CLINICAL PHARMACOLOGY
Mechanism Of Action
Abiraterone acetate (ZYTIGA) is
converted in vivo to abiraterone, an androgen biosynthesis inhibitor, that
inhibits 17 α-hydroxylase/C17,20-lyase (CYP17). This enzyme is expressed
in testicular, adrenal, and prostatic tumor tissues and is required for
androgen biosynthesis.
CYP17 catalyzes two sequential
reactions: 1) the conversion of pregnenolone and progesterone to their
17α-hydroxy derivatives by 17α-hydroxylase activity and 2) the
subsequent formation of dehydroepiandrosterone (DHEA) and
androstenedione, respectively, by C17, 20 lyase activity. DHEA and
androstenedione are androgens and are precursors of testosterone. Inhibition of
CYP17 by abiraterone can also result in increased mineralocorticoid production
by the adrenals [see WARNINGS AND PRECAUTIONS].
Androgen sensitive prostatic carcinoma responds to
treatment that decreases androgen levels. Androgen deprivation therapies, such
as treatment with GnRH agonists or orchiectomy, decrease androgen production in
the testes but do not affect androgen production by the adrenals or in the
tumor.
ZYTIGA decreased serum testosterone and other androgens
in patients in the placebo-controlled clinical trial. It is not necessary to
monitor the effect of ZYTIGA on serum testosterone levels.
Changes in serum prostate specific antigen (PSA) levels
may be observed but have not been shown to correlate with clinical benefit in
individual patients.
Pharmacodynamics
Cardiac Electrophysiology
In a multi-center, open-label, single-arm trial, 33
patients with metastatic CRPC received ZYTIGA orally at a dose of 1,000 mg once
daily at least 1 hour before or 2 hours after a meal in combination with
prednisone 5 mg orally twice daily. Assessments up to Cycle 2 Day 2 showed no
large changes in the QTc interval (i.e., >20 ms) from baseline. However,
small increases in the QTc interval (i.e., <10 ms) due to abiraterone
acetate cannot be excluded due to study design limitations.
Pharmacokinetics
Following administration of abiraterone acetate, the
pharmacokinetics of abiraterone and abiraterone acetate have been studied in
healthy subjects and in patients with metastatic CRPC. In vivo, abiraterone
acetate is converted to abiraterone. In clinical studies, abiraterone acetate
plasma concentrations were below detectable levels (<0.2 ng/mL) in >99%
of the analyzed samples.
Absorption
Following oral administration of abiraterone acetate to
patients with metastatic CRPC, the median time to reach maximum plasma
abiraterone concentrations is 2 hours. Abiraterone accumulation is observed at
steady-state, with a 2-fold higher exposure (steady-state AUC) compared to a
single 1,000 mg dose of abiraterone acetate.
At the dose of 1,000 mg daily in patients with metastatic
CRPC, steady-state values (mean ± SD) of Cmax were 226 ± 178 ng/mL and of AUC
were 993 ± 639 ng.hr/mL. No major deviation from dose proportionality was
observed in the dose range of 250 mg to 1,000 mg. However, the exposure was not
significantly increased when the dose was doubled from 1,000 to 2,000 mg (8%
increase in the mean AUC).
Systemic exposure of abiraterone is increased when
abiraterone acetate is administered with food. In healthy subjects abiraterone
Cmax and AUC0-∞ were approximately 7-and 5-fold higher, respectively,
when a single dose of abiraterone acetate was administered with a low-fat meal
(7% fat, 300 calories) and approximately 17-and 10-fold higher, respectively,
when a single dose of abiraterone acetate was administered with a high-fat (57%
fat, 825 calories) meal compared to overnight fasting. Abiraterone AUC0-∞
was approximately 7-fold or 1.6-fold higher, respectively, when a single dose
of abiraterone acetate was administered 2 hours after or 1 hour before a medium
fat meal (25% fat, 491 calories) compared to overnight fasting.
Systemic exposures of abiraterone in patients with
metastatic CRPC, after repeated dosing of abiraterone acetate were similar when
abiraterone acetate was taken with low-fat meals for 7 days and increased
approximately 2-fold when taken with high-fat meals for 7 days compared to when
taken at least 2 hours after a meal and at least 1 hour before a meal for 7
days.
Given the normal variation in the content and composition
of meals, taking ZYTIGA with meals has the potential to result in increased and
highly variable exposures. Therefore, ZYTIGA must be taken on an empty stomach,
at least one hour before or at least two hours after a meal. The tablets should
be swallowed whole with water [see DOSAGE AND ADMINISTRATION].
Distribution And Protein Binding
Abiraterone is highly bound (>99%) to the human plasma
proteins, albumin and alpha-1 acid glycoprotein. The apparent steady-state
volume of distribution (mean ± SD) is 19,669 ± 13,358 L. In vitro studies show
that at clinically relevant concentrations, abiraterone acetate and abiraterone
are not substrates of P-glycoprotein (P-gp) and that abiraterone acetate is an
inhibitor of P-gp.
Metabolism
Following oral administration of 14C-abiraterone
acetate as capsules, abiraterone acetate is hydrolyzed to abiraterone (active
metabolite). The conversion is likely through esterase activity (the esterases
have not been identified) and is not CYP mediated. The two main circulating
metabolites of abiraterone in human plasma are abiraterone sulphate (inactive)
and N-oxide abiraterone sulphate (inactive), which account for about 43% of
exposure each. CYP3A4 and SULT2A1 are the enzymes involved in the formation of
N-oxide abiraterone sulphate and SULT2A1 is involved in the formation of
abiraterone sulphate.
Excretion
In patients with metastatic CRPC, the mean terminal
half-life of abiraterone in plasma (mean ± SD) is 12 ± 5 hours. Following oral
administration of 14C-abiraterone acetate, approximately 88% of the
radioactive dose is recovered in feces and approximately 5% in urine. The major
compounds present in feces are unchanged abiraterone acetate and abiraterone
(approximately 55% and 22% of the administered dose, respectively).
Patients With Hepatic Impairment
The pharmacokinetics of abiraterone was examined in
subjects with baseline mild (N=8) or moderate (N=8) hepatic impairment
(Child-Pugh Class A and B, respectively) and in 8 healthy control subjects with
normal hepatic function. Systemic exposure to abiraterone after a single oral
1,000 mg dose given under fasting conditions increased approximately 1.1-fold
and 3.6-fold in subjects with mild and moderate baseline hepatic impairment,
respectively. The mean half-life of abiraterone is prolonged to approximately
18 hours in subjects with mild hepatic impairment and to approximately 19 hours
in subjects with moderate hepatic impairment.
In another trial, the pharmacokinetics of abiraterone
were examined in subjects with baseline severe (N=8) hepatic impairment
(Child-Pugh Class C) and in 8 healthy control subjects with normal hepatic
function. The systemic exposure (AUC) of abiraterone increased by approximately
7-fold in subjects with severe baseline hepatic impairment compared to subjects
with normal hepatic function. In addition, the mean protein binding was found to
be lower in the severe hepatic impairment group compared to the normal hepatic
function group, which resulted in a two-fold increase in the fraction of free
drug in patients with severe hepatic impairment [see DOSAGE AND
ADMINISTRATION and Use In Specific Populations].
Patients With Renal Impairment
The pharmacokinetics of abiraterone were examined in
patients with end-stage renal disease (ESRD) on a stable hemodialysis schedule
(N=8) and in matched control subjects with normal renal function (N=8). In the
ESRD cohort of the trial, a single 1,000 mg ZYTIGA dose was given under fasting
conditions 1 hour after dialysis, and samples for pharmacokinetic analysis were
collected up to 96 hours post dose. Systemic exposure to abiraterone after a
single oral 1,000 mg dose did not increase in subjects with end-stage renal
disease on dialysis, compared to subjects with normal renal function [see Use
In Specific Populations].
Drug Interactions
In vitro studies with human hepatic microsomes showed that
abiraterone has the potential to inhibit CYP1A2, CYP2D6, CYP2C8 and to a lesser
extent CYP2C9, CYP2C19 and CYP3A4/5.
In an in vivo drug-drug interaction trial, the Cmax and
AUC of dextromethorphan (CYP2D6 substrate) were increased 2.8-and 2.9-fold,
respectively when dextromethorphan 30 mg was given with abiraterone acetate
1,000 mg daily (plus prednisone 5 mg twice daily). The AUC for dextrorphan, the
active metabolite of dextromethorphan, increased approximately 1.3 fold [see DRUG
INTERACTIONS].
In a clinical study to determine the effects of
abiraterone acetate 1,000 mg daily (plus prednisone 5 mg twice daily) on a
single 100 mg dose of the CYP1A2 substrate theophylline, no increase in
systemic exposure of theophylline was observed.
Abiraterone is a substrate of CYP3A4, in vitro. In a
clinical pharmacokinetic interaction study of healthy subjects pretreated with
a strong CYP3A4 inducer (rifampin, 600 mg daily for 6 days) followed by a
single dose of abiraterone acetate 1,000 mg, the mean plasma AUC∞ of
abiraterone was decreased by 55% [see DRUG INTERACTIONS].
In a separate clinical pharmacokinetic interaction study
of healthy subjects, co-administration of ketoconazole, a strong inhibitor of
CYP3A4, had no clinically meaningful effect on the pharmacokinetics of
abiraterone [see DRUG INTERACTIONS].
In a CYP2C8 drug-drug interaction trial in healthy
subjects, the AUC of pioglitazone was increased by 46% when pioglitazone was
given together with a single dose of 1,000 mg abiraterone acetate [see DRUG
INTERACTIONS].
In vitro, abiraterone and its major metabolites were
shown to inhibit the hepatic uptake transporter OATP1B1. There are no clinical
data available to confirm transporter based interaction.
Animal Toxicology And/Or Pharmacology
A dose-dependent increase in cataracts was observed in
rats after daily oral abiraterone acetate administration for 26 weeks starting
at ≥50 mg/kg/day (similar to the human clinical exposure based on AUC).
In a 39-week monkey study with daily oral abiraterone acetate administration, no
cataracts were observed at higher doses (2 times greater than the clinical
exposure based on AUC).
Clinical Studies
The efficacy and safety of ZYTIGA with prednisone was
established in three randomized placebo-controlled international clinical
studies. All patients in these studies received a GnRH analog or had prior
bilateral orchiectomy. Patients with prior ketoconazole treatment for prostate
cancer and a history of adrenal gland or pituitary disorders were excluded from
these trials. Concurrent use of spironolactone was not allowed during the study
period.
COU-AA-301: Patients With Metastatic CRPC Who Had
Received Prior Docetaxel Chemotherapy
In COU-AA-301 (NCT00638690), a total of 1195 patients
were randomized 2:1 to receive either ZYTIGA orally at a dose of 1,000 mg once
daily in combination with prednisone 5 mg orally twice daily (N=797) or placebo
once daily plus prednisone 5 mg orally twice daily (N=398). Patients randomized
to either arm were to continue treatment until disease progression (defined as
a 25% increase in PSA over the patient’s baseline/nadir together with
protocol-defined radiographic progression and symptomatic or clinical
progression), initiation of new treatment, unacceptable toxicity or withdrawal.
The following patient demographics and baseline disease
characteristics were balanced between the treatment arms. The median age was 69
years (range 39-95) and the racial distribution was 93% Caucasian, 3.6% Black,
1.7% Asian, and 1.6% Other. Eighty-nine percent of patients enrolled had an
ECOG performance status score of 0-1 and 45% had a Brief Pain Inventory-Short
Form score of ≥4 (patient’s reported worst pain over the previous 24
hours). Ninety percent of patients had metastases in bone and 30% had visceral
involvement. Seventy percent of patients had radiographic evidence of disease
progression and 30% had PSA-only progression. Seventy percent of patients had
previously received one cytotoxic chemotherapy regimen and 30% received two
regimens.
The protocol pre-specified interim analysis was conducted
after 552 deaths and showed a statistically significant improvement in overall
survival (OS) in patients treated with ZYTIGA with prednisone compared to
patients in the placebo with prednisone arm (Table 9 and Figure 1). An updated
survival analysis was conducted when 775 deaths (97% of the planned number of
deaths for final analysis) were observed. Results from this analysis were
consistent with those from the interim analysis (Table 7).
Table 7: Overall Survival of Patients Treated with
Either ZYTIGA or Placebo in Combination with Prednisone in COU-AA-301
(Intent-to-Treat Analysis)
Primary Survival Analysis |
ZYTIGA with Prednisone
(N=797) |
Placebo with Prednisone
(N=398) |
Deaths (%) |
333 (42%) |
219 (55%) |
Median survival (months) (95% CI) |
14.8 (14.1, 15.4) |
10.9 (10.2, 12.0) |
p-value1 |
<0.0001 |
Hazard ratio (95% CI)2 |
0.646 (0.543, 0.768) |
Updated Survival Analysis |
Deaths (%) |
501 (63%) |
274 (69%) |
Median survival (months) (95% CI) |
15.8 (14.8, 17.0) |
11.2 (10.4, 13.1) |
Hazard ratio (95% CI)2 |
0.740 (0.638, 0.859) |
1 p-value is derived from a log-rank test
stratified by ECOG performance status score (0-1 vs. 2), pain score (absent vs.
present), number of prior chemotherapy regimens (1 vs. 2), and type of disease
progression (PSA only vs. radiographic).
2 Hazard Ratio is derived from a stratified proportional hazards
model. Hazard ratio <1 favors ZYTIGA with prednisone. |
Figure 1: Kaplan-Meier Overall Survival Curves in
COU-AA-301 (Intent-to-Treat Analysis)
COU-AA-302: Patients With
Metastatic CRPC Who Had Not Received Prior Cytotoxic Chemotherapy
In COU-AA-302 (NCT00887198),
1088 patients were randomized 1:1 to receive either ZYTIGA orally at a dose of
1,000 mg once daily (N=546) or Placebo orally once daily (N=542). Both arms
were given concomitant prednisone 5 mg twice daily. Patients continued
treatment until radiographic or clinical (cytotoxic chemotherapy, radiation or
surgical treatment for cancer, pain requiring chronic opioids, or ECOG
performance status decline to 3 or more) disease progression, unacceptable
toxicity or withdrawal. Patients with moderate or severe pain, opiate use for
cancer pain, or visceral organ metastases were excluded.
Patient demographics were balanced between the treatment
arms. The median age was 70 years. The racial distribution of patients treated
with ZYTIGA was 95% Caucasian, 2.8% Black, 0.7% Asian and 1.1% Other. The ECOG
performance status was 0 for 76% of patients, and 1 for 24% of patients.
Co-primary efficacy endpoints were overall survival and radiographic
progression-free survival (rPFS). Baseline pain assessment was 0-1
(asymptomatic) in 66% of patients and 23 (mildly symptomatic) in 26% of
patients as defined by the Brief Pain Inventory-Short Form (worst pain over the
last 24 hours).
Radiographic progression-free survival was assessed with
the use of sequential imaging studies and was defined by bone scan
identification of 2 or more new bone lesions with confirmation (Prostate Cancer
Working Group 2 criteria) and/or modified Response Evaluation Criteria In Solid
Tumors (RECIST) criteria for progression of soft tissue lesions. Analysis of
rPFS utilized centrally-reviewed radiographic assessment of progression.
The planned final analysis for OS, conducted after 741
deaths (median follow up of 49 months) demonstrated a statistically significant
OS improvement in patients treated with ZYTIGA with prednisone compared to
those treated with placebo with prednisone (Table 8 and Figure 2). Sixty-five
percent of patients on the ZYTIGA arm and 78% of patients on the placebo arm
used subsequent therapies that may prolong OS in metastatic CRPC. ZYTIGA was
used as a subsequent therapy in 13% of patients on the ZYTIGA arm and 44% of
patients on the placebo arm.
Table 8: Overall Survival of Patients Treated with
Either ZYTIGA or Placebo in Combination with Prednisone in COU-AA-302
(Intent-to-Treat Analysis)
|
ZYTIGA with Prednisone
(N=546) |
Placebo with Prednisone
(N=542) |
Overall Survival |
Deaths (%) |
354 (65%) |
219 (55%) |
Median survival (months) (95% CI) |
34.7 (32.7, 36.8) |
30.3 (28.7, 33.3) |
p-value1 |
0.0033 |
Hazard ratio2 (95% CI) |
0.81 (0.70, 0.93) |
1 p-value is derived from a log-rank test
stratified by ECOG performance status score (0 vs. 1).
2 Hazard Ratio is derived from a stratified proportional hazards
model. Hazard ratio <1 favors ZYTIGA with prednisone. |
Figure 2: Kaplan Meier
Overall Survival Curves in COU-AA-302
At the pre-specified rPFS
analysis, 150 (28%) patients treated with ZYTIGA with prednisone and 251 (46%)
patients treated with placebo with prednisone had radiographic progression. A
significant difference in rPFS between treatment groups was observed (Table 9
and Figure 3).
Table 9: Radiographic Progression-free Survival of
Patients Treated with Either ZYTIGA or Placebo in Combination with Prednisone
in COU-AA-302 (Intent-to-Treat Analysis)
|
ZYTIGA with Prednisone
(N=546) |
Placebo with Prednisone
(N=542) |
Radiographic Progression-free Survival |
Progression or death |
150 (28%) |
251 (46%) |
Median rPFS (months) |
NR |
8.28 |
(95% CI) |
(11.66, NR) |
(8.12, 8.54) |
p-value1 |
<0.0001 |
Hazard ratio2 (95% CI) |
0.425 (0.347, 0.522) |
NR=Not reached.
1 p-value is derived from a log-rank test stratified by ECOG
performance status score (0 vs. 1).
2 Hazard Ratio is derived from a stratified proportional hazards
model. Hazard ratio <1 favors ZYTIGA with prednisone. |
Figure 3: Kaplan Meier
Curves of Radiographic Progression-free Survival in COU-AA-302 (Intent-to-Treat
Analysis)
The primary efficacy analyses
are supported by the following prospectively defined endpoints. The median time
to initiation of cytotoxic chemotherapy was 25.2 months for patients in the
ZYTIGA arm and 16.8 months for patients in the placebo arm (HR=0.580; 95% CI:
[0.487, 0.691], p < 0.0001).
The median time to opiate use
for prostate cancer pain was not reached for patients receiving ZYTIGA and was
23.7 months for patients receiving placebo (HR=0.686; 95% CI: [0.566, 0.833],
p=0.0001). The time to opiate use result was supported by a delay in patient
reported pain progression favoring the ZYTIGA arm.
LATITUDE: Patients With
Metastatic High-Risk CSPC
In LATITUDE (NCT01715285), 1199
patients with metastatic high-risk CSPC were randomized 1:1 to receive either
ZYTIGA orally at a dose of 1,000 mg once daily with prednisone 5 mg once daily
(N=597) or placebo orally once daily (N=602). High-risk disease was defined as
having at least two of three risk factors at baseline: a total Gleason score of
≥8, presence of ≥3 lesions on bone scan, and evidence of measurable
visceral metastases. Patients with significant cardiac, adrenal, or hepatic
dysfunction were excluded. Patients continued treatment until radiographic or
clinical disease progression, unacceptable toxicity, withdrawal or death.
Clinical progression was defined as the need for cytotoxic chemotherapy,
radiation or surgical treatment for cancer, pain requiring chronic opioids, or
ECOG performance status decline to≥3.
Patient demographics were balanced between the treatment
arms. The median age was 67 years among all randomized subjects. The racial
distribution of patients treated with ZYTIGA was 69% Caucasian, 2.5% Black, 21%
Asian, and 8.1% Other. The ECOG performance status was 0 for 55%, 1 for 42%,
and 2 for 3.5% of patients. Baseline pain assessment was 0-1 (asymptomatic) in
50% of patients, 2-3 (mildly symptomatic) in 23% of patients, and ≥4 in
28% of patients as defined by the Brief Pain Inventory-Short Form (worst pain
over the last 24 hours).
A major efficacy outcome was overall survival. The
pre-specified interim analysis after 406 deaths showed a statistically
significant improvement in OS in patients on ZYTIGA with prednisone compared to
those on placebos. Twenty-one percent of patients on the ZYTIGA arm and 41% of
patients on the placebos arm received subsequent therapies that may prolong OS
in metastatic CRPC. An updated survival analysis was conducted when 618 deaths
were observed. The median follow-up time was 52 months. Results from this
analysis were consistent with those from the pre-specified interim analysis
(Table 10 and Figure 4). At the updated analysis, 29% of patients on the ZYTIGA
arm and 45% of patients on the placebos arm received subsequent therapies that
may prolong OS in metastatic CRPC.
Table 10: Overall Survival of Patients Treated with
Either ZYTIGA or Placebos in LATITUDE (Intent-to-Treat Analysis)
|
ZYTIGA with Prednisone
(N=597) |
Placebos
(N=602) |
Overall Survival1 |
|
Deaths (%) |
169 (28%) |
237 (39%) |
Median survival (months) (95% CI) |
NE (NE, NE) |
34.7 (33.1, NE) |
p-value2 |
<0.0001 |
Hazard ratio (95% CI)3 |
0.62 (0.51, 0.76) |
Updated Overall Survival |
|
Deaths (%) |
275 (46%) |
343 (57%) |
Median survival (months)(95% CI) |
53.3(48.2, NE) |
36.5(33.5, 40.0) |
Hazard ratio (95% CI)3 |
0.66 (0.56, 0.78) |
NE=Not estimable
1 This is based on the pre-specified interim analysis
2 p value is from log-rank test stratified by ECOG PS score (0/1 or
2) and visceral (absent or present).
3 Hazard Ratio is derived from a stratified proportional hazards
model. Hazard ratio <1 favors ZYTIGA with prednisone. |
Figure 4: Kaplan-Meier Plot
of Overall Survival; Intent-to-treat Population in LATITUDE Updated Analysis
The major efficacy outcome was
supported by a statistically significant delay in time to initiation of
chemotherapy for patients in the ZYTIGA arm compared to those in the placebos
arm. The median time to initiation of chemotherapy was not reached for patients
on ZYTIGA with prednisone and was 38.9 months for patients on placebos (HR =
0.44; 95% CI: [0.35, 0.56], p < 0.0001).