CLINICAL PHARMACOLOGY
Mechanism Of Action
Enzalutamide is an androgen receptor inhibitor that acts
on different steps in the androgen receptor signaling pathway. Enzalutamide has
been shown to competitively inhibit androgen binding to androgen receptors; and
consequently, inhibits nuclear translocation of androgen receptors and their
interaction with DNA. A major metabolite, N-desmethyl enzalutamide, exhibited
similar in vitro activity to enzalutamide. Enzalutamide decreased proliferation
and induced cell death of prostate cancer cells in vitro, and decreased tumor
volume in a mouse prostate cancer xenograft model.
Pharmacodynamics
Cardiac Electrophysiology
The effect of enzalutamide 160 mg/day at steady-state on
the QTc interval was evaluated in 796 patients with metastatic CRPC. No large
difference (i.e., greater than 20 ms) was observed between the mean QT interval
change from baseline in patients treated with XTANDI and that in patients
treated with placebo, based on the Fridericia correction method. However, small
increases in the mean QTc interval (i.e., less than 10 ms) due to enzalutamide
cannot be excluded due to limitations of the study design.
Pharmacokinetics
The pharmacokinetics of enzalutamide and its major active
metabolite (N-desmethyl enzalutamide) were evaluated in patients with
metastatic CRPC and healthy male volunteers. The plasma enzalutamide
pharmacokinetics are adequately described by a linear two-compartment model
with first-order absorption.
Absorption
Following oral administration (XTANDI 160 mg daily) in
patients with metastatic CRPC, the median time to reach maximum plasma
enzalutamide concentrations (Cmax) is 1 hour (range 0.5 to 3 hours). At
steady-state, the plasma mean Cmax values for enzalutamide and N-desmethyl
enzalutamide are 16.6 μg/mL (23% CV) and 12.7 μg/mL (30% CV), respectively,
and the plasma mean predose trough values are 11.4 μg/mL (26% CV) and 13.0
μg/mL (30% CV), respectively.
With the daily dosing regimen, enzalutamide steady-state
is achieved by Day 28, and enzalutamide accumulates approximately 8.3-fold
relative to a single dose. Daily fluctuations in enzalutamide plasma
concentrations are low (mean peak-to-trough ratio of 1.25). At steady-state,
enzalutamide showed approximately dose proportional pharmacokinetics over the
daily dose range of 30 to 360 mg.
A single 160 mg oral dose of XTANDI was administered to
healthy volunteers with a high-fat meal or in the fasted condition. A high-fat
meal did not alter the AUC to enzalutamide or N-desmethyl enzalutamide. The
results are summarized in Figure 1.
Distribution And Protein Binding
The mean apparent volume of distribution (V/F) of
enzalutamide in patients after a single oral dose is 110 L (29% CV).
Enzalutamide is 97% to 98% bound to plasma proteins,
primarily albumin. N-desmethyl enzalutamide is 95% bound to plasma proteins. In
vitro, there was no protein binding displacement between enzalutamide and other
highly protein bound drugs (warfarin, ibuprofen, and salicylic acid) at
clinically relevant concentrations.
Metabolism
Following single oral administration of 14C-enzalutamide
160 mg, plasma samples were analyzed for enzalutamide and its metabolites up to
77 days post dose. Enzalutamide, N-desmethyl enzalutamide, and a major inactive
carboxylic acid metabolite accounted for 88% of the 14C-radioactivity
in plasma, representing 30%, 49%, and 10%, respectively, of the total
14C-AUC0-inf.
In vitro, human CYP2C8 and CYP3A4 are responsible for the
metabolism of enzalutamide. Based on in vivo and in vitro data, CYP2C8 is
primarily responsible for the formation of the active metabolite (N-desmethyl
enzalutamide). In vitro data suggest that carboxylesterase 1 metabolizes
N-desmethyl enzalutamide and enzalutamide to the inactive carboxylic acid
metabolite.
In vitro, N-desmethyl enzalutamide is not a substrate of
human CYP1A1, CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C18, CYP2C19, CYP2D6,
CYP2E1 and CYP3A4/5.
Elimination
Enzalutamide is primarily eliminated by hepatic
metabolism. Following single oral administration of 14C-enzalutamide 160 mg,
85% of the radioactivity is recovered by 77 days post dose: 71% is recovered in
urine (including only trace amounts of enzalutamide and N-desmethyl
enzalutamide), and 14% is recovered in feces (0.4% of dose as unchanged enzalutamide
and 1% as N-desmethyl enzalutamide).
The mean apparent clearance (CL/F) of enzalutamide in
patients after a single oral dose is 0.56 L/h (range 0.33 to 1.02 L/h).
The mean terminal half-life (t½) for enzalutamide in
patients after a single oral dose is 5.8 days (range 2.8 to 10.2 days). Following
a single 160 mg oral dose of enzalutamide in healthy volunteers, the mean
terminal t½ for N-desmethyl enzalutamide is approximately 7.8 to 8.6 days.
Pharmacokinetics In Special Populations
Renal Impairment
A population pharmacokinetic analysis (based on
pre-existing renal function) was carried out with data from 59 healthy male
volunteers and 926 patients with metastatic CRPC enrolled in clinical trials,
including 512 with normal renal function (CrCL ≥ 90 mL/min), 332 with
mild renal impairment (CrCL 60 to < 90 mL/min), 88 with moderate renal impairment
(CrCL 30 to < 60 mL/min), and 1 with severe renal impairment (CrCL < 30
mL/min). The apparent clearance of enzalutamide was similar in patients with pre-existing
mild and moderate renal impairment (CrCL 30 to < 90 mL/min) compared to
patients and volunteers with normal renal function. The potential effect of
severe renal impairment or end-stage renal disease on enzalutamide
pharmacokinetics cannot be determined as clinical and pharmacokinetic data are available
from only one patient [see Use In Specific Populations].
Hepatic Impairment
The plasma pharmacokinetics of enzalutamide and
N-desmethyl enzalutamide were examined in volunteers with normal hepatic
function (N = 22) and with pre-existing mild (N = 8, Child-Pugh Class A)
moderate (N = 8, Child-Pugh Class B), or severe (N = 8, Child-Pugh Class C)
hepatic impairment. XTANDI was administered as a single 160 mg dose. The composite
AUC of enzalutamide plus N-desmethyl enzalutamide was similar in volunteers
with mild, moderate, or severe baseline hepatic impairment compared to
volunteers with normal hepatic function. The results are summarized in Figure 1
[see Use In Specific Populations].
Body Weight And Age
Population pharmacokinetic analyses showed that weight
(range: 46 to 163 kg) and age (range: 41 to 92 yr) do not have a clinically
meaningful influence on the exposure to enzalutamide.
Gender
The effect of gender on the pharmacokinetics of enzalutamide
has not been evaluated.
Race
The majority of XTANDI-treated patients in the randomized
clinical trials were Caucasian (74%). Based on pharmacokinetic data from
studies in Japanese and Chinese patients with prostate cancer, there were no
clinically relevant differences in exposure among the populations. There are
insufficient data to evaluate potential differences in the pharmacokinetics of
enzalutamide in other races.
Drug Interactions
Effect Of Other Drugs On XTANDI
In a drug-drug interaction trial in healthy volunteers, a
single 160 mg oral dose of XTANDI was administered alone or after multiple oral
doses of gemfibrozil (strong CYP2C8 inhibitor). Gemfibrozil increased the
AUC0-inf of enzalutamide plus N-desmethyl enzalutamide by 2.2-fold with minimal
effect on Cmax. The results are summarized in Figure 1 [see DOSAGE AND
ADMINISTRATION and DRUG INTERACTIONS].
In a drug-drug interaction trial in healthy volunteers, a
single 160 mg oral dose of XTANDI was administered alone or after multiple oral
doses of rifampin (strong CYP3A4 and moderate CYP2C8 inducer). Rifampin
decreased the AUC0-inf of enzalutamide plus N-desmethyl enzalutamide by 37%
with no effect on Cmax. The results are summarized in Figure 1 [see DOSAGE
AND ADMINISTRATION and DRUG INTERACTIONS].
In a drug-drug interaction trial in healthy volunteers, a
single 160 mg oral dose of XTANDI was administered alone or after multiple oral
doses of itraconazole (strong CYP3A4 inhibitor). Itraconazole increased the
AUC0-inf of enzalutamide plus N-desmethyl enzalutamide by 1.3-fold with no
effect on Cmax. The results are summarized in Figure 1.
Figure 1: Effects of Other Drugs and
Intrinsic/Extrinsic Factors on XTANDI
# PK parameters (Cmax and AUC0-inf) are for enzalutamide plus
N-desmethyl enzalutamide, except in the food-effect trial, where they are for enzalutamide
alone.
* See DOSAGE AND ADMINISTRATION.
Effect Of XTANDI On Other Drugs
In an in vivo phenotypic cocktail drug-drug interaction
trial in patients with metastatic CRPC, a single oral dose of the CYP probe
substrate cocktail (for CYP2C8, CYP2C9, CYP2C19, and CYP3A4) was administered
before and concomitantly with XTANDI (following at least 55 days of dosing at
160 mg daily). The results are summarized in Figure 2. Results showed that in
vivo, at steady-state, XTANDI is a strong CYP3A4 inducer and a moderate CYP2C9
and CYP2C19 inducer [see DRUG INTERACTIONS]. XTANDI did not cause
clinically meaningful changes in exposure to the CYP2C8 substrate.
In an in vivo phenotypic cocktail drug-drug interaction
trial in patients with CRPC, a single oral dose of the CYP probe substrate
cocktail for CYP1A2 and CYP2D6 was administered before and concomitantly with
XTANDI (following at least 49 days of dosing at 160 mg daily). The results are
summarized in Figure 2. Results showed that in vivo, at steady-state, XTANDI
did not cause clinically meaningful changes in exposure to the CYP1A2 or CYP2D6
substrates.
Figure 2: Effect of XTANDI on Other Drugs
*See DRUG INTERACTIONS.
In vitro, enzalutamide, N-desmethyl enzalutamide, and the
major inactive carboxylic acid metabolite caused direct inhibition of multiple
CYP enzymes including CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4/5; however,
subsequent clinical data showed that XTANDI is an inducer of CYP2C9, CYP2C19,
and CYP3A4 and had no clinically meaningful effect on CYP2C8 (see Figure 2). In
vitro, enzalutamide caused time-dependent inhibition of CYP1A2.
In vitro studies showed that enzalutamide induces CYP2B6
and CYP3A4 and does not induce CYP1A2 at therapeutically relevant
concentrations.
In vitro, enzalutamide, N-desmethyl enzalutamide, and the
major inactive carboxylic acid metabolite are not substrates for human
P-glycoprotein. In vitro, enzalutamide and N-desmethyl enzalutamide are
inhibitors of human P-glycoprotein, while the major inactive carboxylic acid
metabolite is not.
In vitro, enzalutamide and N-desmethyl enzalutamide do
not appear to be substrates of human breast cancer resistance protein (BCRP); however,
enzalutamide and N-desmethyl enzalutamide are inhibitors of human BCRP at
clinically relevant concentrations.
Clinical Studies
The efficacy and safety of XTANDI in 4692 patients with
CRPC were demonstrated in four randomized, multicenter clinical trials. All
patients continued on GnRH therapy or had prior bilateral orchiectomy. Patients
were allowed, but not required, to continue or initiate glucocorticoids.
AFFIRM (NCT00974311): XTANDI Versus Placebo In Metastatic
CRPC Following Chemotherapy
In AFFIRM, a total of 1199 patients who had received
prior docetaxel-based chemotherapy were randomized 2:1 to receive either XTANDI
orally at a dose of 160 mg once daily (N = 800) or placebo orally once daily (N
= 399). Study treatment continued until disease progression (evidence of
radiographic progression, a skeletal-related event, or clinical progression),
initiation of new systemic antineoplastic treatment, unacceptable toxicity, or
withdrawal. Patients with a previous history of seizure, taking medicines known
to decrease the seizure threshold, or with other risk factors for seizure were
not eligible [see WARNINGS AND PRECAUTIONS].
The following patient demographics and baseline disease
characteristics were balanced between the treatment arms. The median age was 69
years (range 41-92) and the racial distribution was 92.7% Caucasian, 3.9%
Black, 1.1% Asian, and 2.1% Other. Ninety-two percent of patients had an ECOG
performance status score of 0-1 and 28% had a mean Brief Pain Inventory score
of ≥ 4. Ninety-one percent of patients had metastases in bone and 23% had
visceral involvement in the lung and/or liver. Fifty-nine percent of patients
had radiographic evidence of disease progression and 41% had PSA-only progression
on study entry. All patients had received prior docetaxel-based therapy and 24%
had received two cytotoxic chemotherapy regimens. During the trial, 48% of
patients on the XTANDI arm and 46% of patients on the placebo arm received
glucocorticoids.
A statistically significant improvement in overall survival
was demonstrated at the pre-specified interim analysis at the time of 520
deaths in patients on the XTANDI arm compared to patients on the placebo arm
(Table 6 and Figure 3).
Table 6: Overall Survival of Patients Treated with
Either XTANDI or Placebo in AFFIRM
|
XTANDI
(N = 800) |
Placebo
(N = 399) |
Number of Deaths (%) |
308 (38.5) |
212 (53.1) |
Median Survival, months (95% CI) |
18.4 (17.3, NR) |
13.6 (11.3, 15.8) |
P-value1 |
p < 0.0001 |
Hazard Ratio (95% CI)2 |
0.63 (0.53, 0.75) |
NR = Not reached.
1. P-value is derived from a log-rank test stratified by baseline ECOG
performance status score (0-1 vs. 2) and mean baseline pain score (BPISF score
< 4 vs. ≥ 4).
2. Hazard Ratio is derived from a stratified proportional hazards model. Hazard
Ratio < 1 favors XTANDI. |
Figure 3: Kaplan-Meier Curves of Overall Survival in
AFFIRM
PREVAIL (NCT01212991): XTANDI Versus Placebo In Chemotherapy-naïve
Metastatic CRPC
In PREVAIL, 1717 chemotherapy-naïve patients were
randomized 1:1 to receive either XTANDI orally at a dose of 160 mg once daily
(N = 872) or placebo orally once daily (N = 845). Patients with visceral
metastases, patients with a history of mild to moderate heart failure (NYHA
class I or II), and patients taking medications associated with lowering the seizure
threshold were allowed. Patients with a previous history of seizure or a
condition that might predispose to seizure and patients with moderate or severe
pain from prostate cancer were excluded. Study treatment continued until
disease progression (evidence of radiographic progression, a skeletal-related
event, or clinical progression) and the initiation of a cytotoxic chemotherapy
or an investigational agent, unacceptable toxicity, or withdrawal. Overall
survival and radiographic progression-free survival (rPFS) were assessed.
Radiographic progression was assessed with the use of sequential imaging and
was defined by bone scan identification of 2 or more new bone lesions with
confirmation (Prostate Cancer Clinical Trials Working Group 2 criteria) and/or
Response Evaluation Criteria in Solid Tumors (RECIST v 1.1) criteria for
progression of soft tissue lesions. The primary analysis of rPFS utilized
centrally reviewed radiographic assessment of progression.
Patient demographics and baseline disease characteristics
were balanced between the treatment arms at entry. The median age was 71 years
(range 42-93) and the racial distribution was 77% Caucasian, 10% Asian, 2%
Black and 11% Other. The ECOG performance status score was 0 for 68% of
patients, and 1 for 32% of patients. Baseline pain assessment was 0-1 (asymptomatic)
in 67% of patients, and 2-3 (mildly symptomatic) in 32% of patients as defined
by the Brief Pain Inventory Short Form (worst pain over past 24 hours at study
entry). Fifty-four percent of patients had radiographic evidence of disease
progression and 43% had PSA-only progression. Twelve percent of patients had
visceral (lung and/or liver) disease involvement. During the study, 27% of
patients on the XTANDI arm and 30% of patients on the placebo arm received
glucocorticoids for varying reasons.
A statistically significant improvement in overall
survival was demonstrated at the pre-specified interim analysis, conducted
after 540 deaths in patients treated with XTANDI compared to those treated with
placebo (Table 6). Forty percent of XTANDI-treated and 70% of placebo-treated
patients received subsequent therapies for metastatic CRPC that may prolong overall
survival. An updated survival analysis was conducted when 784 deaths were
observed. The median follow-up time was 31 months. Results from this analysis
were consistent with those from the pre-specified interim analysis (Table 7,
Figure 4). At the updated analysis, 52% of XTANDI-treated and 81% of
placebo-treated patients had received subsequent therapies that may prolong
overall survival in metastatic CRPC. XTANDI was used as a subsequent therapy in
2% of XTANDI-treated patients and 29% of placebo-treated patients.
Table 7: Overall Survival of Patients Treated with
Either XTANDI or Placebo in PREVAIL
|
XTANDI
(N = 872) |
Placebo
(N = 845) |
Pre-specified Interim Analysis1 |
Number of Deaths (%) |
241 (28) |
299 (35) |
Median Survival, months (95% CI) |
32.4 (30.1, NR) |
30.2 (28.0, NR) |
P-value2 |
p < 0.0001 |
Hazard Ratio (95% CI)3 |
0.71 (0.60, 0.84) |
Updated Survival Analysis4 |
Number of Deaths (%) |
368 (42) |
416 (49) |
Median Survival, months (95% CI) |
35.3 (32.2, NR) |
31.3 (28.8, 34.2) |
Hazard Ratio (95% CI)3 |
0.77 (0.67, 0.88) |
NR = Not reached.
1. The data cutoff date is 16 Sep 2013.
2. P-value is derived from an unstratified log-rank test.
3. Hazard Ratio is derived from an unstratified proportional hazards model.
Hazard Ratio < 1 favors XTANDI.
4. The data cutoff date is 1 Jun 2014. The planned number of deaths for the
final overall survival analysis was ≥ 765. |
Figure 4: Kaplan-Meier Curves of Overall Survival in
PREVAIL
A statistically significant improvement in rPFS was
demonstrated in patients treated with XTANDI compared to patients treated with
placebo (Table 8, Figure 5).
Table 8: Radiographic Progression-free Survival of
Patients Treated with Either XTANDI or Placebo in PREVAIL
|
XTANDI
(N = 832) |
Placebo
(N = 801) |
Number of Progression or Deaths (%) |
118 (14) |
320 (40) |
Median rPFS (months) (95% CI) |
NR (13.8, NR) |
3.7 (3.6, 4.6) |
P-value1 |
p < 0.0001 |
Hazard Ratio (95% CI)2 |
0.17 (0.14, 0.21) |
NR = Not reached.
Note: As of the cutoff date for the rPFS analysis, 1633 patients had been
randomized.
1. P-value is derived from an unstratified log-rank test.
2. Hazard Ratio is derived from an unstratified proportional hazards model.
Hazard Ratio < 1 favors XTANDI. |
Figure 5: Kaplan-Meier Curves of Radiographic
Progression-free Survival in PREVAIL
Time to initiation of cytotoxic chemotherapy was
prolonged after XTANDI treatment, with a median of 28.0 months for patients on
the XTANDI arm versus a median of 10.8 months for patients on the placebo arm
[HR = 0.35 (95% CI: 0.30, 0.40), p < 0.0001].
The median time to first skeletal-related event was 31.1
months for patients on the XTANDI arm versus 31.3 months for patients on the
placebo arm [HR = 0.72 (95% CI: 0.61, 0.84), p < 0.0001]. A skeletal-related
event was defined as radiation therapy or surgery to bone for prostate cancer,
pathologic bone fracture, spinal cord compression, or change of antineoplastic
therapy to treat bone pain.
TERRAIN (NCT01288911): XTANDI Versus Bicalutamide In Chemotherapy-naïve
Metastatic CRPC
TERRAIN was conducted in 375 chemotherapy-naïve patients
who were randomized 1:1 to receive either XTANDI orally at a dose of 160 mg
once daily (N = 184) or bicalutamide orally at a dose of 50 mg once daily (N =
191). Patients with a previous history of seizure or a condition that might
predispose to seizure and patients with moderate to severe pain from prostate
cancer were excluded. Patients could have received prior bicalutamide, but
those whose disease had progressed on prior antiandrogen therapy (e.g.,
bicalutamide) were excluded. Study treatment continued until disease
progression (evidence of radiographic progression, a skeletal-related event),
the initiation of subsequent antineoplastic agent, unacceptable toxicity, or
withdrawal. Radiographic disease progression was assessed by Independent
Central Review (ICR) using the Prostate Cancer Clinical Trials Working Group 2
criteria and/or Response Evaluation Criteria in Solid Tumors (RECIST v 1.1)
criteria for progression of soft tissue lesions. Radiographic progression-free
survival (rPFS) was defined as the time from randomization to the first
objective evidence of radiographic progression as assessed by ICR or death,
whichever occurred first.
Patient demographics and baseline disease characteristics
were balanced between the treatment arms at entry. The median age was 71 years
(range 48-96) and the racial distribution was 93% Caucasian, 5% Black, 1% Asian
and 1% Other. The ECOG performance status score was 0 for 74% of patients and 1
for 26% of patients. Baseline pain assessment was 0-1 (asymptomatic) in 58% of
patients, and 2-3 (mildly symptomatic) in 36% of patients as defined by the
Brief Pain Inventory Short Form Question 3 (worst pain over past 24 hours at
study entry). Ninety-eight percent of patients had objective evidence of
disease progression at study entry. Forty-six percent of patients had received
prior treatment with bicalutamide while no patients received prior treatment
with XTANDI.
An improvement in rPFS was demonstrated in patients
treated with XTANDI compared to patients treated with bicalutamide (Table 9,
Figure 6).
Table 9: Â Radiographic Progression-free Survival of
Patients in TERRAIN
|
XTANDI
(N = 184) |
Bicalutamide
(N = 191) |
Number of Progression or Deaths (%) |
72 (39) |
74 (39) |
Median rPFS (months) (95% CI) |
19.5 (11.8, NR) |
13.4 (8.2, 16.4) |
Hazard Ratio (95% CI)1 |
0.60 (0.4 |
13, 0.83) |
NR = Not reached.
1. Hazard Ratio is derived from an unstratified proportional hazards model.
Hazard Ratio < 1 favors XTANDI |
Figure 6: Kaplan-Meier Curves of Radiographic
Progression-free Survival in TERRAIN
PROSPER (NCT02003924): XTANDI Versus Placebo In Non-metastatic
CRPC
PROSPER enrolled 1401 patients with non-metastatic CRPC
who were randomized 2:1 to receive either XTANDI orally at a dose of 160 mg
once daily (N = 933) or placebo orally once daily (N = 468). All patients in
the PROSPER trial received a gonadotropin-releasing hormone (GnRH) analog or
had a prior bilateral orchiectomy. Patients were stratified by Prostate
Specific Antigen (PSA) Doubling Time (PSADT) and the use of bone-targeting
agents. Patients were required to have a PSA doubling time ≤ 10 months,
PSA ≥ 2 ng/mL, and confirmation of non-metastatic disease by blinded independent
central review (BICR). PSA results were blinded and were not used for treatment
discontinuation. Patients randomized to either arm discontinued treatment for
radiographic disease progression confirmed by BICR, initiation of new
treatment, unacceptable toxicity, or withdrawal.
The following patient demographics and baseline
characteristics were balanced between the two treatment arms. The median age at
randomization was 74 years (range 50-95) and 23% were 80 years of age or older.
The racial distribution was 71% Caucasian, 16% Asian, and 2% Black. A majority
of patients had a Gleason score of 7 or higher (77%). The median PSADT was 3.7
months. Fifty-four percent (54%) of patients received prior treatment for
prostate cancer with either surgery or radiation. Sixty-three percent (63%) of
patients received prior treatment with an anti-androgen; 56% of patients
received bicalutamide and 11% of patients received flutamide. All patients had
an Eastern Cooperative Oncology Group Performance Status (ECOG PS) score of 0
or 1 at study entry.
The major efficacy outcome of the study was
metastasis-free survival (MFS), defined as the time from randomization to whichever
of the following occurred first 1) loco-regional and/or distant radiographic
progression per BICR or 2) death up to 112 days after treatment discontinuation
without evidence of radiographic progression. A statistically significant improvement
in MFS was demonstrated in patients randomized to receive XTANDI compared with
patients randomized to receive placebo. Consistent MFS results were observed
when considering only distant radiographic progression events or deaths
regardless of the cut-off date. Consistent MFS results were also observed in
pre-specified and stratified patient sub-groups of PSADT (< 6 months or
≥ 6 months) and use of a prior bone-targeting agent (yes or no). Overall
survival (OS) data were not mature at the time of final MFS analysis (28% of
the required number of events had been reported). The efficacy results for MFS
from PROSPER are summarized in Table 10 and Figure 7.
Table 10: Summary of Efficacy Results in PROSPER
(Intent-to-treat Population)
|
XTANDI
(N = 933) |
Placebo
(N = 468) |
Metastasis-free survival |
Number of Events (%) |
219 (23.5) |
228 (48.7) |
Median, months (95% CI)1 |
36.6 (33.1, NR) |
14.7 (14.2, 15.0) |
Hazard Ratio (95% CI)2 |
0.29 (0.24, 0.35) |
P-value3 |
p < 0.0001 |
NR = Not reached.
1. Based on Kaplan-Meier estimates.
2. Hazard Ratio is based on a Cox regression model (with treatment as the only
covariate) stratified by PSA doubling time and prior or concurrent use of a
bone-targeting agent. The HR is relative to placebo with < 1 favoring
XTANDI.
3. P-value is based on a stratified log-rank test by PSA doubling time (< 6
months, ≥ 6 months) and prior or concurrent use of a bonetargeting agent
(yes, no). |
Figure 7: Kaplan-Meier Curves of Metastasis-free Survival
in PROSPER
The primary efficacy outcome was supported by a
statistically significant delay in time to first use of new antineoplastic therapy
(TTA) for patients in the XTANDI arm compared to those in the placebo arm. The
median TTA was 39.6 months for patients on XTANDI and was 17.7 months for
patients on placebo (HR = 0.21; 95% CI: [0.17, 0.26], p < 0.0001).