CLINICAL PHARMACOLOGY
Mechanism Of Action
Methylnaltrexone is a selective antagonist of opioid
binding at the mu-opioid receptor. As a quaternary amine, the ability of
methylnaltrexone to cross the blood-brain barrier is restricted. This allows
methylnaltrexone to function as a peripherally-acting mu-opioid receptor
antagonist in tissues such as the gastrointestinal tract, thereby decreasing
the constipating effects of opioids without impacting opioid-mediated analgesic
effects on the central nervous system.
Pharmacodynamics
Cardiac Electrophysiology
In a randomized, double-blind placebo-and (open-label)
moxifloxacin-controlled 4-period crossover study, 56 healthy subjects were
administered methylnaltrexone bromide 0.3 mg/kg and methylnaltrexone bromide
0.64 mg/kg by intravenous infusion over 20 minutes (RELISTOR is not approved
for intravenous use), placebo, and a single oral dose of moxifloxacin. At a
dose approximately 4.3 times the maximum recommended dose (7.5 times the mean
peak plasma concentration for RELISTOR injection and 22 times the peak plasma
concentration for RELISTOR tablets), methylnaltrexone does not prolong the QTc
interval to any clinically relevant extent.
Pharmacokinetics
Between the oral dosage range of 150 mg to 450 mg for
RELISTOR tablets and the subcutaneous dosage range of 0.15 mg/kg to 0.50 mg/kg
for RELISTOR injection, the mean Cmax and area under the plasma
concentration-time curve (AUC) of methylnaltrexone increased in a
dose-proportional manner. There was no significant accumulation of
methylnaltrexone following once-daily oral dosing of 450 mg RELISTOR tablets or
subcutaneous dosing of 12 mg RELISTOR injection for seven consecutive days in
healthy subjects.
Absorption
Tablets
Following administration of a single 450 mg dose of
RELISTOR tablets in OIC patients or healthy subjects, peak concentrations (Cmax)
of methylnaltrexone were observed at approximately 1.5 hours. The absolute
bioavailability of oral methylnaltrexone bromide has not been determined. The Cmax
and AUC in healthy subjects were 48.1 ng/mL and 382 ng·hr/mL, respectively,
following a single 450 mg dose of RELISTOR tablets. Exposure in the OIC patient
population was approximately 27% lower than in healthy subjects.
Food Effect
Administration of a single 450 mg dose of RELISTOR
tablets to healthy subjects with a high-fat breakfast (containing approximately
800 to 1000 total calories, with 60%, 25% and 15% of calories derived from fat,
carbohydrate and protein, respectively) resulted in a decrease in the Cmax of
methylnaltrexone by 60%, the AUC by 43% and delayed the Tmax by 2 hours [see
DOSAGE AND ADMINISTRATION].
Injection
Following administration of RELISTOR injection
subcutaneously, methylnaltrexone achieved peak concentrations (Cmax) at
approximately 0.5 hours (see Table 7).
Table 7: Pharmacokinetic Parameters of
Methylnaltrexone Following Subcutaneous Doses
Parameter |
0.15 mg/kg Single Dose |
12 mg Single Dose |
12 mg at Steady-State |
Cmax (ng/mL)i |
117 (32.7) |
140 (35.6) |
119 (27.2) |
Tmax (hr)ii |
0.5 (0.25 to 0.75) |
0.25 (0.25 to 0.5) |
0.25 (0.25 to 0.5) |
AUC24 (nghr/mL) |
175 (36.6) |
218 (28.3) |
223 (28.2) |
i Expressed as mean (SD).
ii Expressed as median (range). |
Distribution
The steady-state volume of distribution (Vss) of
methylnaltrexone is approximately 1.1 L/kg. The fraction of methylnaltrexone
bound to human plasma proteins is 11% to 15%, as determined by equilibrium
dialysis.
Elimination
Following oral administration of a single 450 mg dose of
RELISTOR tablets, concentrations of methylnaltrexone declined in multiphasic
manner with a terminal half-life (t½) of approximately 15 hours.
Metabolism
In an intravenous mass balance study, approximately 44%
of the administered radioactivity was recovered in the urine over 24 hours with
5 distinct metabolites. None of the detected metabolites was in amounts over 6%
of administered radioactivity. Conversion to methyl-6-naltrexol isomers (5% of
total) and methylnaltrexone sulfate (1% of total) appear to be the primary
pathways of metabolism. N-demethylation of methylnaltrexone to produce
naltrexone is not significant.
Systemic exposure of methylnaltrexone metabolites after
oral administration of a single 450 mg dose of RELISTOR tablets are greater
than the systemic exposure of methylnaltrexone metabolites after subcutaneous
administration of a single 12 mg dose of RELISTOR injection. Subcutaneous
administration is not subject to first-pass hepatic metabolism prior to
appearance in the systemic circulation. After 12 mg subcutaneous once daily
dosing the mean AUC0-24 ratio of metabolites to methylnaltrexone at
steady-state was 30%, 19%, and 9% for methylnaltrexone sulfate,
methyl-6α-naltrexol, and methyl-6Ã-naltrexol, respectively. After 450 mg
oral once daily dosing, the ratio of the mean AUC0-24 of metabolites to
methylnaltrexone at steady-state was 79%, 38%, and 21% for methylnaltrexone
sulfate, methyl-6α-naltrexol, and methyl-6Ã-naltrexol, respectively.
Methylnaltrexone sulfate is a weak mu-opioid receptor antagonist;
methyl-6α-naltrexol, and methyl-6Ãnaltrexol are active mu-opioid receptor
antagonists.
Methylnaltrexone is conjugated by sulfotransferase
SULT1E1 and SULT2A1 isoforms to methylnaltrexone sulfate. Conversion to
methyl-6-naltrexol isomers is mediated by aldo-keto reductase 1C enzymes.
Excretion
In an intravenous mass balance study, approximately half
of the dose was excreted in the urine (54%), and 17% of administered dose was
excreted in the feces up to 168 hours postdose; however, radiolabeled recovery
in this study was only 71% after 7 days. Methylnaltrexone is excreted primarily
as the unchanged drug in the urine and feces. Active renal secretion of
methylnaltrexone is suggested by renal clearance of methylnaltrexone that is
approximately 4-to 5-fold higher than creatinine clearance.
No mass balance clinical studies were conducted with oral
administration of methylnaltrexone bromide. However, following once daily
dosing of 450 mg RELISTOR tablets for 1 week, the percentage of dose recovered
in the urine as the parent methylnaltrexone was low (approximately 1% on both
Day 1 and Day 7).
Specific Populations
Age: Geriatric Population
A study was conducted to characterize the
pharmacokinetics of methylnaltrexone after a single dose of 24 mg
methylnaltrexone bromide via intravenous infusion over 20 min in healthy adults
between 18 and 45 years of age and in healthy adults aged 65 years and older.
In elderly subjects (mean age 72 years old), mean clearance was about 20% lower
(56 L/h versus 70 L/h) and AUC∞ was 26% higher than in subjects between
18 and 45 years of age (mean age 30 years old) [see Use In Specific
Populations].
Renal Impairment
Administration of a single subcutaneous dose of 0.3 mg/kg
of RELISTOR injection in subjects with varying degrees of renal impairment (8
subjects each cohort) resulted in a 1.3-, 1.7-and 1.9-fold higher AUC0-∞ of
methylnaltrexone, respectively, compared to 8 subjects with normal renal
function [see DOSAGE AND ADMINISTRATION]. The mean Cmax did not change
significantly with renal impairment. No studies were performed in patients with
end-stage renal impairment requiring dialysis.
Hepatic Impairment
Administration of a single 450 mg dose of RELISTOR
tablets in subjects with mild, moderate, and severe hepatic impairment (6
subjects each of Child-Pugh Class A, B, and C) resulted in a 1.7-, 4.8-and
3.8-fold higher Cmax of methylnaltrexone, respectively, compared to 6 subjects
with normal liver function. The AUC0-∞, was comparable between healthy
subjects and subjects with mild hepatic impairment, but increased approximately
2.1-fold in subjects with moderate and severe hepatic impairment [see DOSAGE
AND ADMINISTRATION].
Administration of a single dose of 0.3 mg/kg of RELISTOR
injection subcutaneously in 8 subjects with mild hepatic impairment (Child-Pugh
Class A) and 8 subjects with moderate hepatic impairment (Child-Pugh Class B)
did not result in any meaningful change in the AUC or Cmax when compared to 8
healthy subjects with normal hepatic function. The effect of severe (Child-Pugh
Class C) hepatic impairment on the pharmacokinetics of RELISTOR injection has
not been studied [see DOSAGE AND ADMINISTRATION, Use In Specific
Populations].
Drug Interaction Studies
Effect Of Methylnaltrexone And Its Metabolites On CYP
Enzymes
In vitro, methylnaltrexone did not significantly inhibit
or induce the activity of cytochrome P450 (CYP) isozymes CYP1A2, CYP2A6,
CYP2B6, CYP2C9, CYP2C19, or CYP3A4. In addition, methylnaltrexone did not
induce CYP2E1.
In vitro, the methylnaltrexone metabolites,
methylnaltrexone sulfate, methyl-6α-naltrexol and methyl-6βnaltrexol
did not inhibit CYP isozymes CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6,
or CYP3A4. The metabolites of methylnaltrexone did not induce activity of
CYP1A2, CYP2B6, or CYP3A4.
Effect Of Methylnaltrexone And Its Metabolites On Transporters
Based on the in vitro studies, methylnaltrexone and its
three major metabolites, methylnaltrexone sulfate, methyl-6α-naltrexol and
methyl-6β-naltrexol, are unlikely to have clinically meaningful in vivo drug-drug
interactions via inhibition of P-glycoprotein (P-gp), Breast Cancer Resistance Protein
(BCRP), Multidrug Resistance Protein 2 (MRP2), Organic Anion-Transporting
Polypeptide (OATP)1B1, OATP1B3, Organic Cation Transporter (OCT)1, OCT2,
Organic Anion Transporter (OAT)1, OAT3, Multidrug and Toxic Extrusion
Transporter (MATE)1 and MATE2-K at the recommended dosage of 450 mg orally or
12 mg subcutaneously once daily.
Methylnaltrexone
Methylnaltrexone was a substrate of OCT1, OCT2, MATE1,
and MATE2-K, but not a substrate for P-gp, BCRP, MRP2, OATP1B1, OATP1B3, OAT1
and OAT3.
Methylnaltrexone Sulfate
Methylnaltrexone sulfate was a substrate for MATE2-K and
a potential substrate of BCRP, but was not a substrate of P-gp, MRP2, OATP1B1,
OATP1B3, OCT1, OCT2, OAT1, OAT3, and MATE1.
Methyl-6α-naltrexol
Methyl-6α-naltrexol was a substrate of BCRP, OCT1,
OCT2, MATE1, and MATE2-K, but was not a substrate of P-gp, MRP2, OATP1B1,
OATP1B3, OAT1 and OAT3.
Methyl-6β-naltrexol
Methyl-6β-naltrexol was a substrate of OCT1, OCT2,
MATE1, and MATE2-K, but was not a substrate of P-gp, BCRP, MRP2, OATP1B1,
OATP1B3, OAT1 and OAT3.
Cimetidine
A clinical drug interaction study in healthy adult
subjects evaluated the effects of cimetidine, a drug that inhibits the active
renal secretion of organic cations, on the pharmacokinetics of methylnaltrexone
(24 mg administered as an intravenous infusion over 20 minutes). A single dose
of methylnaltrexone bromide was administered before cimetidine dosing and with
the last dose of cimetidine (400 mg every 8 hours for 6 days). Mean Cmax and
AUC of methylnaltrexone increased by 10% with concomitant cimetidine
administration. The renal clearance of methylnaltrexone decreased about 40%.
This change is not considered to be clinically meaningful.
Animal Toxicology And/Or Pharmacology
In an in vitro human cardiac potassium ion channel (hERG)
assay, methylnaltrexone caused concentration-dependent inhibition of hERG
current (1%, 12%, 13% and 40% inhibition at 30, 100, 300 and 1000 micromolar
concentrations, respectively). Methylnaltrexone had a hERG IC50 of more
than1000 micromolar. In isolated dog Purkinje fibers, methylnaltrexone caused
prolongations in action potential duration (APD). The highest tested
concentration (10 micromolar) in the dog Purkinje fiber study was about 18 and
37 times the Cmax at human subcutaneous doses of 0.3 and 0.15 mg/kg,
respectively. In isolated rabbit Purkinje fibers, methylnaltrexone (up to 100
micromolar) did not have an effect on APD, compared to vehicle control. The
highest methylnaltrexone concentration (100 micromolar) tested was about 186
and 373 times the human Cmax at subcutaneous doses of 0.3 and 0.15 mg/kg,
respectively. In anesthetized dogs, methylnaltrexone bromide caused decreases
in blood pressure, heart rate, cardiac output, left ventricular pressure, left
ventricular end diastolic pressure, and +dP/dt at 1 mg/kg or more. In conscious
dogs, methylnaltrexone bromide caused a dose-related increase in QTc interval.
After a single intravenous dosage of 20 mg/kg to beagle dogs, predicted Cmax and
AUC values were approximately 482 and 144 times, respectively, the exposure at
human subcutaneous dose of 0.15 mg/kg and 241 times and 66 times, respectively,
the exposure at a human subcutaneous dose of 0.3 mg/kg. In conscious guinea
pigs, methylnaltrexone bromide caused mild prolongation of QTc (4% over
baseline) at 20 mg/kg, intravenous. A thorough QTc assessment was conducted in
humans [see CLINICAL PHARMACOLOGY].
In juvenile rats administered intravenous
methylnaltrexone bromide for 13 weeks, adverse clinical signs such as
convulsions, tremors and labored breathing occurred at dosages of 3 and 10
mg/kg/day (about 2.4 and 8 times, respectively, the subcutaneous MRHD of 12
mg/day; about 0.06 and 0.22 times, respectively, the oral MRHD of 450 mg/day).
Similar adverse clinical signs were seen in adult rats at 20 mg/kg/day (about
16 times the subcutaneous MRHD of 12 mg/day; about 0.43 times the oral MRHD of
450 mg/day). Juvenile rats were found to be more sensitive to the toxicity of
methylnaltrexone bromide when compared to adults. The no observed adverse
effect levels (NOAELs) in juvenile and adult rats were 1 and 5 mg/kg/day,
respectively (about 0.8 and 4 times, respectively, the subcutaneous MRHD of 12
mg/day; about 0.02 and 0.11 times, respectively, the oral MRHD of 450 mg/day).
Juvenile dogs administered intravenous methylnaltrexone
bromide for 13 weeks had a toxicity profile similar to adult dogs. Following
intravenous administration of methylnaltrexone bromide for 13 weeks, decreased
heart rate (13.2% reduction compared to pre-dose) in juvenile dogs and
prolonged QTc interval in juvenile (9.6% compared to control) and adult (up to
15% compared to control) dogs occurred at 20 mg/kg/day (about 54 times the
subcutaneous MRHD of 12 mg/day; about 1.5 times the oral MRHD of 450 mg/day).
Clinical signs consistent with effects on the CNS (including tremors and
decreased activity) occurred in both juvenile and adult dogs. The NOAELs in
juvenile and adult dogs were 5 mg/kg/day (about 14 times the subcutaneous MRHD
of 12 mg/day; about 0.4 times the oral MRHD of 450 mg/day).
Clinical Studies
Opioid-Induced Constipation In Adult Patients With Chronic
Non-Cancer Pain
RELISTOR Tablets
The efficacy of RELISTOR tablets in the treatment of OIC
in patients with chronic non-cancer pain was evaluated in a randomized,
double-blind, placebo-controlled study (Study 1). This study compared 4-week
treatment of RELISTOR tablets 450 mg orally once daily with placebo.
A total of 401 patients (200 RELISTOR, 201 placebo) were
enrolled and treated in the double-blind period. Patients had a history of
chronic non-cancer pain for which they were taking opioids. The most common
pain condition requiring opioid use was back pain. Other frequently reported
primary pain conditions were arthritis, neurologic/neuropathic pain,
joint/extremity pain, and fibromyalgia. Prior to screening, patients were
receiving opioid therapy for pain for 1 month or longer (median daily baseline
oral morphine equivalent dose of 156 mg) and had OIC (less than 3 spontaneous
bowel movements per week during the screening period). Constipation due to
opioid use had to be associated with 1 or more of the following: A Bristol
Stool Form Scale score of 1 or 2 for at least 25% of the bowel movements (BM),
straining during at least 25% of the BMs or a sensation of incomplete
evacuation after at least 25% of the BMs.
Patients were required to be on a stable opioid regimen
(daily dose 50 mg or more of oral morphine equivalents per day) a minimum of 2
weeks prior to the screening visit and received their opioid medication during
the study as clinically needed. The median duration of OIC at baseline was 53
months (4 years). The mean patient age was 52 years (range 23 to 78 years), 64%
were female, and 84% of patients were Caucasian.
Eligible patients were required to discontinue all
previous laxative therapy and use only the study-permitted rescue laxative
(bisacodyl tablets). If patients did not have a bowel movement for 3
consecutive days during the study, they were permitted to use rescue medication
(up to 3 bisacodyl tablets taken orally once during a 24hour period). Bisacodyl
tablets were taken 5 hours or longer and up to 8 hours after study drug
administration. If rescue treatment with bisacodyl tablets did not result in a
bowel movement, a second dose of bisacodyl or an enema 24 hours after rescue
was permitted. Enema use was permitted after rescue with bisacodyl tablets had
failed at least once.
A responder analysis was performed which defined the
proportion of patients with 3 or more spontaneous bowel movements (SBMs)/week,
with an increase of 1 or more SBM/week over baseline, for 3 or more out of the
first 4 weeks of the treatment period. A SBM was defined as a bowel movement
that occurred without laxative use during the previous 24 hours. Table 8 presents
the proportion of patients who responded during the double-blind treatment
period in the intent-to-treat (ITT) population, which included all randomized
patients who received at least one dose of double-blind study medication.
Table 8: Proportion of Responders* in the ITT
Population in Study 1 of RELISTOR Tablets for the Treatment of OIC in Patients
with Chronic Non-Cancer Pain
Treatment |
N |
n (%) |
Percent Differencea (2-sided 95% CI) |
RELISTOR Tablets 450 mg Once Daily |
200 |
103 (52%) |
13% (3%, 23%) |
Placebo |
201 |
77 (38%) |
CI = confidence interval; ITT = intent-to-treat;
a Difference for active treatment vs. placebo;
*A responder is defined as a patient with 3 or more SBMs/week, with an increase
of 1 or more SBM/week over baseline, for 3 or more out of the first 4 weeks of
the treatment period. |
RELISTOR Injection
The efficacy of RELISTOR injection in the treatment of
OIC in patients with chronic non-cancer pain were evaluated in a randomized,
double-blind, placebo-controlled study (Study 2). This study compared 4-week
treatment of RELISTOR injection 12 mg administered subcutaneously once daily
with placebo.
A total of 312 patients (150 RELISTOR, 162 placebo) were
enrolled and treated in the double-blind period. Patients had a history of
chronic non-cancer pain for which they were taking opioids. The majority of
patients had a primary diagnosis of back pain; other primary diagnoses included
joint/extremity pain, fibromyalgia, neurologic/neuropathic pain, and rheumatoid
arthritis. Prior to screening, patients had been receiving opioid therapy for
pain for 1 month or longer (median daily baseline oral morphine equivalent dose
of 161 mg) and had OIC (less than 3 spontaneous bowel movements per week during
the screening period). Constipation due to opioid use had to be associated with
1 or more of the following: A Bristol Stool Form Scale score of 1 or 2 for at
least 25% of the bowel movements (BM), straining during at least 25% of the BMs
or a sensation of incomplete evacuation after at least 25% of the BMs.
Patients were required to be on a stable opioid regimen
(daily dose 50 mg or more of oral morphine equivalents per day) for at least 2
weeks before the screening visit and received their opioid medication during
the study as clinically needed. The median duration of OIC at baseline was 59
months (5 years). The median patient age at baseline was 49 years, 62% were
females and 90% were Caucasian.
Eligible patients were required to discontinue all
previous laxative therapy and use only the study-permitted rescue laxative
(bisacodyl tablets). If patients did not have a bowel movement for 3
consecutive days during the study, they were permitted to use rescue medication
(up to 4 bisacodyl tablets taken orally once during a 24hour period). Rescue
laxatives were prohibited until at least 4 hours after taking an injection of
study medication.
A responder analysis was performed which defined the
proportion of patients with 3 or more (SBMs) per week for each of the 4 weeks
of the double-blind period. A SBM was defined as a bowel movement that occurred
without laxative use during the previous 24 hours. Table 9 presents the
proportion of patients who responded during the double-blind treatment period
in the modified intent-to-treat (mITT) population, which included all
randomized subjects who received at least one dose of double-blind study
medication.
Table 9: Proportion of Responders* in the mITT
Population in Study 2 of RELISTOR Injection for the Treatment of OIC in
Patients with Chronic Non-Cancer Pain
Treatment |
N |
n (%) |
Percent Differencea (2-sided 95% CI) |
P-valueb |
RELISTOR Injection 12 mg Once Daily |
150 |
88 (59%) |
20%
(10%, 31%) |
<0.001 |
Placebo |
162 |
62 (38%) |
CI = confidence interval; mITT = modified
intent-to-treat;
a Difference for active treatment vs. placebo;
* A responder is defined as a patient with 3 or more SBMs per week for each of
the 4 weeks in the double-blind period.
b P-value for active treatment vs. placebo based on 2-sided
Chi-square test. |
Following the first dose, 33% of patients in the RELISTOR
treatment group had a SBM within 4 hours and approximately half of patients had
a SBM prior to the second dose of RELISTOR injection.
Opioid-Induced Constipation In Adult Patients With Advanced
Illness
The efficacy of RELISTOR injection in the treatment of
OIC in advanced illness patients receiving palliative care was demonstrated in
two randomized, double-blind, placebo-controlled studies. In these studies, the
median age was 68 years (range 21 to 100 years); 51% were females. In both
studies, patients had advanced illness and received care to control their symptoms.
The majority of patients had a primary diagnosis of incurable cancer; other
primary diagnoses included end-stage COPD/emphysema, cardiovascular
disease/heart failure, Alzheimer's disease/dementia, HIV/AIDS, or other
advanced illnesses. Prior to screening, patients had been receiving palliative
opioid therapy (median daily baseline oral morphine equivalent dose of 172 mg),
and had OIC (either less than 3 bowel movements in the preceding week or no
bowel movement for 2 or more days). Patients were on a stable opioid regimen 3
or more days prior to randomization (not including PRN or rescue pain
medication) and received their opioid medication during the study as clinically
needed. Patients maintained their regular laxative regimen for at least 3 days
prior to study entry, and throughout the study. Rescue laxatives were
prohibited from 4 hours before to 4 hours after taking an injection of study
medication.
Study 4 was a double-blind, placebo-controlled study
which compared a single, subcutaneous dose of RELISTOR injection 0.15 mg/kg,
and RELISTOR injection 0.3 mg/kg versus placebo. The double-blind dose was
followed by an open-label 4-week dosing period, where RELISTOR injection could
be used as needed, no more frequently than one dose in a 24-hour period.
Throughout both study periods, patients maintained their regular laxative
regimen. A total of 154 patients (47 RELISTOR 0.15 mg/kg, 55 RELISTOR 0.3
mg/kg, 52 placebo) were enrolled and treated in the double-blind period. The
primary endpoint was the proportion of patients with a rescue-free laxation
within 4 hours of the double-blind dose of study medication. RELISTOR-treated
patients had a significantly higher rate of laxation within 4 hours of the
double-blind dose (62% for 0.15 mg/kg and 58% for 0.3 mg/kg) than did
placebo-treated patients (14%); p < 0.0001 for each dose versus placebo (Figure
1).
Study 5 was a double-blind, placebo-controlled study
which compared subcutaneous doses of RELISTOR injection given every other day
for 2 weeks versus placebo. Patients received opioid medication for at least 2
weeks prior to receiving study medication. During the first week (Days 1, 3, 5,
7) patients received either 0.15 mg/kg RELISTOR injection or placebo. In the
second week the patient's assigned dose could be increased to 0.3 mg/kg if the
patient had 2 or fewer rescue-free laxations up to Day 8. At any time, the
patient's assigned dose could be reduced based on tolerability. Data from 133
(62 RELISTOR, 71 placebo) patients were analyzed. There were 2 primary
endpoints: proportion of patients with a rescue-free laxation within 4 hours of
the first dose of study medication and proportion of patients with a
rescue-free laxation within 4 hours after at least 2 of the first 4 doses of
study medication. RELISTOR-treated patients had a higher rate of laxation
within 4 hours of the first dose (48%) than placebo-treated patients (16%); p
<0.0001 (Figure 1). RELISTOR-treated patients also had significantly higher
rates of laxation within 4 hours after at least 2 of the first 4 doses (52%)
than did placebo-treated patients (9%); p <0.0001. In both studies, in
approximately 30% of patients, laxation was reported within 30 minutes of a
dose of RELISTOR.
Figure 1: Laxation Response within 4 Hours of the
First Dose in Studies 4 and 5
In both studies, there was no evidence of differential
effects of age or gender on safety or efficacy. No meaningful subgroup analysis
could be conducted on race because the study population was predominantly
Caucasian (88%).
Durability Of Response
Durability of response was explored in Study 5, and the
laxation response rate was consistent from dose 1 through dose 7 over the
course of the 2-week, double-blind period.
The efficacy of RELISTOR injection was also demonstrated
in open-label treatment administered from Day 2 through Week 4 in Study 4, and
in two open-label extension studies (Study 4 EXT and Study 5 EXT) in which
RELISTOR was given as needed for up to 4 months. During open-label treatment,
patients maintained their regular laxative regimen. A total of 136, 21, and 82
patients received at least one open-label dose in Studies 4, 4 EXT, and 5 EXT,
respectively. Laxation response was also explored in this open-label setting
and appeared to be maintained over the course of 3 to 4 months of open-label
treatment.
Opioid Use And Pain Scores
No relationship between baseline opioid dose and laxation
response in RELISTOR-treated patients was identified in exploratory analyses of
these studies. In addition, median daily opioid dose did not vary meaningfully
from baseline in either RELISTOR-treated patients or in placebo-treated
patients. There were no clinically relevant changes in pain scores from
baseline in either the RELISTOR or placebo-treated patients.