Clinical Development of An Everolimus Pediatric Formulation

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

The Journal of Clinical

Pharmacology
http://www.jclinpharm.org

Clinical Development of an Everolimus Pediatric Formulation: Relative Bioavailability, Food Effect, and
Steady-State Pharmacokinetics
John M. Kovarik, Adele Noe, Stephane Berthier, Louis McMahon, Wayne K. Langholff, Alan S. Marion, Peter Friedrich
Hoyer, Robert Ettenger and Christiane Rordorf
J. Clin. Pharmacol. 2003; 43; 141
DOI: 10.1177/0091270002239822

The online version of this article can be found at:


http://www.jclinpharm.org/cgi/content/abstract/43/2/141

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 21, 2009


KOVARIK ET AL OF EVEROLIMUS PEDIATRIC FORMULATION
ARTICLE
DEVELOPMENT
PEDIATRICS

Clinical Development of an Everolimus Pediatric


Formulation: Relative Bioavailability, Food
Effect, and Steady-State Pharmacokinetics
John M. Kovarik, PhD, FCP, Adele Noe, PhD, Stephane Berthier, PharmD,
Louis McMahon, PhD, Wayne K. Langholff, PhD, Alan S. Marion, MD, PhD,
Peter Friedrich Hoyer, MD, Robert Ettenger, MD, and Christiane Rordorf, MD

The immunosuppressant everolimus used in organ trans- from the dispersible tablet was 10% lower relative to the con-
plantation is formulated as a conventional tablet for adults ventional tablet, with a ratio (90% confidence interval) of
and a dispersible tablet that can be administered in water for 0.90 (0.76-1.07). After a high-fat meal, tmax was delayed by a
pediatric use. As part of the pediatric clinical development median 2.5 hours, and Cmax was reduced by 50%. Overall ab-
program, the relative bioavailability and food effect for the sorption, however, was not affected by food inasmuch as the
dispersible tablet were evaluated in healthy adult subjects as fed/fasting AUC ratio was 0.99 (0.83-1.17). In pediatric pa-
a prelude to characterizing the steady-state pharmaco- tients, steady state was reached between days 3 and 5. The
kinetics in pediatric kidney allograft recipients. In a random- corresponding steady-state parameters were as follows: Cmin,
ized, open-label, three-way crossover study, 24 healthy adults 4.4 ± 1.7 ng/ml; Cmax, 13.6 ± 4.2 ng/ml; and AUC, 87 ± 27
received single 1.5-mg oral doses of everolimus as (1) six 0.25- ng•h/ml. Steady-state concentration-time profiles in pediat-
mg dispersible tablets in water, (2) two 0.75-mg conventional ric transplant patients receiving the dispersible tablet were
tablets, and (3) six 0.25-mg dispersible tablets in water after a comparable to those of adult patients receiving the conven-
high-fat breakfast. Cmax and AUC were evaluated by standard tional tablet when both were dosed to yield similar trough
bioequivalence testing to determine relative bioavailability concentrations. If a pediatric patient is converted from the
and to quantify the effect of food. In a multicenter open-label everolimus dispersible tablet to the conventional tablet, this
efficacy/safety trial, pediatric renal allograft recipients re- should be based on a 1:1 milligram switch with subsequent
ceived 0.8 mg/m2 (maximum 1.5 mg) bid everolimus as therapeutic drug monitoring to further individualize the dose
dispersible tablets in water. Serial trough concentrations over as needed. The dispersible tablet formulation should be
the first week and a steady-state pharmacokinetic profile on taken consistently either with or without food to minimize
day 7 posttransplant were collected in 19 patients ranging fluctuations in exposure over time.
from ages 2 to 16 years old. The bioavailability of everolimus Journal of Clinical Pharmacology, 2003;43:141-147
©2003 the American College of Clinical Pharmacology

C hildren who receive an organ allograft face a life-


time on immunosuppressive therapy. Conse-
quently, this patient population is particularly likely to
benefit from an expansion of their therapeutic options.
Everolimus (Certican, Novartis Pharmaceuticals) is a
cell proliferation inhibitor in development for
immunosuppression after solid organ transplantation.
From Novartis Pharmaceuticals, Basel, Switzerland, and East Hanover, It is intended to be used in combination with
New Jersey (Dr. Kovarik, Dr. Noe, Dr. Berthier, Dr. McMahon, Dr. Langholff, cyclosporine and corticosteroids. In two international
Dr. Rordorf); MDS Pharma Services, Lincoln, Nebraska (Dr. Marion); De- trials in adult kidney transplantation, everolimus was
partment of Pediatric Nephrology, Universitätsklinik Essen, Essen, Ger- equivalent in efficacy to the comparator myco-
many (Dr. Hoyer); and Mattel Children’s Hospital at UCLA, Division of Pe- phenolate mofetil.1,2 In an international trial in adult
diatric Nephrology, Los Angeles (Dr. Ettenger). Submitted for publication
heart transplantation, everolimus was superior in effi-
September 29, 2002; revised version accepted November 19, 2002. Ad-
dress for reprints: John M. Kovarik, Novartis Pharma AG, Building WSJ cacy to the comparator azathioprine.3
27.4093, 4002 Basel, Switzerland. In parallel with the development of everolimus in
DOI: 10.1177/0091270002239822 adult organ transplantation, a pediatric development

J Clin Pharmacol 2003;43:141-147 141


KOVARIK ET AL

program was initiated in renal transplantation. The men and women. The mean age was 28 ± 8 years (range:
everolimus pediatric program was designed in accor- 19-42 years), and the mean weight was 75.8 ± 12.1 kg
dance with the guidelines set forth in the International (range: 53.1-97.6 kg). They received three 1.5-mg oral
Conference on Harmonization’s Guidance for Industry doses of everolimus in a randomized sequence as fol-
E11 on clinical investigations in pediatric populations4 lows: six 0.25-mg dispersible tablets in water under
and the Food and Drug Administration’s Draft Guid- fasting conditions (treatment A), two 0.75-mg conven-
ance for Industry on pediatric pharmacokinetic stud- tional tablets under fasting conditions (treatment B),
ies.5 The pediatric program components included (1) and six 0.25-mg dispersible tablets in water after a high-
developing a pediatric oral dosage form, (2) determin- fat breakfast (treatment C). In each of the three study pe-
ing the relative bioavailability of everolimus from the riods, subjects remained at the clinical center for 48
pediatric formulation compared with the conventional hours after each treatment and returned on days 4 and 5
tablet, (3) quantifying the influence of a high-fat meal for pharmacokinetic blood draws. Consecutive treat-
on absorption from the pediatric formulation to pro- ments were separated by a 14-day washout. An end-of-
vide guidance to prescribers and patients on adminis- study clinical evaluation was performed 7 days after re-
tration with respect to meals, (4) characterizing the ceiving the last dose.
single-dose pharmacokinetics of the formulation in The patient pharmacokinetic data were collected
maintenance pediatric renal transplant patients to de- as part of an international, 1-year, open-label safety/
rive a dose regimen for clinical use, and (5) performing efficacy trial in 19 pediatric de novo kidney allograft re-
a multicenter clinical trial in de novo pediatric renal cipients. The 9 boys and 10 girls were 9.9 ± 4.4 years of
transplant patients to document safety, efficacy, and age and categorized as 1 infant (1.9 years), 10 children
longitudinal steady-state pharmacokinetics. (3.4-10.3 years), and 8 adolescents (12.9-16.7 years).
For patients who cannot swallow tablets, such as They weighed 32.7 ± 19.7 kg (range: 10.7-76.8 kg) with
children, everolimus is formulated as a dispersible tab- body surface areas (BSAs) of 1.06 ± 0.43 m2 (range: 0.49-
let in dose strengths of 0.1 and 0.25 mg. These tablets 1.92 m2). Everolimus was begun on the day of trans-
are placed in a small amount of water in an oral syringe, plantation at 0.8 mg/m2 (maximum 1.5 mg) twice daily
on a spoon, or in a plastic cup; the tablets are allowed to in addition to cyclosporine microemulsion (Neoral,
disperse, and the contents are administered to the pa- Novartis) and corticosteroids, with the optional use of a
tient. Before proceeding to studies with this formula- CD25 monoclonal antibody. Cyclosporine was initi-
tion in pediatric transplant patients, a relative ated at 6 to 12 mg/kg/day in two divided doses and sub-
bioavailability link to the conventional adult tablet sequently individualized to yield morning predose
needed to be established to extrapolate safety and ex- trough levels of 200 to 350 ng/ml in month 1 and 100 to
posure data from adult studies with the conventional 300 ng/ml thereafter. Oral prednisone or equivalent
tablet and thereby design appropriate pediatric studies was dosed according to local practice through at least
with the dispersible tablet. Reported herein is a clinical month 6 posttransplant and a minimum dose of 0.1 mg/
pharmacology study in healthy adults addressing the kg/day.
relative bioavailability and food effect of the Both study protocols were reviewed by investiga-
dispersible tablet (the second and third components of tional review boards at the participating study centers.
the development program). Of the two clinical studies The healthy subject study was conducted at MDS
in pediatric transplant patients, the single-dose Pharma Services (Lincoln, NE). The patient study was
pharmacokinetic study (the fourth component) has conducted at 14 clinical centers in Belgium, Brazil,
been summarized elsewhere.6 Pertinent pharmaco- France, Germany, Spain, the United Kingdom, and the
kinetic aspects of the multicenter trial (the fifth compo- United States. The healthy subjects gave written con-
nent) are presented here to demonstrate some of the sent for participation in the relative bioavailability,
biopharmaceutical performance characteristics of the food effect study. The patients gave informed assent,
dispersible tablet in children compared with the con- and their parents or legal guardians gave written con-
ventional tablet in adults. sent for participation in the clinical trial.

MATERIALS AND METHODS Drug Administration and Assessments

Study Designs The healthy subjects reported to the study center the
evening before each treatment period and received a
The study in healthy subjects was a randomized, open- standard dinner. After a 10-hour overnight fast, they re-
label, three-way crossover trial enrolling 27 healthy ceived the scheduled treatment. For treatment A, the

142 • J Clin Pharmacol 2003;43:141-147


DEVELOPMENT OF EVEROLIMUS PEDIATRIC FORMULATION

everolimus dispersible tablets (Certican, Novartis) containing collection tubes before the morning
were placed in a 20-ml oral syringe, and 15 ml water everolimus dose on days 3, 5, 6, and 7. Additional
was added. The tablets were dispersed by gentle shak- blood samples were obtained on day 7 at 1, 2, 5, and 8
ing of the syringe for about 90 seconds, and the con- hours postdose for a steady-state profile. Blood sam-
tents were administered directly into the subject’s ples were transferred to polypropylene tubes and
mouth. The residue at the syringe tip was retracted into frozen at –20°C.
the syringe with an additional 5 ml of water and the
contents administered orally. Thereafter, the subject Bioanalytical Methods
drank 200 ml water. For treatment B, the everolimus
conventional tablets (Certican, Novartis) were swal- Everolimus concentrations were determined in blood
lowed with 200 ml water. For treatment C, subjects re- by a validated liquid chromatography method. Briefly,
ceived a standardized, high-fat breakfast with 250 car- after addition of ammonium hydroxide, samples were
bohydrate calories, 600 fat calories, and 150 protein extracted with tert-butylmethylether. The organic ex-
calories. The everolimus dispersible tablets were pre- tract was evaporated to dryness and reconstituted in
pared as described for treatment A and administered the chromatographic mobile phase. Samples were ana-
within 5 minutes after completing the breakfast. lyzed using liquid chromatography with atmospheric
Scheduled, standardized meals were served beginning pressure chemical ionization and mass spectrometry.
4 hours postdose in all three treatment arms. Assay performance was assessed on the basis of cali-
To characterize the single-dose pharmacokinetics of bration concentrations ranging from 0.26 to 51.4 ng/ml
everolimus, 2-ml venous blood samples were collected and quality control concentrations ranging from 0.29 to
into EDTA-coated collection tubes predose and 0.5, 1, 38.7 ng/ml determined in duplicate with patient sam-
1.5, 2, 2.5, 3, 4, 6, 8, 12, 24, 36, 48, 60, 72, and 96 hours ples. The lower limit of quantification was 0.26 ng/ml.
postdose. Samples were transferred to polypropylene
tubes and frozen at –20°C. Safety assessments included Pharmacokinetic and Statistical Evaluation
physical examinations, electrocardiograms, vital signs
measurements (blood pressure, pulse rate, tempera- Conventional noncompartmental pharmacokinetic pa-
ture), laboratory evaluations (biochemistry, hematol- rameters were derived for single-dose everolimus, in-
ogy, urinalysis), and adverse event monitoring at base- cluding the peak blood concentration (Cmax), time to
line (the day before each treatment period), at multiple reach the peak (tmax), area under the concentration-time
times during the treatments, and at study completion. curve extrapolated to infinity (AUC), and half-life (t1/2)
The assessments during treatment consisted of routine by unweighted log-linear regression on the terminal
laboratory parameters predose and at 24 and 96 hours portion of the profile. For the multiple-dose profiles in
postdose and vital signs predose and at 2, 24, 48, 72, patients, parameters included the predose concentra-
and 96 hours postdose. Subjects were discharged from tion (Cminss), tmax, Cmaxss, AUCτss, the average concentra-
the study center after the 48-hour safety and tion over the dosing interval (Cavgss = AUCτss/12), the
pharmacokinetic blood draws. The study completion percent swing (Swing = [Cmaxss – Cminss]/Cminss), and the
evaluation was performed 7 days after the last peak-trough fluctuation (PTF = [Cmaxss – Cminss]/Cavgss).7,8
treatment. To calculate the AUC over the full 12-hour dosing inter-
In the pediatric clinical trial, everolimus dispersible val, the measured concentrations at 5 and 8 hours in the
tablets (0.1- and 0.25-mg strengths) were placed in a 10- terminal phase were extrapolated by log-linear regres-
ml oral syringe or spoon. The tablets were dispersed in sion to yield the estimated 12-hour concentration.
5 ml water over 90 seconds and administered to the The single-dose pharmacokinetic parameters were
child. For administration by syringe, the residue at the compared among treatments in an ANOVA with se-
syringe tip was retracted with an additional 5 ml water quence, subject within sequence, treatment, and pe-
and the contents administered. After administration by riod as sources of variation on log-transformed data.
either route, the child drank an additional 10 to 100 ml Relative bioavailability was quantified as the AUC ratio
water. The dose could also be delivered via nasogastric of the dispersible tablet/conventional tablet with its
tube in the early days posttransplant, prepared in a sy- 90% confidence interval (CI). The presence of a food ef-
ringe. Everolimus was given within 10 minutes after fect was based on the Cmax and AUC parameter ratios
the morning and evening cyclosporine dose, separated (fed/fasting) with 90% confidence intervals. If the ratio
by at least 1 hour before or after the nearest meal. Ve- and its confidence interval were within the bounds of
nous blood samples of 1.2 ml were obtained in EDTA- 0.80 to 1.25, lack of a food effect was concluded for a

PEDIATRICS 143
KOVARIK ET AL

Table I Relative Bioavailability and Food Effect 3.0

Dispersible Dispersible 2.5


Tablet Tablet Tablet

Parameter ratio
Parameter Fasting Fasting Fed 2.0

tmax (h) 1.0 (0.5-2.0) 1.0 (0.5-1.5) 3.0 (2.0-6.1) 1.5


Cmax (ng/ml) 12.3 ± 4.8 9.3 ± 3.3 4.6 ± 1.8
1.0
AUC (ng•h/ml) 99 ± 28 92 ± 37 94 ± 39
t1/2 (h) 41.0 ± 11.2 39.1 ± 11.7 37.7 ± 10.9 0.5
Values are mean ± standard deviation except for tmax, which is median
(range). tmax, time to reach peak exposure; Cmax, peak exposure; AUC, area 0.0
under the curve; t1/2, half-life. C max AUC

Figure 1. Individual Cmax and AUC ratios from the dispersible


9 ss tablet/conventional tablet (filled circles). Bars designate the geomet-
given parameter. In the patient study, serial C val- min ric mean, and dashed lines demarcate the equivalence boundaries of
ues were assessed by ANOVA, with patient and visit as 0.8 to 1.25.
sources of variation; lack of a visit effect was consid-
ered evidence for attainment of steady state. An un-
paired t-test was used to compare steady-state parame-
dian 1 hour postdose and was not different between
ters between pediatric and adult patients from the
formulations (p = 0.12). The Cmax ratio was 0.76 (90%
phase 3 trials in renal transplantation (data on file,
CI, 0.64-0.91), indicating that the peak concentration
Novartis). Data are presented as mean ± standard devia-
was 24% lower on average from the dispersible tablet
tion unless otherwise stated.
relative to the tablet. Interindividual variability in Cmax
was similar for both formulations: 39.0% for the tablet
RESULTS versus 35.5% for the dispersible tablet.
Inspection of Figure 1 indicates that the AUC from
Single-Dose Study: Subject Disposition the dispersible tablet for half of the subjects (n = 13) was
and Clinical Observations within ±20% of that from the tablet. Of the remaining
11 subjects, 7 had a lower and 4 had a higher AUC from
Of the 27 subjects enrolled, 24 completed the study. the dispersible tablet. The overall average bioavail-
One subject discontinued due to pharyngolaryngeal ability of everolimus from the dispersible tablet was
pain that the investigator did not consider treatment re- 0.90 (0.76-1.07) relative to the tablet. There was a trend
lated; 1 subject discontinued due to urticaria, which toward higher interindividual variability in AUC from
was suspected by the investigator as treatment related; the dispersible tablet, but this was not statistically sig-
and 1 subject withdrew consent to participate. Over the nificant and appeared to be influenced by a single out-
full course of the study, 17 subjects reported 65 adverse lier value: 28.3% for the tablet versus 40.2% for the
events, mostly of mild intensity. These consisted pri- dispersible tablet (p = 0.09). The half-life of everolimus
marily of headache, rhinorrhea and cold-related symp- from both formulations was similar.
toms, general body aches, and tiredness. There were no
clinically relevant changes in vital signs, electrocardio- Food Effect on the Dispersible Tablet
grams, or laboratory parameters over the course of the
study. Pharmacokinetic parameters after a fasting and fed ad-
ministration of the dispersible tablet are summarized
Relative Bioavailability from in Table I, and the mean concentration-time profiles are
the Dispersible Tablet compared in Figure 2. After a high-fat meal, tmax was de-
layed in all subjects; the median delay was 2.5 hours,
Pharmacokinetic parameters from the conventional with a range of 1.0 to 5.6 hours (p = 0.001). Cmax was no-
tablet and the dispersible tablet under fasting condi- tably decreased in the majority of subjects, on average,
tions are compared in Table I, and the corresponding by 50% in the presence of food; the Cmax ratio was 0.50
parameter ratios (dispersible tablet/tablet) are shown (0.42-0.59). Interindividual variability in Cmax was not
in Figure 1. The peak concentration occurred at a me- influenced by a high-fat meal: 35.5% for fasting admin-

144 • J Clin Pharmacol 2003;43:141-147


DEVELOPMENT OF EVEROLIMUS PEDIATRIC FORMULATION

Table II Steady-State Pharmacokinetics


10
10 in Renal Transplant Patients
8
8
Concentration (ng/ml)

6 Pediatric Patients Adult Patients


4 Parameter Dispersible Tablet Tablet p-Value
6
2
0
Number 19 116 —
4
0 2 4 6 Dose 0.8 mg/m2 bid 0.75 mg bid —
Cminss (ng/ml) 4.4 ± 1.7 4.6 ± 2.1 0.97
2
tmax (h) 1 (1-5) 2 (1-5) 0.93
Cmaxss (ng/ml) 13.6 ± 4.2 11.0 ± 4.5 0.02
0
AUCτss (ng•h/ml) 87 ± 27 76 ± 31 0.08
0 12 24 36 48 60 72
Cavgss (ng/ml) 7.2 ± 2.2 6.3 ± 2.6 0.08
Time (h)
Swing (%) 242 ± 159 158 ± 94 0.14
PTF (%) 132 ± 64 117 ± 49 0.44
Figure 2. Mean concentration-time profiles from the dispersible
tablet under fasting conditions (open squares) and after a high-fat Values are mean ± standard deviation except tmax, which is median (range).
Cmin, predose trough concentration; tmax, time to reach peak; Cmax, peak
meal (filled squares). Bars represent the 95% confidence interval. concentration; AUC, area under the concentration curve; PTF, peak-trough
The inset compares the initial portion of the profiles to 6 hours fluctuation.
postdose.

Everolimus was well tolerated by pediatric patients


istration versus 39.1% after a high-fat meal. There was over the year-long study. Full clinical results will be re-
no overall food effect on the extent of absorption, with ported elsewhere.
an AUC ratio of 0.99 (0.83-1.17). Nonetheless, inspec-
tion of the individual fed/fasting AUC ratios indicated Steady-State Pharmacokinetics
that occasionally a notable food effect occurred. Spe- in Pediatric Patients
cifically, 1 subject had a 145% increase and another
had an 88% decrease in AUC after a high-fat meal. Mean serial trough blood concentrations on day 3 aver-
Interindividual variability in AUC was not influenced aged 3.6 ± 1.7 ng/ml, which was 84% of the subsequent
by food: 40.2% for fasting administration versus 41.5% steady-state level. Steady state itself was reached be-
after a high-fat meal. Elimination half-lives were simi- tween the day 3 and 5 visits, with concentrations on
lar under both administration conditions. day 5 averaging 4.3 ± 1.8 ng/ml. These levels were
maintained at subsequent visits, with values of 4.2 ± 2.1
Dosing and Administration ng/ml on day 6 and 4.4 ± 1.7 ng/ml (predose) and 4.3 ±
in Pediatric Patients 2.4 ng/ml (12 h postdose) on day 7. Attainment of
steady state was supported by lack of a visit effect on
Everolimus doses averaged 0.9 ± 0.3 mg twice daily, Cminss from days 5 through 7 (p = 0.93).
with individual doses ranging from 0.4 to 1.5 mg twice Table II and Figure 3 compare the steady-state
daily. This complied with the protocol-specified dos- pharmacokinetics in pediatric patients receiving the
ing inasmuch as the average BSA-adjusted dose was dispersible tablet with those of adults receiving 0.75
0.81 ± 0.03 mg/m2 bid, ranging from 0.77 to 0.88 mg/m2 mg twice daily with the conventional tablet, which was
bid. The small deviations from 0.8 mg/m2 in individual a safe and effective regimen in the international pivotal
cases were due to rounding the calculated dose to the trials.1,2 At doses yielding similar trough concentra-
available tablet strengths. In the first few postsurgical tions, tmax and AUCτss were not different between the
days, 4 patients received the dose via nasogastric tube groups; however, Cmaxss was significantly higher in pe-
and all others orally. The preferred device for oral ad- diatric patients by an average 19%. The profile shapes,
ministration was the syringe used for 15 patients; only as indicated by percent swing and percent peak-trough
4 patients used a spoon. When the pharmacokinetic fluctuation, were comparable. When AUC was plotted
profile was obtained on day 7, a single patient was re- against age in the pediatric group, the slope of 2.3 ng•h/
ceiving everolimus via nasogastric tube. The pharma- ml per year was not different from zero (p = 0.11), indi-
cokinetic parameters of this 1-year-old girl (tmax, 1 h; cating that there was no age-related trends in exposure
Cmaxss, 15.2 ng/ml; AUCτss, 71 ng•h/ml) were similar to when pediatric patients were dosed on a BSA-adjusted
those from the rest of the population (see Table II). basis.

PEDIATRICS 145
KOVARIK ET AL

16 absorption of everolimus from the dispersible tablet, as


14
evidenced by a median 2.5-hour delay in tmax and an av-
erage 50% decrease in the peak concentration. None-
Concentration (ng/ml)

12
theless, the overall extent of absorption (AUC) was not
10
affected, on average. Similar results were found for the
8
conventional tablet in a previous development study in
6 which Cmax was reduced by 60% and AUC reduced by
4 16%.10 While the average food effect on AUC for both
2 formulations was generally modest, there were a few
0 subjects who displayed a notable food effect, with up to
0 2 4 6 8 10 12 twofold changes in AUC after a high-fat meal. Based on
Time (h) the individual responses to food, it would appear pru-
dent to administer the dispersible tablet to an individ-
Figure 3. Mean steady-state concentration-time profiles in pediat-
ric renal transplant patients receiving the dispersible tablet (filled
ual patient on a consistent basis either with or without
circles) and adult patients receiving the conventional tablet (open food to avoid unnecessary fluctuations in exposure
circles) at doses yielding similar trough blood concentrations. Bars over time. This conforms to the recommendation for
represent 95% confidence intervals. the conventional tablet.10
Pharmacokinetic evaluations in the context of the
international pediatric clinical trial demonstrated that
steady state was reached on or before day 5, as is also
DISCUSSION the case in adult patients11 and consistent with the fact
that pediatric and adult patients have similar elimina-
Everolimus is formulated as a conventional tablet for tion half-lives.6 The steady-state concentration-time
use in the general organ transplant population. For pa- profiles indicated that exposure, which is safe and ef-
tients unable to swallow tablets—such as children, the fective in adults receiving the conventional tablet,1,2,12
elderly, or adults in the immediate postoperative pe- can be achieved in pediatric patients with the
riod—a dispersible tablet was developed. This tablet dispersible tablet. When dosed to yield comparable
disintegrates in water in an oral syringe, on a spoon, or trough levels, the shape of the profiles was generally
in a plastic cup and is then administered by mouth. The similar between formulations and age groups, with the
two studies described herein were components of a pe- exception that Cmaxss was 19% higher in pediatric pa-
diatric clinical development program for everolimus in tients. The extent to which biopharmaceutical charac-
renal transplantation using this formulation. teristics of the two formulations or physiologic differ-
The relative bioavailability study was an essential ences in the patient populations may have contributed
first clinical study with the dispersible tablet in order to this cannot be definitively answered from these data.
to link the safety and exposure data generated in adult Based on the results from the formulation crossover in
trials using the conventional tablet to appropriate dos- healthy adults, a lower Cmaxss from the dispersible tablet
ing for a single-dose pharmacokinetic study in pediat- would be anticipated when Cminss is similar between
ric maintenance renal transplant patients.6 It also pro- formulations. The higher Cmaxss actually measured sug-
vided useful information for converting between gests that age-related differences in distribution vol-
formulations during patient care. Specifically, the ume, for example, would be a more likely explanation
crossover evaluation in healthy adults demonstrated for the observed differences in the early portion of the
that the bioavailability of everolimus from the concentration-time profile between populations. In
dispersible tablet was 10% lower relative to the con- any case, the average Cmaxss in pediatric patients receiv-
ventional tablet, and the Cmax was 24% lower. This im- ing the dispersible tablet was only modestly higher
plies that when a child or adolescent, for example, than that in adults receiving the conventional tablet.
switches from the dispersible tablet to the conventional Furthermore, the range of individual Cmaxss values in
tablet, the switch should be on a 1:1 milligram basis. pediatric patients was 4.7 to 18.4 ng/ml, which fits
When the new regimen reaches steady state (after 4-5 comfortably in the adult range of 2.2 to 24.6 ng/ml.
days), therapeutic drug monitoring should be per- In sum, steady-state concentration-time profiles in
formed to further individualize the dose as necessary. pediatric transplant patients receiving the dispersible
The food effect part of the healthy subject study pro- tablet were comparable to those of adult patients re-
vided guidelines for administering the dispersible tab- ceiving the conventional tablet when both were dosed
let with respect to meals. A high-fat meal slowed the to yield similar trough concentrations. If a pediatric pa-

146 • J Clin Pharmacol 2003;43:141-147


DEVELOPMENT OF EVEROLIMUS PEDIATRIC FORMULATION

tient is converted from the everolimus dispersible tab- 5. Food and Drug Administration: Draft Guidance for Industry: Gen-
let to the conventional tablet, this should be based on a eral Considerations for Pediatric Pharmacokinetic Studies for Drugs
and Biological Products. Rockville, MD: Food and Drug Administra-
1:1 milligram switch with subsequent therapeutic drug tion, 1998.
monitoring to further individualize the dose as needed. 6. Webb N, Mahan J, Vandamme-Lombaerts R, Lemire J, Ettenger R,
The dispersible tablet formulation should be taken Hoyer PF, et al: Pharmacokinetics and tolerability of RAD001 in pedi-
consistently either with or without food to minimize atric stable renal transplant patients [abstract]. Pediatr Transplant
fluctuations in exposure over time. The availability of a 2000;4(suppl. 2):123.
dispersible tablet formulation should facilitate the use 7. Food and Drug Administration: Draft Guidance for Industry:
of this drug in patients not able to take a conventional Bioavailability and Bioequivalence Studies for Orally Administered
tablet, such as children, the elderly, and patients with a Drug Products—General Considerations. Rockville, MD: Food and
Drug Administration, 2002.
nasogastric or nasoduodenal tube.
8. Sauter R, Steinijans VW, Diletti E, Boehm A, Schulz HU: Presenta-
tion of results from bioequivalence studies. Int J Clin Pharmacol Ther
REFERENCES Toxicol 1992;30(suppl. 1):S7-S30.
9. Schuirmann DJ: Comparison of the two one-sided tests procedure
1. Vitko S, Margreiter R, Weimar W, Dantal J, Viljoen H, Cambon N, and the power approach for assessing the bioequivalence of average
et al: International, double-blind, parallel-group study of the safety bioavailability. J Pharmacokinet Biopharm 1987;15:657-680.
and efficacy of Certican versus mycophenolate mofetil in combina- 10. Kovarik JM, Hartmann S, Figueiredo J, Rordorf C, Golor G, Lison A,
tion with Neoral and steroids [abstract]. Am J Transplant et al: Effect of food on everolimus absorption: quantification in
2001;1(suppl. 1):474. healthy subjects and a confirmatory screening in patients with renal
2. Kaplan B, Tedesco Silva H, Mendez R, Kahan B, Van Buren D, transplants. Pharmacother 2002;22:154-159.
Boger R: North/South American, double-blind, parallel-group study 11. Kahan BD, Wong RL, Carter C, Katz SH, von Fellenberg J,
of the safety and efficacy of Certican versus mycophenolate mofetil in van Buren CT, et al: Phase I study of a 4-week course of RAD in quies-
combination with Neoral and corticosteroids [abstract]. Am J Trans- cent cyclosporine-prednisone-treated renal transplant patients.
plant 2001;1(suppl. 1):475. Transplantation 1999;68:1100-1106.
3. Valantine H, Eisen H, Dorent R, Mancini D, Vigano M, Starling R, 12. Kovarik JM, Kaplan B, Tedesco Silva H, Kahan BD, Dantal J, Vitko S,
et al: Twelve-month results of a multicenter study comparing efficacy et al: Exposure-response relationships for everolimus in de novo kid-
and safety of everolimus to azathioprine in de novo cardiac trans- ney transplantation: defining a therapeutic range. Transplantation
plant recipients [abstract]. Am J Transplant 2002;2(suppl. 3):247. 2002;73:920-925.
4. International Conference on Harmonization: Guidance for Indus-
try E11: Clinical Investigations of Medicinal Products in Pediatric
Populations. Rockville, MD: Food and Drug Administration, 2000.

PEDIATRICS 147

You might also like