abiraterone acetate
`(AA) or JNJ-212082 is a 3ß-O-acetated version and a prodrug of CB7598, with
`better bioavailability and easier formulation than the 3ß-hydroxy compound
`(Figs. 9.1 and 9.2) and its administration in mice was able to decrease circulating
`levels of testosterone [10].
`
`Fig. 9.1 Abiraterone structure. Abiraterone (CB7598) for R=H and abiraterone acetate (CB7630)
`for R=Ac. Adapted from O’Donnell [16]. With permission from Nature Publishing Group
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`127
`
`Fig. 9.2 The parent ABCD
`steroid ring system
`(hydrocarbon framework) is
`shown with IUPAC-approved
`ring lettering and atom
`numbering
`
`Phase I and II Development
`
`In 2004 the first in human trial of AA in PCa was reported and consisted of three
`small phase I studies of AA without the use of concomitant corticosteroids in
`castrate and non-castrate patients [16]. Administration was continuous in one of the
`arms and once-off in the other two and AA was dose-escalated from 10 to 800 mg
`with pharmacokinetic (PK) studies indicating good bioavailability and supporting
`once-daily dosing. AA was well tolerated without documented grade 3–4 toxicities.
`Abiraterone plasma concentrations were detected at doses of AA ≥ 200 mg with
`consistent effects on testosterone levels when dose-escalated to the 500 and 800 mg
`dose level. In castrate males suppression of testosterone was sustained, while in
`non-castrate males a compensatory increase in LH levels overcame inhibition of
`gonadal testosterone synthesis. Boehringer Ingelheim, to which abiraterone was
`licensed at that time, suspended further clinical development despite these initial
`data. Later on, the interest in abiraterone was re-sparkled, probably because of
`accumulating data supporting an important
`role of CYP171A in castration
`resistance.
`In 2008 and 2010 two additional phase I trials were reported respectively
`[17, 18]. In the first study, twenty-one chemotherapy-naive CRPC patients were
`dose-escalated from 250 to 2000 mg once daily, with 3 patients per cohort and 6
`additional patients in the 1000 mg group to complete PK and pharmacodynamic
`(PD) studies. The recommended dose for the phase II development was determined
`at 1000 mg OD as above that level there was a plateau of the endocrine effect of
`AA. In the 2010 study 33 chemotherapy-naïve CRPC patients were enrolled, of
`which 19 had received prior ketoconazole therapy. Patients were dose-escalated
`from 250 to 1000 mg once daily; all patients received a single dose at day 1, and
`continuous dosing from day 7 with fed and fasted cohorts at each dose level. PSA
`
`

`
`128
`
`Z. Zafeiriou et al.
`
`and objective responses were commonly witnessed and warranted further
`evaluation.
`The first phase II study on AA was reported in 2009 on 42 chemotherapy-naïve
`patients [19], and was a phase II expansion of the UK phase I study reported in
`2008 [17]. Encouraging PSA declines of ≥50 % were seen in 28 of 42 CRPC
`patients, with objective responses by RECIST in 9 of 24 patients with measurable
`disease. In this study the median time to PSA progression was 225 days (95 % CI,
`162 to 287 days). Subsequently further phase II trials were performed which are
`summarized in Table 9.1, both in the pre- as well as in the post-chemotherapy
`setting and strengthened proof of activity across the whole spectrum of mCRPC
`with a frequency of PSA declines of ≥50 % between 36 % and 51 % which
`appeared higher in ketoconazole-naïve rather than in ketoconazole-pretreated
`patients. In the first reported study AA was given without the use of concomitant
`steroids [20] whereas later prednisone 5 mg was prescribed twice daily from the
`beginning of the treatment [21].
`
`Pharmacokinetics and Pharmacodynamics of Abiraterone
`and Interaction with Other Drugs
`
`Pharmacokinetics
`
`PK data of Abiraterone are available from the first phase I studies [17, 18] as well as
`from two post-licensing studies: a PK study in healthy men [22] and a population
`PK analysis with 359 subjects including 62 healthy volunteers [23].
`Pharmacokinetics of Abiraterone are strongly influenced by food and therefore it
`is administered routinely in the fasted state, 2 h after a previous meal and 1 h before
`the next meal. Following oral administration AA is converted to its active
`metabolite Abiraterone by rapid hydrolysis taking place in the intraluminal envi-
`ronment of the intestine [24] but also in the liver involving hydroxylation mediated
`by esterases [25]. Therefore, AA is below detectable levels in the plasma [22] and
`Abiraterone is detectable instead at doses of AA ≥200 mg. Abiraterone reaches its
`maximum concentration (Cmax) of 1.2–5 μΜ in approximately 1–2 h in fasting
`patients and 4 h following a high-fat meal (Tmax) [17] and subsequently follows a
`biphasic elimination [22] with a terminal half-life of 5–16 h [17, 18, 22]. Further
`metabolic reactions generate its two main but inactive circulating metabolites,
`abiraterone sulphate and N-oxide abiraterone sulphate. The main metabolite
`excreted in urine is N-oxide abiraterone sulfate. Faeces is the primary route of
`excretion (87.9 %), with major components unchanged AA (55.3 %) and abi-
`raterone (22.3 %) [23, 26]. Interestingly, the apparent clearance of Abiraterone is
`lower in CRPC patients compared to healthy subjects [23].
`There is a significant inter-subject and within-subject variability regarding Cmax
`and drug exposure, as measured with the area under the concentration-time curve
`(AUC) of approximately 40–70 % [22, 23]. There is also variability between cycles,
`with the Cmax observed in cycle 1 being higher by 10–15 % than the Cmax at later
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`129
`
`Danila (2010)
`[21]
`
`Satoh (2014) [28]
`
`Kwak (2014)
`
`Prior
`ketoconazole
`(%)
`
`2 (4.8 %)
`
`0
`
`NR
`
`PSA ≥50 %
`(%)
`
`28 (67 %)
`
`26 (79 %)
`
`30 (63 %)
`
`ORR (%,
`evaluable
`patients)
`
`Time to PSA progression,
`median days (95 % CI)
`
`Population
`
`First author (year
`of publication)
`
`9/24 (37.5 %)
`
`225 (162–287)
`
`caucasian
`
`Attard (2009) [19]
`
`9/13 (69 %)
`
`4/18 (22 %)
`
`496 (280–NE)
`NEa
`
`caucasian
`
`Ryan (2011) [41]
`
`asian
`
`Matsubara (2014)
`[27]
`
`8 (17 %)
`
`29 (51 %)
`
`8/30 (27 %)
`
`169 (113–281)
`
`caucasian
`
`Reid (2010) [20]
`
`27 (46.6 %)
`
`22 (36 %)
`
`4/22 (18 %)
`
`169 (82–200)
`
`caucasian
`
`Table 9.1 Phase II studies of AA in CRPC
`
`Phase II studies of abiraterone acetate in CRPC
`
`Patients
`
`Chemotherapy
`setting
`
`Naïve
`
`Naïve
`
`Naïve
`
`Pretreated
`
`Pretreated
`
`42
`
`33
`
`48
`
`47
`
`58
`
`46
`
`Pretreated
`
`0
`
`Pretreated
`35
`aMedian time not reached
`Abbreviations: NE not estimable; NR not reported
`
`0
`
`16 (35 %)
`
`35 (43 %)
`
`1/22 (4 %)
`
`2/50 (4 %)
`
`142 (85–NE)
`
`143 (113–252)
`
`asian
`
`asian
`
`

`
`130
`
`Z. Zafeiriou et al.
`
`cycles [17, 18]. AUC and Cmax increase with incremental dose proportionally
`between 750 and 1000 mg but more than proportionally between 500 and 750 mg
`[17, 22]. When administered with high-fat content food, drug exposure is increased
`by 4.4-fold compared to fasting achieved by extended absorption without any
`observed increase of Cmax [17, 18]. Beyond food intake and cancer status, other
`covariates such as age, prior chemotherapy status, testosterone level, body mass
`index and total plasma proteins were not found to impact the PK of Abiraterone [23].
`
`Pharmacodynamics
`
`The changes of hormone levels of the steroidogenesis pathway upstream and
`downstream of CYP17 (Fig. 9.3) served as pharmacodynamic markers for the
`phase I trials. Both testosterone and androstenedione were suppressed to unde-
`tectable levels (<1 ng/dL) at all Abiraterone dose levels already by day eight for the
`study by Attard et al. and by day 28 in Ryan et al. (Fig. 9.4) [17]. By day 28,
`DHEA was decreased by a median of 3.4-fold but was still at detectable levels in
`contrast to the other androgens; this was caused by interference of DHEA with the
`used assay. ACTH was increased up to six-fold, as well as the levels of steroid
`precursors upstream of CYP17A1, namely deoxycorticosterone (median 10-fold)
`and corticosterone (median 40-fold) with a plateau of endocrine effects observed at
`the dose level of 750 mg. Administration of dexamethasone, given at PSA pro-
`gression, resulted in suppression of ACTH and decrease of upstream steroids. The
`recommended phase II dose was selected at 1000 mg once daily based on these PD
`endocrine studies [17]. The phase I study by Ryan et al. confirmed the aforemen-
`tioned endocrine changes and concluded to the same recommended dose for the
`phase II [29].
`
`Drug Interactions
`
`AA has been shown to be a strong inhibitor of CYP1A2 and CYP2D6 and a
`moderate inhibitor of CYP2C9, CYP2C19, CYP3A4 and CYP3A5 in vitro [30].
`A phase I drug-drug interaction (DDI) study in CRPC indicated only a relevant
`in vivo interaction between AA-P and CYP2D6, but not with CYP1A2, after single
`dose co-administration with dextromethorphan, a CYP2D6 substrate, and theo-
`phylline, a CYP1A2 substrate [31]. An in vitro study indicated Abiraterone could
`its substrates 1α,25-dihydrox-
`decrease CYP3A4-dependent hydroxylation of
`yvitamin D3 (calcitriol, active vitamin D) and midazolam [32]. In view of the
`reported in vivo interactions caution especially is indicated with concomitant
`administration of AA with known CYP2D6 substrates.
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`131
`
`Fig. 9.3 CYP17 has 17a-hydroxylase and C17,20-lyase activity necessary for the conversion of
`21-carbon pregnanes into 19-carbon sex steroid precursors. a Androgen biosynthesis pathway.
`b Abiraterone inhibits 17a-hydroxylase resulting in feedback increase in ACTH with subsequent
`increase in deoxycorticosterone and corticosterone and symptoms of mineralocorticoid excess.
`Inhibition of C17,20-lysase results in suppression of DHEA, androstenedione and testosterone.
`Adapted from Attard et al. [17]. With permission from American Society of Clinical Oncology
`
`Mechanisms of Action of Abiraterone
`
`Abiraterone blocks the CYP17A1 17α-hydroxylase/17,20-lyase enzyme via for-
`mation of a covalent bond between the nitrogen of its pyridine group and the haem
`iron of CYP17 (Fig. 9.5a,b) while a hydrogen bond network contributes to the
`interaction [33]. Inhibition of the hydroxylase activity suppresses hydroxylation of
`pregnenolone and progesterone at C17 whereas inhibition of the lyase activity limits
`
`

`
`132
`
`Z. Zafeiriou et al.
`
`Fig. 9.4 Pharmacodynamic end points of the treatment with single agent Abiraterone Acetate.
`Treatment with Abiraterone Acetate results in significant suppression of testosterone, dehy-
`droepiandrostenedione (DHEA), and androstenedione (a–c) and an increase of corticosterone and
`deoxycorticosterone (d–e). Plateau of endocrine effect after the dose of 750 mg (f). Adapted from
`Attard et al. [17]. With permission from American Society of Clinical Oncology
`
`Fig. 9.5 Representation of CYP17A1-Abiraterone bound structure. a CYP17A1 from the N
`terminus (blue) to the C terminus (red). b Abiraterone binds at an angle of approximately 60° from
`haem against helix I (yellow). Adapted from DeVore and Scott [33]. With permission from Nature
`Publishing Group
`
`the subsequent conversion of the hydroxylated metabolites to dehydroepiandros-
`terone and androstenedione, respectively, resulting in decreased testosterone and
`In addition to its 17α-hydroxylase/C17,20-lyase
`DHT levels (Fig. 9.3a, b).
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`133
`
`Fig. 9.6 D4A, an Abiraterone metabolite, has the ability to interact with multiple enzymes of the
`androgen biosynthesis pathway. a Conversion from Abiraterone to D4A by 3ßHSD. b D4A IC50
`inhibitory activity in comparison to Abiraterone and Enzalutamide. Abbreviations: AD
`androstenedione; A5diol ∆5-androstenediol; D4A ∆4-Abiraterone. Adapted from Li et al. [36].
`With permission from Nature Publishing Group
`
`inhibiting activity, Abiraterone also was shown to block 3-hydroxysteroid dehy-
`drogenase (3ßHSD) [34] which transforms DHEA to androstenedione. Abiraterone
`can also interact with the AR directly acting in vitro as an antagonist of both the wild
`type and mutant AR but at the dose of 5 μM which is not achieved in plasma under
`normal administration [35]. It was also found to undergo a 3ßHSD enzymatic
`conversion into ∆4-Abiraterone, in which the double bond is moved from C5 to C4
`(D4A; Figs. 9.2, 9.6a) [36]. D4A shares identical steroid A and B rings with
`testosterone, enabling a strong antagonistic interaction with wildtype and mutant
`AR. D4A also inhibits both 3ßHSD and 5α-reductases (SRD5A) (Fig. 9.6b) while it
`retains CYP171A inhibitory activity comparable to Abiraterone.
`Mineralocorticoids are able to inhibit AR transcriptional activity in vitro in the
`presence of
`androgens,
`at
`concentrations
`similar
`to those measured in
`Abiraterone-treated patients [37]. In the phase III studies a significant proportion of
`patients experienced symptoms associated with mineralocorticoid overload, and it
`is not inconceivable, that in this group of patients the mineralocorticoid antagonistic
`effects on AR could contribute to Abiraterone’s clinical activity.
`
`Efficacy of Abiraterone
`
`After the promising results of the phase I and II trials AA was further evaluated in
`the phase III trials COU-AA-301 and COU-AA-302 in patients with mCRPC who
`had progressed on docetaxel in the former and were chemotherapy naive in the
`latter. In both trials patients were blindly randomized between the combination of
`AA 1000 mg once a day with prednisone 5 mg twice a day in the experimental arm
`—from now on in this work designated as AAP—and placebo plus prednisone
`
`

`
`134
`
`Z. Zafeiriou et al.
`
`5 mg twice a day in the control arm respectively—from now on designated as
`PP. Inclusion criteria, randomization scheme, numbers of patients and stratification
`factors are summarized in Table 9.2.
`The first interim analysis of COU-AA-301 was announced after 552 events and a
`median follow-up of 12.8 months (m) and showed an absolute improvement in the
`median Overall Survival (mOS) of 3.9 m in AAP compared to PP which remained
`significant after adjusting for stratification factors [38]. Subsequently the trial was
`un-blinded, crossover of patients from placebo to active drug was allowed and the
`drug gained regulatory approval for mCRPC patients who progressed after treatment
`with docetaxel. In the final survival analysis performed with the data available before
`crossover [39], after a median follow up of 20.2 months and 775 death events both
`the primary and secondary endpoints remained in favour of the AAP arm with an
`absolute benefit of 4.6 m in mOS (Table 9.3; Fig. 9.7). In multivariate analysis all
`the pre-specified stratification factors proved to be prognostic for survival and AAP
`showed superior survival compared to PP in all subgroups, while in some of them it
`did not reach statistical significance probably due to their small size (Fig. 9.8).
`The COU-AA-302 trial enrolled chemo-naive patients with good prognostic
`parameters (Table 9.2) i.e. absence of significant pain, liver disease, low hae-
`moglobin or albumin. The primary endpoints of the trial were radiographic PFS and
`OS and clinically meaningful secondary endpoints were captured: time to cytotoxic
`chemotherapy initiation, time to opiate use for cancer related pain, time to prostate
`specific antigen progression and time to performance status deterioration.
`Two interim analyses (IA) were published on this trial and neither could
`demonstrate a survival benefit consistent with the pre-specified criterion for efficacy
`[40, 41]. This was subsequently met in the final overall analysis [42]. The first IA
`was performed after 43 % of the death events and showed a beneficial effect of
`AAP in the risk for radiographic progression or death with a hazard ratio of 0.49 as
`well as in the risk for death with a HR of 0.75[41] (Table 9.5). All clinically
`meaningful secondary endpoints-median time to cytotoxic chemotherapy, median
`TTPP, median time to performance status deterioration by 1 grade [41]
`(Table 9.4)-favoured AAP despite the fact that the median OS of 27.2 m reported in
`the PP group was the largest ever reported in that patient population. As a result the
`study was un-blinded and crossover allowed and AA’s regulatory approval in the
`US and Europe was expanded to the pre-chemotherapy setting. The second pub-
`lished IA was performed shortly after the un-blinding of the study when only 3
`patients had crossed over to active treatment from placebo. Again AAP showed an
`improved trend in OS compared to PP and an impressively improved time to rPFS
`of 16.5 m compared to 8.2 m of PP and this favourable effect was consistent in all
`stratification subgroups irrespective of age, baseline PSA, serum LDH or ALP,
`ECOG PS, presence or not at entry of pain or bone metastases [40]. In the final
`overall analysis after 49.2 months of follow up, 65 % of patients had died in AAP
`compared to 71 % in PP [42]. Only 67 % of the patients in the Abiraterone group
`compared to 80 % of patients in the placebo group had received further treatments.
`The final analysis showed increased mOS in the Abiraterone group of 34.7 months
`versus 30.3 months in the placebo group which met the pre-specified statistical
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`135
`
`Table 9.2 Inclusion criteria and stratification factors in COU-AA-301 and COU-AA-302
`
`COU-AA-301
`
`Number of patients
`
`1195
`
`Randomization
`ratio
`
`Inclusion criteria
`
`2:1
`
`Prior treatment
`
`Post docetaxel
`
`ECOG PS
`
`≤2
`
`ALT and AST
`
`<2.5 times ULN or < 5 times the ULN if liver
`lesions present
`
`Albumin
`
`Haemoglobin
`
`>3 g/dL
`
`≥9.0 g/dL
`
`Platelet Count
`
`≥100,000/µL
`
`Visceral disease
`
`Allowed
`
`Score of BPI-SF
`question 3
`
`Any
`
`Type of progression
`to prior treatment
`
`Primary endpoints
`
`Radiologic or PSA progression
`
`Overall survival
`
`Secondary endpoints
`
`Time to PSA progression
`
`Radiologic progression free survival
`
`PSA response rate
`
`Stratification factors
`
`ECOG performance status 0–1 versus 2
`
`Presence of significant pain in the past 24 h before
`randomization as captured by the Brief Pain
`Inventory-Short Form (BPI-SF) question 3 (Yes vs.
`No)
`
`Number of previous chemotherapy regimens (1 vs.
`2)
`
`PSA progression only versus radiographic
`progression regardless of PSA progression
`
`COU-AA-302
`
`1088
`
`1:1
`
`Pre docetaxel
`
`≤1
`
`<2.5 times ULN
`
`>3.5 g/dL
`
`≥10.0 g/dL
`
`≥100,000/µL
`
`Not allowed
`
`0–3
`
`Radiologic or PSA
`progression
`
`Overall survival
`
`Radiologic progression
`free survival
`
`Time to PSA progression
`(TTPP)
`
`Time to opiate use for
`cancer-related pain
`
`Time to initiation of
`cytotoxic chemotherapy
`
`Time to ECOG
`performance status
`deterioration
`
`ECOG performance
`status 0 versus 1
`
`

`
`136
`
`Z. Zafeiriou et al.
`
`Table 9.3 Outcomes of efficacy endpoints in COU-AA-301
`
`COU-AA-301
`
`Duration of drug exposure
`(months)
`
`Median overall survival
`(months)
`
`Time to PSA progression
`(months)
`
`rPFS (months)
`
`PSA response rate
`
`Response rate by RECIST in
`patients with measurable
`disease
`
`COU-AA-301 1st
`analysis [38]
`
`interim
`
`COU-AA-301 final analysis [39]
`
`AAP PP HR [95 % CI]
`
`AAP
`
`PP
`
`HR [95 % CI]
`
`8
`
`4
`
`7.4
`
`3.6
`
`14.8
`
`10.9 0.65 [0.54–0.77]
`
`15.8
`
`11.2
`
`0.74[0.64–0.86]
`
`10.2
`
`6.6
`
`0.58; [0.46–0.73]
`
`8.5
`
`6.6
`
`0.63[0.52–0.78]
`
`5.6
`
`3.6
`
`0.67[0.58–0.78]
`
`5.6
`
`3.6
`
`0.66[0.58–0.76]
`
`29 % 6 %
`
`14 % 3 %
`
`29.5 % 5.5 %
`
`14.8 % 3.3 %
`
`Fig. 9.7 Overall survival in the COU-AA-301 trial as evaluated in the last analysis at the time
`point of crossover. Adapted from Fizazi et al. [39]. With permission from Elsevier
`
`boundary for efficacy. The radiographic progression free survival endpoint was also
`met by the trial [42] (Table 9.5).
`It is noteworthy that the Kaplan-Meier curves in COU-AA-302 started sepa-
`rating at 12 months and most clearly so after 18 months [42] which also explains
`why there was no significant survival benefit evident between AAP and P in the
`interim analyses (Fig. 9.9). This can be attributed to the inclusion of patients with
`good prognosis and the resulting small number of death events in the beginning of
`the trial. Still, during the first 12 months of the trial approximately 10 % of the
`patients died and 25–30 % of these deaths could be attributed to non cancer-related
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`137
`
`Fig. 9.8 Overall survival in COU-AA-301 based on duration of previous docetaxel chemother-
`apy (a ≤3 months and b >3 months). The group of ≤3 months seems to have a non-statistically
`significant HR of 0.76 (95 % CI 0.53–1.08) and could be considered as not deriving any benefit
`from treatment with Abiraterone. Nevertheless, the 209 patients represented in this group are
`substantially less compared to the 981 of the >3 months group. Additionally, the survival curves
`appear to have a divergent course. Adapted from Fizazi et al. [39]. With permission from Elsevier
`
`Table 9.4 Outcomes of secondary endpoints in COU-AA-301 and COU-AA-302
`
`AAP [95 %
`CI] (months)
`
`PP [95 % CI]
`
`COU-AA-301
`
`Improvement of fatigue intensity (%) [43]
`
`Improvement of fatigue interference (%) [43]
`
`Median time to fatigue improvement (days) [43]
`
`Time to fatigue intensity progression [25th percentile]
`(days) [43]
`
`Pain palliation (%) [44]
`
`Median time to palliation of pain intensity (months) [44]
`
`Median duration of palliation of pain intensity (months)
`[44]
`
`Median time to occurrence of first skeletal related event
`(months) [44]2
`
`Improvement in the FACT-P total score (%) [46]
`
`Median time to deterioration of FACT-P(weeks) [46]
`
`COU-AA-302
`
`Median time to cytotoxic chemotherapy [40]
`
`Median time to decline in ECOG PS
`
`Pain
`
`Median time to opiate use (months) [40]
`
`Median time to pain progression (months) [41]
`
`58.1
`
`55
`
`59
`
`232
`
`45
`
`5.6
`
`4.2
`
`25
`
`48
`
`59.9
`
`26.5
`
`12.3
`
`33.4
`
`26.7 [19.3–
`not estimable]
`
`Median time to progression of the pain interference with
`daily activities (months) [45]
`
`(10.3 [9.3–
`13.0]
`
`40.3
`
`38
`
`194
`
`139
`
`28.8
`
`13.7
`
`2.1
`
`20.3
`
`32
`
`36.1
`
`16.8
`
`10.9
`
`23.4
`
`18.4 [14.9–not
`estimable]
`
`7.4 [6.4–8.6]
`
`(continued)
`
`

`
`138
`
`Table 9.4 (continued)
`
`Z. Zafeiriou et al.
`
`AAP [95 %
`CI] (months)
`
`PP [95 % CI]
`
`Median time to progression of the worst pain (months)
`[45]
`
`26.7 [19.4–not
`estimable]
`
`(19.4 [16.6–not
`estimable])
`
`QOL
`
`Median time to deterioration of QOL (as measured by
`FACT-P) (months) [45]
`
`Median time to deterioration of QOL (as measured by
`PCa specific FACT-P subscale) (months) [45]
`
`12.7 [11.1–
`14.0]
`
`11.1 [8.6–
`13.8]
`
`8.3 [7.4–10.6]
`
`5.8 [5.5–8.3]
`
`Table 9.5 Outcomes of efficacy endpoints in COU-AA-302
`
`COU-AA-302 1st
`analysis [41]
`
`interim
`
`COU-AA-302 2nd
`interim analysis [40]
`
`COU-AA-302 final
`analysis [42]
`
`AAP
`
`PP
`
`HR [95 %
`CI]
`
`AAP PP
`
`HR [95 %
`CI]
`
`AAP PP
`
`HR [95 %
`CI]
`
`Median overall
`survival (months)
`
`Not
`reached
`
`27.2
`
`0.75
`[0.61–0.93]
`
`35.3
`
`30.1
`
`0.79
`[0.66–0.95]
`
`34.7
`
`30.3 0.81 [95 %
`CI 0 ·70–
`0 · 93]
`
`Time to PSA
`progression (months)
`
`11.1
`
`5.6
`
`rPFS (months)
`
`16.5
`
`8.3
`
`0.49
`[0.42–0.57]
`
`0.53
`[0.45–0.62]
`
`NR
`
`NR
`
`NR
`
`NR
`
`NR NR
`
`16.5
`
`8.2
`
`0.52
`[0.45–0.61]
`
`NR
`
`NR NR
`
`62 %
`
`36 %
`
`24 %
`
`16 %
`
`68 % 29 %
`
`NR
`
`NR
`
`NR
`
`NR
`
`NR NR
`
`NR
`
`PSA response rate
`
`Response rate by
`RECIST in patients
`with measurable
`disease
`
`NR not reported
`
`morbidity [42]. The rest of the patients who died in this time period, approximately
`7 % of the total patients, potentially reflect patients with disease of bad prognosis
`that was not captured by the prognostic factors applied in the inclusion criteria and
`who did not respond either to Abiraterone or to prednisone.
`
`Effect of Abiraterone on Secondary Endpoints
`
`In both COU-AA-301 and COU-AA-302 trials secondary endpoints were captured
`to substantiate the clinical effect of Abiraterone and all of them favoured AAP over
`PP (Table 9.4).
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`139
`
`Fig. 9.9 Overall survival curves in COU-AA-302. Noteworthy is the fact that the curves started
`separating only after 12–18 months of treatment. Adapted from Ryan et al. [42]. With permission
`from Elsevier
`
`Fatigue
`
`In COU-AA-301 fatigue was evaluated at each cycle of treatment by assessment of
`the Brief Fatigue Inventory questionnaire which evaluates fatigue intensity and
`fatigue interference with aspects of everyday life. Improvement of fatigue intensity
`and fatigue interference was significantly higher in AAP compared to PP (58.1 %
`vs. 40.3 % and 55 % vs. 38 % respectively). Additionally, in the AAP arm the time
`to improvement of fatigue was substantially shorter and the time to fatigue pro-
`gression delayed compared to the PP arm [43]. In COU-AA-302 fatigue was not
`prospectively captured.
`
`Pain Palliation and Progression
`
`Pain was evaluated throughout COU-AA-301 and COU-AA-302 by means of the
`BPI-SF. Pain intensity was defined as the score of the BPI-SF question 3 (worst
`pain in the previous 24 h) and pain interference was defined as the mean of all
`seven questions assessing pain interference with everyday activities on the BPI-SF.
`Additionally, in COU-AA-301 the mean analgesic use was scored as per WHO
`criteria. In a post hoc analysis of COU-AA-301 with the data available at the
`time-point before the crossover to active drug the median time to pain progression
`was not reached at that time point, yet significantly more patients in the AAP group
`had experienced pain palliation and improvement of pain interference than in the PP
`group and pain palliation occurred faster in AAP (Table 9.4) [44].
`
`

`
`140
`
`Z. Zafeiriou et al.
`
`In COU-AA-302 patients did not have significant pain at treatment initiation but
`AA delayed pain occurrence compared to placebo as indicated by the delayed time
`to all pain related parameters: median time to pain progression, median time to
`opiate use [41, 42], median time to progression of the pain interference with daily
`activities and the median time to progression of the worst pain [45] (Table 9.4).
`
`Skeletal Related Events
`
`In COU-AA-301 the proportion of patients with skeletal related events was similar
`in both groups but there was a significantly longer median time to skeletal related
`events in Abiraterone (25 m) compared to placebo (20.3 m)(Table 9.4) [44].
`Skeletal related events were not captured in COU-AA-302.
`
`Quality of Life
`
`In COU-AA-301 and 302 health related quality of life (HRQoL) was assessed by
`means of the Functional Assessment of Cancer Therapy-Prostate (FACT-P) ques-
`tionnaire which has subscales to assess different aspects of QoL. In COU-AA-301,
`AA improved quality of life in more patients (48 % vs. 32 %) and in a shorter time
`compared to prednisone and delayed functional status deterioration by 6 months
`despite the fact that prednisone was able to produce substantial improvements in
`QOL as well [46]. In the minimally symptomatic population of COU-AA-302, AA
`delayed median time to deterioration of quality of life both as measured by the
`FACT-P total score and by the prostate-cancer-specific subscale [45] (Table 9.4).
`
`Toxicity of Abiraterone
`
`AA is a well tolerated drug as indicated by the fact that in COU-AA-301 the
`frequency of adverse events (AE) leading to treatment discontinuation was lower in
`AAP compared to PP (19 % vs. 23 %) [38] and in COU-AA-302 [42] the same
`frequency was again at the level of 19 % for AAP but only 12 % for PP [41], the
`latter reflecting a population with less advanced and symptomatic disease.
`The most frequent adverse events in both trials were fatigue, back pain, con-
`stipation, diarrhoea, nausea, bone pain and arthralgia. These occurred in similar
`frequency in the experimental and control groups [38, 39, 41, 42] and constitute
`disease related symptoms rather than Abiraterone related AE (Table 9.6). A con-
`tribution of Abiraterone to arthralgia, fatigue and diarrhoea cannot be excluded as
`these AEs showed consistently a mildly increased frequency in both trials in the
`AAP over the PP arm (Table 9.6).
`
`

`
`9 Abiraterone for the Treatment of mCRPC
`
`141
`
`Table 9.6 Adverse events grade 1–4 in AA phase III trials
`
`Adverse event
`
`COU-AA-301
`
`COU-AA-302
`
`AAP (%)
`
`PP (%)
`
`AAP (%)
`
`PP (%)
`
`Anaemia
`
`Fatigue
`
`Back pain
`
`Arthralgia
`
`Nausea
`
`Vomiting
`
`Constipation
`
`Hot flush
`
`Diarrhoea
`
`Bone pain
`
`Pain in extremity
`
`Cough
`
`Urinary tract infection
`
`25
`
`47
`
`33
`
`30
`
`33
`
`24
`
`28
`
`NR
`
`20
`
`27
`
`20
`
`NR
`
`13
`
`Abbreviations: NR not reported
`Based on data from Refs. [38, 41, 42]
`
`28
`
`44
`
`36
`
`24
`
`33
`
`26
`
`32
`
`NR
`
`15
`
`30
`
`21
`
`NR
`
`7
`
`NR
`
`39
`
`32
`
`28
`
`22
`
`NR
`
`23
`
`22
`
`22
`
`20
`
`17
`
`17
`
`2
`
`NR
`
`34
`
`32
`
`24
`
`22
`
`NR
`
`19
`
`18
`
`18
`
`19
`
`16
`
`14
`
`<1
`
`On the other hand, AEs related to mineralocorticoid excess (ME) (hypokalemia,
`fluid retention, hypertension) were more frequent in AAP compared to PP in both
`trials [38, 42] (Table 9.7). ME observed in CRPC patients treated with single agent
`AA can be managed either by the addition of eplerenone, a mineralocorticoid
`receptor antagonist or by exogenous corticosteroids [17, 47] that inhibit secretion of
`corticotrophin-releasing hormone and ACTH, therefore exogenous corticosteroids
`are routinely co-administered with Abiraterone. Use of spironolactone for the
`management of ME should be avoided as it can act as an agonist for wildtype AR
`[35, 48]. Hypertension that arises despite steroid use can be managed symp-
`tomatically by addition of an antihypertensive agent but diuretics should be avoided
`as they may exacerbate Abiraterone induced hypokalemia. Hypokalemia can be
`treated by supplementation with potassium.
`Cardiac events were consistently more frequent in AAP rather than in PP in both
`trials although this difference did not reach statistical significance. Most frequent
`were grade 1 or 2 tachycardia and grade 3 or less atrial fibrillation [39] and fatal
`cardiac events were rare and approximately 1 % for each arm in both trials [38, 42]
`(Table 9.7). No effect of AA could be detected on the QTcF and QRS interval by a
`further small study evaluating ECG changes with AA [49].
`Early in the course of the COU-AA-301 trial an incident of grade 4 liver function
`test (LFT) elevation instigated patients’ vis

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