CLINICAL PHARMACOLOGY
Mechanism Of Action
The mechanism of action of pitolisant in excessive
daytime sleepiness (EDS) in adult patients with narcolepsy is unclear. However,
its efficacy could be mediated through its activity as an antagonist/inverse
agonist at histamine-3 (H3) receptors.
Pharmacodynamics
Pitolisant binds to H3 receptors with a high affinity (Ki
= 1nM) and has no appreciable binding to other histamine receptors (H1, H2, or
H4 receptors; Ki >10 μM).
Cardiac Electrophysiology
WAKIX at the highest recommended dosage (i.e., 35.6 mg
daily) led to a QTc increase of 4.2 msec. Exposures 3.8-fold higher than
achieved at the highest recommended dose increase QTc 16 msec (mean) [see WARNINGS
AND PRECAUTIONS].
Pharmacokinetics
Following oral administration of pitolisant 35.6 mg once
daily, the steady state Cmax and AUC are 73 ng/mL (range: 49.2 to 126 ng/mL)
and 812 ng*hr/mL (range: 518 to 1468 ng*hr/mL), respectively. Pitolisant exposure
(Cmax and AUC) increases proportionally with dose and steady state is reached
by day 7.
Absorption
The median time to maximum plasma concentration (Tmax) of
pitolisant is 3.5 hours (2 to 5 hours). The oral absorption of WAKIX is around
90%.
Food Effect
No clinically significant differences in the
pharmacokinetics of pitolisant were observed following administration with a
high-fat meal.
Distribution
The apparent volume of distribution of pitolisant is
approximately 700 L (5 to 10 L/kg). Serum protein binding is approximately 91%
to 96%. The blood to plasma ratio of pitolisant is 0.55 to 0.89.
Elimination
After a single dose of 35.6 mg, the median half-life of
pitolisant is approximately 20 hours (7.5 to 24.2 hours). The apparent oral
clearance (CL/F) of pitolisant is 43.9 L/hr and renal clearance accounts for
<2% of the total clearance of pitolisant.
Metabolism
Pitolisant is primarily metabolized by CYP2D6 and to a
lesser extent by CYP3A4; these metabolites are further metabolized or
conjugated with glycine or glucuronic acid. None of these metabolites are pharmacologically
active.
Excretion
After a single oral radiolabeled pitolisant 17.8 mg dose,
approximately 90% of the dose was excreted in urine (<2% unchanged) and 2.3%
in feces.
Specific Populations
No clinically significant differences in the
pharmacokinetics of pitolisant were observed based on age (18 to 82 years old),
sex, race/ethnicity (Caucasians or Blacks), or body weight (48 to 103 kg). The effects
of end-stage renal disease and severe hepatic impairment on the
pharmacokinetics of pitolisant are unknown.
Patients With Hepatic Impairment
Six subjects with mild hepatic impairment (Child Pugh A),
6 subjects with moderate hepatic impairment (Child Pugh B), and 12 healthy
subjects matched for age, sex, body mass index and ethnicity received a single
dose of WAKIX 17.8 mg to assess the pharmacokinetics of WAKIX in patients with
hepatic impairment. Exposure of pitolisant in patients with mild or moderate
hepatic impairment is summarized in Figure 1. No studies have been conducted in
patients with severe hepatic impairment.
Figure 1: Effect of Hepatic Impairment on Pitolisant
Pharmacokinetics
Dots = Geometric LSM ratios, Error bars = 90% CI;
reference dashed lines are 0.8 and 1.25.
AUCinf = area under the curve from time 0 to time
infinity; Cmax = maximum plasma concentration.
Patients With Renal Impairment
A single dose of WAKIX 17.8 mg was administered to 4
subjects with mild renal impairment (Egfr of 60 to 89 mL/min/1.73m²), 4
subjects with moderate renal impairment (eGFR of 30 to 59 mL/min/1.73m²), 4
subjects with severe renal impairment (eGFR of 15 to 29 mL/min/1.73m²), and 12
subjects with normal renal function (i.e., eGFR >90 mL/min/1.73m²) to assess
the pharmacokinetics of WAKIX in patients with renal impairment. Exposure of pitolisant
in patients with mild, moderate, and severe renal impairment are summarized in Figure
2. No studies have been conducted in patients with ESRD.
Figure 2: Effect of Renal Impairment on Pitolisant
Pharmacokinetics
Dots = Geometric LSM ratios, Error bars = 90% CI;
reference dashed lines are 0.8 and 1.25.
AUCinf = area under the curve from time 0 to time
infinity; Cmax = maximum plasma concentration.
CYP2D6 Poor Metabolizers
The pharmacokinetics of pitolisant were evaluated in 3
subjects who were CYP2D6 poor metabolizers (PMs) and 5 subjects who were CYP2D6
extensive metabolizers (EMs). All subjects received WAKIX 17.8 mg daily for 7
days. Exposure of pitolisant in CYP2D6 PMs are summarized in Figure 3.
Figure 3: Pitolisant Pharmacokinetics in CYP2D6 Poor
Metabolizers
Dots = Geometric LSM ratios, Error bars = 90% CI;
reference dashed lines are 0.8 and 1.25.
AUC(0-24) = area under the curve from time 0 to 24 hours
post-dose; Cmax = maximum plasma concentration.
Drug-Drug Interactions
Effect Of Other Drugs On The Pharmacokinetics Of WAKIX
The effect of other drugs on the pharmacokinetics of
pitolisant are presented in Figure 4 [see DOSAGE AND ADMINISTRATION, DRUG
INTERACTIONS].
Figure 4: Effect of Concomitant Medications on
Pitolisant
Dots = Geometric LSM ratios, Error bars = 90% CI;
reference dashed lines are 0.8 and 1.25.
AUCinf = area under the curve from time 0 to time
infinity; Cmax = maximum plasma concentration.
Effect Of WAKIX On The Pharmacokinetics Of Other Drugs
The effect of pitolisant on the pharmacokinetics of other
drugs are presented in Figure 5 [see DRUG INTERACTIONS].
Figure 5: Effect of Pitolisant on Concomitant
Medications
Dots = Geometric LSM ratios, error bars = 90% CI;
reference dashed lines are 0.8 and 1.25.
AUCinf = area under the curve from time 0 to time
infinity; AUC0-24 = area under the curve from time 0 to 24 hours;
Cmax = maximum plasma concentration.
Administration of WAKIX concomitantly with oral
contraceptives may reduce their effectiveness [see DRUG INTERACTIONS, Use
In Specific Populations].
Pharmacogenomics
Approximately 3 to 10% of Caucasians and 2 to 7% of
African Americans generally lack the capacity to metabolize CYP2D6 substrates
and are classified as poor metabolizers. The AUC of pitolisant was approximately
2.4 times higher in CYP2D6 poor metabolizers than in normal metabolizers and is
similar to the exposure of pitolisant when WAKIX is administered concomitantly
with a CYP2D6 inhibitor [see DOSAGE AND ADMINISTRATION, DRUG INTERACTIONS].
In CYP2D6 poor metabolizers, the Cmax of pitolisant is
153 (151 to 157) ng/mL and the AUC is 1920 (1854 to 2000) ng*hr/mL after steady
state dosing with 35.6 mg once daily.
Animal Toxicology And/Or Pharmacology
Adverse CNS-related clinical signs including tremors and
convulsions occurred after single and repeated oral administration of
pitolisant across multiple species. In a 9-month repeat-dose toxicity study in
monkeys, sporadic incidences of convulsions occurred at doses corresponding to
exposures approximately 3 times the MRHD based on Cmax and 1 times the MRHD,
based on AUC. Convulsions were first observed close to Tmax and resolved by 2
hours after dosing. Convulsions were not observed after discontinuation of
dosing and were not associated with microscopic findings in the brain. Safety margins
at the no-observed-adverse-effect-level (NOAEL) correspond to 1 times the MRHD
based on Cmax and 0.4 times based on AUC.
Clinical Studies
The efficacy of WAKIX for the treatment of excessive
daytime sleepiness in adult patients with narcolepsy was evaluated in two
multicenter, randomized, double-blind, placebo-controlled studies (Study 1;
NCT01067222 and Study 2; NCT01638403). Patients ≥18 years of age who met
the International Classification of Sleep Disorders (ICSD-2) criteria for
narcolepsy and who had an Epworth Sleepiness Scale (ESS) score ≥14 were
eligible to enroll in the studies. EDS was assessed using the ESS, an 8-item
questionnaire by which patients rate their perceived likelihood of falling asleep
during usual daily life activities. Each of the 8 items on the ESS is rated
from 0 (would never doze) to 3 (high chance of dozing); the maximum score is
24. Study 1 and Study 2 included an 8-week treatment period, a 3-week dose
titration phase followed by a 5-week stable dose phase. These studies compared
WAKIX to both a placebo and an active control.
In Study 1, 95 patients were randomized to receive WAKIX,
placebo, or active control. The dose of WAKIX was initiated at 8.9 mg once
daily and could be increased at weekly intervals to 17.8 mg or 35.6 mg, based
on efficacy response and tolerability. No dose adjustments were permitted
during the 5-week stable dose phase. 61% of patients reached a stable dose of
35.6 mg. Median age in the study was 37 years. More than 90% patients in the
pitolisant and placebo groups were Caucasian and 54% were male. Approximately
80% of the population had a history of cataplexy.
WAKIX demonstrated statistically significantly greater
improvement on the primary endpoint, the least square mean final ESS score
compared to placebo (Table 3).
In Study 2, 166 patients were randomized to receive
WAKIX, placebo, or active control. The dose of WAKIX was initiated at 4.45 mg
and could be increased at weekly intervals to 8.9 mg or 17.8 mg, based on
efficacy response and tolerability. No dose adjustments were permitted during
the 5-week stable-dose phase. 76% of patients reached a stable dose of 17.8 mg.
Median age in the study was 40 years. In pitolisant and placebo groups,
approximately 50% of patients were male, 90% of patients were Caucasian, and
75% of patients had a history of cataplexy. WAKIX demonstrated statistically significantly
greater improvement on the primary endpoint, the least square mean final ESS
score compared to placebo (Table 3). Examination of demographic subgroups by
sex did not suggest differences in response.
The efficacy results from Study 1 and Study 2 are shown
in Table 3.
Table 3: Efficacy Results for Epworth Sleepiness Scale
in Patients with Narcolepsy (Study 1 and Study 2)
Study |
Treatment Group (N) |
Baseline ESS Score Mean (SD) |
Final ESS Scorec LS Mean at Week 8 (SE) |
Placebo Subtracted Difference [95% CI] at Week 8d |
Study 1a |
WAKIX (n=31) |
17.8 (2.5) |
12.4 (1.01) |
-3.1*
[-5.73; -0.46] |
Placebo (n=30) |
18.9 (2.5) |
15.5 (1.03) |
Study 2b |
WAKIX (n=66) |
18.3 (2.4) |
13.3 (1.19) |
-2.2*
[-4.17; -0.22] |
Placebo (n=32) |
18.2 (2.3) |
15.5 (1.32) |
SD = standard deviation; SE = standard error; LS Mean =
least square mean; CI = confidence interval
aMaximum dose randomized to was 35.6 mg
bMaximum dose randomized to was 17.8 mg
cA lower score on the ESS represents improvement; scores range from
0 (no symptoms) to 24 (worst symptoms)
dA negative value for the placebo subtracted difference represents
improvement
* Statistically significant |
Figure 6 shows the ESS score from baseline to Week 8 in
Study 1.
Figure 6: Epworth Sleepiness Scale Score (mean ± SEM)
from Baseline to Week 8 in Study 1
SEM = standard error of the mean (raw mean scores)
ESS scores range from 0 to 24, with 0 being the best
score and 24 being the worst score