CLINICAL PHARMACOLOGY
Midazolam is a short-acting benzodiazepine central nervous system (CNS) depressant.
Pharmacodynamics
Pharmacodynamic properties of midazolam and its metabolites, which are similar
to those of other benzodiazepines, include sedative, anxiolytic, amnesic and
hypnotic activities. Benzodiazepine pharmacologic effects appear to result from
reversible interactions with the γ-amino butyric acid (GABA) benzodiazepine
receptor in the CNS, the major inhibitory neurotransmitter in the central nervous
system. The action of midazolam is readily reversed by the benzodiazepine receptor
antagonist, flumazenil.
Data from published reports of studies in pediatric patients clearly demonstrate
that oral midazolam provides safe and effective sedation and anxiolysis prior
to surgical procedures that require anesthesia as well as before other procedures
that require sedation but may not require anesthesia.The most commonly reported
effective doses range from 0.25 to 1 mg/kg in children (6 months to <16 years).The
single most commonly reported effective dose is 0.5 mg/kg. Time to onset of
effect is most frequently reported as 10 to 20 minutes.
The effects of midazolam on the CNS are dependent on the dose administered,
the route of administration, and the presence or absence of other medications.
Following premedication with oral midazolam, time to recovery has been assessed
in pediatric patients using various measures, such as time to eye opening, time
to extubation, time in the recovery room, and time to discharge from the hospital. Most
placebo-controlled trials (8 total) have shown little effect of oral midazolam on recovery time from general anesthesia; however, a number of other placebo-controlled
studies (5 total) have demonstrated some prolongation in recovery time following
premedication with oral midazolam. Prolonged recovery may be related to duration
of the surgical procedure and/or use of other medications with central nervous
system depressant properties.
Partial or complete impairment of recall following oral midazolam has been
demonstrated in several studies.Amnesia for the surgical experience was greater
after oral midazolam when used as a premedicant than after placebo and was generally
considered a benefit. In one study, 69% of midazolam patients did not remember
mask application versus 6% of placebo patients.
Episodes of oxygen desaturation, respiratory depression, apnea, and airway
obstruction have been reported in <1% of pediatric patients following premedication
(eg, sedation prior to induction of anesthesia) with midazolam HCI syrup; the
potential for such adverse events are markedly increased when oral midazolam
is combined with other central nervous system depressing agents and in patients
with abnormal airway anatomy, patients with cyanotic congenital heart disease,
or patients with sepsis or severe pulmonary disease (see WARNINGS).
Concomitant use of barbiturates or other central nervous system depressants
may increase the risk of hypoventilation, airway obstruction, desaturation or
apnea, and may contribute to profound and/or prolonged drug effect. In one study
of pediatric patients undergoing elective repair of congenital cardiac defects,
premedication regimens (oral dose of 0.75 mg/kg midazolam or IM morphine plus
scopolamine) increased transcutaneous carbon dioxide (PtcCO2), decreased
SpO2 (as measured by pulse oximetry), and decreased respiratory rates
preferentially in patients with pul-monary hypertension. This suggests that
hypercarbia or hypoxia following premedication might pose a risk to children
with congenital heart disease and pulmonary hypertension. In a study of an adult
population 65 years and older, the preinduction administration of oral midazolam
7.5 mg resulted in a 60% incidence of hypoxemia (paO2<90% for
over 30 seconds) at some time during the operative procedure versus 15% for
the nonpremedicated group.
Pharmacokinetics
Absorption: Midazolam is rapidly absorbed after oral administration
and is subject to substantial intestinal and hepatic first-pass metabolism.
The pharmacokinetics of midazolam and its major metabolite, α-hydroxy-midazolam,
and the absolute bioavailability of midazolam HCI syrup were studied in pediatric
patients of different ages (6 months to <16 years old) over a 0.25 to 1 mg/kg
dose range. Pharmacokinetic parameters from this study are pre-sented in Table
1. The mean Tmax values across dose groups (0.25, 0.5, and 1 mg/kg) range from
0.17 to 2.65 hours. Midazolam exhibits linear pharmacokinetics between oral
doses of 0.25 to 1 mg/kg (up to a maximum dose of 40 mg) across the age groups
ranging from 6 months to <16 years. Linearity was also demonstrated across
the doses within the age group of 2 years to <12 years having 18 patients
at each of the three doses. The absolute bioavailability of the midazolam syrup
in pediatric patients is about 36%, which is not affected by pediatric age or
weight. The AUC0-∞ ratio of α-hydroxymidazolam to midazolam
for the oral dose in pediatric patients is higher than for an IV dose (0. 38
to 0.75 versus 0.21 to 0.39 across the age group of 6 months to <16 years),
and the AUC0-∞ ratio of α-hydroxy-midazolam to midazolam
for the oral dose is higher in pediatric patients than in adults (0.38 to 0.75
versus 0.40 to 0.56).
Food effect has not been tested using midazolam HCI syrup. When a 15 mg oral
tablet of midazolam was administered with food to adults, the absorption and
disposition of midazolam was not affected. Feeding is generally contraindicated
prior to sedation of pediatric patients for procedures.
Table1. Pharmacokinetics of Midazolam Following Single Dose
Administration of Midazolam HCI Syrup
Number of Subjects/age group |
Dose (mg/kg) |
Tmax (h) |
Cmax (ng/mL) |
T½ (h) |
AUC0-∞ (ng h/mL) |
6 months to < 2 years old |
1 |
0.25 |
0.17 |
28.0 |
5.82 |
67.6 |
1 |
0.50 |
0.35 |
66.0 |
2.22 |
152 |
1 |
1.00 |
0.17 |
61.2 |
2.97 |
224 |
2 to |
18 |
0.25 |
0.72 ± 0.44 |
63.0 ± 30.0 |
3.16 ± 1.50 |
138 ± 89.5 |
18 |
0.50 |
0.95 ± 0.53 |
126 ± 75.8 |
2.71 ± 1.09 |
306 ± 196 |
18 |
1.00 |
0.88 ± 0.99 |
201 ± 101 |
2.37 ± 0.96 |
743 ± 642 |
12 to < 16 years old |
4 |
0.25 |
2.09 ± 1.35 |
29.1 ± 8.2 |
6.83 ± 3.84 |
155 ± 84.6 |
4 |
0.50 |
2.65 ± 1.58 |
118 ± 81.2 |
4.35 ± 3.31 |
821 ± 568 |
2 |
1.00 |
0.55 ± 0.28 |
191 ± 47.4 |
2.51 ± 0.18 |
566 ± 15.7 |
Distribution: The extent of plasma protein binding of midazolam
is moderately high and concentration independent. In adults and pediatric patients
older than 1 year, midazolam is approximately 97% bound to plasma protein, principally
albumin. In healthy volunteers, α-hydroxymidazolam is bound to the extent
of 89%. In pediatric patients (6 months to <16 years) receiving 0.15 mg/kg
IV midazolam, the mean steady-state volume of distribution ranged from 1.24
to 2.02 L/kg.
Metabolism: Midazolam is primarily metabolized in the liver and
gut by human cytochrome P450 IIIA4 (CYP3A4) to its pharmacologic active metabolite,
α-hydroxymidazolam, followed by glucuronidation of the α-hydroxyl
metabolite which is present in unconjugated and conjugated forms in human plasma.
The α- hydroxymidazolam glucuronide is then excreted in urine. In a study
in which adult volunteers were administered intravenous midazolam (0.1 mg/kg)
and α-hydroxymidazolam (0.15 mg/kg), the pharmacodynamic parameter values
of the maximum effect (Emax) and concentration eliciting half-maximal effect
(EC50) were similar for both compounds. The effects studied were
reaction time and errors in tracing tests. The results indicate that α-hydroxymidazolam
is equipotent and equally effective as unchanged midazolam on a total plasma
concentration basis. After oral or intravenous administration, 63% to 80% of
midazolam is recovered in urine as α-hydroxymidazolam glucuronide. No
significant amount of parent drug or metabo-lites is extractable from urine
before beta-glucuronidase and sulfatase deconjugation, indicating that the urinary
metabolites are excreted mainly as conjugates.
Midazolam is also metabolized to two other minor metabolites: 4-hydroxy metabolite
(about 3% of the dose) and 1,4-dihydroxy metabolite (about 1% of the dose) are
excreted in small amounts in the urine as conjugates.
Elimination: The mean elimination half-life of midazolam ranged
from 2.2 to 6.8 hours following single oral doses of 0.25, 0.5, and 1 mg/kg
of midazolam (midazolam HCI syrup). Similar results (ranged from 2.9 to 4.5
hours) for the mean elimination half-life were observed following IV administration
of 0.15 mg/kg of midazolam to pediatric patients (6 months to <16 years old).
In the same group of patients receiving the 0.15 mg/kg IV dose, the mean total
clearance ranged from 9.3 to 11 mL/min/kg.
Pharmacokinetic-Pharmacodynamic Relationships: The relationship
between plasma concentration and sedation and anxiolysis scores of oral midazolam
syrup (single oral doses of 0.25, 0.5, or 1 mg/kg) was investigated in three
age groups of pediatric patients (6 months to <2 years, 2 to <12 years,
and 12 to <16 years old). In this study, the patient's sedation scores were
recorded at baseline and at 10-minute intervals up to 30 minutes after oral
dosing until satisfactory sedation (“drowsy” or “asleep but
responsive to mild shaking” or “asleep and not responsive to mild
shaking”) was achieved. Anxiolysis scores were measured at the time when
the patient was separated from his/her parents and at mask induction. The results
of the analyses showed that the mean midazolam plasma concentration as well
as the mean of midazolam plus α-hydroxymidazolam for those patients with
a sedation score of 4 (asleep but responsive to mild shaking) is significantly
different than the mean concentrations for those patients with a sedation score
of 3 (drowsy), which is significantly different than the mean concentrations
for patients with a sedation score of 2 (awake/calm). The statistical analysis
indicates that the greater the midazolam, or midazolam plus α-hydroxymidazo-lam
concentration, the greater the maximum sedation score for pediatric patients.
No such trend was observed between anxiolysis scores and the mean midazolam
concentration or mean of midazolam plus α-hydroxymidazolam concentration;however,
anxiolysis is a more variable surrogate measurement of clinical response.
Special Populations
Renal Impairment: Although the pharmacokinetics of intravenous
midazolam in adult patients with chronic renal failure differed from those of
subjects with normal renal function, there were no alterations in the distribution,
elimination, or clearance of unbound drug in the renal failure patients. However,
the effects of renal impairment on the active metabolite α-hydroxymidazolam
are unknown.
Hepatic Dysfunction: Chronic hepatic disease alters the pharmacokinetics
of midazolam. Following oral administration of 15 mg of midazolam, Cmax and
bioavailability values were 43% and 100% higher, respectively, in adult patients
with hepatic cirrhosis than adult subjects with normal liver function. In the
same patients with hepatic cirrhosis, following IV administration of 7.5 mg
of midazolam, the clearance of midazolam was reduced by about 40% and the elimination
half-life was increased by about 90% compared with subjects with normal liver
function. Midazolam should be titrated for the desired effect in patients with
chronic hepatic disease.
Congestive Heart Failure: Following oral administration of 7.5
mg of midazolam, elimination half-life values were 43% higher in adult patients
with congestive heart failure than in control subjects.
Neonates: Midazolam HCI syrup has not been studied in pediatric
patients less than 6 months of age.
Drug-Drug Interactions: See PRECAUTIONS: DRUG
INTERACTIONS.
INHIBITORS OF CYP3A4 ISOZYMES
Table 2 summarizes the changes in the Cmax and AUC of midazolam when drugs
known to inhibit CYP3A4 were con-currently administered with oral midazolam
in adults subjects.
Table 2
Interacting Drug |
Adult Doses Studied |
% Increase in Cmax of Oral
Midazolam |
% Increase in AUC of Oral
Midazolam |
Cimetidine |
800-1200 mg up to qid in divided doses |
6-138 |
10-102 |
Diltiazem |
60 mg tid |
105 |
275 |
Erythromycin |
500 mg tid |
170-171 |
281-341 |
Fluconazole |
200 mg qd |
150 |
250 |
Grapefruit Juice |
200 mL |
56 |
52 |
Itraconazole |
100-200 mg qd |
80-240 |
240-980 |
Ketoconazole |
400 mg qd |
309 |
1490 |
Ranitidine |
150 mg bid or tid; 300 mg qd |
15-67 |
9-66 |
Roxithromycin |
300 mg qd |
37 |
47 |
Saquinavir |
1200 mg tid |
235 |
514 |
Verapamil |
80 mg tid |
97 |
192 |
Other drugs known to inhibit the effects of CYP3A4, such as protease inhibitors,
would be expected to have similar effects on these midazolam pharmacokinetic
parameters.
INDUCERS OF CYP3A4 ISOZYMES
Table 3 summarizes the changes in the Cmax and AUC of midazolam when drugs
known to induce CYP3A4 were con-currently administered with oral midazolam in
adult subjects. The clinical significance of these changes is unclear.
Table 3
Interacting Drug |
Adult Doses Studied |
% Decrease in Cmax of
Oral Midazolam |
% Decrease in AUC of
Oral Midazolam |
Carbamazepine |
Therapeutic Doses |
93 |
94 |
Phenytoin |
Therapeutic Doses |
93 |
94 |
Rifampin |
600 mg/day |
94 |
96 |
Although not tested, phenobarbital, rifabutin and other drugs known to induce
the effects of CYP3A4 would be expected to have similar effects on these midazolam
pharmacokinetic parameters.
Drugs that did not affect midazolam pharmacokinetics are presented in Table
4.
Table 4
Interacting Drug |
Adult Doses Studied |
Azithromycin |
500 mg/day |
Nitrendipine |
20 mg |
Terbinafine |
200 mg/day |
Clinical Trials
Dose Ranging, Safety and Efficacy Study With Midazolam HCI Syrup in Pediatric
Patients: The effectiveness of midazolam HCI syrup as a premedicant
to sedate and calm pediatric patients prior to induction of general anesthesia
was compared among three different doses in a randomized, double-blind, parallel-group
study. Patients of ASA physical status I, II or III were stratified to 1 of
3 age groups (6 months to <2 years, 2 to <6 years, and 6 to <16 years),
and within each age group randomized to 1 of 3 dosing groups (0.25, 0.5, and
1 mg/kg up to a maximum dose of 20 mg). Greater than 90% of treated patients
achieved satisfactory sedation and anxiolysis at at least one timepoint within
30 minutes posttreatment. Similarly high proportions of patients exhibited satisfactory
ease of separation from parent or guardian and were cooperative at the time
of mask induction with nitrous oxide and halothane administra-tion. Onset time
of satisfactory sedation or anxiolysis occurred within 10 minutes after treatment
for >70% of patients who started with an unsatisfactory baseline rating.
Whereas pairwise comparisons (0.25 mg/kg versus 0.5 mg/kg groups, and 0.5 mg/kg
versus 1 mg/kg groups) on satisfactory sedation did not yield significant p-values
(p=0.08 in both cases), comparative analysis of the clinical response between
the high and low doses demonstrated that a higher proportion of patients in
the 1 mg/kg dose group exhibited satisfactory sedation and anxiolysis as compared
to the 0.25 mg/kg group (p<0.05).