You are on page 1of 11

doi: 10.1111/j.1472-8206.2005.00315.

ORIGINAL Population pharmacokinetic analysis of


ARTICLE
indinavir in HIV-infected patient treated
with a stable antiretroviral therapy
Karl Brendela*, Mayeule Legrandb, Anne-Marie Taburetc, Gabriel Barona,
Cécile Goujardd, France Mentréa and the Cophar 1-ANRS 102 Trial Group
a
INSERM E0357, Department of Epidemiology, Biostatistics and Clinical research, AP-HP, Bichat University Hospital,
Paris, France
b
Clinical Pharmacology Department, AP-HP, Pitié-Salpétrière University Hospital, Paris, France
c
Clinical Pharmacy, AP-HP, Bicêtre University Hospital, Bicêtre, France
d
Internal Medicine, AP-HP, Bicêtre University Hospital, Bicêtre, France

Keywords ABSTRACT
indinavir,
population The objectives of this study were to build a population pharmacokinetic model that
pharmacokinetics, describe plasma concentrations of indinavir in human immunodeficiency virus (HIV)-
ritonavir, infected patients with sustained virological response under a stable antiretroviral
therapeutic drug combination, and to characterize the effect of covariates and comedications on
monitoring indinavir pharmacokinetics. Data were obtained from 45 patients who received
different dosages of indinavir: either indinavir alone t.i.d. (mostly 800 mg), either
Received 26 March 2004;
indinavir b.i.d. (mostly 800 mg) with a booster dose of 100 mg of ritonavir. Patients
revised 19 November 2004; were required to have a baseline plasma HIV RNA <200 copies/mL and to have
accepted 30 November 2004 unchanged antiretroviral treatment for 6 months. Indinavir concentrations were
measured at a first visit (one sample before drug administration and five after) and at
a second visit 3 months later (before and 1 or 3 h after drug administration). A one-
*Correspondence and reprints:
karl.brendel@bch.ap- compartment model with first-order absorption and first-order elimination best
hop-paris.fr described indinavir pharmacokinetics. For patients treated with indinavir alone,
absorption rate constant was estimated to be 0.43/h, and oral clearance Cl/F was
33 L/h. For patients treated with indinavir plus ritonavir these estimates were 0.25/h
and 19 L/h, respectively. Cl/F was found to increase by 1.45-fold in men and by
1.18-fold in patients also receiving zidovudine. Oral volume of distribution (V/F) was
24 L. The inter-individual and intra-individual variability were 117 and 205% for
V/F, 42 and 58% for Cl/F, respectively. This population analysis in patients with
sustained virological response, quantified the effect of ritonavir on the absorption rate
constant and on the clearance of indinavir, showed an increase of Cl/F in men and
can be used to draw reference curve for therapeutic drug monitoring.

inhibits the viral protease enzyme, one of the key


INTRODUCTION
enzymes that HIV needs in order to replicate. Results
Since the introduction of highly active antiretroviral from a number of studies suggest that indinavir should
therapy (HAART), life expectancy of human immuno- be used as part of a combination therapy regimen with
deficiency virus (HIV)-infected patients has been largely other anti-HIV drugs [3]. Indinavir plasma concentra-
improved by potent antiretroviral regimens including tions have been shown to be correlated with antiviral
protease inhibitors (PIs) and/or non-nucleoside reverse efficacy [4–9]. Furthermore, several studies showed
transcriptase inhibitors (NNRTIs) [1,2]. Indinavir is a a link between indinavir plasma concentration and
PI prescribed in the treatment of HIV infection which toxicity [8,10,11].

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383 373
374 K. Brendel et al.

Indinavir is metabolized in seven compounds: a by the Ethic committee of Bicêtre Hospital (France). One
glucuronide conjugate and six oxidative metabolites arm of this study was composed of patients who took
(CRIXIVANÒ; Merck & Co., Imc, Whitehouse station, indinavir and the other arm of patients with nelfinavir.
New Jersey, USA, US Package insert Merk RCO, 2002). It Patients from the indinavir arm were analyzed in this
is mainly metabolized by the liver into oxidative metab- paper. Patients were required to have a baseline plasma
olites and in vitro studies indicate that cytochrome P450 HIV RNA value below 200 copies/mL for at least
(CYP 3A4) is the major isoenzyme for oxidative meta- 4 months and to have the same antiretroviral treatment,
bolism. As ritonavir is a very potent CYP 3A4 inhibitor, including indinavir, for at least 6 months. There was no
indinavir plasma concentrations increase when ritonavir indication given to physician on the indinavir dosage
is co-administrated [12–14]. With a booster dose of and indinavir could be associated with ritonavir booster
ritonavir, 100 mg b.i.d., patients usually receive 800 mg (100 mg b.i.d.).
b.i.d. of indinavir instead of the standard regimen of Exclusion criteria were: co-administration of a treat-
800 mg t.i.d. when indinavir is given without ritonavir. ment known for its interaction with indinavir (such as
Although the increase in indinavir peak concentration rifampicin or itraconazole), renal deficiency (creatinemia
with ritonavir enhancement is relatively low, the overall >180 lmol/L), hepatic deficiency (prothrombin time
exposure enhancement appears to increase the risk of <50% or cirrhosis), ongoing opportunistic infection,
renal toxicity significantly in boosted regimens [15]. previous or ongoing chemotherapy.
The population approach is well adapted to describe Inclusion criteria were verified at visit 0 (V0). A first
the pharmacokinetics of indinavir in HIV patients. This indinavir pharmacokinetic profile was performed at visit
method can analyze sparse individual data with few 1 (V1) within a maximum of 30 days after V0. Four
concentrations by patient. In HIV-infected patients, four months after V0, two other plasma samples were collected
population pharmacokinetic analyses of indinavir are at visit 2 (V2). The duration of the trial was 8 months
published [16–19]. They involved data on 25, 22, 11, from V0, to the last visit V3. Routine clinical data, drug-
and 171 HIV-infected patients, respectively. The purpose related adverse event were recorded at each visit.
of the three first studies was to analyze cerebral Adherence was evaluated at V1 and V2 using a validated
penetration of indinavir from the ratio of the area under auto-questionnaire and applying the algorithm proposed
the cerebrospinal fluid and under the plasma concentra- by Carrieri et al. [20] to compute a score from 0 to 100%
tion curves. The last study analyzed the effect of the adherence. Patients were classified as highly adherent if
coadministration of several PIs on the pharmacokinetic they reported taking 100% of their prescribed regimen in
of efavirenz, and did not report a detailed population the last five days, and non-highly adherent otherwise.
pharmacokinetic analysis of indinavir.
The purpose of this paper were to build a population Plasma collection and analysis
pharmacokinetic model that describe plasma concentra- For the pharmacokinetic study at V1, five blood samples
tions of indinavir in HIV-infected patients with sustained were collected: a trough sample before indinavir admin-
virological response under a stable antiretroviral combi- istration and samples at 0.5, 1, 3 and 6 h after indinavir
nation, and to characterize the effect of covariates and administration. The two samples collected at V2 are a
comedications on indinavir pharmacokinetics. The other trough concentration and a peak plasma concentration
aim was to estimate inter-individual and intra-individual (Cmax). Because of the reported impact of ritonavir on
variability of the pharmacokinetic parameters. indinavir absorption, the peak concentration at V2 was
collected 1 h after administration for indinavir alone and
3 h after administration for indinavir associated with a
MATERIALS AND METHODS
booster dose of ritonavir.
Study population Indinavir concentrations were, when possible, assayed
Data were obtained from patients enrolled in the in the laboratory of pharmacology or toxicology of the
COPHAR 1-ANRS 102 study which was an open, clinical site having included the patient or sent to a close
multicentric (18 centers in France) and prospective trial one. A specific quality control was set up before the trial
whose aim was to describe indinavir and nelfinavir to validate the laboratories which were allowed to take
plasma concentrations in HIV-infected patients. This trial part in the study to analyze indinavir concentrations.
complies with the current laws of France: all subjects Twelve laboratories participated to indinavir assay in
gave a written consent and the protocol was approved plasma, after validation by a blind external quality

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


Population pharmacokinetics of indinavir 375

control. Plasma concentrations of indinavir were deter- the center where it was analyzed, we fixed them to LOQ/2
mined by specific high-performance liquid chromato- using the LOQ of the corresponding assay [27].
graphy assays with ultraviolet–photodiode array or
spectrofluorometric detection, according to previously Computational methods
published assays [21–25]. In brief, indinavir was isolated The models were fitted to the data by use of WinNonMix
from alkaline plasma samples by double-step liquid–liquid version 2.0.1 (Pharsight Corporation, Mountain View,
extraction or by solid–liquid extraction. Dry extracts were CA, USA) with the first-order method (FO). Simulations
injected onto C18 reversed phase column. Results of the were also performed using WinNonMix version 2.0.1.
blind interlaboratory quality control at three concentra-
tions were within 15% of the target values for medium Analysis of concentrations at V1
and high values and within 20% for low values [26]. First the indinavir model was built with the data at V1.
Lower limit of quantification (LOQ) were 0.005– Using the best error model, selected using the Akaike
0.1 mg/L for indinavir, depending on each method. criterion (AIC), several models were tested to determine
the basic model that best fit the data. The basic model was
Population pharmacokinetic modeling constructed by testing the inclusion of random effects on
A one-compartment model with first-order absorption Ka, Cl/F and V/F, and by testing the influence of taking or
and first-order elimination was used to fit the concen- not ritonavir on the three parameters. Goodness-of-fit
tration–time data for indinavir and for ritonavir. This plots (observed vs. predicted population and individual
model was parameterized with the oral volume of concentrations, weighted residuals vs. predicted concen-
distribution (V/F), the first-order absorption rate con- trations and vs. time) were examined for each model.
stant (Ka), and the oral clearance (Cl/F). Models were also compared using the AIC. The correlation
The statistical model for the observed plasma concen- between the pharmacokinetic parameters was also tested.
tration of one drug, Ci,j in patient i at time ti,j, is given by: Secondly, the effects of the following covariates were
evaluated from the basic model: age, weight, gender,
Ci;j ¼ f ðti;j ; hi Þ þ eij other antiretroviral drugs and adherence at V1 (as a
categorical variable). The effects of covariates on the
where hi is the pharmacokinetic parameter vector of empirical Bayes estimates of each individual random
patient i, eij the measurement error and f the pharma- effects from the basic model with ritonavir, were tested
cokinetic model. by using a Mann–Whitney non-parametric test for
An exponential random-effect model was chosen to categorical variables and a correlation test for continu-
describe inter-individual variability: ous variables. The covariate model was then built with
the covariates which were found to have a significant
hi ¼ h expðgi Þ
effect in this first step (P < 0.05). All population models
with all the combinations of these covariates were then
where h is the population mean vector of the phar-
evaluated. The combination with the smallest AIC was
macokinetic parameters, and gi represents the random
chosen in order to built the population covariate model.
effect vector. Random effects were assumed to follow a
normal distribution with zero mean and variance matrix
Analysis of concentrations at V1 and V2
X which was supposed to be diagonal.
From the model with covariates obtained with the data
Residual variability was modeled using a combined
at V1, the inter-occasion variability (IOV) was studied by
proportional and additive error model. The measurement
analyzing together the data at V1 and V2. The following
error eij is assumed to follow a normal distribution with
model was used where k ¼ 1 at V1 and k ¼ 2 at V2. Let
zero mean and an heteroscedastic variance r2i;j given by:
bki be the vector of random effects for total variability at
r2i;j ¼ r2 ða þ f ðti;j; hi ÞÞ2 : occasion k, gi for inter-individual variability (IIV) and jki
for inter-occasion variability at occasion k. We have:
This model is often used in population pharmacokinet-
bki ¼ gi þ jki
ics. For high concentrations, variance becomes propor-
tional to the square of the concentration. For the lowest Varðbki Þ ¼ Varðgi Þ þ Varðjki Þ
concentrations, the variance becomes proportional to a2.
When indinavir concentrations were below the LOQ of because gi and jki are assumed to be independent.

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


376 K. Brendel et al.

However, with the software WinNonMix, this more 200 patients for each group of combination of significant
complex random effects model cannot be implemented covariates given the total variability of the random
directly. Because there are only two occasions, we found effects without measurement error. This was performed
a way to implement it using IOV as a covariate. Two in order to illustrate the clinical relevance of the
random effects were modeled in WinNonMix: g0 for significant covariates found in the population analysis
occasion 1 only and g1 added to g0 for occasion 2. The on the real-through concentrations (without measure-
expressions for this implementation of the IOV are given ment error) which are thought to be related both to
in the Appendix. efficacy and toxicity [1,6].
The addition of random effects for IOV on one or
several parameters was evaluated and goodness-of-fit
RESULTS
plots were again used to assess the reliability of the
models and AIC in order to compare them. Patients
From the best model including IOV, a backward Concentration–time data were obtained from 45 patients
elimination procedure was then used to test whether who received indinavir. Among these patients, 14 also
all covariates selected from V1 concentrations should received a booster dose of ritonavir (100 mg b.i.d.).
remain in the final model. The likelihood ratio test (LRT) Indinavir dosage regimens were as follows: 600 mg t.i.d.
was used to compare the corresponding nested models. (one patient), 800 mg t.i.d. (27 patients), 1000 mg t.i.d.
More precisely, if the elimination of a covariate signifi- (two patients) and 1200 mg b.i.d. (one patient) in
cantly (P < 0.05) decreased the log-likelihood, that patients receiving indinavir alone and 400 mg b.i.d.
covariate was kept in the model. (five patients), 600 mg b.i.d. (one patient) and 800 mg
b.i.d. (eight patients) in patients receiving also ritonavir.
Model evaluation Table I describes the main characteristics of these
Simulations of indinavir steady-state concentrations patients. The antiretroviral drugs associated with indi-
profile were performed and compared with the observed navir were lamivudine (3TC, n ¼ 36), stavudine (D4T,
ones to evaluate the predictive performance of the n ¼ 22), zidovudine (AZT, n ¼ 13), didanosine (DDI,
models. We simulated 1000 patients with 800 mg t.i.d. n ¼ 9), nevirapine (NVP, n ¼ 5), abacavir (ABC, n ¼ 3),
without ritonavir and 1000 patients with 800 mg b.i.d. and efavirenz (EFV, n ¼ 2).
of indinavir and 100 mg b.i.d. of ritonavir. The simula- Among the 45 patients included in the trial who
tions were done using the final model of indinavir. We received indinavir, three patients experienced virological
used, in the simulations the total variability (inter plus failure during the trial and all received indinavir alone.
intra) of the random effects and the residual error One patient experienced nephrolithiasis (at V1 but not at
variability in order to mimic the observations. V2) which led to treatment discontinuation after V1.
We kept in each group of simulation the same Liver function remained unchanged during the trial. No
distribution of the significant covariates as in the study clinically relevant grade III or IV laboratory abnormal-
population. ities were observed.
The 10th and 90th predicted percentiles for the 1000
simulations in each group were compared with the Basic model
observed concentrations at V1 and V2 for the patients Figure 1 displays indinavir plasma concentrations at V1
receiving the corresponding dosage. It should be noted and V2 vs. time for patients with or without ritonavir.
that in order to evaluate the non-parametric 10th and
90th percentiles, a rather large sample of simulated
patients is needed. Table I Patient characteristics at baseline.

Patients (n ¼ 45) Median (range)


Simulation of trough concentrations
with the final model Age (years) 40.3 (21–65)
From the final population model of indinavir, we Weight (kg) 72 (41–110)
simulated the range of individual steady-state trough Sex (male/female) 35/10
Time since first ARV treatment (years) 4.9 (0.9–10.7)
concentrations for the two usual dosage regimen:
CD4 cell count (/mm3) 599.6 (150–1425)
800 mg t.i.d. for indinavir alone and 800 mg b.i.d. for
Adherence (%) 100 (44–100)
indinavir with 100 mg b.i.d. of ritonavir. We simulated

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


Population pharmacokinetics of indinavir 377

(a) 18 ritonavir. From both the Akaike criterion and the LRT,
the best model was the model with a ritonavir effect
Indinavir concentration (mg/L)

16

14 on Ka and on Cl/F. With that model (AIC ¼ 800.8),


12
the variability was low and poorly estimated for gKa. A
10

8
model with random effect only on Cl/F and V/F was
6 then tested (AIC ¼ 800.5) and kept as the basic
4
model.
2

0
The equations of this model are:
0 2 4 6 8 10 12 14 16
Time (h) Ka;i ¼ ðKa  ð1  ritonaviri Þ þ Ka;rito  ritonaviri Þ
(b) 18
Indinavir concentration (mg/L)

16
Cl=Fi ¼ ðCl=F  ð1  ritonaviri Þ þ Cl=Frito  ritonaviri Þ
14

12  expðgCl;i Þ
10

8 V=Fi ¼ V=F  expðgV;i Þ


6

4
where ritonaviri ¼ 0 if patient took indinavir alone and
2

0
ritonaviri ¼ 1 if it was coadministrated.
0 2 4 6 8 10 12 14 16 The population estimates for Ka and Cl/F were
Time (h)
0.43/h and 34.22 L/h, respectively, for patients who
Figure 1 Observed plasma indinavir concentration vs. time for received indinavir without ritonavir, 0.26/h and
patients without ritonavir (a) and with ritonavir (b) at the two visits 28.88 L/h for patients with ritonavir. The estimated
V1 and V2. volume of distribution was 21.45 L. Large inter-indi-
vidual variabilities were observed for Cl/F, 47.5%, and
It shows that the decrease of concentrations was for V/F, 134% and r in the residual error model was
slower when ritonavir was co-administered with indina- estimated to be 55%. The goodness-of-fit plots of that
vir and that indinavir Cmax was obtained later. That is model were satisfactory.
why at V2 the first sample was collected at 1 h for The standard error of estimation expressed as
indinavir alone and at 3 h for indinavir with ritonavir coefficient of variations (CV) were lower than 25%
(except for three patients where it was collected at 1 h). for all population parameters. The model with a
Among the 305 samples, two indinavir plasma concen- correlation (estimated to be 0.22) between Cl/F and
trations in two different patients, were below the limit of V/F had the same likelihood than the model without
quantification at 10 and 10.6 h and were fixed to the correlation but a higher AIC. This correlation was
LOQ/2 of the assay of the corresponding center, i.e. therefore not kept in the model and the random effects
0.025 mg/L in both cases. were assumed to be independent. The inter-individual
First, the combined error model with random effects variability of Cl/F and V/F was estimated globally from
on Ka, Cl/F and V/F was used. The initial value for a2 all patients with and without ritonavir. When we
was the average (LOQ/2)2 ¼ 0.0004 (mg/L)2 and the considered different variances for inter-individual vari-
model converged to a2 ¼ 0.029. This model (AIC ¼ ability in patients with and without ritonavir, that
867.8) was better than a model with only a proportional model had a higher AIC than the model with only 1
error model (AIC ¼ 876.6). Because of the high gain in variance.
the AIC when a combined error variance was used on So the basic model for indinavir concentrations was
the data at V1, and because the subsequent models with described by a one-compartment model with first-order
combined error variance did not converge during the absorption and first-order elimination with random
tests of covariates and of the inter-occasion variability, effects on Cl/F and V/F and with a different Ka and
this value of a2, 0.029, was fixed and kept in the Cl/F in patients who took ritonavir.
following analyses.
Significant effects of ritonavir on empirical Bayes Covariates model building
estimates of Ka (P < 0.001) and Cl/F (P < 0.003) were From that basic model, we first tested the effects of the
found. We then implemented and tested in the covariates on the individual estimates of the random
population model the effect of co-administration of effects. Significant effects of AZT, D4T, and gender on gCl

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


378 K. Brendel et al.

(P ¼ 0.011, 0.011 and 0.049, respectively) and of AZT estimated to be 19.53 L and increased by 1.60 with
on gV (P ¼ 0.038) were found. AZT. The inter-individual variability was 39% for
The model with the lowest AIC (732.1) had AZT, D4T clearance and 113% for volume of distribution. They
and gender effects on Cl/F and AZT effect on V/F. The were slightly decreased from the basic model by the
equations are: incorporation of the covariates. Residual variability
was 47%.
Ka;i ¼ ðKa  ð1  ritonaviri Þ þ Ka;rito  ritonaviri Þ
Inter-occasion variability
Cl=Fi ¼ ðCl=F  ð1  ritonaviri Þ þ Cl=Frito  ritonaviri Þ
We then built the model of inter-occasion variability
 ðbAZT  bD4T  bmale Þ  expðgCl;i Þ based on the model at V1 with covariates and with the
data of the second visit V2. The best model had an IOV on
V=Fi ¼ ðV=F  aAZT Þ  expðgV;i Þ:
both Cl/F and V/F (AIC ¼ 1035.6). Performing back-
When a patient receives zidovudine (AZTi ¼ 1), his ward elimination of the covariates from that model, the
clearance is multiplied by a factor bAZT and his volume best AIC was obtained when the effects of only AZT
by a factor aAZT; when a patient receives stavudine and gender on Cl/F were kept. Elimination of one of
(D4Ti ¼ 1), his clearance is multiplied by a factor bD4T; these covariates decreased significantly the likelihood
when a patient is a male (genderi ¼ 1), his clearance is (P < 0.001 for gender, P ¼ 0.01 for AZT).
multiplied by a factor bmale. The equations of that final model are:
The population parameters of this model and their
Ka;i ¼ ðKa  ð1  ritonaviri Þ þ Ka;rito  ritonaviri Þ
standard error are given in the first column of Table II.
Absorption rate constant was estimated to be 0.27/h
Cl=Fi ¼ ðCl=F  ð1  ritonaviri Þ þ Cl=Frito  ritonaviri Þ
for patients who received ritonavir and 0.49/h for
patients without ritonavir. Oral clearance was 23.43  ðbAZT  bmale Þ  expðg0Cl;i þ IOV  g1Cl;i Þ
and 30.33 L/h in women with and without ritonavir,
respectively. It increased by 1.47-fold in men, V=Fi ¼ V=F  expðg0V;i þ IOV  g1V;i Þ
decreased by 1.22-fold in patients who received AZT
where IOV ¼ 0 for data at V1 and IOV ¼ 1 for data at
and increased by 0.68-fold in patients who received
V 2.
D4T. Oral volume of distribution without AZT was
Table II shows the parameters estimated for this final
model with IOV. The same estimations were approxi-
Table II Population pharmacokinetic parameters of indinavir mately observed for the three fixed effects. Using equa-
(estimate and CV of estimation) for the intermediate model with tions (1) and (2) in the appendix, the inter-individual
data at V1 only and for the final model with data at V1 and V2. and inter-occasion variability were derived from the
V1 only V1 and V2
estimated variances of g0i and g1i provided by WinNon-
Mix. They were 42 and 58% for Cl/F, 110 and 205% for
Parameters Estimate CV (%) Estimate CV (%) V/F. The total variability was 72% for Cl/F and 232% for
Ka (/h) 0.49 6 0.43 5 V/F. r in the error model was estimated to be 47%. We
Ka,rito (/h) 0.27 5 0.25 9 tried to perform the estimation using the FOCE method of
Cl/F (L/h) 30.33 15 33.41 15 WinNonMix for this final model but the run did not
Cl/Frito (L/h) 23.43 17 19.35 18 converge.
V/F (L) 19.53 18 24.21 15
The goodness-of-fit plots of this final model with IOV
bmale 1.47 11 1.45 15
bD4T 0.68 14 – –
and covariates was satisfactory. The evaluation proce-
bAZT 1.22 12 1.18 11 dure was applied to the 27 patients receiving 800 mg
aAZT 1.60 20 – – t.i.d. alone and the eight patients receiving 800 mg b.i.d.
x0Cl (%) 39 30a 36 31a with ritonavir. The simulated 10th, 90th percentiles
x1Cl (%) – – 82 53a and the median are displayed in Figure 2, together with
a
x0V (%) 113 2 110 34a all the observed concentrations of these patients. The
x1V (%) – – 285 72a
evaluation results of this indinavir population model
r (%) 47 22a 47 24a
were very satisfactory both when indinavir is adminis-
a
CV for x2V , x2Cl and r2. trated alone or with ritonavir.

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


Population pharmacokinetics of indinavir 379

(a) (a)
18 3
16
2.5
14

Trough concentration (mg/L)


12
2
10

8 1.5
6
1
4

2
0.5
0
0 2 4 6 8 10 12 14 16
0
Time (h)
Women Women Men Men
(b)
18 without AZT with AZT without AZT with AZT
16
(b) 3
14

12
2.5

Trough concentration (mg/L)


10

8
2
6

4 1.5
2

0
1
0 2 4 6 8 10 12 14 16
Time (h) 0.5

Figure 2 Indinavir model evaluation: comparison between the 0


10th, 90th (dashed lines) and 50th (continued line) predicted Women Women Men Men
percentiles for the 1000 simulations and the observed concentra- without AZT with AZT without AZT with AZT

tions at V1 and V2 for the patients receiving the corresponding


Figure 3 Box plots of trough indinavir concentrations simulated
dosage in each group for patients with 800 mg t.i.d. of indinavir
from the population model for 800 mg of indinavir t.i.d. without
alone (a) and for patient with 800 mg b.i.d. of indinavir associated
ritonavir (a) and for 800 mg b.i.d. of indinavir with 100 mg of
with a booster dose of ritonavir (b).
ritonavir (b). The influence of gender and of the co-administration
of zidovudine (AZT) which were the two significant covariates in
Simulation of indinavir trough concentrations the population analysis is illustrated. The boxes represent 25th to
Figure 3 displays the steady-state trough concentrations, 75th percentiles, with the 50th percentile shown within the boxes;
simulated with this final model and the total variability the 10th and 90th percentiles are shown as capped bars.
for gCl and gV, in the four groups of patients as defined
by the covariates included in the final model: male or therefore an effect with a rather small clinical rele-
female with or without AZT. vance. With respect to gender effect, median trough
The simulations were done with the two most usual concentrations increased by 1.47-fold in women com-
indinavir dosages: 800 mg t.i.d. alone or 800 mg b.i.d. pared with men without AZT and by 1.83-fold with
with 100 mg b.i.d. of ritonavir. The trough concentra- AZT. Gender effect on trough concentrations had
tions were the highest for women who did not take AZT therefore a clear clinical relevance. For instance,
and the lowest for men who took AZT, both with and median trough concentrations (25th and 75th percen-
without ritonavir. When the dosage regimen of indinavir tiles) was 1.22 mg/L (0.81, 1.75) in women receiving
is 800 mg b.i.d. with ritonavir, trough concentrations 800 mg indinavir plus 100 mg ritonavir with AZT,
were higher than for 800 mg t.i.d. of indinavir alone. which is very high.
However, in both cases the variability was very large.
Median trough concentrations obtained with 800 mg
DISCUSSION
of indinavir with 100 mg ritonavir b.i.d., showed
an increase with AZT compared to without AZT by The pharmacokinetics of indinavir was described by a
1.05-fold in women and by 1.36-fold in men. AZT had one-compartment model with first-order absorption and

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


380 K. Brendel et al.

first-order elimination with random effects on Cl/F and frequent regimens in this trial. In these simulations, the
on V/F but not on Ka when the effect of ritonavir was impact of gender appeared especially important when
incorporated. Although the mechanism of interaction patients also took zidovudine. However, it should be
between ritonavir and indinavir is well known [28–30], noticed that in this trial only three women received
the effect of ritonavir was tested for the first time in a zidovudine and this result should be further confirmed.
pharmacokinetic model of indinavir. We found that The high trough levels predicted with 800 mg b.i.d. and
when patients receive ritonavir, they have an absorption ritonavir might explain the high level of nephrolithiasis
rate constant lower than patients without ritonavir found in the BEST study [15]. This result is important
(0.25 and 0.43/h, respectively). With respect to elimin- and suggests to decrease indinavir initial dosage in
ation, the oral clearance of indinavir was found to women compared with men.
decrease when patients take ritonavir; this was expected With respect to the effect of co-administration of
as ritonavir is a CYP3A4 inhibitor. The absence of zidovudine, Cl/F was estimated to increase by 1.18-fold
significant effect on V/F found here suggests that there is in patients who received AZT. The P-value was signifi-
no effect of ritonavir on the bioavailability of the drug cant, but the clinical relevance of this effect was low in
which is also supported by the absence of correlation the simulations of trough concentrations. A pharmaco-
found between Cl/F and V/F. kinetic interaction between indinavir and AZT has been
In this study, a gender effect was found on the oral reported in the US package insert of indinavir because
clearance of indinavir which was estimated to increase in a study, after 1 week of co-administration of AZT,
by 1.45-fold in men. To further study whether this indinavir clearance was found to decrease. In this paper,
gender difference was due to a weight effect or was a real we found an opposite interaction with a slight increase of
gender difference in metabolism, we also tested the indinavir clearance.
weight effect on the individual indinavir clearances. We The individual pharmacokinetic parameters of a lot of
studied both weight as a continuous covariate or as a drugs can change between two periods of time [33–35].
categorical covariate with a cut off of 67.5 kg (the mean Some of these variations are due to the fact that
between median weight in men, 75 kg, and in women, pharmacokinetic parameters are linked with covariates,
60 kg). By using a Mann–Whitney non-parametric test which can vary physiologically with time, as serum
or a correlation test, no significant weight effect was creatinine. However, the biggest variability is not fore-
found on Cl/F. This strongly suggests that the gender seeable and might be due to changes in bioavailability.
effect is more the result of the difference of metabolism Modeling the inter-occasion variability (also called intra-
between men and women than a weight effect. Few individual variability) is very important in population
other studies also reported similar results. Csaika et al. pharmacokinetics [36], and this is why patients were
[31], in a study with 239 HIV-infected patients, found sampled at two occasions (V1 and V2) in the COPHAR
that indinavir clearance increased by 1.64-fold in men 1-ANRS 102 trial. A population model including inter-
very close to the increase of 1.45 found here. In a recent occasion variability has never been implemented in the
in vivo and in vitro animal study [32], indinavir software WinNonMix before this study, and we had to
clearance was found to increase by 2.17-fold in male develop a method to do it. The quality control procedure
rats. Consistent with the in vivo observations, hepatic implemented before the trial was very important to
microsomes from male rats had a substantially higher minimize the inter-laboratory variability (below 20%).
metabolizing activity toward indinavir than that from We did not test the assay as a covariate in the model,
females. Cytochrome 3A isoforms may contribute to the because assays differed mostly in their LOQ and only two
observed gender-related differences in indinavir metabo- concentrations below the same LOQ (0.05 mg/L) were
lism in rat [32]. These results are in contradiction with found.
the US package insert, where AUC was found to be lower In this study, we found both large inter and intra-
in 10 HIV seropositive women who received 800 mg patient variabilities of indinavir pharmacokinetic param-
indinavir t.i.d. in addition to zidovudine and lamivudine eters. These variabilities estimated here confirm the need
compared with a pool of historical control data in men. of drug monitoring of indinavir in patients [37]. The
But we think that the results found here and by Csajka potential benefit of therapeutic drug monitoring (TDM)
et al. [31] in a prospective study are more reliable. The of PIs has been shown by the ATHENA study based
clinical relevance of this gender effect was illustrated in on concentration ratio [38]. This ratio is obtained by
the simulation of the trough levels for the two most dividing the observed concentration by the correspond-

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


Population pharmacokinetics of indinavir 381

ing time-adjusted value of a reference pharmacokinetic (Kremlin Bicêtre), Dr C. Solas (Marseille), Dr D. Hillaire
curve. More precisely, this reference curve is based on (Montpellier), Dr E. Dailly (Nantes), Pr B. Diquet, Dr G.
the fit of a pharmacokinetic model to the median values Peytavin, Dr J.M. Poirier, Dr E. Rey, Dr H. Sauvageon
of concentrations grouped by 30-min time intervals. In (Paris), Dr J.C. Alvarez (Versailles). We also thank the
the ATHENA study, after 1 year of follow up of 55 HIV- patients for their participation.
infected patients, significantly fewer patients in the TDM
group had discontinued indinavir than in the control
REFERENCES
group (25% vs. 48%) because of virological failure or
indinavir toxicity. Also the TDM group showed a 1 Palella F.J., Delaney K.M., Moorman A.C. et al. Declining
significantly higher proportion of patients (78% vs. morbidity and mortality among patients with advanced human
immunodeficiency virus infection. N. Engl. J. Med. (1998) 338
55%) with a viral load below 500 copies/mL after
853–860.
12 months of treatment. The present prospective trial,
2 Jordan R., Gold L., Cummins C., Hyde C. Systematic review and
performed in patients with a sustained virological meta-analysis of evidence for increasing numbers of drugs in
response, could be used to define more realistic reference antiretroviral combination therapy. Br. Med. J. (2002) 324 1–10.
concentrations. The reference curve should be derived 3 Yeni P., Hammer S.M., Carpenter C.J. et al. Antiretroviral
from the mean population PK parameters using the treatment for adult HIV infection: updated recommendations of
model at steady state. For 800 mg of indinavir without the International AIDS Society-USA Panel. JAMA (2002) 288
ritonavir and for 800 mg b.i.d. of indinavir with ritona- 222–235.
4 Lichterfeld M., Nischalke H.D., Bergmann F. et al. Long-term
vir (100 mg b.i.d.), the most frequent regimen now-
efficacy and safety of ritonavir/indinavir at 400/400 mg twice
adays, the reference curve is similar to the median curve a day in combination with two nucleoside reverse transcriptase
displayed in Figure 2. Furthermore, the population inhibitors as first line antiretroviral therapy. HIV Med. (2002) 3
pharmacokinetic parameter estimates obtained here 37–43.
allow for the adaptative adjustment of indinavir regimen 5 Polis M.A., Sidorov I.A., Yoder C. et al. Correlation between
based on Bayesian therapeutic drug monitoring. reduction in plasma HIV-1 RNA concentration one week after
start of antiretroviral treatment and long-term efficacy. Lancet
(2001) 358 1760–1765.
ACKNOWLEDGEMENTS 6 Condra J.H., Petropoulos C.J., Ziermann R., Schleif W.A.,
Shivaprakash M., Emini E.A. Drug resistance and predicted
This study was financially supported by the Agence virologic responses to human immunodeficiency virus type 1
Nationale de la Recherche sur le SIDA (ANRS), Paris, protease inhibitor therapy. J. Infect. Dis. (2000) 182 758–765.
France. 7 Salzberger B., Rockstroh J., Wieland U. et al. Clinical efficacy of
We gratefully acknowledge all the members of the protease inhibitor based antiretroviral combination therapy – a
scientific committee of the Cophar 1-ANRS 102 trial: Dr V. prospective cohort study. Eur. J. Med. Res. (1999) 4 449–455.
8 Launay O., Gerard L., Morand-Joubert L. et al. Nevirapine or
Calvez (Hôpital Pitié-Salpétrière, Paris), Pr G. Chêne
lamivudine plus stavudine and indinavir: examples of 2-class
(INSERM U330, Bordeaux) Pr B. Diquet (Hôpital Pitié-
vs. 3-class regimen for the treatment of human immunodefi-
Salpétrière, Paris), Pr C. Katlama (Hôpital Pitié- ciency virus type I. Clin. Infect. Dis. (2002) 35 1096–1105.
Salpétrière, Paris), Pr C. Leport (Hôpital Bichat, Paris), 9 Duval X., Peytavin G., Albert I. et al. Determination of indinavir
Dr A. Metro (ANRS, Paris), Dr G. Peytavin (Hôpital Bichat, and nelfinavir trough plasma concentration efficacy thresholds
Paris), Pr F. Raffi (Hôpital Hôtel Dieu, Nantes), Dr A. Roux according to virological response in HIV-infected patients.
(Hôpital Ambroise Paré, Boulogne), and Pr D. Salmon- HIV Med. (2004) 5 307–313.
Ceron (Hôpital Cochin, Paris). We would like to thank the 10 Dieleman J.P., Gyssens I.C., Van der Ende M.E., De Marie S.,
Burger D.M. Urological complaints in relation to indinavir
investigators in the clinical centres for including the
plasma concentrations in HIV-infected patients participating in
patients at Pr J.M. Estavoyer, Pr R. Laurent (Besançon), Pr the ADAM study. AIDS (2000) 14 59–98.
X. Bazin (Caen), Pr C. Perrone (Garches), Pr J.F. Delfraissy 11 Reijers M.H., Weigel H.M., Hart A.A. et al. Toxicity and drug
(Kremlin Bicêtre), Pr X. Gallais, Pr J.A. Gastaut (Marseille), exposure in a quadruple drug regimen in HIV-1 infected
Pr J. Reynes (Montpellier), Pr F. Raffi (Nantes), Pr E. patients participating in the ADAM study. AIDS (2000) 14
Bouvet, Pr F. Bricaire, Pr S. Herson, Pr W. Rozenbaum, Pr 59–68.
D. Séréni, Pr D. Sicard, Dr A. Simon, Pr J.L. Vildé (Paris), Pr 12 Von Moltke L.L., Greenblatt D.J., Grassi J.M., Granda B.W.,
Duan S.X., Fogelman S.M. Protease inhibitors as inhibitors of
X. Lang (Strasbourg) and the pharmacological laborator-
human cytochromes P450: high risk associated with ritonavir.
ies for performing drug assays at Dr B. Royer, Dr P. Muret
J. Clin. Pharmacol. (1998) 38 106–111.
(Besançon), Dr M. Tod (Bobigny), Dr A.M. Taburet

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


382 K. Brendel et al.

13 Hsu A., Granneman G.R., Cao G., Carothers L., Japour A., 27 Beal S.L. Ways to fit a PK model with some data below the
Shourbagy T. Pharmacokinetic interaction between ritonavir quantification limit. J. Pharmacokinet. Pharmacodyn. (2001)
and indinavir in healthy volunteers. Antimicrob. Agents 28 481–504.
Chemother. (1998) 42 2784–2791. 28 Lee C.G., Gottesman M.M., Cardarelli C.O. et al. HIV-1 protease
14 Saah A., Winchell G., Nessly M., Seniouk E., Rhodes R. inhibitors are substrates for the MDR1 multidrug transporter.
Pharmacokinetic profile and tolerability of indinavir–ritonavir Biochemistry (1998) 37 3594–3601.
combinations in healthy volunteers. Antimicrob. Agents 29 Washington C.B., Duran G.E., Man M.C., Sikic B.I., Blaschke T.F.
Chemoter. (2001) 45 2710–2721. Interaction of anti-HIV protease inhibitor with the multidrug
15 Arnaiz J.A., Mallolas J., Podzamczer D. et al. Continued transporter P-glycoprotein (P-gp) in human cultured cells. J. Acq.
indinavir vs. switching to indinavir/ritonavir in HIV-infected Immun. Def. Synd. Hum. Retrovirol. (1998) 19 203–209.
patients with suppressed viral load. AIDS (2001) 17 831– 30 Raffi F. Resistance to protease inhibitors: the role of the MDR
840. gene. Presse Med. (2000) 29 2036–2037.
16 Zhou X.J., Havlir D.V., Richman D.D. et al. Plasma population 31 Csaika C., Marzolini C., Fattinger K. et al. Population pharma-
pharmacokinetics and penetration into cerebrospinal fluid of cokinetics and effects of indinavir in HIV infected patients. Clin.
indinavir in combination with zidovudine and lamivudine in Pharmacol. Ther. (2003) 73 69.
HIV-1-infected patients. AIDS (2000) 14 2869–2876. 32 Lin J.H., Chiba M., Chen I.W., Nishime J.A., Vastag K.J. Sex
17 Letendre S.L., Capparelli E.V., Ellis R.J., McCutchan J.A. dependant pharmacokinetics of indinavir: in vivo and in vitro
Indinavir population pharmacokinetics in plasma and cere- evidence. Drug Metab. Dispos. (1996) 24 1298–1306.
brospinal fluid. Antimicrob. Agents Chemother. (2000) 44 33 De Maat M.M., Huitema A.D., Mulder J.W., Meenhorst P.L.,
2173–2175. Van Gorp E.C., Beijnen J.H. Population pharmacokinetics of
18 Martin C., Sonnerborg A., Olof Svensson J., Stahle L. Indinavir- nevirapine in an unselected cohort of HIV-1-infected individ-
based treatment of HIV-1 infected patients: efficacy in the uals. Br. J. Clin. Pharmacol. (2002) 54 378–385.
central nervous system. AIDS (1999) 13 1227–1232. 34 Huitema A.D., Mathot R.A., Tibben M.M., Schellens J.H.,
19 Pfister M., Labbé L., Hammer S.M. et al. Population pharma- Rodenhuis S., Beijnen J.H. Population pharmacokinetics of
cokinetics and pharmacodynamics of efavirenz, nelfinavir, thioTEPA and its active metabolite TEPA in patients undergoing
and indinavir: Adult AIDS Clinical Trial Group Study 398. high-dose chemotherapy. Br. J. Clin. Pharmacol. (2001) 51
Antimicrob. Agents Chemoter. (2003) 47 130–137. 61–70.
20 Carrieri P., Cailleton V., Le Moing V. et al. The dynamic of 35 Frame B., Koup J., Miller R., Lalonde R. Population pharma-
adherence to highly active antiretroviral therapy: results from cokinetics of clinafloxacin in healthy volunteers and patients
the French National APROCO Cohort. J. Acq. Immun. Def. with infections: experience with heterogeneous pharmaco-
Synd. (2001) 28 232–239. kinetic data. Clin. Pharmacokinet. (2001) 40 307–315.
21 Aymard G., Legrand M., Trichereau N., Diquet B. Determin- 36 Karlsson M.O., Sheiner L.B. The importance of modeling
ation of twelve antiretroviral agents in human plasma sample interoccasion variability in population pharmacokinetic
using reverse-phase high-performance liquid chromatography. analyses. J. Pharmacokinet. Biopharm. (1993) 21 735–750.
J. Chromatogr. B (2000) 744 227–240. 37 Back D., Gatti G., Fletcher C. et al. Therapeutic drug monitoring
22 Proust V., Toth K., Hulin A., Taburet A.M., Gimenez F., Singlas in HIV infection: current status and future direction. AIDS
E. Simultaneous high-performance liquid chromatographic (2002) 16 S5–S37.
determination of the antiretroviral agents amprenavir, nelfin- 38 Burger D., Hugen P., Reiss P. et al. Therapeutic drug monit-
avir, ritonavir, saquinavir, delavirdine and efavirenz in human oring of nelfinavir and indinavir in treatment-naı̈ve HIV-1-
plasma. J. Chromatogr. B. (2000) 742 453–458. infected individuals. AIDS (2003) 17 1157–1165.
23 Dailly E., Thomas L., Kergueris M.F., Jolliet P., Bourin M. High-
performance liquid chromatographic assay to determine the
plasma levels of HIV-protease inhibitors (amprenavir, nelfina- APPENDIX
vir, ritonavir, saquinavir) and the non-nucleoside reverse
transcriptase inhibitor (nevirapine) after liquid. J. Chromatogr. The following model was implemented for incorporation
B. (2001) 758 129–35. of IOV in WinNonMix, using IOV as a covariate:
24 Poirier J.M., Robidou P., Jaillon P. Determination of indinavir in
plasma by solid-phase extraction and column liquid chromato- bi ¼ g0i þ IOV  g1i
graphy. Ther. Drug Monit. (1999) 21 404–410.
25 Bouley M., Briere C., Padoin C., Petitjean O., Tod M. Sensitive where IOV ¼ 0 for occasion 1 (V1) and IOV ¼ 1 for
and rapid method for the simultaneous quantification of the occasion 2 (V2).
HIV-protease inhibitors indinavir, nelfinavir, ritonavir and Therefore,
saquinavir in human plasma by reversed-phase liquid chro-
matography. Ther. Drug Monit. (2001) 23 56–60. g0i ¼ gi þ j1i
26 The United States Pharmacopoeia XXII: validation of compen-
dial methods. USP Convention Inc., Rockville, 1990, pp. 1710. g0i þ g1i ¼ gi þ j2i

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383


Population pharmacokinetics of indinavir 383

and g0i and g1i are correlated. We estimated Var(g0i ), To derive the variance of inter-individual variability
Var(g1i ) and the covariance between g0i and g1i with we used:
WinNonMix.
Then to derive the variance of inter-occasion variab- 2g0i þ g1i ¼ 2gi þ j1i þ j2i
ility from these values, we used: so that
g1i ¼ j2i  j1i 4Varðg0i Þ þ Varðg1i Þ þ 4Covðg0i ; g1i Þ
so that ¼ 4Varðgi Þ þ Varðg1i Þ:

Varðg1i Þ ¼ 2  Varðjki Þ: The inter-individual variability is therefore:

The inter-occasion variability is therefore: Varðgi Þ ¼ Varðg0i Þ þ Covðg0i ; g1i Þ: ð2Þ

Varðjki Þ ¼ 1=2 Varðg1i Þ: ð1Þ Total variance was estimated by Var(gi) + Var(jki ).

Ó 2005 Blackwell Publishing Fundamental & Clinical Pharmacology 19 (2005) 373–383

You might also like