You are on page 1of 8

Antiviral Therapy 2015; 20:425–432 (doi: 10.

3851/IMP2932)

Original article
A novel ritonavir paediatric powder formulation
is bioequivalent to ritonavir oral solution with a
similar food effect
Ahmed Hamed Salem1,2*, Yi-Lin Chiu1, Joaquin M Valdes3, Angela M Nilius3, Cheri E Klein1
1
Clinical Pharmacology and Pharmacometrics, AbbVie, North Chicago, IL, USA
2
Clinical Pharmacy, Ain Shams University, Cairo, Egypt
3
Infectious Diseases Development, AbbVie, North Chicago, IL, USA

*Corresponding author e-mail: sale0074@umn.edu

Background: A novel ritonavir oral powder formulation Bioavailability comparisons were assessed by the 90% CIs
has been developed to eliminate the alcohol and propyl- for the geometric least-squares mean ratios.
ene glycol contents in the current ritonavir oral solution Results: Ritonavir powder formulation in water was
for paediatric use. Two clinical studies were conducted to found to be bioequivalent to the marketed oral solution.
assess the bioequivalence of the powder formulation to Ritonavir powder formulation administered in chocolate
the marketed oral solution and to evaluate the effect of milk, pudding, infant formula or apple sauce was bio-
food and vehicles on bioavailability. equivalent to the powder formulation administered in
Methods: Study 1 was a randomized, partial-crossover, water. Compared with fasting conditions, moderate-fat
4-period study in 48 subjects. Regimens included: oral and high-fat meals were associated with approximately
solution under moderate-fat conditions, powder formu- 25–40% and 35–50% reduction in ritonavir concentra-
lation in water under fasting, moderate-fat or high-fat tions, respectively.
conditions, and powder formulation in chocolate milk Conclusions: The novel ritonavir powder formulation is
or pudding under moderate-fat conditions. Study 2 was bioequivalent to marketed ritonavir oral solution under
a randomized, crossover, 4-period study in 24 subjects. moderate-fat conditions with a similar effect of meals.
Subjects were randomized to a sequence of the oral solu- None of the vehicles tested negatively affected the bioa-
tion and powder formulation in water, infant formula vailability, which suggests the potential for use of a broad
and apple sauce, all under moderate-fat conditions. range of vehicles for dose preparation.

Introduction

Ritonavir is a peptidomimetic inhibitor of HIV-1 and used as a pharmacokinetic enhancer, ritonavir blocks
HIV-2 proteases. Ritonavir (marketed as Norvir®) was the cytochrome P450 (CYP) 3A-mediated metabolism
first approved in the United States (US) and the European of the other drug, leading to increased serum levels and
Union (EU) in 1996 and is indicated in combination with prolonged half-life of the other drug [1].
other antiretroviral (ARV) agents for the treatment of Ritonavir is available as 100 mg film-coated tablets,
HIV-1-infected patients (adults and children older than 1 100 mg soft capsules and as an 80 mg/ml oral solution.
month of age in the US and 2 years of age and older in the A ritonavir oral powder formulation (AbbVie, North
EU). It is marketed in 159 countries globally. Chicago, IL, USA) for paediatric use was developed to
In recent years, ritonavir has been independently eliminate the alcohol and propylene glycol contents in
approved to be administered as a pharmacokinetic the marketed ritonavir oral solution. Feasibility stud-
enhancer at doses up to 200 mg once or twice daily for ies initially focused on an improved oral solution by
the adult as well as paediatric populations, according examining the minimum amount of alcohol and pro-
to the approved labels of the following HIV protease pylene glycol co-solvent required to solubilize the drug
inhibitors (PIs): amprenavir, atazanavir, darunavir, fosa- substance. However, even formulation variations based
mprenavir, lopinavir, saquinavir and tipranavir. When on lower ritonavir concentrations of 10 to 20 mg/ml,

©2015 International Medical Press 1359-6535 (print) 2040-2058 (online) 425

AVT-14-OA-3347_Salem.indd 425 26/06/2015 16:30:06


AH Salem et al.

rather than the current 80 mg/ml, still required rela- high-fat regimens, study drug was administered
tively high levels of alcohol co-solvents to maintain rito- approximately 30 min after the start of the meal. The
navir solubility and would have potentially increased moderate-fat breakfast consisted of approximately 600
daily alcohol intake for an equivalent therapeutic dose. Kcal, with approximately 30% from fat. The high-fat
Therefore, an alternative ritonavir oral powder formu- breakfast consisted of approximately 900 Kcal, with
lation that eliminates alcohol and propylene glycol co- approximately 60% from fat. On the other study days,
solvents was explored to better meet the needs of the subjects received a standardized diet during confine-
paediatric population. The two clinical studies reported ment, providing approximately 30% of the daily calo-
here aimed to assess the bioequivalence of the oral ries from fat, 50% from carbohydrates and 20% from
powder to the marketed ritonavir oral solution, assess protein (adding up to approximately 2,000 Kcal/day).
the bioavailability of the new formulation under vari- In study 2, single doses of 100 mg ritonavir were
ous meal conditions and evaluate the bioavailability of separated by at least 5 days in a 4-period, randomized,
the oral powder when administered in chocolate milk, complete crossover, open-label design in 24 healthy
pudding, infant formula or apple sauce. subjects according to a complete-crossover design.
The four regimens, all administered under moderate-
Methods fat meal conditions were: ritonavir powder in water,
infant formula and apple sauce, and ritonavir oral solu-
Design of the studies tion. In addition to the respective vehicle, each subject
Two studies (studies 1 and 2) were performed to assess consumed approximately 240 ml of water. The caloric
the bioavailability of the ritonavir oral powder (AbbVie) intake came primarily from carbohydrates (54%) while
compared to the marketed Norvir® oral solution (AbbVie) fat and protein accounted for 29% and 17%, respec-
under moderate-fat conditions. Study 1 also assessed the tively, of the calories. The sequences of regimens were
bioavailability of the oral powder in water under various such that each subject was to receive all four regimens
meal conditions (dosing following a 10-h fast or following upon completion of the study.
a moderate-fat or high-fat meal) and assessed the effect Study subjects were HIV-negative adults in general
of two vehicles (chocolate milk and vanilla pudding) on good health who did not require any concomitant med-
the bioavailability of the oral powder. Study 2 assessed ication. These studies were performed in accordance
the bioavailability of the oral powder in three vehicles with the Declaration of Helsinki and its amendments,
(water, infant formula, apple sauce). The bioequivalence and in compliance with the guidelines of Good Clini-
and vehicle effect assessments were performed on study cal Practice. The study protocols and informed consent
drug administered following a moderate-fat meal, as the forms were approved by the Institutional Review Board
marketed oral solution is recommended to be taken with of Research Consultants Review Committee (Austin,
food. Safety and tolerability were assessed throughout the TX, USA). Written informed consent was obtained
studies based on reported adverse events, vital signs, elec- from all subjects prior to entry into the study.
trocardiograms and clinical laboratory measurements.
In study 1, single doses of 100 mg ritonavir were sepa- Sample collection and assay
rated by at least 5 days in a 4-period, randomized, par- In each study, blood samples were drawn for pharma-
tial-crossover, open-label design in 48 healthy subjects. In cokinetic analysis immediately prior to dosing (0 h) and
period 1 and 2, all 48 subjects received a single 100-mg at approximately 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 18, 24,
dose of marketed ritonavir oral solution and a single 100- 30 and 36 h post-dosing. The concentrations of rito-
mg dose of ritonavir oral powder in water administered navir in plasma were analysed using a validated liquid
under moderate-fat conditions in a crossover fashion. In chromatography method with tandem mass spectrom-
period 3 and 4, 24 subjects received single 100-mg doses etry (MS/MS) detection [2]. The correlation coefficients
of ritonavir oral powder in water administered under were equal to or greater than 0.990 and 0.995 for the
high-fat conditions and under fasting conditions, while respective study 1 and study 2 standard curves. The
the other 24 subjects received single 100-mg doses of rito- coefficients of variation ranged from -2.8% to 2.2%
navir oral powder in chocolate milk administered under for study 1 and from -1.4% to 3.1% for study 2. The
moderate-fat conditions and in vanilla pudding adminis- lower limits of quantitation for study 1 and study 2
tered under moderate-fat conditions, in a crossover fash- were approximately 9.49 and 9.58 ng/ml, respectively.
ion. Each dose of study drug was administered orally in Values that were less than the lower limit of quantita-
the morning on day 1 of each period. tion for a given run were reported as zero.
Approximately 240 ml of water was administered
following each dose. For the fasting regimen, study Pharmacokinetic and statistical analyses
drug was administered following an approximate 10-h Ritonavir pharmacokinetic parameters were calculated
fast and at least 4 h before lunch. For the moderate- and with standard non-compartmental analyses using Phoenix

426 ©2015 International Medical Press

AVT-14-OA-3347_Salem.indd 426 26/06/2015 16:30:07


Ritonavir paediatric powder formulation

WinNonlin version 5.2.1 (Pharsight Corp., Mountain subject discontinued due to a positive cotinine test that
View, CA, USA). The maximum observed plasma concen- was assessed as not related to study drug.
tration (Cmax), time to Cmax (Tmax), area under the plasma
concentration–time curve (AUC) to the last measured Analyses from study 1
plasma concentration (AUCt) and extrapolated to infinite The mean concentration–time profiles for ritonavir
time (AUC∞) were calculated. 100 mg dosed as the oral powder and oral solution
All available data were included in the analyses for are shown in Figure 1. The point estimates of relative
both studies. Three types of assessments were per- bioavailability and the 90% CIs for ritonavir AUC and
formed: bioequivalence assessment (oral powder in Cmax values for the assessments in study 1 are presented
water versus marketed oral solution), vehicle effect in Table 2.
assessment (oral powder in chocolate milk, vanilla
pudding, apple sauce or infant formula versus oral Bioequivalence assessment for study 1
powder in water) and food effect assessment (oral The oral powder was bioequivalent to the reference
powder in water following a moderate-fat or high-fat marketed oral solution when administered following
breakfast versus oral powder in water under fasting a moderate-fat meal as the 90% CIs of the geometric
conditions). For the bioequivalence assessment, bio- mean ratios for Cmax, AUCt and AUC∞ were contained
equivalence was declared if the 90% CIs of the ratios within the range of 0.80 to 1.25.
of AUCt, AUC∞ and Cmax for the oral powder relative
to the oral solution were contained within the 0.80 to Food effect assessment for study 1
1.25 range. For the oral powder under moderate-fat conditions ver-
For all three assessments, a mixed effects analysis sus fasting conditions, a moderate-fat meal decreased
was performed for Tmax, and the natural logarithms of the bioavailability with respect to Cmax, AUCt and AUC∞
Cmax, AUCt and AUC∞ using the SAS system version 9.2 by approximately 39%, 25% and 23%, respectively.
software (SAS Institute, Cary, NC, USA). The model Likewise, comparing administration under high-fat
included effects for sequence, period and regimen. conditions with fasting conditions, a high-fat meal
decreased the bioavailability with respect to Cmax, AUCt
Results and AUC∞ by approximately 49%, 34% and 32%,
respectively. In Table 3, food effect results from study
Demographic data for the subjects enrolled in these 1 are compared to those observed with currently mar-
studies are shown in Table 1. For study 1, all 48 sub- keted oral solution and tablets.
jects completed study drug administration for all 4
study periods, and all 48 subjects completed all study Vehicle effect assessment for study 1
procedures. For study 2, 24 subjects were enrolled in Under moderate-fat conditions, administration of the
the study and 23 completed all study procedures. One oral powder in chocolate milk and in pudding demon-
strated bioequivalent peak and total ritonavir expo-
sures when compared with administration in water
Table 1. Demographics of the healthy adult subjects enrolled as the 90% CIs of the geometric mean ratios for Cmax,
in the studies
AUCt and AUC∞ were contained within the range of
Study 1 (n=48) Study 2 (n=24) 0.80 to 1.25.

Sex Analyses from study 2


Male, n (%) 30 (63) 15 (63) The mean concentration–time profiles for ritonavir 100
Female, n (%) 18 (37) 9 (37) mg in each regimen are shown in Figure 2. The overall
Ethnicity/race concentration profiles are similar for the oral solution and
White, n (%) 30 (63) 19 (79)
the oral powder. The point estimates of relative bioavail-
Black, n (%) 13 (27) 5 (21)
ability and the 90% CIs for ritonavir AUC and Cmax values
Asian, n (%) 2 (4) –
for the assessments in study 2 are presented in Table 4.
Multi-race, n (%) 2 (4) –
Native Hawaiian or other 1 (2) – Administration of the oral powder formulation in
Pacific Islander, n (%) water was bioequivalent to the oral solution under
Mean age, years ±sd 31.6 ±8.4 37.3 ±10.9 moderate-fat conditions. Likewise, oral powder formu-
(range) (20–53) (19–55) lation in infant formula and apple sauce under moder-
Mean weight, kg ±sd 75.4 ±11.7 72.2 ±12.0 ate-fat conditions met bioequivalence criteria relative
(range) (53–115) (55–99) to the oral powder in water because the 90% CIs of
Mean height, cm ±sd 170 ±9.8 170 ±10.0 the ratio of central values for AUCs and Cmax were con-
(range) (152–200) (153–197) tained within the range of 0.80 to 1.25.

Antiviral Therapy 20.4 427

AVT-14-OA-3347_Salem.indd 427 26/06/2015 16:30:07


AH Salem et al.

Figure 1. Mean (+sd) ritonavir plasma concentration–time profiles from study 1

A 0.6
Marketed ritonavir oral solution, moderate-fat (n=48)
Ritonavir oral powder in water, moderate-fat (n=48)

Ritonavir concentration, µg/ml 0.5

0.4

0.3

0.2

0.1

0.0
0 4 8 12 16 20 24 28 32 36
Time, h
B 1.2
Ritonavir oral powder in water, moderate-fat (n=24)
Ritonavir oral powder in water, high-fat (n=24)
1.0 Ritonavir oral powder in water, fasting (n=24)
Ritonavir concentration, µg/ml

0.8

0.6

0.4

0.2

0.0
0 4 8 12 16 20 24 28 32 36
Time, h
C 0.7
Marketed ritonavir solution, moderate-fat (n=24)
Ritonavir oral powder in water, moderate-fat (n=24)
0.6 Ritonavir oral powder in chocolate milk, moderate-fat (n=24)
Ritonavir oral powder in pudding, moderate-fat (n=24)
Ritonavir concentration, µg/ml

0.5

0.4

0.3

0.2

0.1

0.0
0 4 8 12 16 20 24 28 32 36
Time, h

(A) Bioequivalence assessment. (B) Food effect assessment. (C) Vehicle assessment.

428 ©2015 International Medical Press

AVT-14-OA-3347_Salem.indd 428 26/06/2015 16:30:07


Ritonavir paediatric powder formulation

Table 2. Relative bioavailability and 90% CIs for the bioequivalence assessment of ritonavir from study 1

Central valueb Relative bioavailability


Regimens and pharmacokinetic parameters a
Test Reference Point estimatec 90% CI

Bioequivalence assessment (under moderate-fat conditions)


Oral powder versus oral solution
Cmax 0.39 0.39 1.017 0.947, 1.092
AUCt 4.15 4.02 1.032 0.982, 1.084
AUC∞ 4.36 4.19 1.039 0.992, 1.087
Food effect assessment
Oral powder: high-fat versus fasting
Cmax 0.36 0.72 0.506 0.440, 0.581
AUCt 4.11 6.25 0.658 0.608, 0.711
AUC∞ 4.32 6.39 0.677 0.629, 0.728
Oral powder: moderate-fat versus fasting
Cmax 0.43 0.72 0.606 0.535, 0.686
AUCt 4.68 6.25 0.750 0.692, 0.812
AUC∞ 4.90 6.39 0.767 0.709, 0.829
Vehicle effect assessment (under moderate-fat conditions)
Oral powder in chocolate milk versus oral powder in water
Cmax 0.32 0.36 0.897 0.803, 1.001
AUCt 3.62 3.68 0.983 0.899, 1.074
AUC∞ 3.78 3.88 0.976 0.896, 1.063
Oral powder in pudding versus oral powder in water
Cmax 0.38 0.36 1.064 0.953, 1.188
AUCt 4.07 3.68 1.106 1.012, 1.209
AUC∞ 4.24 3.88 1.094 1.004, 1.191
a
Units for maximum observed plasma concentration (Cmax) and area under the plasma concentration–time curve (AUC) are µg/ml and µg•h/ml, respectively. bAnti-
logarithm of the least squares means for logarithms. cAnti-logarithm of the difference (test minus reference) of the least squares means of the logarithms. AUCt, AUC
to the last measured plasma concentration; AUC∞, AUC extrapolated to infinite time.

Table 3. Effect of food on the bioavailability of the marketed tablet, marketed oral solution and oral powder

Marketed tablet [3] (100 mg) Marketed oral solution [4] (600 mg) Oral powder (study 1; 100 mg)
Parametera PEb 90% CIc PEb 90% CI PEb 90% CI

Cmax 0.782d 0.675, 0.907d 0.769e 0.705, 0.838e 0.606f 0.535, 0.686f
AUC∞ 0.795d 0.724, 0.873d 0.930
e
0.847, 1.021e 0.767f 0.709, 0.829f
Cmax 0.766g 0.659, 0.891g – – 0.506h 0.440, 0.581h
AUC∞ 0.775g 0.704, 0.853g – – 0.677h 0.629, 0.728h
a
Units for maximum observed plasma concentration (Cmax) and area under the plasma concentration–time curve extrapolated to infinite time (AUC∞) are µg/ml and
µg•h/ml, respectively. bPoint estimate (PE) for relative bioavailability determined as the anti-logarithm of the difference of least squares means for logarithmically
transformed values, with ‘fasting’ as the reference. cDue to the discreteness of the sign test statistic, the actual confidence level was 94.8% for the marketed
tablet, which was higher than the necessary 90%. dModerate fat (approximately 30% fat of 857 Kcal). eLow fat (approximately 10% fat of 514 Kcal). fModerate fat
(approximately 30% fat of 600 Kcal). gHigh fat (approximately 50% fat of 907 Kcal). hHigh fat (approximately 60% fat of 900 Kcal).

Safety analysis 1 subject each. Seven subjects in study 1 reported only


All treatment-emergent adverse events (TEAE) reported AEs that were determined by the investigator as having
in both studies were mild in severity. No deaths or dis- no reasonable possibility of relationship to study drug,
continuations due to adverse events occurred during while one subject reported events of headache and nau-
the studies. No clinically significant or potentially clini- sea that were determined as having a reasonable pos-
cally significant clinical laboratory, vital signs, ECG and sibility of a relationship to study drug. In the post-study
physical examination findings were observed after dos- period of study 1, one serious adverse event (SAE) of
ing in either of the two studies. ‘induced abortion’ was reported in a 21-year-old female
In study 1, 8 of 48 subjects (16.7%) reported ≥1 subject who underwent an elective abortion. This event
TEAE. The most common TEAE was headache reported was evaluated by the investigator as having no reason-
by 4 of 48 subjects. All other TEAEs were reported by able possibility of relationship to the study drug.

Antiviral Therapy 20.4 429

AVT-14-OA-3347_Salem.indd 429 26/06/2015 16:30:07


AH Salem et al.

Figure 2. Mean (+sd) ritonavir plasma concentration–time profiles from study 2 under moderate-fat conditions

0.8
Ritonavir oral powder in water
Ritonavir oral powder in infant formula
Ritonavir oral powder in apple sauce
Marketed ritonavir oral solution

0.6
Ritonavir concentration, µg/ml

0.4

0.2

0.0
0 4 8 12 16 20 24 28 32 36
Time, h

Table 4. Relative bioavailability and 90% CI for the central values of ritonavir pharmacokinetic parameters from study 2
Regimens (test versus reference under moderate-fat Central valueb Relative bioavailability
conditions) and pharmacokinetic parametera Test Reference Point estimatec 90% CI

Oral powder in water versus oral solution


Cmax 0.49 0.49 1.001 0.901, 1.112
AUCt 4.26 4.24 1.005 0.938, 1.077
AUC∞ 4.42 4.43 0.998 0.934, 1.066
Oral powder in infant formula
versus oral powder in water
Cmax 0.51 0.49 1.053 0.947, 1.17
AUCt 4.48 4.26 1.054 0.985, 1.128
AUC∞ 4.66 4.42 1.054 0.988, 1.124
Oral powder in apple sauce versus
oral powder in water
Cmax 0.47 0.49 0.964 0.868, 1.071
AUCt 4.08 4.26 0.959 0.897, 1.026
AUC∞ 4.25 4.42 0.962 0.902, 1.026

a
Units for maximum observed plasma concentration (Cmax) and area under the plasma concentration–time curve (AUC) are µg/ml and µg•h/ml, respectively. bAnti-
logarithm of the least squares means for logarithms. cAnti-logarithm of the difference (test minus reference) of the least squares means for logarithms. AUCt, AUC to
the last measured plasma concentration; AUC∞, AUC extrapolated to infinite time.

In study 2, 5 of 24 subjects (20.8%) reported ≥1 The most common TEAE was headache, reported by
TEAEs. The proportions of subjects reporting ≥1 three subjects (12.5%). All other TEAEs were reported
TEAE summarized by regimen were 3/23 subjects by one subject each. TEAEs of abdominal pain, nau-
(13.0%) in regimen A and 4/23 (17.4%) in regimen D. sea and headache were assessed by the investigator as

430 ©2015 International Medical Press

AVT-14-OA-3347_Salem.indd 430 26/06/2015 16:30:08


Ritonavir paediatric powder formulation

possibly related to study drug. No SAEs were reported 20–23% lower when dosed following a meal compared
in study 2. to administration under fasting conditions [3]. When
the oral solution was given under non-fasting condi-
Discussion tions, peak ritonavir concentrations decreased by 23%
and the extent of absorption decreased 7% relative
Here we describe the bioavailability of a novel ritonavir to fasting conditions [4]. Both tablet and solution are
formulation for paediatric use. The new formulation is labelled to be taken with food and hence the new pow-
an oral powder that is to be suspended in liquid vehicles der formulation is proposed to be taken with food.
or sprinkled on food for administration. In comparison Both studies were conducted at a dose of 100 mg.
to the marketed ritonavir oral solution, the new pow- The 100 mg dose is the most commonly administered
der formulation provides longer expiry dating through dose of ritonavir as a pharmacokinetic enhancer of
slower degradation and an alcohol and propylene gly- other PIs [5,6]. In addition, the 100 mg dose lies in the
col-free formulation which may be desired by some non-linear range of ritonavir pharmacokinetics where
physicians and other health-care providers. In addition, potential differences in exposure between formula-
the new powder formulation would retain the flexibility tions are likely to be magnified. Therefore, the 100 mg
of the ritonavir oral solution to accommodate ARV or dose is considered to be a sensitive dose to detect dif-
pharmacokinetic-enhancing doses, including weight or ferences in bioavailability and bioequivalence results
body surface area doses as indicated in other approved at this dose level can be confidently extrapolated to
package inserts. It will also provide a dosage form for other doses.
patients who may have difficulty swallowing a tablet. The effect of food on the ritonavir oral powder bioa-
Two clinical studies were conducted to assess the bio- vailability may be attributed to the change in physiolog-
equivalence of the new formulation to the marketed oral ical pH of the upper intestinal tract after meals. Due to
solution and to evaluate the effect of food and adminis- the gastric emptying of acidic chime in the postprandial
tration vehicles on its bioavailability. The results of study state, the physiological pH of the upper intestinal tract
1 demonstrated the bioequivalence of the powder for- is lower, at pH 5, when compared with the pre-prandial
mulation to the oral solution. The bioequivalence dem- state, pH 6.5 [7,8]. Ritonavir is a weak base and hence
onstrated in the study allows the extrapolation of the will be ionized to a higher extent in the fed state. Such
existing safety, clinical pharmacology and efficacy data ionization may result in slightly lower permeability
obtained with the oral solution to the new formulation. and absorption when compared with the fasted state
Five different vehicles for administration were [9]. When co-administered with other PIs, effect of acid
assessed including two soft foods and three liquids. Soft reducing agents on ritonavir varied according to the co-
food options included apple sauce and pudding which administered PI [10,11].
allow for administration of the powder as a sprinkle. The safety results were consistent with the known
The other tested vehicles were water, infant formula safety profile of ritonavir. Headache and gastrointesti-
and chocolate milk which allow for administration as a nal events were the most frequently reported TEAEs in
liquid. Administration of the new formulation in infant these studies. No clinically significant values in clini-
formula, chocolate milk, apple sauce or pudding was cally laboratory evaluations or ECGs were observed.
shown to be bioequivalent to administration in water. In summary, the new ritonavir powder formulation
Therefore, the new ritonavir paediatric formulation may is bioequivalent to the currently marketed ritonavir
be taken in a variety of vehicles to allow convenience in oral solution under moderate-fat conditions. Ritonavir
various patient settings and to meet patient preference. exposure following administration of the new formu-
In study 1, we also compared the bioavailability of lation is decreased by 23% to 49% in the fed condi-
the new formulation under moderate-fat and high-fat tion, compared with the fasted condition. Consistent
conditions to that under fasting condition in order with the recommendation for the marketed tablets
to assess the effect of food on the new formulation. and oral solution, the new powder formulation should
Results demonstrated that food significantly decreased be administered with food. The data from this study
both Cmax and AUC of ritonavir by 23% to 49% follow- suggest that a variety of vehicles may produce similar
ing powder formulation administration under either bioavailability as none of the vehicles tested negatively
moderate-fat or high-fat meal conditions. Tmax was affected the bioavailability of the ritonavir oral powder.
longer when the formulation was given with food. This
increase in Tmax is consistent with delayed gastric emp- Acknowledgements
tying following a meal. Reductions in ritonavir expo-
sures with food have also been reported with currently The authors gratefully acknowledge Sonja Kemmis
marketed oral solution and tablets. For the ritonavir Causemaker and Mary Woulfe, both from AbbVie
tablets, ritonavir Cmax and AUC were approximately (North Chicago, IL, USA), for their assistance with

Antiviral Therapy 20.4 431

AVT-14-OA-3347_Salem.indd 431 26/06/2015 16:30:08


AH Salem et al.

study conduct, data analysis and manuscript prepara- 4. Norvir (ritonavir). Package insert 2013. AbbVie Inc., North
Chicago, IL, USA.
tion. The findings reported in this article were presented
in part at the 15th International Workshop on Clinical 5. Reyataz (atazanavir). Package insert 2014. Bristol–Myers
Squibb, Princeton, NJ, USA.
Pharmacology of HIV & Hepatitis Therapy in Washing- 6. Prezista (darunavir). Package insert 2014. Janssen
ton DC, USA from 19–21 May 2014 (Abstract P_01). Therapeutics, Titusville, NJ, USA.
7. Shiau YF, Fernandez P, Jackson MJ, McMonagle S.
Mechanisms maintaining a low-pH microclimate in the
Disclosure statement intestine. Am J Physiol 1985; 248:G608–G617.
8. Charman WN, Porter CJ, Mithani S, Dressman JB.
The design, study conduct and financial support for the Physicochemical and physiological mechanisms for the
effects of food on drug absorption: the role of lipids and
clinical trials were provided by AbbVie. AbbVie partici- pH. J Pharm Sci 1997; 86:269–282.
pated in the interpretation of data, review and approval 9. Marasanapalle VP, Crison JR, Ma J, Li X, Jasti BR.
of the publication. All authors are employees of AbbVie Investigation of some factors contributing to negative food
and have no additional conflicts of interest to disclose. effects. Biopharm Drug Dispos 2009; 30:71–80.
10. Klein CE, Chiu YL, Cai Y, et al. Effects of acid‐reducing
agents on the pharmacokinetics of lopinavir/ritonavir
References and ritonavir‐boosted atazanavir. J Clin Pharmacol 2008;
48:553–562.
1. Hull MW, Montaner JS. Ritonavir-boosted protease 11. Sekar VJ, Lefebvre E, De Paepe E, et al. Pharmacokinetic
inhibitors in HIV therapy. Ann Med 2011; 43:375–388. interaction between darunavir boosted with ritonavir and
2. Myasein F, Kim E, Zhang J, Wu H, El-Shourbagy TA. Rapid, omeprazole or ranitidine in human immunodeficiency virus-
simultaneous determination of lopinavir and ritonavir negative healthy volunteers. Antimicrob Agents Chemother
in human plasma by stacking protein precipitations and 2007; 51:958–961.
salting-out assisted liquid/liquid extraction, and ultrafast
LC–MS/MS. Anal Chim Acta 2009; 651:112–116.
3. Klein C, Chiu Y, Awni W, et al. The effect of food on
ritonavir bioavailability following administration of
ritonavir 100 mg film-coated tablet in healthy adult subjects.
Seventh International Congress on Drug Therapy in HIV
Infection. 9–13 November 2008, Glasgow, Scotland.
Abstract P247.
Accepted 24 November 2014; published online 9 January 2015

432 ©2015 International Medical Press

AVT-14-OA-3347_Salem.indd 432 26/06/2015 16:30:08

You might also like