Quick viewing(Text Mode)

Original Article Pharmacokinetic Interaction Between Ethinyl Estradiol, Norethindrone and Darunavir with Low-Dose Ritonavir in Healthy Women

Original Article Pharmacokinetic Interaction Between Ethinyl Estradiol, Norethindrone and Darunavir with Low-Dose Ritonavir in Healthy Women

Sekar 11/6/08 16:41 Page 563

Antiviral Therapy 13:563–569

Original article Pharmacokinetic interaction between ethinyl , norethindrone and darunavir with low-dose ritonavir in healthy women

Vanitha J Sekar1*, Eric Lefebvre1, Sabrina Spinosa Guzman2, Elise Felicione1, Martine De Pauw2, Tony Vangeneugden2 and Richard MW Hoetelmans2

1Tibotec Inc, Yardley, PA, USA 2Tibotec BVBA, Mechelen, Belgium

*Corresponding author: E-mail: [email protected]

Background: An open-label, randomized, crossover concentration (Cmin), the maximum plasma concentration

study was performed to investigate the effect of (Cmax), and the area under the curve (AUC24h) of EE multiple doses of darunavir co-administered with low- (which decreased by 62%, 32% and 44%, respectively) dose ritonavir (DRV/r) on the steady-state and NE (which decreased by 30%, 10% and 14%, pharmacokinetics of the oral contraceptives ethinyl respectively) compared with administration of EE and NE estradiol (EE) and norethindrone (NE) (commercial alone. Five participants discontinued the study due to name of the combined Ortho-Novum 1/35) in 19 grade 2 cutaneous events, as required per protocol, HIV-negative healthy women. during treatment with EE and NE in combination with Methods: In session 1, participants received 35 μg EE and DRV/r. There were no clinically relevant findings for 1.0 mg NE from days 1 to 21. In session 2, participants laboratory and cardiovascular parameters. received the same oral contraceptive treatment as in Conclusions: The pharmacokinetic interaction observed session 1 on days 1 to 21 plus DRV/r (600 mg/100 mg here is considered to be clinically relevant as EE concen- twice daily) on days 1 to 14. Pharmacokinetic assessments trations are considerably reduced when DRV/r is were performed on day 14 for each session. co-administered with EE and NE. Alternative or addi- Results: Steady-state systemic exposure to EE and NE tional contraceptive measures should be used when decreased when DRV/r was co-administered, based on oestrogen-based contraceptives are co-administered the ratio of least square means of the minimum plasma with DRV/r.

Introduction

Oral contraceptives (OCs) are one of the most (DRV) and ritonavir (RTV) inhibit CYP3A4, while commonly used contraceptive methods worldwide and RTV also induces CYP3A4 and other enzymes, are used by both HIV-1-negative and HIV-1-infected including glucuronosyl transferase [8,9]. Therefore, as women [1]. OCs most commonly comprise a fixed a result of drug–drug interactions, there is the potential combination of ethinyl estradiol (EE) and norethin- for an alteration of drug exposure for OCs. drone (NE) [2]. As guidelines for the treatment of DRV (TMC114, Prezista®) is a novel PI administered HIV-infected, antiretroviral-naive individuals recom- in combination with low-dose ritonavir (DRV/r). DRV mend the first-line use of two nucleoside reverse binds to the HIV protease and is highly active against transcriptase inhibitors (NRTIs) with either a protease both wild-type and resistant HIV-1 strains [8,10]. DRV inhibitor (PI; generally boosted) or a non-nucleoside has received its first regulatory approval for the treat- reverse transcriptase inhibitor (NNRTI) [3–5], PIs and ment of HIV infection in treatment-experienced adult OCs are likely to be combined in HIV-1-infected patients, such as those with HIV-1 strains resistant to women. Both PIs and OCs are metabolized by the more than one PI [8]. cytochrome P450 3A4 isoenzyme (CYP3A4) and EE is Drug–drug interaction studies with PIs and OCs also metabolized by glucuronidation [5–7]. Darunavir have shown changes (both increases and decreases) in

© 2008 International Medical Press 1359-6535 563 Sekar 11/6/08 16:41 Page 564

VJ Sekar et al.

pharmacokinetic parameters of EE and NE [5,11]. (DRV was taken as two 300 mg tablets; RTV as a 100 mg Co-administration with atazanavir (ATV) and indi- capsule). Intake of DRV/r was within 15 min after navir (IDV) led to plasma area under the curve (AUC) completing a meal. When DRV, RTV, EE and NE were increases of EE (48% and 24%, respectively) and NE co-administered (session 2), the order of intake was: first (110% and 26%, respectively). Co-administration EE/NE, then RTV, followed by DRV. During session 1, with fosamprenavir (FPV) resulted in increases in the participants were admitted to the study unit from day 13

minimum plasma concentration (Cmin) of EE and NE to day 15; during session 2, participants were admitted to (32% to 45%), but no significant changes in the AUC the study unit from day 1 to day 15. On day 14 of EE and NE. This co-administration also affected the (pharmacokinetic sampling day) of both sessions 1 and 2, pharmacokinetic parameters of amprenavir (APV): a participants received a standard breakfast in the testing

decrease in AUC of 22% and a decrease in Cmin of facility (a standard breakfast consisted of four slices of 20% was observed. Decreases in the AUC of both EE bread, two slices of ham or cheese, butter, jelly and two (42–47%) and NE (17–18%) have been observed with cups of decaffeinated coffee or tea with milk and/or co-administration of lopinavir/ritonavir (LPV/r) and sugar). The time of each intake of study medication and nelfinavir (NFV) and decreases in the AUC of EE start and stop times of meals served in the testing facility (40–50%) have been observed with co-administration were recorded in the case report forms. of RTV and tipranavir/ritonavir (TPV/r). For patients The study protocol was reviewed and approved by who use one of these four PIs and, more importantly, the appropriate Institutional Review Board (IRB) and ritonavir-boosted PIs, which decrease the plasma health authorities, and was conducted in accordance concentrations of EE and NE (LPV, NFV, RTV and with the Declaration of Helsinki. Written informed TPV), the use of alternative or an additional method of consent was obtained from all volunteers. contraception is recommended. In the present study, the effect of multiple doses of Pharmacokinetic assessments DRV/r was investigated on the steady-state Serial venous blood samples were drawn over the pharmacokinetics of EE and NE in HIV-negative dosing interval for EE, NE, DRV and RTV. Exact times healthy women. Short-term safety and tolerability of of blood sampling for pharmacokinetics were recorded the co-administration of DRV/r with EE and NE was and plasma samples were stored at ≤-18˚C until also assessed. analysis. Plasma concentrations of EE, DRV and RTV were determined by validated liquid chromatography Methods tandem mass spectrometry (LC-MS/MS) methods. The lower limit of quantification was 3.00 pg/ml for EE, Study population and study design 10.0 ng/ml for DRV, and 5.00 ng/ml for RTV. Plasma This was an open-label, single centre, randomized, concentrations of NE were determined using a validated crossover trial to investigate the effect of multiple doses gas chromatography mass spectrometry (GC-MS) of DRV/r on the steady-state pharmacokinetics of EE method. The lower limit of quantification was 0.0500 and NE. The study participants were 19 HIV-negative, ng/ml. The precision and accuracy of the analytical healthy, non-smoking women, aged 18 to 43 years with method for plasma DRV and RTV was within the normal weight (body mass index 18–30 kg/m2, acceptable limit of 15%: coefficients of variation for the extremes included) receiving oral contraceptives EE low-, medium- and high-quality control samples were 35 μg/NE 1.0 mg for at least 2 weeks before screening <13% [12]. The precision and accuracy of the analytical and until 1 month after receiving the last dose of method for plasma EE and NE was within the accept- DRV/r. Study participants consented to use a double able limit of 20%: coefficients of variation for the low-, barrier method of birth control (that is, condom plus medium- and high-quality control samples were <18%. diaphragm or cervical cap) from screening until 1 In session 1, plasma samples were taken on day 1 month after the last study medication intake. (pre-dose) and on day 14 at pre-dose and 0.5, 1, 2, 3, 4, Individuals were randomized to two panels to receive 6, 8, 10, 12, 16, 20 and 24 h post-dose for the measure- two consecutive 28-day sessions (each session is a full ment of plasma concentrations of EE and NE. In session menstrual cycle): session 1 followed by session 2 (panel I; 2, samples for the measurement of plasma concentra- n=10) or session 2 followed by session 1 (panel II; n=9). tions of EE and NE were taken on day 1 (pre-dose), During session 1, participants received EE 35 μg/NE 1.0 days 11 to 13 (pre-dose), and on day 14 at pre-dose, 0.5, mg (capsule) from day 1 to day 21, there was no OC 1, 2, 3, 4, 6, 8, 10, 12, 16, 20 and 24 h post-dose. In treatment on days 22–28. In session 2, participants session 2, samples for the measurement of plasma received the same OC treatment (from day 1 to day 21) concentrations of DRV and RTV were taken on day 1 as in session 1, but in addition from day 1 to day 14 (pre-dose), days 11 to 13 (pre-dose), and on day 14 at DRV/r 600 mg/100 mg twice daily was co-administered pre-dose, 0.5, 1, 1.5, 2, 3, 4, 5, 6, 10 and 12 h post-dose.

564 © 2008 International Medical Press Sekar 11/6/08 16:41 Page 565

Pharmacokinetic interaction of oral contraceptives and darunavir/r

Safety and pharmacodynamic evaluations participants discontinued the study before completing Safety and tolerability were assessed throughout the session 1 of panel I (2 participants were lost to follow up study by adverse events (AEs), clinical laboratory eval- and 1 participant withdrew consent) and 5 participants uations, physical examinations, vital signs, and 12-lead discontinued owing to AEs during session 2 of panel II electrocardiograms (ECG). All safety evaluations were (for details, see results for safety and tolerability below). graded according to the Division of AIDS (DAIDS) Full pharmacokinetic profiles of EE and NE were avail- grading scale [13]. able for 13 participants in session 1 and for 11 Pharmacodynamic assessments of OC efficacy, that participants in session 2; full pharmacokinetic profiles of is, serum concentrations of , luteinising DRV and RTV were available for 11 participants (in (LH) and follicle-stimulating hormone (FSH), session 2). The median age was 34 years (range 18–43). were determined before drug administration on day 1 and day 14 of each session. For these assessments, no Pharmacokinetic analysis formal statistical analysis was performed as this The linear mean plasma concentration–time curves of analysis was only exploratory in nature. EE and NE are shown in Figures 1A and 1B, respec- tively. Visual inspection of the mean plasma Pharmacokinetic and statistical methods concentration–time curves of EE and NE revealed that Pharmacokinetic analysis was done using WinNonlin at steady-state, co-administration of EE/NE and DRV/r Professional™ (version 4.1; Pharsight Corporation, (session 2, day 14) resulted in lower mean plasma Mountain View, CA, USA). Noncompartmental analysis concentrations of EE and NE than following adminis- model 200 (extravascular input, plasma data) was tration of EE/NE alone (session 1, day 14) in the entire applied for the pharmacokinetic analysis. Descriptive dosing interval. Mean pharmacokinetic parameters statistics were calculated for the plasma concentrations of determined for EE and NE are shown in Table 1. EE, NE, DRV and RTV, and the derived pharmacokinetic Individual pre-dose plasma concentrations of EE parameters. Furthermore, Microsoft Excel® (version and NE for days 11, 12, 13 and 14 of session 2 showed 2000; Microsoft, Redmond, WA, USA) and/or SAS that steady-state conditions were achieved prior to full (version 9.1.3, SAS Institute Inc., Cary, NC, USA) were pharmacokinetic blood sampling on day 14. used for statistical analyses. When DRV/r was added to treatment with EE/NE, Statistical analysis was performed comparing systemic exposure to EE decreased. The ratios of the LS session 2 (test) versus session 1 (reference) for EE and means (90% CI) of EE were 0.38 (0.27–0.53), 0.68

NE. The primary pharmacokinetic parameters were (0.61–0.74) and 0.56 (0.50–0.63), for Cmin, Cmax and

Cmin, maximum plasma concentration (Cmax) and AUC24h, respectively, comparing administration of

AUC24h for EE and NE. The least square (LS) means of EE/NE in the presence of DRV/r 600 mg/100 mg twice the primary parameters for each treatment group were daily to administration of EE/NE alone. The median estimated with a linear mixed effects model, controlling time to reach maximum plasma concentrations of EE for treatment, period and sequence as fixed effects and was 2.0 to 3.0 h after dosing. participant as a random effect. A 90% confidence Systemic exposure to NE also decreased when interval (CI) was constructed around the difference DRV/r was added to treatment with EE/NE. The ratios between the LS means of test and reference. Both the of the LS mean (90% CI) of NE were 0.70 (0.51–0.97),

difference between the LS means and the 90% CIs were 0.90 (0.83–0.97) and 0.86 (0.75–0.98) for Cmin, Cmax

retransformed to the original scale. Tmax (time to and AUC24h, respectively, comparing administration of maximal plasma concentrations) was subjected to the EE/NE in the presence of DRV/r to administration of Mann–Whitney U-test as described by Koch [14]. EE/NE alone. The median time to reach maximum Pharmacokinetic parameters of DRV and RTV were plasma concentrations of NE was 2.0 h after dosing for derived and were compared with historical data. Pre- both treatments (Table 1). ± dose plasma concentrations on days 11, 12, 13 and 14 The mean SD values of Cmax, Cmin and AUC12h of in session 2 were compared graphically to verify the DRV (8,073 ±988 ng/ml, 2,594 ±1,090 ng/ml and achievement of steady-state conditions for EE, NE, 55,920 ±11,290 ng h/ml) and ritonavir (1,780 ±864 DRV and RTV. Frequency tabulations were constructed ng/ml, 285 ±129 ng/ml and 8,548 ±3,112 ng h/ml) for the incidence of AEs and laboratory abnormalities. were compared with those obtained in previously performed Phase I studies (TMC114-C138 [15] and Results TMC114-C150 [16] see Table 2; and interaction trials between DRV/r and, respectively, saquinavir In total, 19 healthy women were assigned to treatment. and digoxin). Overall, there is no indication that the Eleven participants completed both sessions and 8 presence of EE and NE had an influence on DRV or participants prematurely discontinued the study: 3 RTV pharmacokinetics.

Antiviral Therapy 13:4 565 Sekar 11/6/08 16:41 Page 566

VJ Sekar et al.

Safety, tolerability and pharmacodynamic assessments (reported by 3 participants), and headache, diarrhoea No serious adverse events (SAEs) or grade 3 or 4 AEs and nausea (all reported by 2 participants). AEs were reported. Nine participants (47%) reported at reported by 1 participant only were vomiting, drug least one AE during the trial, all during treatment with hypersensitivity, pain in extremity and papular rash. EE/NE co-administered with DRV/r 600 mg/100 mg Five participants prematurely discontinued treatment twice daily No AEs were reported when EE/NE was due to an AE (all during session 2 of Panel II). All 5 administered alone. AEs reported in more than 1 partic- participants discontinued the trial due to grade 2 cuta- ipant during combined treatment were drug eruption neous events, as required per protocol (3 participants

Figure 1. Linear plasma concentration–time curves of ethinyl estradiol and norethindrone in the absence and presence of ritonavir-boosted darunavir

A B Session 1: EE plus NE alone Session 1: EE plus NE alone Session 2: EE plus NE with DRV/r Session 2: EE plus NE with DRV/r

140 25 120 20 100 80 15 60 10 40 5

ethinyl estradiol, pg/ml 20 ethinyl estradiol, pg/ml Plasma concentration of 0 Plasma concentration of 0 0 4 8 12 16 20 24 0 4 8 12 16 20 24 Time, h Time, h

(A) Linear mean (±SD) plasma concentration–time curves of ethinyl estradiol (EE) on day 14 following oral administration of EE and norethindrone (NE) alone (session 1) and in combination with ritonavir-boosted darunavir (DRV/r) 600 mg/100 mg twice daily (session 2). (B) Linear mean (±SD) plasma concentration–time curves of NE on day 14 following oral administration of EE and NE alone (session 1) and in combination with DRV/r 600 mg/100 mg twice daily (session 2).

Table 1. Pharmacokinetic results for ethinyl estradiol and norethindrone after oral administration alone (session 1; day 14) and in combination with ritonavir-boosted darunavir 600 mg/100 mg twice daily (session 2; day 14) Parameter EE and NE EE and NE plus DRV/r LS means ratio (90% CI)

Ethinyl estradiol Participants, n 13 11 –

Median Tmax, h (range) 2.0 (1.0–4.0) 3.0 (1.0–4.0) –

Mean C0h, pg/ml (±SD) 23.66 (±11.91) 9.628 (±4.683) –

Mean Cmin, pg/ml (±SD) 22.27 (±11.11) 8.082 (±3.856) 0.38 (0.27–0.53)

Mean Cmax, pg/ml (±SD) 104.7 (±29.01) 73.74 (±16.96) 0.68 (0.61–0.74)

Mean AUC24h, pg h/ml (±SD) 1,095 (±400.4) 608.7 (±151.0) 0.56 (0.50–0.63) Norethindrone Participants, n 13 11 –

Median Tmax, h (range) 2.0 (1.0–3.0) 2.0 (1.0–4.0) –

Mean C0h, ng/ml (±SD) 2.381 (±1.254) 1.578 (±0.9786) –

Mean Cmin, ng/ml (±SD) 2.241 (±1.267) 1.419 (±0.9034) 0.70 (0.51–0.97)

Mean Cmax, ng/ml (±SD) 17.16 (±4.718) 15.36 (±3.845) 0.90 (0.83–0.97)

Mean AUC24h, ng h/ml (±SD) 144.2 (±54.57) 112.5 (±33.73) 0.86 (0.75–0.98)

AUC, area under plasma concentration–time curve; C0h, pre-dose plasma concentration; Cmin, minimum plasma concentration; Cmax, maximum plasma concentration; CI, confidence interval; DRV/r, darunavir with low-dose ritonavir; EE, ethinyl estradiol; LS, least square; NE, norethindrone; Tmax, time to maximum plasma concentration.

566 © 2008 International Medical Press Sekar 11/6/08 16:41 Page 567

Pharmacokinetic interaction of oral contraceptives and darunavir/r

had drug eruption [2 of these participants used hydro- small. There were no clinically relevant changes in cortisone butyrate propionate as concomitant vital sign or ECG parameters at any time point during medication]; 1 participant had drug hypersensitivity; any treatment and no AEs related to vital signs or and 1 participant had papular rash). All other AEs were ECG parameters were reported. grade 1. With the exception of one unrelated AE, all AEs were considered at least possibly related to DRV/r. Discussion No clinically relevant changes over time in laboratory parameters were noted. Except for increased low-density The main pathways of metabolism for OCs, including lipoprotein (reported in 3 participants during combined EE and NE, occur via the cytochrome P450 enzyme treatment: grade 3 in 1 participant and grade 1 in 2 system, predominantly in the and participants), all other treatment-emergent abnormalities , and through glucuronidation. In particular, during combined treatment were reported in 1 or 2 CYP3A4 is the dominant enzyme involved in the metab- participants and were grade 1 or 2 in severity. olism of OCs [7]. There is potential for significant For pharmacodynamic assessments, FSH and LH drug–drug interactions in the use of OCs and antiretro- concentrations decreased versus pre-dose after 14 days of viral medications, because NNRTIs and PIs also treatment with EE/NE, as expected. The median decreases undergo metabolism via the CYP3A4 enzyme [5,6]. were less when DRV/r was co-administered (Table 3). NRTIs do not undergo metabolism via the cytochrome The fluctuations in median vital sign and ECG P450 system, therefore, there is less of a concern for parameter values noted during the trial were generally drug–drug interactions between OCs and NRTIs [5].

Table 2. Comparison of darunavir and ritonavir PK parameters obtained in this study with historical data Clinical study* TMC114-C131 TMC114-C138 TMC114-C150 DRV/r plus EE and NE DRV/r alone† DRV/r with digoxin

PK darunavir 600 mg twice daily Participants, n 11 14 14‡

Mean Cmin, ng/ml (±SD) 2,594 (±1,090) 3,017 (±1,064) 3,426 (±900)

Mean Cmax, ng/ml (±SD) 8,073 (±988) 7,283 (±2,030) 7,729 (±1,072)

Mean AUC12h, ng h/ml (±SD) 55,920 (±11,290) 56,288 (±16,395) 63,830 (±10,830) PK ritonavir 100 mg twice daily Participants, n 11 14 14§

Mean Cmin, ng/ml (±SD) 285 (±129) 221 (±87.9) 372 (±282)

Mean Cmax, ng/ml (±SD) 1,780 (±864) 1009 (±468) 1,436 (±682)

Mean AUC12h, ng h/ml (±SD) 8,548 (±3,112) 6,362 (±3,147) 8,582 (±4,486)

*C131, this study (that is, ritonavir-boosted darunavir [DRV/r] and oral contraceptive study); C138, trial number for the DRV/r saquinavir interaction study; C150, trial † ‡ § number for the DRV/r digoxin study. 400 mg darunavir administered, pharmacokinetic (PK) parameters were dose-normalized to 600 mg. n=13 for AUC12h. n=13 for AUC12h. AUC12h, area under plasma concentration–time curve after 12 h; Cmin, minimum plasma concentration; Cmax, maximum plasma concentration.

Table 3. Median values and changes from reference for contraceptive efficacy parameters EE and NE alone EE and NE plus DRV/r Day 1 pre-dose Day 14 pre-dose Day 1 pre-dose Day 14 pre-dose Actual Actual Changes Actual Actual Changes Parameter values (n=14) values (n=13) versus day 1 (n=13) values (n=16) values (n=12) versus day 1 (n=12)

Median 17-OH-progesterone, nmol 3.0 2.1 1.9 2.0 2.0 0.1 (range) (1.1–46.1) (0.5–26.2) (–42.4–23.2) (1.1–43.7) (0.1–13.7) (–42.3–10.5) Median FSH, U/l 6.1 1.6 3.0 5.4 4.0 1.8 (range) (1.8–8.3) (0.9–6.1) (–6.1–3.2) (1.5–13.5) (1.5–6.5) (–9.5–2.9) Median LH, U/l 5.0 4.1 2.4 4.8 5.8 1.4 (range) (1.2–13.7) (0.5–9.5) (–11.3–6.7) (0.7–23.4) (1.0–9.6) (–15.8–6.0)

Reference: day 1 pre-dose in the same session as the reassessments. n, number of participants at the scheduled timepoints with available data; , decrease. FSH, follicle-stimulating hormone; LH, lutenising hormone.

Antiviral Therapy 13:4 567 Sekar 11/6/08 16:41 Page 568

VJ Sekar et al.

Like all PIs, DRV is a substrate for CYP3A4 HIV-infected women taking DRV/r and OCs: 12 TITAN metabolism [8]. DRV is co-administered with low- patients (4%) and 31 ARTEMIS patients (9%) [19]. dose RTV, because DRV/r has an improved However, in the ARTEMIS trial, for example, rash (all pharmacokinetic profile compared with DRV alone types) was observed with a comparable incidence in HIV- [8]. RTV is a potent inhibitor of both CYP3A4 and infected men (14%) and women (15%) (unpublished CYP2C9 and is also an inducer of both CYP3A4 and data). There have been very few publications on the inci- glucuronidation [9]. Therefore, drug–drug interac- dence of rash in women receiving other PIs and OCs, tions between OCs and DRV/r are likely. The present although it has been suggested that women receiving study was designed to investigate the effect of oestrogen and TPV/r could have an increased risk of non- multiple doses of DRV co-administered with low- serious rash [20]. The incidence of laboratory, vital signs dose RTV (DRV/r 600 mg/100 mg twice daily) on the and ECG abnormalities was generally low. No labora- steady-state pharmacokinetics of EE and NE. Steady- tory, vital signs or ECG abnormalities were reported as state plasma concentrations of EE, NE, DRV and AEs. None of the women presented dysfunctional RTV were achieved prior to full pharmacokinetic bleeding during the trial. Concentrations of FSH and LH blood sampling on day 14 of combined treatment. decreased between pre-dose at day 1 and day 14 in In the presence of DRV/r and based on the ratio of patients receiving EE/NE alone and also in those receiving

LS means, Cmin, Cmax and AUC24h of EE decreased by EE/NE and DRV/r, indicating reliable intake of OCs via a

62%, 32% and 44%, respectively, and Cmin, Cmax and negative feedback loop and as demonstrated elsewhere

the AUC24h of NE decreased by 30%, 10% and 14%, [21]. Comparison of FSH and LH between those respectively, compared with administration of EE/NE receiving EE/NE and those receiving EE/NE and DRV/r alone. This pharmacokinetic interaction is considered revealed a decrease in these hormone levels. This change to be clinically relevant as EE concentrations are is thought to be attributed to metabolic enzyme induction considerably reduced when DRV/r is co-administered by RTV, which in turn increases the metabolism of OCs with Ortho-Novum® (the commercially available and lessens the extent of the negative feedback combined form of EE/NE). mechanism responsible for the secretion of FSH and LH. The effect of DRV/r on the pharmacokinetics of EE In conclusion, the results of this trial demonstrated and NE is consistent with what has been observed for that plasma exposures of EE and NE were decreased other RTV-boosted PIs. For example, an interaction when DRV/r 600 mg/100 mg twice daily was co-admin- study between LPV/r 400 mg/100 mg twice daily and istered. These decreases in EE and NE exposure are EE/NE showed that for EE the AUC decreased by 42%, considered clinically relevant, as they could be associ-

Cmax decreased by 41% and Cmin decreased by 58% ated with an increased risk of pregnancy in HIV-infected upon co-administration, whereas for NE the AUC women of childbearing age. Alternative or additional

decreased by 17%, Cmax decreased by 16% and Cmin contraceptive measures are recommended when decreased by 32% [17]. The observed decrease in the oestrogen-based contraceptives are co-administered exposure to EE and NE in this study may be a result of with DRV/r. metabolic enzyme induction by RTV [18]. The clinical relevance of these decreases depends on their effects on Acknowledgements the hormone levels of FSH and LH, as discussed below. The presence of EE and NE did not seem to have a Some data in this article was previously been presented clinically relevant influence on the pharmacokinetics of at the 46th Interscience Conference on Antimicrobial DRV or RTV, as the PK parameters of both compounds Agents and Chemotherapy, September 27–30, 2006, were generally comparable to those observed in other San Francisco, CA, USA. trials with DRV/r in healthy volunteers. Owing to AES, 5 out of the 19 healthy female Disclosure statement participants (26%) discontinued treatment with DRV/r 600 mg/100 mg twice daily in combination with EE This study was financially supported by Tibotec and NE. These discontinuations were attributable to Pharmaceuticals Inc. and all authors are employees grade 2 cutaneous events, as required per protocol. of Tibotec. Although results of other Phase I (pooled Phase I repeated dose trials) trials with DRV/r performed in References healthy volunteers showed that women were more 1. United Nations, Department of Economics and Social likely to develop rash-related events than men [19], this Affairs, Population Division. World Contraceptive Use observation was not found in HIV-infected participants 2005. (Accessed 30 May 2008). Available at: http://www.un.org/esa/population/publications/contracepti participating in large-scale clinical trials [8]. In Phase ve2005/2005_World_Contraceptive_files/WallChart_WCU III trials of DRV/r, there was a small proportion of 2005.pdf

568 © 2008 International Medical Press Sekar 11/6/08 16:41 Page 569

Pharmacokinetic interaction of oral contraceptives and darunavir/r

2. Hatcher RA, Nelson A. Combined hormonal contraceptive 12. Bouche MP, Michielsen L, Piot M, Timmerman P. Swift methods. In Contraceptive Technology, 18th revised and simultaneous determination of darunavir (TMC114) edition. Edited by Hatcher RA, Robert A. New York: and ritonavir in human plasma using LC-MS/MS. 17th Ardent Media 2004; pp. 391–460. International. Mass Spectrometry Conference. 27 August 3. Hammer SM, Saag MS, Schechter M, et al. Treatment for to 1 September 2006, Prague, Czech Republic. Abstract adult HIV infection: 2006 recommendations of the TuP-042. International AIDS Society-USA Panel. JAMA 2006; 13. Division of AIDS Table for Grading the Severity of Adult 296:827–843. and Pediatric Adverse Events Publish date: December, 4. Pozniak A, Gazzard B, Anderson J, et al. British HIV 2004 Association (BHIVA) guidelines for the treatment of HIV- infected adults with antiretroviral therapy. HIV Med 2003; 14. Koch G. The use of nonparametric methods in the 4 Suppl 1:1–41. statistical analysis of the two-period change-over design. Biometrics 1972; 28:577–584. 5. DHHS Panel on Antiretroviral Guidelines for Adults and Adolescents – a Working Group of the Office of AIDS 15. Sekar V, Lefebvre E, Mariën K, et al. Pharmacokinetic Research Advisory Council (OARAC). Guidelines for the interaction between the HIV protease inhibitors TMC114 Use of Antiretroviral Agents in HIV-1-Infected Adults and and saquinavir, in the presence of low-dose ritonavir. Adolescents, October 10, 2006; 1-113. (Accessed 6 44th Annual Meeting of the Infectious Diseases Society September 2007). Available at: http://aidsinfo.nih.gov/ of America. October 12–15, 2006, Toronto, Ontario, 6. de Maat MMR., Ekhart GC, Huitema ADR, Koks CHW, Canada. Abstract 959. Mulder JW, Beijnen JH. Drug interactions between 16. Sekar V, El Malt M, De Paepe E, et al. Effect of the HIV antiretroviral and co-medicated agents. Clin Pharmacokinet 2003; 42:223–282. protease inhibitor TMC114, coadministered with low- dose ritonavir, on the pharmacokinetics of digoxin in 7. Zhang H, Cui D, Wang B, et al. Pharmacokinetic drug healthy volunteers. American Society for Clinical interactions involving 17alpha-. Clin Pharmacology and Therapeutics Annual Meeting. March Pharmacokinet 2007; 46:133–157. 21–24 2007, Anaheim, CA, USA. Abstract PII 104. 8. PREZISTA™ (darunavir). US Prescribing Information. 17. Kaletra® (NDC 0074-3959-77) Product Information. October 2006. Tibotec Inc., Yardley, PA, USA. (Accessed 6 Abbott Laboratories, North Chicago USA. (Accessed 6 September, 2007). Available at: http://www.tibotectherapeutics.com/ September 2007; Revised January 2007). Available at: http://www.rxabbott.com/pdf/kaletrapi.pdf 9. Norvir® (ritonavir). US Prescribing Information, January 2006. Abbott Laboratories., North Chicago, IL, USA. 18. Ouellet D, Hsu A, Qian J, et al. Effect of ritonavir on the pharmacokinetics of ethinyl oestradiol in healthy female 10. De Meyer S, Azijn H, Surleraux D, et al. TMC114, a novel volunteers. Br J Clin Pharmacol 1998; 46:111–116. human immunodeficiency virus type 1 protease inhibitor active against protease inhibitor-resistant viruses, including 19. Aptivus® (tipranavir). US Prescribing Information, 2007. a broad range of clinical isolates. Antimicrob Agents Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Chemother 2005; 49:2314–2321. CT, USA. 11. United States, Department of Veterans Affairs. National 20. Fröhlich M, Burhenne J, Martin-Facklam M et al. Oral HIV/AIDS Program. Antiretroviral Medications and Oral contraception does not alter single dose saquinavir Contraceptive Agents, July 2006; updated July 2007. pharmacokinetics in women. Br J Clin Pharmacol 2004; Available at: http://www.hiv.va.gov/vahiv?page=cm- 57:244–252. 307_women (Accessed 6 September 2007). Accepted for publication 3 March 2008

Antiviral Therapy 13:4 569