Effect of Different Durations of Ketoconazole Dosing On The Single-Dose Pharmacokinetics of Midaz

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

The Journal of Clinical

Pharmacology
http://www.jclinpharm.org

Effect of Different Durations of Ketoconazole Dosing on the Single-Dose Pharmacokinetics of


Midazolam: Shortening the Paradigm
S. A. Stoch, E. Friedman, A. Maes, K. Yee, Y. Xu, P. Larson, M. Fitzgerald, J. Chodakewitz and J. A. Wagner
J. Clin. Pharmacol. 2009; 49; 398 originally published online Feb 26, 2009;
DOI: 10.1177/0091270008331133

The online version of this article can be found at:


http://www.jclinpharm.org/cgi/content/abstract/49/4/398

Published by:

http://www.sagepublications.com

On behalf of:
American College of Clinical Pharmacology

Additional services and information for The Journal of Clinical Pharmacology can be found at:

Email Alerts: http://www.jclinpharm.org/cgi/alerts

Subscriptions: http://www.jclinpharm.org/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

Downloaded from http://www.jclinpharm.org by Luciana Oliveira on April 19, 2009


title

Effect of Different Durations of Ketoconazole


Dosing on the Single-Dose Pharmacokinetics of
Midazolam: Shortening the Paradigm
S. A. Stoch, MD, E. Friedman, MS, A. Maes, PhD, K. Yee, PhD, Y. Xu, PhD,
P. Larson, MS, M. Fitzgerald, MD, J. Chodakewitz, MD, and J. A. Wagner, MD, PhD

Given the prominent role of cytochrome P450 3A (CYP3A) zolamday 1 and ketoconazole + midazolamday 5/ketoco-
in the metabolism of drugs, it is critical to determine nazole + midazolamday 2) for midazolam AUC0-∞ were
whether new chemical entities will be affected by the inhi- 1.36 and 1.06 with corresponding 90% confidence inter-
bition of this enzyme system and result in clinically rele- vals of (1.17, 1.57) and (0.83, 1.23), respectively. These
vant drug interactions. Ketoconazole interaction studies findings suggest that short-term drug-drug interaction
are frequently performed to determine a given compound’s studies can predict the magnitude of change in AUC as
sensitivity to CYP3A metabolism. The present study evalu- reliably as studies using longer duration treatments.
ated whether probing a sensitive substrate (midazolam)
with a potent inhibitor (ketoconazole) at earlier time points Keywords:  Midazolam; ketoconazole; drug-drug inter-
(days 1 or 2) might be used to reliably gauge the magnitude actions; cytochrome P450 3A
of a meaningful interaction. The geometric mean ratios Journal of Clinical Pharmacology, 2009;49:398-406
(ketoconazole + midazolamday 5/ketoconazole + mida- © 2009 the American College of Clinical Pharmacology

T he CYP3A family of enzymes plays a major role


in the biotransformation of many commonly
used drugs1 and is present in the liver and gut.
unique opportunity to prioritize compounds and
select molecules with a lower likelihood of being a
sensitive CYP3A substrate. If an investigational drug
Therefore, the importance of assessing the impact of is a putative substrate, an assessment is made based
CYP3A inhibition on the metabolism of a new chem- both on in vitro and in vivo interaction studies.
ical entity (NCE) cannot be overemphasized. Drug- Based on these data, clinical studies are performed
drug interactions (DDIs) can lead to important severe to assess the likelihood and the magnitude of the
side effects and have previously resulted in the effect that may be anticipated in the clinic. From a
withdrawal of approved drugs.2 The relevance of drug development standpoint, it is preferable to
assessing this early in drug development cannot be make an informed decision as early as possible as to
underestimated, particularly in evaluating NCEs whether a large drug interaction liability may exist.
with a projected narrow therapeutic index. Therefore, It may be possible to evaluate this after a single or
the ability to reliably gauge the magnitude of a clini- limited multiple-dose administration.
cally meaningful DDI early in development affords a To date, no prospective within-subject clinical study
data have been reported in the literature to evaluate the
changes in the pharmacokinetics of midazolam, a
From Merck & Co, Inc, Whitehouse Station, New Jersey (Dr Stoch, E. sensitive CYP3A substrate, following the administration
Friedman, Dr Maes, Dr Yee, Dr Xu, P. Larson, Dr Chodakewitz, Dr of different durations of ketoconazole, a strong CYP3A
Wagner) and ProMedica Clinical Research Center, Inc, Boston,
inhibitor. The present study was designed to explore
Massachusetts (Dr Fitzgerald). Submitted for publication July 28, 2008;
revised version accepted December 19, 2008. Address for correspond-
whether shortening the “traditional” duration of
ence: S. Aubrey Stoch, MD, Merck & Co, Inc, RY34-A500, PO Box ketoconazole dosing from 5 days to either 2 days or a
2000, Rahway, NJ 07065-0900; e-mail: aubrey_stoch@merck.com. single-dose administration could be used to reliably
DOI: 10.1177/0091270008331133 predict clinically meaningful effects on midazolam

398 • J Clin Pharmacol 2009;49:398-406


EFFECT OF KETOCONAZOLE DOSING ON MIDAZOLAM

pharmacokinetics. A 5-day ketoconazole dosing Treatment C: multiple doses of 400 mg ketoconazole for 2
paradigm with the administration of a 2 mg3 dose of days with 2 mg midazolam coadministered on day 2
midazolam on day 5 has been used by certain sponsors Treatment D: multiple doses of 400 mg ketoconazole for 5
to study the effect of a potent CYP3A inhibitor on the days with 2 mg midazolam coadministered on day 5
pharmacokinetics of a sensitive CYP3A substrate. It is
important to ascertain whether these trials can be Participants were randomly assigned to 1 of the 4
shortened without compromising their predictive treatment sequences, and there was a minimum of 7
value. This would minimize the exposure of healthy days between each treatment period. All doses of
volunteers while at the same time realizing more midazolam were administered in the fasted state.
nimble drug development and more efficient decision Ketoconazole was administered without regard to
making. This study was also simulated using a food except on days when it was coadministered
ketoconazole-interaction model developed by Chien with midazolam, in which case ketoconazole was
et al,4 in which the inhibitory effect of ketoconazole given fasted. All doses of ketoconazole and midazo-
was explicitly accounted for by changes in the lam were administered in the morning with 240 mL
substrate’s bioavailability and systemic clearance of water. No acidification was employed in the
due to ketoconazole concentrations in the gut, plasma, administration of either study drug.5
and liver. If the model is able to successfully replicate Midazolam
results observed clinically, it may provide insight into
the mechanisms behind the experimentally observed Midazolam (Roxane Laboratories, Columbus, Ohio) was
results and serve as a predictive tool to supplement administered as a 2 mg oral dose (2.0 mg/mL midazo-
abbreviated ketoconazole DDI studies. lam HCL syrup). All doses of midazolam were adminis-
tered with 240 mL of water. On days of coadministration
METHODS with ketoconazole, midazolam was given 5 minutes
after the ketoconazole dose. All doses of midazolam
Participants were administered following an 8-hour fast.
Ketoconazole
A total of 12 healthy, white, male subjects were
Ketoconazole (Teva Pharmaceuticals, Petah Tikva,
enrolled in this study. All participants were non-
Israel) was administered as oral 400 mg doses (1 ×
smokers with a mean age of 38.8 years (range, 21-54
400‑mg ketoconazole tablet). All doses of ketocon-
years) and a body mass index of 18.5 to 27.0 kg/m2
azole were administered with 240 mL of water and
with corresponding body weights between 64 and 89
without regard to food except when coadministered
kg. Participants were in good general health accord-
with midazolam, in which case both drugs were
ing to routine medical history, physical examina-
given after an 8-hour fast.
tion, vital signs, 12-lead electrocardiogram, and
laboratory data.
Every participant gave written informed consent to Pharmacokinetic Assessments
participate in this study. The protocol was approved
by the institutional review board (New England Blood samples for midazolam pharmacokinetics were
Institutional Review Board) and was conducted in collected predose and at various time points post-
accordance with the guidelines on good clinical dose. Immediately after collection, blood samples
practice and with ethical standards for human were kept on ice, for no more than 30 minutes, until
experimentation established by the Declaration of centrifugation at 1500 g for 15 minutes at 4°C. The
Helsinki Principles. plasma fraction was removed and stored at –20°C
until analyzed for midazolam concentrations.
Study Design When midazolam was administered alone, blood
samples were drawn, and plasma was collected
This was a 4-period, open-label, randomized, cross- predose and at 0.25, 0.5, 1, 2, 4, 6, 8, and 12 hours after
over study in 12 healthy male participants. The 4 dosing. When midazolam was coadministered with
treatments were as follows: ketoconazole, on day 1, 2, or 5, blood samples were
drawn, and plasma was collected predose and at 0.25,
Treatment A: a single 2 mg dose of midazolam 0.5, 1, 2, 4, 6, 8, 12, 16, and 24 hours after dosing.
Treatment B: a single 2 mg dose of midazolam coadmin- Plasma concentration of midazolam was determined
istered simultaneously with a single dose of 400 by reverse-phase high-performance liquid chroma­
mg ketoconazole tography (HPLC) coupled with tandem quadrupole

DRUG INTERACTIONS 399


STOCH ET AL

mass spectrometry (MS/MS). The sample analysis was contained within the interval (0.50, 2.00). To address
conducted at Pharmaceutical Product Development this hypothesis, we followed a closed testing procedure
(Wilmington, North Carolina). Midazolam and its starting with comparison of ketoconazole administra­
isotopically labeled internal standard were extracted tion on day 5 versus day 2, then day 5 versus day 1.
from human plasma through liquid-liquid extraction Two-sided 90% confidence intervals for the midazolam
using methyl-t-butyl ether and separated by reverse- AUC0-∞ geometric mean ratios (GMRs, midazolam +
phase chromatography using a Thermo Aquasil C18 ketoconazoleMD, day 5/midazolam + ketoconazoleMD, day 1 or 2)
column (2.1 × 100 mm, 5 mm) with a gradient composed were calculated from the above mixed effects model. If
of acetonitrile, water, and ammonium acetate. The the 90% confidence interval for the AUC0-∞ GMR was
analytes were detected using an MDS Sciex API 3000 contained within the prespecified bounds of (0.50,
triple quadrupole mass spectrometer system. Ionization 2.00), then no clinically meaningful decrease in AUC0-∞
occurred using positive ion Turbo IonSpray, and the after ketoconazole administration on day 2 (day 1) was
compounds were detected at the mass transitions m/z = concluded.
326 → 291 and m/z = 330 → 295 for midazolam and its
internal standard, respectively. The data system was Modeling and Simulation
configured to calculate and annotate the areas of
midazolam and internal standard peaks automatically. Observed concentration-time profiles from adminis-
A calibration curve was constructed using peak area tration of a single 2 mg dose of midazolam without
ratios of the calibration standards by applying a linear ketoconazole were fit to a 2-compartment model to
weighted (1/concentration squared) least squares determine mean absorption rate (Ka), clearance
regression algorithm. The midazolam concentrations (CL/F), central compartment volume (Vc/F), periph-
in clinical samples were then back-calculated from eral compartment volume (Vp/F), and transport
their peak area ratios of midazolam versus internal between central and peripheral compartments (Q)
standard against the calibration curve. The lower limit using NONMEM (Version VI, GloboMax, Ellicot
of quantitation (LLOQ) for midazolam was 0.1 ng/mL. City, Maryland). The demographics of this popula-
Calibration standards ranged from 0.1 to 100 ng/mL. tion are described in the “Participants” section. The
With 0.3 ng/mL (3× LLOQ) midazolam quality control optimal parameter fit was chosen based on visual
samples, the accuracy and precision were 91.24% and goodness of fit and minimization of the standard
7.17% (% coefficient of variation [CV]), respectively, error of the estimates. Due to the lack of pharma-
across the analytical runs (n = 5). cokinetic sampling points beyond 12 hours postdose
for this data set, good estimates for intersubject vari-
Statistical Analyses ability could not be obtained. Nevertheless, based
on visual inspection, these rough parameter esti-
The effects of multiple oral doses of 400 mg ketocon- mates (see Table III) adequately capture the observed
azole once daily on the midazolam single-dose phar- midazolam pharmacokinetics (Figure 2) and are suf-
macokinetic parameters (AUC0-∞, Cmax, tmax, and ficient for our purposes because the focus of the
apparent terminal t1/2) were evaluated using a linear simulation model is on ketoconazole and its inhibi-
mixed effects model with fixed effects for period and tory effects rather than on midazolam population
duration of ketoconazole administration (0, 1, 2 or 5 pharmacokinetics.
days) and a random effect for subject. The assump- The midazolam 2-compartment model was then inter­
tions of the linear model were tested and found to be faced with a model for ketoconazole pharmacokinetics
satisfied. AUC0-∞ and Cmax were log-transformed, tmax and inhibition effects previously developed by Chien et
was ranked, and the reciprocal of apparent terminal al.4 This model incorporates the effects of changing
t1/2 was taken prior to conducting the analyses. Back- ketoconazole concentrations over time in the gut,
transformed summary statistics and inferential plasma, and hepatic enzyme site due to ketoconazole
results are reported. dosing, absorption, and clearance by compartmental
The primary hypothesis of this study was that the modeling of ketoconazole pharmacokinetics. The
midazolam AUC0-∞ following a single 2 mg dose unbound concentrations of ketoconazole in the
coadministered on day 5 of once-daily dosing of 400 gut and hepatic compartments at any given time
mg ketoconazole is less than 2-fold than that of a 2-mg are used to estimate a corresponding midazolam
dose coadministered on day 1 or 2. That is, the clearance. Parameter values for the ketoconazole
midazolam AUC0-∞ geometric mean ratio (ketoconazole pharmacokinetic and inhibitory effect model were
+ midazolamday 5/ketoconazole + midazolamday 1 or 2) is the same as those used by Chien et al,4 whereas

400  •  J Clin Pharmacol 2009;49:398-406


EFFECT OF KETOCONAZOLE DOSING ON MIDAZOLAM

Table I Single-Dose Pharmacokinetics of Midazolam With and Without 400 mg Ketoconazole


Pharmacokinetic Least Squares Geometric
parameter Treatment n Mean (SD) Mean Ratioa (95% CI)

AUC0-∞, ng/mL⋅h 400 mg ketoconazole + 2 mg midazolamday 5 10 265.9 (82.9) 13.96 (11.79, 16.53)
400 mg ketoconazole + 2 mg midazolamday 2 10 250.3 (105.6) 13.14 (11.08, 15.58)
400 mg ketoconazole + 2 mg midazolamday 1 10 195.9 ( 76.7) 10.28 (8.68, 12.19)
2 mg midazolam 11 19.0 (10.6)
Cmax, ng/mL 400 mg ketoconazole + 2 mg midazolamday 5 10 39.1(7.5) 5.42 (4.25, 6.90)
400 mg ketoconazole + 2 mg midazolamday 2 10 38.1 (12.3) 5.29 (4.15, 6.75)
400 mg ketoconazole + 2 mg midazolamday 1 10 36.1 (6.8) 5.01 (3.93, 6.39)
2 mg midazolam 11 7.2 (3.4)
tmax, hb 400 mg ketoconazole + 2 mg midazolamday 5 10 1.00 (0.50, 1.00)
400 mg ketoconazole + 2 mg midazolamday 2 10 0.75 (0.50, 2.00)
400 mg ketoconazole + 2 mg midazolamday 1 10 1.00 (0.50, 2.00)
2 mg midazolam 11 0.50 (0.25, 1.00)
t1/2, hc 400 mg ketoconazole + 2 mg midazolamday 5 10 6.89 (3.56)
400 mg ketoconazole + 2 mg midazolamday 2 10 6.07 (3.23)
400 mg ketoconazole + 2 mg midazolamday 1 10 4.91 (2.11)
2 mg midazolam 11 2.61 (1.07)
CI, confidence interval.
a. Ketoconazole + midazolamday x/midazolam alone.
b. Median (range).
c. Harmonic mean ± jackknife standard deviation.

gender, weight, and age distributions for the subject Pharmacokinetics


population were based on our clinical study
population. Concentration-time profiles for a single Effect of different durations of ketoconazole dosing on
dose of midazolam administered in the absence of the single-dose pharmacokinetics of midazolam. The
ketoconazole and with 1, 2, and 5 days of ketoconazole effects of 400 mg ketoconazole, administered for 1, 2,
treatment were simulated in Trial Simulator 2.2.1 or 5 days, on the single-dose pharmacokinetics of
(Pharsight Corp, Mountainview, California) using midazolam are summarized in Table I and Figure 1.
the 2-compartment pharmacokinetic parameters Compared with a single dose of 2 mg midazolam given
estimated in NONMEM and the ketoconazole alone, administration of 400 mg ketoconazole for 1, 2,
inhibition model described. Because the midazolam or 5 days resulted in a 10.28-, 13.14-, and 13.96-fold
pharmacokinetic parameters represent a composite increase in midazolam AUC0–∞, respectively. The max-
subject based on the population in the clinical study, imum concentration (Cmax) of midazolam was increased
only intrasubject variability terms were included in 5.01-, 5.29-, and 5.42-fold, respectively, when midazo-
the ketoconazole-midazolam interaction model. lam was administered on day 1, 2, or 5 of once-daily
Intrasubject variability could not be estimated from dosing of 400 mg ketoconazole. Ketoconazole, regard-
the midazolam data and was assumed to be normally less of the dosing duration, did not affect the time to
distributed with a CV of 30%. Cmax (tmax) of midazolam. Coadministration of ketocon-
azole for 1, 2, and 5 days increased the apparent termi-
RESULTS nal half-life (t1/2) of midazolam to 4.9, 6.1, and 6.9
hours, respectively, compared with 2.6 hours when
midazolam was dosed alone.
Participant Demographics
Comparative effect of 1, 2, and 5 days of ketocon-
Of the 12 participants enrolled in the study, 3 with- azole dosing on the single-dose pharmacokinetics of
drew consent for personal reasons prior to complet- midazolam. The comparative effect that different
ing all 4 treatment periods. Data from 11 participants durations of ketoconazole dosing had on the single-
who completed at least 2 treatments were used in the dose pharmacokinetics of midazolam is summarized
evaluation of midazolam pharmacokinetics. Of these in Table II. The geometric mean ratio (ketoconazole
11 participants, 9 completed all 4 treatment periods, + midazolamday 5/ketoconazole + midazolamday 2) for
1 completed 3 periods, and 1 completed 2 periods. midazolam AUC0-∞ was 1.06 with a corresponding

DRUG INTERACTIONS 401


STOCH ET AL


administered on day 2 or day 5 of once-daily dosing

of 400 mg ketoconazole. However, the comparison of
  midazolam AUC0-∞ after a single dose of midazolam
0LGD]RODPSODVPDFRQFHQWUDWLRQ QJP/

coadministered on day 5 of dosing with ketoconazole


  versus a single dose of midazolam coadministered
with a single dose of ketoconazole reached statistical
 
significance (P > .002). Similarly, a statistically sig-
     
nificant treatment difference in midazolam AUC0-∞
 0'=RQO\ Q 
0'=RQ'D\RI.HWR Q  was also observed for a single dose of midazolam
0'=RQ'D\RI.HWR Q 
0'=RQ'D\RI.HWR Q  coadministered on day 2 of ketoconazole dosing ver-

sus a single dose of midazolam coadministered with
a single dose of ketoconazole (P = .007). There were

     
no statistically significant between-treatment differ-
7LPH K ences in midazolam Cmax and tmax, but a statistically
significant difference in midazolam t1/2 was observed
Figure 1. Mean midazolam plasma concentration (ng/mL) ver- when midazolam was given on day 1 versus day 5 of
sus time following single-dose administration of 2 mg midazolam
with and without 400 mg ketoconazole in the fasted state to
once-daily dosing with 400 mg ketoconazole.
young, healthy male participants.
Model Validation and Application to
Experimental Results
90% confidence interval of (0.92, 1.23). The geomet-
ric mean ratio (ketoconazole + midazolamday 5/keto- The 2-compartment pharmacokinetic model for
conazole + midazolamday 1) for midazolam AUC0-∞ midazolam described previously was interfaced with
was 1.36 with a corresponding 90% confidence the ketoconazole interaction model and used to
interval of (1.17, 1.57). Based on the prespecified simulate the present experimental study (see Table
AUC0-∞ comparability bounds for the 90% confi- III). In general, the model was able to reproduce the
dence interval (0.5, 2.00), there was no meaningful quantitative and qualitative results of the experi-
difference observed for midazolam AUC0-∞ following mental study, where the effects of 5 days of ketocon-
a single 2-mg oral dose of midazolam coadminis- azole administration on the substrate AUC0-∞ and
tered on day 1, 2, or 5 of once-daily dosing with 400 Cmax may be determined after 1 or 2 days of ketocon-
mg ketoconazole. There was no difference in mida- azole administration. The simulated effects of keto-
zolam AUC0-∞ when a single dose of midazolam was conazole administration on single-dose midazolam

Table II Comparative Effect of Different Durations of Ketoconazole Dosing


on the Single-Dose Pharmacokinetics of Midazolam
AUC0-∞ Cmax tmax Apparent
Between- Between- Between- t1/2 Between-
AUC0-∞ GMR Treatment Cmax GMR Treatment Treatment Treatment
Treatment Comparison (90% CI) P Valuea (90% CI) P Valuea P Valuea P Valuea

Ketoconazole + 1.06 (0.92, 1.23) >.200 1.02 (0.83, 1.26) >.200 >.200 .164
midazolamday 5 vs
ketoconazole +
midazolamday 2
Ketoconazole + 1.36 (1.17, 1.57) .002 1.08 (0.88, 1.33) >.200 >.200 .005
midazolamday 5 vs
ketoconazole +
midazolam day 1
Ketoconazole + 1.28 (1.11, 1.47) .007 1.05 (0.86, 1.30) >.200 >.200 .097
midazolamday 2 vs
ketoconazole +
midazolamday 1
CI, confidence interval; GMR, geometric mean ratio.
a. P value < .050 indicates a statistically significant difference between treatment regimens.

402  •  J Clin Pharmacol 2009;49:398-406


EFFECT OF KETOCONAZOLE DOSING ON MIDAZOLAM

Table III   Estimated Midazolam 2-Compartment


Model Pharmacokinetic Parameters
Standard
Error (%
Parameter Estimate of Estimate)

Ka, h–1 0.797 17


CL/F, L/h 111 16
Vc/F, L 76.7 24
Vp/F, L 47.6 24
Q, L/h 174 13
Proportional residual error 0.076 26
CL/F intersubject variability 0.186 51
Vc/F intersubject variability 0.569 66

pharmacokinetics were in good agreement with


experimentally observed concentration-time profiles
(Figure 2) and fold-changes in AUC0-∞ and Cmax fol-
lowing 1, 2, and 5 days of ketoconazole administra-
tion (Figure 3). The ability of the model to
quantitatively reproduce the experimental results
suggests that the model adequately captures the
Figure 2. Experimental and simulated midazolam plasma con-
mechanisms of ketoconazole inhibition of midazo- centration (ng/mL) versus time following single-dose administra-
lam metabolism and therefore may be a source of tion of 2 mg midazolam with and without 400 mg ketoconazole in
insight into the mechanisms behind the results of the fasted state to young, healthy male participants. The solid
the experimental study. and dotted lines are the median and 5th and 95th percentile
In both our within-subject clinical study and the simulated profiles for n = 1000 runs. The points correspond to
predicted results from the ketoconazole inhibition experimentally observed midazolam concentrations.
model, ketoconazole has exerted its maximal inhibitory
effect by day 2 of ketoconazole dosing. The effect of 5
consecutive days of ketoconazole dosing on systemic
clearance in our simulations is shown in Figure 4,
providing insight into the potential mechanism for the
time course of ketoconazole’s inhibitory activity. In our
simulations, it would appear that clearance has already
reached steady-state levels following a single dose
of ketoconazole administration despite predicted
accumulation of ketoconazole at the site of midazolam
metabolism in the liver throughout the 5 days of
dosing. Moreover, large fluctuations in gut ketoconazole
concentration following each daily dose of ketoconazole
have little effect on the overall clearance of midazolam.
Based on the ketoconazole interaction model, the Figure 3. Midazolam AUC0-∞ and Cmax ratios after 1, 2, and 5
concentration of ketoconazole in the gut after each days of once-daily ketoconazole administration compared to a
dose of ketoconazole increases the gut bioavailability single dose of midazolam only. Experimental results from the cur-
rent study are plotted as individual points representing geometric
(Fgut) of midazolam nearly 2-fold by inhibition of mean ratios (GMRs) and 95% confidence intervals (CIs) for each
CYP3A activity in the gut. However, given the rapid ketoconazole dosing regimen, whereas simulated results are plot-
absorption of ketoconazole, there is no accumulation ted as solid (median), dotted (25th and 75th percentile), and
in the gut, and the fluctuations in Fgut due to daily dashed (5th and 95th percentile) lines.

DRUG INTERACTIONS 403


STOCH ET AL

50 50 predict the data obtained from more “traditional,”


Median
5th and 95th
Inset: t = 0 – 24hours
lengthier interaction studies. The goal was to evaluate
40

Midazolam Plasma
whether a strong CYP3A inhibitor, ketoconazole,
Midazolam Plasma Clearance (L/hr)

Clearance (L/hr)
Percentiles
40 30
would affect the pharmacokinetics of a sensitive
20 substrate, midazolam, to a similar extent when
30
10 midazolam was coadministered with ketoconazole
on either day 1 or 2 when compared to probing on
0
20
0 5 10 15 20 day 5 to assess the magnitude of key drug-drug
Time (hr)
interactions. Given the importance of evaluating drug-
10
drug interaction potential in early development to
inform critical decision making, this study focused on
the timing of substrate administration relative to
0
0 20 40 60 80 100 120 140 160
interacting drug (single dose and multiple dose).
Time (hr) Midazolam, a short-acting benzodiazepine that is
selectively metabolized by CYP3A and widely used
Figure 4. Simulated midazolam plasma clearance after 5 days of as a phenotyping probe for CYP3A activity, was
ketoconazole administration (inset: t = 0-24 hours). The solid line
administered at a dose of 2 mg orally to investigate the
represents the median (n = 1000) predicted plasma clearance,
whereas the dotted lines represent the 5th and 95th percentiles.
maximal inhibitory effect in vivo.3,6 Ketoconazole, a
strong CYP3A inhibitor, was used at a dose of 400 mg
daily as an in vivo probe to evaluate the magnitude of
CYP3A-mediated drug interactions with midazolam on
ketoconazole administration are nearly identical days 1, 2, and 5. Midazolam pharmacokinetic parameters
following 1, 2, 5, or more days of dosing. Therefore, have been shown to change dramatically during the
administering additional doses of ketoconazole administration of ketoconazole with increases in
through days 2 and 5 will likely have minimal additional AUC0-∞ of ~16-fold and Cmax of ~5-fold7-10 and may be
effect on the inhibitory potential of ketoconazole accompanied by an almost 5-fold prolongation of the
because maximum changes in substrate bioavailability midazolam elimination half-life with ketoconazole.9
and clearance are achieved by day 1. This is consistent Consistent with data reported in the literature,
with what was observed in our clinical study. probing with midazolam on day 5 of multiple-dose
ketoconazole administration resulted in a 13.96- and
Safety and Tolerability 5.4-fold increase in midazolam AUC0-∞ and Cmax,
respectively. When comparing these findings to the
Administration of ketoconazole and midazolam, magnitude of response on day 2 of coadministration of
either separately or together, was well tolerated by ketoconazole with midazolam, the increase in AUC0-∞
the study participants. Of the 11 participants and Cmax was indistinguishable at 13.14- and 5.3-fold,
included in the pharmacokinetic analysis, none respectively. Day 2 and day 5 were com­parable based
reported any side effects. on the prespecified bounds (0.5 to 2.00) and would
also have met the more stringent bioequivalence (0.8,
DISCUSSION 1.25) bounds for AUC0-∞ and closely for Cmax had they
been employed. Similarly, if one coadministered
Given the prominent role of CYP3A in the metabo- ketoconazole 400 mg and midazolam 2 mg on day 1, a
lism of drugs, it is important to identify early in 10.28- and 5.0-fold increase in midazolam AUC0-∞ and
development whether new chemical entities will be Cmax, respectively, would be observed. Although the
affected by this enzyme system and produce clini- AUC0-∞ data on day 1 do not exhibit the full degree of
cally relevant drug interactions. Because the expres- inhibition as that observed on days 2 and 5, the
sion and function of CYP3A enzymes are highly magnitude of effect seen on day 1 is likely clinically
variable, large intersubject variability in pharmacoki- relevant. The alteration in midazolam pharmaco­
netic parameters can result. This could pose chal- kinetics seen with coadministration on day 1 is in
lenges for a drug with a narrow therapeutic index, keeping with data reported by others following
such as midazolam. Conducting DDI studies early in pretreatment with ketoconazole for 4 days, which
development and exercising confident decision mak- resulted in a 10-fold increase in AUC0-∞.7 These data
ing as early as possible cannot be overemphasized. support conducting shorter drug-drug interaction
The objective of this study was to determine whether studies using strong CYP3A inhibitors without
performing shorter DDI studies may be used to reliably compromising data quality.

404  •  J Clin Pharmacol 2009;49:398-406


EFFECT OF KETOCONAZOLE DOSING ON MIDAZOLAM

The notable strengths of this study include the suggests that despite continued accumulation of
within-subject treatment comparisons and low ketoconazole in the liver over 5 days of dosing, the
variability estimates resulting from the randomized maximum change in midazolam clearance caused by
crossover design. Potential limitations of our study are ketoconazole is achieved within 1 day of ketoconazole
discussed below. This study lacks the use of midazolam dosing. Furthermore, daily fluctuations in ketoconazole
pharmacodynamic testing and 1′-hydroxymidazolam level in the gut give rise to large changes in the
analysis. The 1-hydroxy metabolite accounts for 50% bioavailability of midazolam over the course of 24
to 70% of metabolism of midazolam and is reported to hours but are consistent between consecutive days
be pharmacologically active.11,12 Various authors have of dosing, so that the steady-state inhibitory effects of
investigated the ratio of 1′-hydroxymidazolam to ketoconazole are achieved after approximately 1 day
midazolam in plasma as an index for CYP3A4 activity of dosing.
but with inconsistent results13,14 because the hydroxyl- However, the enzyme site with the saturable
metabolites of midazolam undergo extensive glucuro­ efflux model used here predicts that significant
nidation.15 Ketoconazole administration was given in concentrations of ketoconazole remain in the enzyme
the fasted state on days when it was coadministered site for a number of days following administration of
with midazolam, but on all other study days, the last dose of ketoconazole. The present clinical
ketoconazole was given without regard to food. study results suggest that the accumulation of
Although the potential impact of food on ketoconazole ketoconazole in the enzyme site does not play a role
pharmacokinetics was not assessed in this study, a in the observed midazolam pharmacokinetics as
previous report concluded that only tmax is significantly there were minimal carryover effects. Thus, although
affected by food, and AUC, Cmax, and t1/2 the saturable efflux mechanism may be sufficient to
pharmacokinetics are not altered to a statistically predict the inhibitory effects of ketoconazole, it may
significant extent.5 One should also take into not accurately reflect the true accumulation of
consideration the relative small, homogeneous ketoconazole in the hepatic enzyme site in vivo.
population restricted to white men employed in the Given the good agreement between simulated results
present study (n = 10 or 11), particularly when from the ketoconazole inhibition model and the clinical
compared with those evaluated in model-based study, the model may also serve as a predictive tool to
simulation work (n = 1000); therefore, it is possible complement the abbreviated clinical study proposed
that both reported exposure (AUC0-∞) and Cmax observed here. With clinical concentration-time profiles for a
in an experimental study may not necessarily reflect single dose of substrate without ketoconazole and after
the true population mean. As a consequence, some 1 day of ketoconazole administration, substrate
discrepancy may exist between experimental and pharmacokinetic parameters may be estimated and the
simulated data, and a direct comparison between the predictive ability of the model validated. The
two may not be possible. concentration-time profile of the substrate after 2, 5, or
Simulation models to predict the effect of more days of ketoconazole dosing may then be predicted.
ketoconazole inhibition of CYP3A on midazolam Moreover, with estimates for the Ki for ketoconazole
pharmacokinetics4,16-19 have the potential to be a inhibition (fraction unbound for ketoconazole and the
complementary tool to the clinical study described substrate) and the Vmax and Km for CYP3A metabolism
here. In the ketoconazole interaction model reported of the substrate (the fraction of metabolism attributed to
by Chien et al,4 biological details such as saturable CYP3A [fm] and the bioavailability in the gut), the
efflux from the hepatic enzyme site, gut wall ketoconazole-inhibition model potentially may be
permeability, and metabolism in the gut were applied to substrates other than midazolam.
considered. Based on this model, the inhibitory activity
of ketoconazole is mediated by the ketoconazole Conclusion
concentration in the gastrointestinal and hepatic
compartments, which increases the oral bioavailability Although extensive CYP3A inhibition with <24 hours
and decreases the hepatic clearance of midazolam. of ketoconazole exposure is well recognized,20 this
The observed agreement between our simulated and study demonstrated that earlier administration of a
experimental data suggests that this reliably captures sensitive substrate (midazolam) relative to a potent
the effects of ketoconazole. The compartment-based interacting drug (ketoconazole) may be used to reli-
ketoconazole inhibition model also provides a view ably predict the magnitude of drug-drug interaction
into the mechanisms in the gut and liver that may potential, which may be very enabling in early drug
contribute to the clinically observed result. This model development. The extent of the ­interaction observed

DRUG INTERACTIONS 405


STOCH ET AL

after earlier administration (day 1 or 2) was consis- 7. Olkkola K, Backman, J, Neuvonen M, et al. Midazolam should be
tent with that reported in the literature of key drug- avoided in patients receiving the systemic antimycotics ketocon-
azole and itraconazole. Clin Pharmacol Ther. 1994;55:481-485.
drug interaction studies of longer (5 days) duration.
8. Gorski J, Jones D, Haehner-Daniels B, et al. The contribution of
It was remarkable how predictive day 2 data were of
intestinal and hepatic CYP3A4 to the interaction between midazo-
day 5; in addition, data obtained on day 1 were also lam and clarithromycin. Clin Pharmacol Ther. 1998;64:133-143.
a close approximate to those obtained on day 5. The 9. Tsunoda S, Vlez R, Moltke L, et al. Differentiation of intestinal
decision whether to implement probing on day 1 and hepatic cytochrome P450 3A activity with use of midazolam
versus day 2 may rest on the degree of confidence as an in vivo probe: effect of ketoconazole. Clin Pharmacol Ther.
that would be required for decision making. Based 1999;66:461-471.
on this study, there is no difference between days 2 10. McCrea J, Prueksaritanont T, Gertz B, et al. Concurrent admin-
and 5, and day 1 would be a close approximate. istration of the erythromycin breath test (EBT) and oral midazo-
lam as in vivo probes for CYP3A activity. J Clin Pharmacol.
Although midazolam was selected as the sensitive 1999;39:1212-1220.
CYP3A probe substrate and ketoconazole as the
11. Zieler WH, Schalch E, Leishman B, et al. Comparison of the
inhibitor, one could speculate that a similar relation- effects of intravenously administered midazolam, triazolam and
ship may extend to other less sensitive CYP3A sub- their hydroxy metabolites. Br J Clin Pharmacol. 1983;19:271-278.
strates but not necessarily to less potent inhibitors. 12. Mandema JW, Tuk B, van Steneninck AL, et al. Pharmacokinetic-
pharmacodynamic modeling of the central nervous system effects
The authors acknowledge Rita Chiou and Jin Zhang for coor- of midazolam and its main metabolite alpha-hydroxymidazolam
dination of midazolam sample analysis. The authors wish to in healthy volunteers. Clin Pharmacol Ther. 1992;51:715-728.
thank Susi Li for her constructive input and review of this manu- 13. Lee LS, Bertino JS Jr, Nafziger AN. Limited sampling models
script. The authors thank the participants and study personnel at for oral midazolam: midazolam plasma concentrations, not the
Promedica CRC, Inc, who made this study possible. ratio of 1-hydroxymidazolam to midazolam plasma concentra-
Financial disclosure: This study was funded by Merck & Co, tions, accurately predicts AUC as a biomarker of CYP3A activity.
Inc. Authors who are employees of Merck may potentially own J Clin Pharmacol. 2006;46:229-234.
stock and/or hold stock options in the company. Authors who are 14. Rogers JF, Nafziger AN, Kashuba AD, et al. Single plasma
not employees of Merck have received grant support, consultant concentrations of 1′-hydroxymidazolam or the ratio of 1′-
fees, and/or lecture honoria. hydroxymidazolam:midazolam do not predict midazolam clear-
ance in healthy subjects. J Clin Pharmacol. 2002;42:1079-1082.
15. Link B, Haschke M, Grignaschi N, et al. Pharmacokinetics of
intravenous and oral midazolam in plasma and saliva in humans:
REFERENCES
usefulness of saliva as matrix for CYP3A phenotyping. Br J Clin
Pharmacol. 2008;66:473-484.
1. Kashuba A, Bertino J. Mechanisms of drug interactions I. In:
16. Zhou SF, Chan SY, Goh BC, et al. Mechanism-based inhibition
Pisceteill SC, Rodvold KA, eds. Drug Interactions in Infectious
of cytochrome P450 3A4 by therapeutic drugs. Clin Pharmacokinet.
Diseases. 2nd ed. Totowa, NJ: Humana; 2005:13-39.
2005;44:279-304.
2. Honig P, Wortham D, Zamani K, et al. Terfenadine-ketoconazole
17. Brown HS, Galetin A, Hallifax D, et al. Prediction of in vivo
interaction, pharmacokinetic and electrocardiographic conse-
drug-drug interactions from in vitro data: factors affecting proto-
quences. JAMA. 1993;269:1513-1518.
typic drug-drug interactions involving CYP2C9, CYP2D6 and
3. Lin Y, Lockwood G, Graham M, et al. In-vivo phenotyping for CYP3A4. Clin Pharmacokinet. 2006;45:1035-1050.
CYP3A by a single-point determination of midazolam plasma
18. Shou MG. Prediction of pharmacokinetics and drug-drug
concentration. Pharmacogenetics. 2001;11:781-791.
interactions from in vitro metabolism data. Curr Opin Drug
4. Chien J, Lucksiri A, Ernest C, et al. Stochastic prediction of Discov Dev. 2005;8:66-77.
CYP3A-mediated inhibition of midazolam clearance by ketocon-
19. Yamano K, Yamamoto K, Katashima M, et al. Prediction of
azole. Drug Metab Dispos. 2006;34:1208-1219.
midazolam-CYP3A inhibitors interaction in the human liver from
5. Daneshmend TK, Warnock DW, Ene MD, et al. Influence of food in vivo/in vitro absorption, distribution, and metabolism data.
on the pharmacokinetics of ketoconazole. Antimicrob Agents Drug Metab Dispos. 2001;29:443-452.
Chemother. 1984;25:1-3.
20. von Moltke L, Greenblatt D, Harmatz J, et al. Triazolam
6. Bjornsson T, Callaghan J, Einolf H, et al. The conduct of in vitro biotransformation by human microsomes in vitro: effects of
and in vivo drug-drug interaction studies: a Pharmaceutical metabolic inhibitors and clinical confirmation of a predicted
Research and Manufacturers of America (PhRMA) perspective. interaction with ketoconazole. J Pharmacol Exp Ther. 1996;
Drug Metab Dispos. 2003;31:815-832. 276:370-379.

406  •  J Clin Pharmacol 2009;49:398-406

You might also like