CLINICAL PHARMACOLOGY
Mechanism Of Action
Umeclidinium is a long-acting
antimuscarinic agent, which is often referred to as an anticholinergic. It has
similar affinity to the subtypes of muscarinic receptors M1 to M5. In the
airways, it exhibits pharmacological effects through inhibition of M3 receptor
at the smooth muscle leading to bronchodilation. The competitive and reversible
nature of antagonism was shown with human and animal origin receptors and
isolated organ preparations. In preclinical in vitro as well as in vivo studies,
prevention of methacholine-and acetylcholine-induced bronchoconstrictive
effects was dose-dependent and lasted longer than 24 hours. The clinical relevance
of these findings is unknown. The bronchodilation following inhalation of
umeclidinium is predominantly a site-specific effect.
Pharmacodynamics
Cardiac Electrophysiology
QTc interval prolongation was studied in a double-blind,
multiple dose, placebo-and positive-controlled, crossover trial in 86 healthy
subjects. Following repeat doses of umeclidinium 500 mcg once daily (8 times
the recommended dosage) for 10 days, umeclidinium does not prolong QTc to any
clinically relevant extent.
Pharmacokinetics
Linear pharmacokinetics was observed for umeclidinium
(62.5 to 500 mcg).
Absorption
Umeclidinium plasma levels may not predict therapeutic
effect. Following inhaled administration of umeclidinium in healthy subjects, Cmax
occurred at 5 to 15 minutes. Umeclidinium is mostly absorbed from the lung
after inhaled doses with minimum contribution from oral absorption. Following
repeat dosing of inhaled INCRUSE ELLIPTA, steady state was achieved within 14
days with 1.8-fold accumulation.
Distribution
Following intravenous administration to healthy subjects,
the mean volume of distribution was 86 L. In vitro plasma protein binding in
human plasma was on average 89%.
Metabolism
In vitro data showed that umeclidinium is primarily
metabolized by the enzyme cytochrome P450 2D6 (CYP2D6) and is a substrate for
the P-glycoprotein (P-gp) transporter. The primary metabolic routes for
umeclidinium are oxidative (hydroxylation, O-dealkylation) followed by
conjugation (e.g., glucuronidation), resulting in a range of metabolites with
either reduced pharmacological activity or for which the pharmacological
activity has not been established. Systemic exposure to the metabolites is low.
Elimination
Following intravenous dosing with radiolabeled
umeclidinium, mass balance showed 58% of the radiolabel in the feces and 22% in
the urine. The excretion of the drug-related material in the feces following
intravenous dosing indicated elimination in the bile. Following oral dosing to
healthy male subjects, radiolabel recovered in feces was 92% of the total dose
and that in urine was less than 1% of the total dose, suggesting negligible
oral absorption. The effective half-life after once-daily dosing is 11 hours.
Special Populations
Population pharmacokinetic analysis showed no evidence of
a clinically significant effect of age (40 to 93 years) (Figure 1), gender (69%
male) (Figure 1), inhaled corticosteroid use (48%), or weight (34 to 161 kg) on
systemic exposure of umeclidinium. In addition, there was no evidence of a
clinically significant effect of race.
Hepatic Impairment: The impact of hepatic
impairment on the pharmacokinetics of INCRUSE ELLIPTA has been evaluated in
subjects with moderate hepatic impairment (Child-Pugh score of 7-9). There was
no evidence of an increase in systemic exposure to umeclidinium (Cmax and AUC) (Figure
1). There was no evidence of altered protein binding in subjects with moderate
hepatic impairment compared with healthy subjects. INCRUSE ELLIPTA has not been
evaluated in subjects with severe hepatic impairment.
Renal Impairment: The pharmacokinetics of INCRUSE
ELLIPTA has been evaluated in subjects with severe renal impairment (creatinine
clearance less than 30 mL/min). There was no evidence of an increase in
systemic exposure to umeclidinium (Cmax and AUC) (Figure 1). There was no
evidence of altered protein binding in subjects with severe renal impairment
compared with healthy subjects.
Figure 1: Impact of Intrinsic and Extrinsic Factors on
the Systemic Exposure of Umeclidinium
Drug Interactions
Umeclidinium and
P-glycoprotein Transporter: Umeclidinium is a substrate of P-gp. The effect of the
moderate P-gp transporter inhibitor verapamil (240 mg once daily) on the
steady-state pharmacokinetics of umeclidinium was assessed in healthy subjects.
No effect on umeclidinium Cmax was observed; however, an approximately 1.4-fold
increase in umeclidinium AUC was observed (Figure 1).
Umeclidinium and Cytochrome
P450 2D6: In vitro metabolism of umeclidinium is mediated primarily by CYP2D6.
However, no clinically meaningful difference in systemic exposure to
umeclidinium (500 mcg) (8 times the approved dose) was observed following repeat
daily inhaled dosing to normal (ultrarapid, extensive, and intermediate
metabolizers) and CYP2D6 poor metabolizer subjects (Figure 1).
Clinical Studies
The safety and efficacy of
umeclidinium 62.5 mcg were evaluated in 3 dose-ranging trials, 2
placebo-controlled clinical trials (one 12-week trial and one 24-week trial),
and a 12-month long-term safety trial. The efficacy of INCRUSE ELLIPTA is based
primarily on the dose-ranging trials in 624 subjects with COPD and the 2
placebo-controlled confirmatory trials in 1,738 subjects with COPD.
The safety and efficacy of
INCRUSE ELLIPTA in combination with an ICS/LABA were also evaluated in four
12-week clinical trials. The efficacy of INCRUSE ELLIPTA in combination with an
ICS/LABA is based on 1,637 subjects with COPD.
Dose-Ranging Trials
Dose selection for umeclidinium
in COPD was supported by a 7-day, randomized, double-blind, placebo-controlled,
crossover trial evaluating 4 doses of umeclidinium (15.6 to 125 mcg) or placebo
dosed once daily in the morning in 163 subjects with COPD. A dose ordering was observed,
with the 62.5-and 125-mcg doses demonstrating larger improvements in FEV1 over
24 hours compared with the lower doses of 15.6 and 31.25 mcg (Figure 2).
The differences in trough FEV1 from baseline after 7 days
for placebo and the 15.6-, 31.25-, 62.5-, and 125-mcg doses were -74 mL (95%
CI: -118, -31), 38 mL (95% CI: -6, 83), 27 mL (95% CI: -18, 72), 49 mL (95% CI:
6, 93), and 109 mL (95% CI: 65, 152), respectively. Two additional dose-ranging
trials in subjects with COPD demonstrated minimal additional benefit at doses
above 125 mcg. The dose-ranging results supported the evaluation of 2 doses of
umeclidinium, 62.5 and 125 mcg, in the confirmatory COPD trials to further
assess dose response.
Evaluations of dosing interval by comparing once-and
twice-daily dosing supported selection of a once-daily dosing interval for
further evaluation in the confirmatory COPD trials.
Figure 2: Adjusted Mean Change from Baseline in
Postdose Serial FEV1 (mL) on Days 1 and 7
Maintenance Treatment:
Confirmatory Trials
The clinical development
program for INCRUSE ELLIPTA included 2 randomized, double-blind,
placebo-controlled, parallel-group trials in subjects with COPD designed to
evaluate the efficacy of INCRUSE ELLIPTA on lung function. Trial 1 was a
24-week placebo-controlled trial, and Trial 2 was a 12-week placebo-controlled
trial. These trials treated subjects that had a clinical diagnosis of COPD,
were 40 years of age or older, had a history of smoking greater than or equal
to 10 pack-years, had a post-albuterol FEV1 less than or equal to 70% of
predicted normal values, had a ratio of FEV1/FVC of less than 0.7, and had a
Modified Medical Research Council (mMRC) score greater than or equal to 2.
Subjects in Trial 1 had a mean age of 63 years and an average smoking history
of 46 pack-years, with 50% identified as current smokers. At screening, the
mean postbronchodilator percent predicted FEV1 was 47% (range: 13% to 74%), the
mean postbronchodilator FEV1/FVC ratio was 0.47 (range: 0.20 to 0.74), and the
mean percent reversibility was 15% (range: -35% to 109%). Baseline demographics
and lung function for subjects in Trial 2 were similar to those in Trial 1.
Trial 1 evaluated umeclidinium
62.5 mcg and placebo. The primary endpoint was change from baseline in trough
(predose) FEV1 at Day 169 (defined as the mean of the FEV1 values obtained at
23 and 24 hours after the previous dose on Day 168) compared with placebo.
INCRUSE ELLIPTA 62.5 mcg demonstrated a larger increase in mean change from
baseline in trough (predose) FEV1 relative to placebo (Table 2). Similar
results were obtained from Trial 2.
Table 2: Least Squares Mean Change from Baseline in
Trough FEV1 (mL) at Day 169 in the Intent-to-Treat Population (Trial 1)
Treatment |
n |
Trough FEV1 (mL) at Day 169 |
Difference from Placebo (95% CI)
n = 280 |
INCRUSE ELLIPTA |
n = 418 |
115 (76, 155) |
n = Number in intent-to-treat
population. |
Serial spirometric evaluations
throughout the 24-hour dosing interval were performed in a subset of subjects
(n = 54, umeclidinium 62.5 mcg; n = 36, placebo) at Days 1, 84, and 168 in
Trial 1, and for all patients at Days 1 and 84 in Trial 2. Results from Trial 1
at Day 1 and Day 168 are shown in Figure 3.
Figure 3: Least Squares (LS) Mean Change from Baseline
in FEV1 (mL) over Time (0-24 h) on Days 1 and 168 (Trial 1 Subset Population)
In Trial 1, the mean peak FEV1 (over
the first 6 hours relative to baseline) at Day 1 and at Day 168 for the group
receiving umeclidinium 62.5 mcg compared with placebo was 126 and 130 mL,
respectively.
Health-related quality of life
was measured using St. George's Respiratory Questionnaire (SGRQ). Umeclidinium
demonstrated an improvement in mean SGRQ total score compared with placebo
treatment at Day 168: -4.69 (95% CI: -7.07,-2.31). The proportion of patients
with a clinically meaningful decrease (defined as a decrease of at least 4
units from baseline) at Week 24 was greater for INCRUSE ELLIPTA 62.5 mcg (42%;
172/410) compared with placebo (31%; 86/274).
Maintenance Treatment:
Combination With An ICS/LABA Trials
The efficacy of INCRUSE ELLIPTA
in combination with an ICS/LABA was evaluated in 4 randomized, double-blind,
parallel-group trials in subjects with COPD. These trials, all of similar study
design, were of 12-weeks' treatment duration. Subjects were randomized to
INCRUSE ELLIPTA 62.5 mcg + ICS/LABA or placebo + ICS/LABA. Entry criteria for
subjects enrolled in these trials were similar to those described above in
Section 14.2. The primary endpoint for these trials was change from baseline in
trough (predose) FEV1 at Day 85 (defined as the mean of the FEV1 values
obtained at 23 and 24 hours after the previous dose on Day 84). Baseline FEV1 was
measured while subjects were on background ICS/LABA.
Combination With Fluticasone
Furoate + Vilanterol
Trials 1 and 2 randomized
subjects to INCRUSE ELLIPTA 62.5 mcg + FF/VI 100 mcg/25 mcg administered once
daily or placebo + FF/VI 100 mcg/25 mcg administered once daily. Trial
population demographics and results for Trials 1 and 2 were similar; therefore,
only Trial 1 results are presented below.
Subjects in Trial 1 across all
treatment arms had a mean age of 64 years and an average smoking history of 50
pack-years, with 42% identified as current smokers. At screening, the mean postbronchodilator
percent predicted FEV1 was 45% (range: 13% to 76%), the mean postbronchodilator
FEV1/FVC ratio was 0.48 (range: 0.22 to 0.70), and the mean percent
reversibility was 14% (range: -20% to 71%).
The primary endpoint was change from baseline in trough
(predose) FEV1 at Day 85 (defined as the mean of the FEV1 values obtained at 23
and 24 hours after the previous dose on Day 84) compared with placebo (INCRUSE
ELLIPTA + FF/VI vs. placebo + FF/VI). INCRUSE ELLIPTA + FF/VI demonstrated a
larger mean change from baseline in trough (predose) FEV1 relative to placebo +
FF/VI (Table 3).
Table 3: Least Squares Mean Change from Baseline in
Trough FEV1 (mL) at Day 85 in the Intent-to-Treat Population (Trial 1)
Treatment |
n |
Trough FEV1 (mL) at Day 85 |
Difference from Placebo + FF/VI (95% CI)
n = 206 |
INCRUSE ELLIPTA + FF/VI |
n = 206 |
124 (93, 154) |
FF/VI = Fluticasone
furoate/vilanterol.
n = Number in intent-to-treat population. |
Combination With Fluticasone Propionate + Salmeterol
Trials 3 and 4 randomized subjects to INCRUSE ELLIPTA 62.5 mcg + FP/SAL 250 mcg/50 mcg or placebo +
FP/SAL 250 mcg/50 mcg. The treatments with INCRUSE ELLIPTA and placebo were
administered once daily, while the FP/SAL treatment was administered twice
daily. Trial population demographics and results for Trials 3 and 4 were
similar; therefore, only Trial 3 results are presented below.
Subjects in Trial 3 across all
treatment arms had a mean age of 63 years and an average smoking history of 50
pack-years, with 54% identified as current smokers. At screening, the mean postbronchodilator
percent predicted FEV1 was 47% (range: 12% to 70%), the mean postbronchodilator
FEV1/FVC ratio was 0.47 (range: 0.22 to 0.69), and the mean percent
reversibility was 16% (range: -36% to 79%).
The primary endpoint was change
from baseline in trough (predose) FEV1 at Day 85 (defined as the mean of the
FEV1 values obtained at 23 and 24 hours after the previous dose on Day 84)
compared with placebo (INCRUSE ELLIPTA + FP/SAL vs. placebo + FP/SAL). INCRUSE
ELLIPTA + FP/SAL demonstrated a larger mean change from baseline in trough
(predose) FEV1 relative to placebo + FP/SAL (Table 4).
Table 4: Least Squares Mean Change from Baseline in
Trough FEV1 (mL) at Day 85 in the Intent-to-Treat Population (Trial 3)
Treatment |
n |
Trough FEV1 (mL) at Day 85 |
Difference from Placebo + FP/SAL (95% CI)
n = 205 |
INCRUSE ELLIPTA+FP/SAL |
n = 204 |
147 (107, 187) |
FP/SAL = Fluticasone propionate/salmeterol.
n = Number in intent-to-treat population. |