Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2008 Sep 29;52(12):4300–4307. doi: 10.1128/AAC.00363-08

Pharmacokinetic and Safety Evaluation of BILR 355, a Second-Generation Nonnucleoside Reverse Transcriptase Inhibitor, in Healthy Volunteers

Fenglei Huang 1,*, Michael Koenen-Bergmann 2, Thomas R MacGregor 1, Arne Ring 2, Susan Hattox 1, Patrick Robinson 1
PMCID: PMC2592860  PMID: 18824608

Abstract

BILR 355 is a second-generation nonnucleoside reverse transcriptase inhibitor (NNRTI) under clinical development for the treatment of human immunodeficiency virus infection, particularly in those who harbor virus resistant to the currently available NNRTIs. Two single-center, double-blinded, placebo-controlled, parallel dose-escalation studies were conducted to evaluate the pharmacokinetics and safety of oral BILR 355 administration alone and after coadministration with ritonavir (RTV) at 100 mg. Following a single dose of BILR 355 in oral solution, the mean half life (t1/2) was 2 to 4 h, with peak concentrations occurring at 0.5 to 1 h postadministration. The mean apparent clearance (CL/F) ranged from 79.2 to 246 liters/h for administered doses of 12.5 mg to 100 mg. This observed nonlinearity in CL/F resulted from the increased bioavailability attributed to a saturated absorption and/or elimination process at higher doses. In contrast, after the coadministration of single doses of 5 mg to 87.5 mg of BILR 355 with RTV, the mean CL/F ranged from 5.88 to 8.47 liters/h. Over the dose range (5 to 87.5 mg) studied, systemic BILR 355 exposures were approximately proportional to the doses administered when they were coadministered with RTV. With RTV coadministration, the mean t1/2 increased to 10 to 16 h, and the mean time of the maximum concentration in plasma lengthened to 1.5 to 5 h. Compared to the values for BILR 355 given alone, the mean area under the concentration-time curve from time zero to infinity, the maximum concentration in plasma, and the t1/2 of BILR 355 achieved after coadministration with RTV increased 15- to 30-fold, 2- to 5-fold, and 3- to 5-fold, respectively. In both studies, BILR 355 appeared to be safe and well tolerated in healthy volunteers when the outcomes in the treated volunteers were compared with those in the placebo group.


Since the introduction of highly active antiretroviral therapy in the mid-1990s, there have been marked reductions in the rates of mortality and morbidity among individuals infected with human immunodeficiency virus (HIV) (19), and HIV infection is now considered a manageable chronic disease (14). However, the emergence of viral resistance to the approved therapies, mainly due to suboptimal drug potency and pharmacokinetics (PK), poor adherence to complex highly active antiretroviral therapy regimens, and/or compromised immune function, has challenged the early success of treatment of HIV infection by severely limiting the treatment options available (8). Viruses are particularly sensitive to the development of resistance to members of the first generation of nonnucleoside reverse transcriptase inhibitors (NNRTIs) because of their low genetic barrier to the development of drug-resistant reverse transcriptase (RT) variants. Mutant viruses resistant to one member of the current NNRTIs are also usually cross-resistant to the whole class. Thus, patients may be left without a treatment option among this class of compounds (1, 2). Recent surveys of the status of HIV infection alarmingly revealed that the prevalence of drug-resistant variants, particularly those resistant to NNRTIs in newly infected (treatment-naïve) patients, has increased significantly in the past few years (3, 11, 18), suggesting that the resistance issue is even more critical than before. Therefore, there is an urgent need to develop new antiretroviral therapies that combine potency against resistant and wild-type viruses with safety in a convenient dosing regimen.

BILR 355 {11-ethyl-5,11-dihydro-5-methyl-8-[2-(1-oxido-4-quinolinyl)oxy]ethyl)-6H-dipyrido[3,2-b:2′,3′-e][1,4]dizepin-6- one; molecular weight, 477.52} is a second-generation NNRTI under development for the treatment of HIV infections, particularly those caused by strains resistant to current NNRTIs, in adults and children (5, 22). BILR 355 is highly specific toward HIV type 1 (HIV-1) RT and exhibits activity toward HIV-1 RT several magnitudes greater than that toward than other viral and mammalian polymerases, including β-human polymerase, γ-human polymerase, and hepatitis C virus polymerase. The in vitro data show that the 50% effective concentration of BILR 355 against wild-type HIV-1 is 0.26 ng/ml, while the 50% effective concentrations against common NNRTI-resistant viruses range from 1.5 to 13 ng/ml. BILR 355 exhibits an attractive resistance profile against both laboratory strains and clinical isolates of HIV-1. In vitro data indicate that BILR 355 exhibits a low level of cross-resistance to a broad spectrum of viruses that are highly resistant to nevirapine, efavirenz, and delavirdine (5).

Single-dose PK studies with several animal species have indicated that BILR 355 has a short half-life (t1/2; <2 h) and volume of distribution (V; 1 to 2 liters/kg). In preclinical studies, the absolute bioavailability of BILR 355 varied widely among animal species, from 4.2% in the rhesus monkey to 100% in the rat. The plasma protein binding for BILR 355 is 95.8% (4).

The purposes of the two studies described here were to assess the safety, tolerability, and preliminary PKs (including a preliminary assessment of the dose linearity) of BILR 355 after the administration of single ascending oral doses alone or in combination with ritonavir (RTV) at 100 mg.

MATERIALS AND METHODS

Subjects.

The studies were conducted at the Human Pharmacology Centre of Boehringer Ingelheim Pharma KG in Ingelheim, Germany, following the provision of approval by the local ethics committee. The study was conducted by following the guidance of the Declaration of Helsinki. After signing the written inform consent, the healthy male volunteers (age range, 21 to 50 years; body mass index range, 18.5 to 29.9 kg/m2) entered into the studies. The subjects were in generally good health, as judged by the medical history, a physical examination, and clinical laboratory data. Clinically abnormal laboratory results, evidence of existing diseases or disorders, or any observations or conditions (e.g., smoking of more than 10 cigarettes per day, excessive consumption of alcohol, and drug abuse) which might interfere with the PKs of the study drug were reasons for exclusion. The subjects abstained from all grapefruit products and other foods or beverages that might affect enzyme function. The subjects could be withdrawn from the study at any time due to the occurrence of adverse events (AEs), inclusion or exclusion criteria violations and a failure to report for the study, withdrawal of consent, smoking, and/or the onset of illness. Volunteers dropped from the trial were not replaced unless the number of volunteers in a dose group was less than six.

Study design.

Two studies with similar designs were conducted to evaluate the safety and PKs of BILR 355 after BILR 355 was administered alone and after it was coadministered with RTV.

BILR 355 alone in single ascending doses.

The single-center, randomized, double-blind, placebo-controlled, single dose-escalation (1, 5, 12.5, 25, 50, 75, and 100 mg) study of BILR 355 alone was designed to evaluate the PKs of BILR 355 and document the safety and tolerability of BILR 355 in healthy male volunteers. For each dose level, six volunteers received active drug dissolved in a polyethylene glycol (PEG) solution and two received a placebo PEG control. One dose level was tested within each group. The subject groups were dosed and evaluated sequentially, beginning with the lowest-dose group and proceeding stepwise to the highest-dose group. The next higher dose was administered only if no safety concerns arose during treatment of the preceding group. The placebo or BILR 355 in PEG solution was administered with 200 ml of tap water on the morning of each study day after an overnight fast; a standard breakfast was served 60 to 90 min after study drug administration. BILR 355 did not change the taste of the PEG solution.

BILR 355 coadministered with RTV in single ascending doses.

The study of BILR 355 coadministered with RTV was similar to the single-ascending-dose study, except that each dose group received one dose of RTV at 100 mg 10 h prior to the administration of BILR 355 and a second dose of RTV at 100 mg with the BILR 355 dose on the morning of the study day (day 1). Additionally, the effect of food on the PKs of BILR 355 in PEG solution was assessed in the study cohort that received the 50-mg dose of BILR 355, after completion of the study with the highest-dose (87.5 mg) group. The subjects in this fed cohort were given a high-fat breakfast 30 min before drug administration. The recipe of the high-fat meal was obtained from FDA guidance for food-effect bioavailability and fed bioequivalence studies. This meal derived approximately 150, 250, and 500 to 600 cal from proteins, carbohydrate, and fat, respectively.

Sampling and analytical methods. (i) Blood sampling.

To study the effects of a single dose of BILR 355 administered alone, blood samples were taken predosing (0 h) and at 0.5, 1, 2, 4, 6, 8, 10, 12, 24, 32, 48, 72, 96, 120, and 144 h after drug administration. For dose groups receiving 100 mg or more of BILR 355 alone or BILR 355 with RTV, blood samples were drawn before drug administration on day 1 (predosing) and at 0.25, 0.50, 0.75, 1, 2, 4, 6, 8, 10, 12, 24, 32, 48, 96, and 144 h postdosing. The total amount of blood collected from any individual in each study during the trial was approximately 300 ml. At each time point, about 9 ml of blood was drawn into evacuated collection tubes containing EDTA anticoagulant and labeled with sample identification information. For the preparation of plasma, the samples were centrifuged within 60 min after blood sampling at 4°C (refrigerator) and 2,000 × g (equivalent to 3,000 rpm) for 10 min. Two aliquots of plasma (about 2 ml in polypropylene tubes) were available for the drug assays. The plasma was stored in individually labeled polypropylene tubes at <−18°C (sample stability data support the stability of the drug for up to 1 year of storage under this condition).

(ii) Urine sampling.

Urine was collected quantitatively by weight. Two 10-ml aliquots of urine were stored at −20°C until analysis (stability data support the stability of the drug when it is stored for up to 1 year under this condition). Urine was collected from 0 to 72 h after drug administration over the following collection intervals: prior to drug administration (blank sample) and at 0 to 6 h, 6 to 24 h, and 24 to 72 h following drug administration. The bladder was voided before the beginning of each collection interval.

(iii) Bioanalytical method.

A validated high-pressure liquid chromatography-tandem mass spectrometry/mass spectrometry method was used for the assay of plasma and urine samples. An aliquot of human plasma or urine containing BILR 355 with an internal standard was extracted by a protein precipitation/online switch-valve procedure. The extracted samples were analyzed with a high-pressure liquid chromatograph equipped with a PE Sciex API 3000 mass spectrometer. Positive ions were monitored in the selected reaction-monitoring mode. Quantification was performed by use of the peak area ratio. The calibration range was 1.99 ng/ml to 497.00 ng/ml. The lower limit of quantification was 2 ng/ml for both urine and plasma samples. For plasma, the between-batch precision and accuracy were 5.3% to 5.5% and 95.8% to 98.0%, respectively; the within-batch precision and accuracy were 1.9% to 9.3% and 91.1% to 100.3%, respectively. For urine, the between-batch precision and accuracy were 2.7% to 5.4% and 110.5% to 113.7%, respectively; the within-batch precision and accuracy were 2.1% to 4.6% and 103.2% to 106.4%, respectively.

PK analyses.

Plasma BILR 355 concentration-time data were analyzed by a noncompartmental approach with the WinNonlin program (version 4.01; Cary, NC). Standard formulas for noncompartmental PK analysis provided by WinNonlin were used to determine the maximum concentration of drug in serum (Cmax), the time to Cmax (Tmax), oral clearance (CL/F), mean residence time after oral administration (MRTp.o.), and the apparent V in the terminal phase (Vz/F). The area under the plasma concentration-time curve (AUC) was calculated by using a linear up/log down trapezoidal algorithm. The last predicted concentration was used for extrapolation of the AUC and the area under the first moment of the concentration-time curve. The V at steady state (Vss) was calculated as CL/F·(MRTp.o. − 1/Ka), where Ka (the absorption rate constant) was determined by compartmental modeling of the data. The renal clearance (CLR) was calculated as the ratio of the amount of unchanged drug excreted in urine (Ae) over a time interval to the AUC over the same time interval. The fraction excreted as unchanged drug in urine (fe) was calculated as the percentage of Ae compared to the administered dose. The plasma concentrations below the lower limit of quantification were ignored during the PK analysis and calculation of the mean plasma concentration, unless they were in the lag phase between the predose sampling time (0 h) and the time of the first measurable concentration, which were treated as 0. For a scheduled sampling time point of any treatment group, if less than two-thirds of the subjects had concentrations above the lower limit of quantification, then the mean concentration of this time point was not presented in the mean plasma-time plots.

Statistical analysis.

The power model was used to analyze dose proportionality in terms of the AUC from time zero to infinity (AUC0-∞) and Cmax. The model is described by the following equation: Ykm = α·Dkβ·ekm, where Ykm is the response (AUC0-∞, Cmax) measured for subject m receiving dose k; α is the intercept; β is the slope; Dk is the effect of the kth dose, where k is equal to 1, 2,…; and ekm is the random error associated with the mth subject who received dose k. Logarithmic transformation yields the linear regression equation, as follows: ln Ykm = ln α + β·ln Dk + ln ekm. Dose linearity requires that β equal 1. Statistical analysis was performed with SAS program (version 8.2, Cary, NC).

RESULTS

Subjects.

A total of 54 eligible subjects participated in the study of single ascending doses of BILR 355 given alone. The PK evaluation included 40 subjects treated with active drug. The median age, weight, and height of the volunteers were 31 years (range, 21 to 48 years), 79 kg (range, 60 to 106 kg), and 179 cm (range, 169 to 195 cm), respectively. There were no major differences in demographic or baseline data among the subjects in the different treatment groups (including the placebo group).

Sixty-two subjects entered the study of single ascending doses of BILR 355 coadministered with RTV; all subjects were treated and completed the planned assessments according to the protocol. In all, eight dose groups were studied. In all dose groups except the 5-mg and 62.5-mg dose groups, eight subjects were treated. Only seven subjects were treated in the 5-mg and 62.5-mg groups because of screening failures. The median age, weight, and height of the subjects were 33 years (range, 21 to 49 years), 77 kg (range, 63 to 102 kg), and 178 cm (range, 168 to 195 cm), respectively. There were no major differences in the demographic or baseline data between treatment groups in this study.

Safety and tolerability.

The AEs are summarized in Tables 1 and 2 for BILR 355 alone and BILR 355 plus RTV treatment, respectively. For both studies, all AEs were, overall, of mild to moderate intensity and transient in nature, and in the majority of cases they resolved completely without treatment. There were no severe or serious AEs, and no subject withdrew from the trial because of the occurrence of an AE. The AEs were almost evenly distributed over all treatment groups, including the placebo group (or the group receiving placebo plus RTV), with a slightly higher incidence of six AEs in four subjects (66.7%) in the 75-mg group of BILR 355 alone in the dose-escalation study. There was no evidence of any clinically relevant, drug-related changes according to physical examination, skin inspection, vital signs, neurological assessment, and testing for occult blood in both studies. Additionally, no clinically relevant electrocardiographic changes were detected in either study. No drug- or dose-related changes were observed in the global tolerability assessment. Six days after dosing of BILR 355, one subject (16.7%) in the 5-mg dose group in the study of BILR 355 alone had an increase in the alanine aminotransferase level of more than double the normal upper limit. It was considered unlikely to have been due to the BILR 355 treatment.

TABLE 1.

AEs observed after administration of single oral ascending doses of BILR 355 or placebo by treatment

AE No. (%) of subjects with AEs or no. (%) of AEs observed after administration of:
BILR 1 mg BILR 5 mg BILR 12.5 mg BILR 25 mg BILR 50 mg BILR 75 mg BILR 100 mg Placebo
Subjects with at least one AE 1 (16.7) 2 (33.3) 1 (20.0) 3 (50.0) 1 (16.7) 4 (66.7) 1 (20.0) 2 (14.3)
Fatigue 1 (16.7) 0 1 (20.0) 0 1 (16.7) 1 (16.7) 1 (20.0) 0
Headache 0 0 0 1 (16.7) 1 (16.7) 0 0 1 (7.1)
Abdominal pain 0 0 0 1 (16.7) 0 1 (16.7) 0 0
Nause 0 0 0 0 0 0 0 1 (7.1)
Vomiting 0 0 0 0 0 0 0 1 (7.1)
Migraine 0 0 0 0 0 0 0 1 (7.1)
Increased alanine aminotransferase level 0 1 (16.7) 0 0 0 0 0 0
Maculopapular rash 0 0 0 1 (16.7) 0 0 0 0
Constipation 0 0 0 1 (16.7) 0 0 0 0
Neurological eyelid disorder 0 0 0 0 0 1 (16.7) 0 0
Flatulence 0 0 0 0 0 1 (16.7) 0 0
Palpitations 0 0 0 0 0 1 (16.7) 0 0
Herpes simplex 0 1 (16.7) 0 0 0 0 0 0
Frequent bowel movements 0 0 0 0 0 1 (16.7) 0 0

TABLE 2.

AEs observed after single oral ascending doses of BILR 355 or placebo coadministered with 100 mg RTV by treatment

AE No. (%) of subjects with AEs or no. (%) of AEs observed after administration of:
BILR 5 mg BILR 12.5 mg BILR 25 mg BILR 37.5 mg BILR 50 mg BILR 62.5 mg BILR 75 mg BILR 87.5 mg BILR 50 mg (fed) RTV only
Subjects with at least one AE 1 (14.3) 0 1 (16.7) 1 (16.7) 0 0 0 2 (33.3) 1 (16.7) 1 (8.3)
Aphthous stomatitis 1 (14.3) 0 1 (16.7) 0 0 0 0 0 0 0
Diarrhea 0 0 0 0 0 0 0 1 (16.7) 0 1 (8.3)
Fatigue 0 0 0 0 0 0 0 0 1 (16.7) 0
Maculopapular rash 0 0 1 (16.7) 1 (16.7) 0 0 0 1 (16.7) 0 0

Pharmacokinetics.

The mean plasma concentration-time profiles for BILR 355 after oral administration of single doses of BILR 355 alone and coadministration of 100 mg RTV are depicted in Fig. 1 and Fig. 2, respectively; the mean (median and percent coefficient of variation [CV]) PK parameters of BILR 355 alone and of BILR 355 coadministered with RTV are summarized in Table 3 and Table 4, respectively. Dose-normalized plots of AUC0-∞ and Cmax versus dose are presented in Fig. 3 and Fig. 4 for BILR 355 alone and BILR 355 coadministered with RTV, respectively. BILR 355 was rapidly absorbed and eliminated following oral administration, with median Tmax values of 0.5 to 2.0 h and mean t1/2 values of 1.86 to 5.02 h. The mean Cmax showed a trend toward more than a dose-proportional increase in relation to the dose. A nonlinear relationship was also observed between the mean exposure (AUC0-∞) and dose (36.3 ng·h/ml at the 5-mg dose to 1,308.4 ng·h/ml at the 100-mg dose). Increasing oral doses led to decreases in CL/F from a mean of 246 liters/h with the 12.5-mg dose to a means of 79.2 liters/h with the 100-mg dose. The very high mean CL/F at the 75-mg dose level was the result of a very large CL/F in a single subject, which might be considered an outlier. Less than 1% of the BILR 355 doses administered were excreted in the urine as unchanged drug. The majority of the urinary excretion of BILR 355 was completed within 6 h after drug administration, and almost all renally excreted BILR 355 was recovered within 24 h postdosing (data not shown). It appears that the fe of BILR 355 increased with an increase in the dose. The median Tmax decreased from 2.0 h to 0.50 h as the dose increased from 5 mg to 100 mg, indicating that Tmax is dose dependent. The mean t1/2 values for the BILR 355 doses of 5, 12.5, 25, 50, 75, and 100 mg administered were 5.0, 4.1, 3.4, 2.5, 2.2, and 1.9 h, respectively; there was a gradual decrease in the t1/2 with an increase in the dose. The mean apparent Vz/F values were 955, 1,425, 1,037, 459, 831, and 213 liters for the BILR 355 dose groups of 5, 12.5, 25, 50, 75, and 100 mg, respectively, indicating a trend of decreasing volume with an increase in the dose. A similar pattern was observed for the apparent Vss/F.

FIG. 1.

FIG. 1.

Mean plasma BILR 355 concentration-time profiles after the administration of single ascending oral doses of BILR 355.

FIG. 2.

FIG. 2.

Mean plasma BILR 355 concentration-time profiles after the administration of single ascending oral doses of BILR 355 with 100 mg RTV.

TABLE 3.

PK parameters for BILR 355 after administration of single oral ascending doses of BILR 355

Dose (mg) Statistic AUC0-∞ (ng·h/ml) AUC0-last (ng·h/ml) Cmax (ng/ml) Tmax (h) t1/2 (h) CL/F (liters/h) MRTp.o. (h) Vz/F (liters) Vss/F (liters) CLR (liters/h) fe (%) % AUC_extrapa
5 Mean 36.3 13.6 4.36 1.50 5.02 149 7.74 956 1020 0.271 0.172 42.3
Median 28.9 16.7 4.20 2.00 4.45 173 6.83 1,110 1,150 0.286 0.130 42.3
CV (%) 37.0 62.8 34.2 47.1 58.3 30.5 51.3 30.7 24.0 47.5 80.8 45.2
12.5 Mean 71.8 56.2 14.8 1.00 4.10 246 6.12 1,430 1,460 0.411 0.212 30.0
Median 49.4 33.5 8.92 1.00 3.23 253 5.18 1,500 1,620 0.363 0.180 29.1
CV (%) 83.9 107 97.3 0.00 46.7 48.6 31.0 54.7 48.7 30.6 58.1 49.3
25 Mean 142 128 41.6 0.833 3.44 204 4.52 1,040 914 0.688 0.405 11.7
Median 123 109 32.7 1.00 3.07 210 4.31 828 846 0.620 0.255 10.0
CV (%) 43.6 49.0 69.9 31.0 40.2 40.0 22.3 62.9 47.7 48.1 78.7 57.9
50 Mean 419 406 164 0.667 2.50 129 3.20 460 390 0.549 0.485 3.09
Median 448 435 174 0.500 2.64 113 3.22 425 383 0.489 0.380 3.20
CV (%) 27.5 27.7 24.4 38.7 10.8 32.2 10.9 30.0 25.3 38.6 61.2 27.4
75 Mean 779 763 348 0.667 2.21 272 2.95 832 608 0.615 0.602 2.98
Median 835 820 349 0.500 2.16 105 2.85 344 282 0.651 0.585 1.83
CV (%) 67.5 67.6 64.1 38.7 11.6 151 12.2 146 133 31.5 71.7 89.3
100 Mean 1,310 1,300 937 0.550 1.86 79.2 2.23 213 163 0.591 0.816 0.941
Median 1,340 1,320 939 0.500 1.89 74.7 2.25 238 176 0.468 0.630 0.934
CV (%) 20.8 20.8 15.1 38.0 12.6 21.3 13.0 24.3 21.6 54.0 68.6 50.8
a

AUC_extrap, extrapolated area for estimation of AUC0-∞.

TABLE 4.

PK parameters of BILR 355 after coadministration of single oral ascending doses of BILR 355 with 100 mg RTV

Dose (mg) Statistic AUC0-∞ (ng·h/ml) AUC0-last (ng·h/ml) Cmax (ng/ml) Tmax (h) t1/2 (h) CL/F (liters/h) MRTp.o. (h) Vz/F (liters) Vss/F (liters) CLR (liters/h) fe (%) % AUC_extrapa
5 Mean 707 615 25.3 5.71 14.7 7.33 24.8 153 166 0.362 4.78 12.7
Median 623 567 22.6 6.00 14.6 8.02 24.5 164 173 0.363 4.57 12.0
CV (%) 22.4 20.7 24.2 42.5 20.8 18.7 14.8 21.6 18.8 20.9 19.2 36.2
12.5 Mean 1,960 1,680 74.1 3.59 15.9 6.70 24.8 149 153 0.438 6.37 13.4
Median 2,030 1,790 80.4 3.00 16.8 6.21 25.3 143 143 0.460 7.00 14.2
CV (%) 22.6 19.8 19.6 67.5 20.9 24.4 14.8 20.7 18.3 37.2 38.1 34.9
25 Mean 4,340 4,140 226 1.75 11.5 5.88 18.1 97.6 102 0.390 6.62 4.41
Median 4,230 4,070 228 1.50 11.8 5.94 18.4 92.5 99.2 0.387 5.89 4.85
CV (%) 16.1 15.0 15.0 71.1 8.48 16.0 5.84 19.4 15.5 29.2 35.7 54.0
37.5 Mean 5,960 5,800 344 1.50 11.0 6.52 16.9 104 105 0.295 4.58 2.89
Median 6,330 6,180 339 1.50 10.6 5.95 17.0 105 104 0.240 4.35 3.19
CV (%) 19.8 21.1 18.3 36.5 17.2 21.5 8.71 27.6 20.6 43.6 41.9 58.3
50 Mean 6,800 6,540 468 0.958 11.5 7.84 16.7 129 125 0.292 3.58 4.08
Median 7,140 6,730 431 0.875 11.3 7.00 16.6 120 113 0.287 3.70 5.15
CV (%) 26.2 27.7 37.8 58.1 6.70 29.2 5.47 27.2 27.6 48.8 35.7 52.0
62.5 Mean 7,630 7,480 653 0.700 9.73 8.47 14.0 118 113 0.430 5.05 1.98
Median 7,790 7,700 579 0.750 9.61 8.03 14.3 110 108 0.401 5.44 1.89
CV (%) 19.9 19.9 24.2 29.9 10.4 21.4 10.2 21.0 16.4 30.8 20.2 57.3
75 Mean 12,300 12,100 812 1.63 11.9 6.35 17.4 108 105 0.351 5.44 1.90
Median 12,100 11,800 817 1.38 11.3 6.18 16.9 103 101 0.347 5.76 1.22
CV (%) 21.2 21.6 15.0 83.6 16.8 22.4 15.2 22.2 16.2 30.1 16.9 91.8
87.5 Mean 11,800 11,700 838 1.50 9.61 8.28 14.6 114 112 0.492 5.96 1.10
Median 11,100 11,000 934 2.00 9.02 8.22 14.8 103 107 0.416 5.91 0.785
CV (%) 35.4 36.1 25.4 51.6 21.5 34.8 14.7 40.8 29.4 46.9 30.5 87.0
50 (fed) Mean 8,300 7,970 430 6.00 11.0 6.26 18.7 100 81.4 0.299 4.62 4.32
Median 8,410 8,120 430 6.00 10.5 6.05 17.3 96.3 77.5 0.289 4.19 4.62
CV (%) 21.1 23.1 16.3 0.00 15.6 21.5 13.3 29.0 18.5 48.1 37.4 65.5
a

AUC_extrap, extrapolated area for estimation of AUC0-∞.

FIG. 3.

FIG. 3.

Plot of dose-normalized (norm) AUC0-∞ (AUC0-inf) and Cmax after the administration of single doses of BILR 355 alone versus the doses.

FIG. 4.

FIG. 4.

Plot of dose-normalized (norm) AUC0-∞ (AUC0-inf) and Cmax after the administration of single doses of BILR 355 with 100 mg RTV versus the doses.

Statistical analyses of the doses versus exposures revealed that over the BILR 355 dose range of 5 to 100 mg, the point estimate and 95% confidence interval (CI) of the regression slopes based on a power model (analysis of covariance) were 1.98 (95% CI, 1.58 to 2.38), 1.58 (95% CI, 1.22 to 1.94), and 1.41 (95% CI, 1.08 to 1.76) for Cmax, AUC from 0 to the last quantifiable plasma concentration (AUC0-last), and AUC0-∞, respectively. As the 95% CI did not contain a value of unity (1.00) for either exposure parameters, a conclusion for dose proportionality could not be determined statistically for the dose range studied.

Following the oral administration of BILR 355 with RTV, BILR 355 was rapidly absorbed and resulted in a median Tmax of 3 to 6 h for subjects given lower dose levels (5 mg and 12.5 mg) and 1 to 2 h for subjects given dose levels of 25 mg and higher (Fig. 2 and Table 4). After the Cmax was reached, the plasma BILR 355 concentration declined at a moderate rate, and the terminal t1/2s were approximately 14 to 16 h and 10 to 12 h for the low dose levels (5 mg and 12.5 mg) and higher dose levels (25 mg and above), respectively. The mean CL/F was relatively constant and ranged from 5.88 to 8.47 liters/h. The plots of the dose-normalized AUC0-∞ and Cmax versus dose (Fig. 4) suggest that increases in the dose of BILR 355 result in approximately linear increases in AUC0-∞ over the dose range of 5 mg to 87.5 mg studied. However, the proportional increases in Cmax with increasing doses were observed only over the dose range of 25 mg to 87.5 mg. Statistical analyses of Cmax and AUC0-∞ versus the doses revealed that over the dose range of 5 mg to 87.5 mg, the 95% CI of the regression slope contained the value of unity only for AUC0-∞ (point estimate, 0.97; 95% CI, 0.90 to 1.04) but not for Cmax (point estimate, 1.27; 95% CI, 1.20 to 1.33), suggesting that dose proportionality is demonstrated only with the extent of exposure for this dose range. In the case of the dose range of 25 mg to 87.5 mg, the 95% CI of the slope attained the value of unity for both AUC0-∞ and Cmax. About 5% of the BILR 355 dose administered was recovered in urine as unchanged drug during the time interval from 0 to 72 h for all dose levels. Table 5 compares the mean PK parameters for BILR 355 after the administration of single doses of BILR 355 with or without RTV boosting. After RTV boosting, the increases in AUC0-∞ ranged from 15-fold (50 mg and 75 mg) to 31-fold (12.5 mg and 25 mg); the mean Cmax increased about 2- to 3-fold and 5- to 6-fold for high (50 and 75 mg) and low (12.5 and 25 mg) dose levels, respectively. The t1/2s of BILR 355 increased from 2 to 4 h (without RTV boosting) to 11 to 16 h (with RTV coadministration).

TABLE 5.

Ratios of mean BILR 355 PK parameters after administration of a single dose of BILR 355 plus RTV compared with that after administration of a single dose of BILR 355 alone

BILR 355 dose (mg) Fold change in PK parameters after administration ofBILR 355 and RTV
AUC Cmax t1/2
12.5 31.0 5.0 3.9
25 31.1 5.4 3.3
50 15.5 2.9 4.6
75 15.0 2.3 5.4

The PK parameters for BILR 355 after the coadministration of BILR 355 at 50 mg with RTV in the fed state are presented in Table 4. The mean t1/2s of BILR 355 were approximately 11 h in subjects receiving treatments in either the fasting or the fed state. The mean Cmax, AUC0-∞, and Tmax values of BILR 355 after administration for subjects in the fed state were 91.9%, 122.0%, and 626.3%, respectively, of the values determined for subjects in the fasted state.

DISCUSSION

The objectives of the two studies described here were (i) to characterize the preliminary PK of BILR 355, a second-generation NNRTI, in healthy male volunteers after the administration of a single oral dose of BILR 355 alone and in combination with low-dose RTV and (ii) to document the safety and tolerability of BILR 355 in these subjects.

In these studies, only male subjects were enrolled because the reproductive toxicological study had not been completed when these studies were initiated, and on the basis of regulatory requirements, no healthy female subjects (with ages comparable to those of the male subjects in the study) were allowed to enroll in the study. It is very important to evaluate the PK in female subjects; this is particularly true when a drug's metabolism is mediated by the cytochrome P450 (CYP) CYP3A4 isozyme (7). We did enroll female subjects in later phase I studies (i.e., drug interaction studies and formulation studies) of BILR 355, and data from those studies will be published at a later date. In the present studies, no heavy smokers (those who smoke <10 cigarettes/day) were allowed to participate in the study; however, during the study, the subjects were not allowed to smoke. Since CYP3A4 is the major enzyme identified to be responsible for the metabolism of BILR 355 and the literature so far contains no data suggesting that smoking has a significant impact on the activity of CYP3A4, it is believed that the enrollment of smokers in this study had a minimal impact on the evaluation of the PK of BILR 355.

For both studies, six subjects received active drugs and two received a placebo. Due to a lack of prior clinical data, it is usually not possible to accurately perform a sample size calculation for a human study conducted for the first time; however, it is generally believed that the sample size selected (six subjects receiving active drugs and two subjects receiving a placebo) is sufficient for the evaluation of the safety and preliminary PK of a new chemical entity (6).

The safety parameters investigated in these two studies included the standard parameters usually used in the first studies to be conducted with humans, as well as other specific tests performed because of the toxicological profiles and experience with the NNRTI class of drugs. With and without coadministration with RTV, BILR 355 displayed an excellent safety profile. Analyses of AEs, laboratory parameters, and other safety data did not raise any concerns over the safety and tolerability of single oral doses of BILR 355. No dose-AE relationship was observed, and BILR 355 did not appear to affect electrocardiographic parameters, including the Q-T interval. No deaths, no severe AEs, and no AEs leading to treatment discontinuation occurred. No drug-related changes in vital signs or laboratory abnormalities were identified.

In the present study, some extreme values for the PK parameters were reported, particularly for those PK parameters obtained after the administration of BILR 355 alone. To better represent the data, both mean and median values are reported in the PK tables (Tables 3 and 4). The percentage of the extrapolated area for estimation of AUC0-∞ was also reported. As showed in Table 3 and 4, the percentages of the extrapolated AUC for 5 mg and 12.5 mg of BILR 355 after BILR 355 was given alone were more than 20%, suggesting a bias in the estimation of AUC0-∞ for these two groups. The analysis of dose proportionality was therefore performed by using both AUC0-∞ and AUC0-last values. The assumption for the power model was checked and confirmed that the required linear relationship between the expected logarithm of the exposures (AUC and Cmax) and the logarithm of the doses was indeed established. The residuals of the fitted analysis of covariance model (power model) on the logarithmic scale were examined and confirmed the normal distribution of the residuals; in addition, no sequence effect was found (data did not show).

By using recombinant CYP enzymes in a metabolism study, BILR 355 was found not to be the substrate of CYP2B6, CYP2D6, CYP1A2, CYP2C9, or CYP2C19; the CYP3A4 isozyme was identified as an isoform of human CYP responsible for the metabolism of BILR 355, with an estimated Km of 377.2 ng/ml obtained when recombinant CYP was used. In a study with human microsomes and specific enzyme inhibitors, CYP3A (CYP3A4/5) was found to be the major isoform of the human CPY3A4 responsible for the metabolism of BILR 355 (Boehringer Ingelheim Pharmaceuticals, Inc., data on file). A preliminary transport study with Caco-2 monolayer cell cultures suggested that BILR 355 is a P-glycoprotein substrate. Given the low Km value for CYP3A4, it is expected that saturated metabolism and/or absorption might occur during a clinical study. Indeed, a pronounced, more than dose-proportional increase in Cmax/AUC0-∞ was observed with increasing single doses of BILR 355 alone, particularly over the dose range of 75 mg to 100 mg (Fig. 1 and Table 3), suggesting saturated absorption and elimination. In addition, a slight increase in the fe of unchanged BILR 355 over the studied dose was observed, indicating a saturation of nonrenal clearance. The saturation of absorption and, possibly, that of elimination after the oral administration of BILR 355 suggest that at least one of the following processes may occur at higher doses: (i) BILR 355 saturated intestinal CPY 3A4/5 and/or other CYP enzymes; (ii) BILR 355 saturated the intestinal transporters, particularly P-glycoprotein; (iii) BILR 355 had a dose-dependent formulation effect; or (iv) BILR 355 saturated the metabolism enzymes (i.e., CYP3A4/5) responsible for BILR 355 elimination. Since after the coadministration of BILR 355 and RTV the nonlinear PKs observed after the administration of BILR 355 alone largely disappeared (Fig. 3 and Fig. 4), even though the same BILR 355 formulation (a powder in a solution for oral consumption) was used, it is hypothesized that the saturated PKs were probably not caused by a formulation effect but were caused by a combination of saturated absorption and elimination processes.

Interestingly, the terminal t1/2 slightly decreased with increasing doses; however, this may be an artifact because of the fewer sampling time points obtained for the low-dose group and the larger number of sampling time points obtained for the high-dose group. A preliminary population PK analysis of these data suggested that the observed change in t1/2 was probably an artifact.

After BILR 355 was given alone, both Vss and Vz/F decreased with increasing doses. This was likely attributed to the fact that the absorption and/or the elimination process was saturated at higher dose, which led to an increase in relative bioavailability (a decrease in the apparent CL/F) with an increase in the dose, since both Vss and Vz/F were related to CL/F and, therefore, the reported values of Vss and Vz/F were probably biased, given the fact that apparent CL/F was changed with the dose administered.

RTV, a mechanism-based CYP3A4/5 inhibitor, inactivates CYP3A by forming a reactive metabolite which irreversibly inactivates cytochrome activity (17). RTV is the most potent inhibitor of CYP3A4 activity of the currently available antiretroviral agents. In addition, RTV is an inhibitor of P-glycoprotein (10, 13). Because of its better safety profile and sufficient inhibitory effects, low-dose RTV (100 or 200 mg once or twice daily) has been widely used as a boosting agent to alter the PK profiles of coadministered antiretroviral agents (20). For example, saquinavir (SQV) and lopinavir (LPV) coadministration with low-dose RTV results in increased plasma concentrations of SQV and LPV, including increased Cmaxs and minimum concentrations of drug in serum (Cmin) and prolonged elimination t1/2s for SQV and LPV (9, 15, 20). The increase in Cmax is possibly the result of the inhibition of first-pass metabolism and/or P-glycoprotein transport by RTV (9, 15, 20). On the other hand, low-dose RTV increased the elimination t1/2s, Cmins, and extent of indinavir and amprenavir bioavailability but had a negligible effect on Cmax, indicating that RTV has little effect on the absorption of these two antiretroviral agents (16, 20).

In the current study, the coadministration of BILR 355 with low-dose RTV resulted in 15- to 30-fold increases in AUC0-∞s, 2- to 5-fold increases in Cmaxs, and 3- to 5-fold increases in elimination t1/2s. The dramatic boosting effects of RTV on the values of AUC0-∞, t1/2, and Cmax for BILR 355 indicate that RTV alters the absorption as well as the metabolism and elimination of BILR 355. The results suggest the impairment of the first-pass metabolism of CYP3A4 (intestinal) and/or the P-glycoprotein transporter, as well as systemic (hepatic) CYP3A4. The prolongation of the BILR 355 t1/2 from 2 to 4 h to 10 to 12 h when BILR 355 is administered with a low dose of RTV may permit a reduction in the dosing frequency required while maintaining the minimum concentration of BILR 355 required for the suppression of HIV replication. An extended dosing interval should improve patient adherence. Prolongation of the t1/2 may also reduce the fluctuations in the steady-state Cmax to Cmin of BILR 355 and thereby potentially improve patient tolerability of the drug.

The plasma concentration of BILR 355 at 12 h postdosing (Cp12h) was also markedly increased after BILR 355 was coadministered with RTV. For example, at the 75-mg dose level, the mean Cp12h was 4.7 ng/ml when BILR 355 was given alone, whereas the mean Cp12h was 343.5 ng/ml after BILR 355 was coadministered with RTV. The increase in Cp12h would likely result in an increase in the Cmin at steady state (Cmin,ss) with a twice-daily dosing regimen. A significant increase in the plasma Cmin,ss for BILR 355 would be expected to increase the rate of virologic success with this agent because high Cmins of NNRTIs have been shown to positively correlate with rapid and sustainable viral suppression (12, 21). However, it should be pointed out that the chronic effects of the coadministration of BILR 355 and RTV need to be further examined in multiple-dosing studies either with healthy volunteers or with patients to confirm the boosting effect of RTV on the PKs of BILR 355 and to obtain more safety data.

Consistent with the belief that metabolism plays a major role in the elimination of BILR 355, the urinary excretion of BILR 355 after the administration of single doses of BILR 355 alone was found to be less than 1% of the administered dose for all dose groups. The urinary excretion of BILR 355 after oral administration of the drug alone gradually increased from 0.172% to 0.816%, a more than fourfold increase, suggesting a saturation of nonrenal clearance; however, since the urinary excretion of BILR 355 was low and the variability (60 to 80%) was quite large, more studies (data) may be needed to confirm this observation. After coadministration with RTV, the urinary recovery of BILR 355 was consistently found to be 5%, suggesting an increase in the systemic availability of BILR 355.

The effect of food on the PKs of BILR 355, after coadministration with RTV was examined with a small number of subjects (n = 6) at the 50-mg dose level. Because of the exploratory nature of the study, a formal inferential statistical analysis was not performed. Comparison of the relative bioavailability of BILR 355 in fed subjects and that in fasting subjects was based on the group arithmetic mean exposure ratios for the fed state versus the fasting state. The mean Cmax, AUC0-∞, and Tmax of BILR 355 in the fed state were 91.9%, 122.0%, and 626.3%, respectively, of those in the fasted state. The high-fat and high-calorie breakfast apparently somewhat decreased the rate of absorption (Cmax, Tmax) but increased the extent of absorption (AUC0-∞) for BILR 355; all mean changes in exposures (AUC0-∞ and Cmax) were less than 25%.

In conclusion, after the administration of single doses of BILR 355 alone, BILR 355 was rapidly absorbed, with a Tmax of 0.5 to 1.5 h and with a short elimination t1/2 of 2 to 4 h. Increased doses led to a more than a dose-proportional increase in BILR 355 exposure. After coadministration with low-dose RTV, the PK profiles of BILR 355 were markedly altered. The values of AUC0-∞, Cmax, and t1/2 were increased 15- to 30-fold, 2- to 3-fold, and 5- to 6-fold, respectively. The urinary recovery of BILR 355 was increased from less than 1% to approximately 5% in the presence of RTV. Also, the administered doses of BILR 355 with RTV were approximately proportional to the systemic exposures observed. BILR 355 is safe and well tolerated in healthy male subjects after the administration of single ascending doses of BILR 355 alone or with RTV.

Acknowledgments

This work was founded by Boehringer Ingelheim Pharmaceuticals, Inc.

Footnotes

Published ahead of print on 29 September 2008.

REFERENCES

  • 1.Antinori, A., M. Zaccarelli, A. Cingolani, F. Forbici, M. G. Rizzo, M. P. Trotta, S. Di Giambendetto, P. Narciso, A. Ammassari, E. Girardi, A. De Luca, and C. F. Perno. 2002. Cross-resistance among nonnucleoside reverse transcriptase inhibitors limits recycling efavirenz after nevirapine failure. AIDS Res. Hum. Retrovir. 18:835-838. [DOI] [PubMed] [Google Scholar]
  • 2.Bacheler, L., S. Jeffrey, G. Hanna, R. D'Aquila, L. Wallace, K. Logue, B. Cordova, K. Hertogs, B. Larder, R. Buckery, D. Baker, K. Gallagher, H. Scarnati, R. Tritch, and C. Rizzo. 2001. Genotypic correlates of phenotypic resistance to efavirenz in virus isolates from patients failing nonnucleoside reverse transcriptase inhibitor therapy. J. Virol. 75:4999-5008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Barbour, J. D., F. M. Hecht, T. Wrin, T. J. Liegler, C. A. Ramstead, M. P. Busch, M. R. Segal, C. J. Petropoulos, and R. M. Grant. 2004. Persistence of primary drug resistance among recently HIV-1 infected adults. AIDS 18:1683-1689. [DOI] [PubMed] [Google Scholar]
  • 4.Bonneau, P., P. A. Robinson, J. Duan, L. Doyon, B. Simoneau, C. Yoakim, M. Bos, M. Cordingley, B. Brenner, B. Spira, M. Wainberg, F. Huang, K. Drda, C. Ballow, M. Koenen-Bergmann, and D. L. Mayers. 2005. Antiviral characterization and human experience with BILR 355 BS, a novel next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) with a broad anti-HIV-1 profile, abstr. 558. Abstr. 12th Conf. Retrovir. Opportunistic Infect.
  • 5.Boone, L. R. 2006. Next-generation HIV-1 non-nucleoside reverse transcriptase inhibitors. Curr. Opin. Investig. Drugs 7:128-135. [PubMed] [Google Scholar]
  • 6.Broom, C. 1990. Design of first-administration studies in healthy man, p. 206-213. In J. O'Grady and O. I. Linet (ed.), Early phase drug evaluation in man. Macmillan Press, London, United Kingdom.
  • 7.Chen, M., L. Ma, G. L. Drusano, J. S. Bertino, Jr., and A. N. Nafziger. 2006. Sex differences in CYP3A activity using intravenous and oral midazolam 1. Clin. Pharmacol. Ther. 80:531-538. [DOI] [PubMed] [Google Scholar]
  • 8.Clotet, B. 2004. Strategies for overcoming resistance in HIV-1 infected patients receiving HAART. AIDS Rev. 6:123-130. [PubMed] [Google Scholar]
  • 9.Cvetkovic, R. S., and K. L. Goa. 2003. Lopinavir/ritonavir: a review of its use in the management of HIV infection. Drugs 63:769-802. [DOI] [PubMed] [Google Scholar]
  • 10.Decker, C. J., L. M. Laitinen, G. W. Bridson, S. A. Raybuck, R. D. Tung, and P. R. Chaturvedi. 1998. Metabolism of amprenavir in liver microsomes: role of CYP3A4 inhibition for drug interactions. J. Pharm. Sci. 87:803-807. [DOI] [PubMed] [Google Scholar]
  • 11.de Mendoza, C., C. Rodriguez, J. Colomina, C. Tuset, F. Garcia, J. M. Eiros, A. Corral, P. Leiva, J. Aguero, J. Torre-Cisneros, J. Pedreira, I. Viciana, J. del Romero, A. Saez, R. Ortiz de Lejarazu, and V. Soriano. 2005. Resistance to nonnucleoside reverse-transcriptase inhibitors and prevalence of HIV type 1 non-B subtypes are increasing among persons with recent infection in Spain. Clin. Infect. Dis. 41:1350-1354. [DOI] [PubMed] [Google Scholar]
  • 12.de Vries-Sluijs, T. E., J. P. Dieleman, D. Arts, A. D. Huitema, J. H. Beijnen, M. Schutten, and M. E. van der Ende. 2003. Low nevirapine plasma concentrations predict virological failure in an unselected HIV-1-infected population. Clin. Pharmacokinet. 42:599-605. [DOI] [PubMed] [Google Scholar]
  • 13.Eagling, V. A., D. J. Back, and M. G. Barry. 1997. Differential inhibition of cytochrome P450 isoforms by the protease inhibitors, ritonavir, saquinavir and indinavir. Br. J. Clin. Pharmacol. 44:190-194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hogg, R. S., K. V. Heath, B. Yip, K. J. Craib, M. V. O'Shaughnessy, M. T. Schechter, and J. S. Montaner. 1998. Improved survival among HIV-infected individuals following initiation of antiretroviral therapy. JAMA 279:450-454. [DOI] [PubMed] [Google Scholar]
  • 15.Kilby, J. M., G. Sfakianos, N. Gizzi, P. Siemon-Hryczyk, E. Ehrensing, C. Oo, N. Buss, and M. S. Saag. 2000. Safety and pharmacokinetics of once-daily regimens of soft-gel capsule saquinavir plus minidose ritonavir in human immunodeficiency virus-negative adults. Antimicrob. Agents Chemother. 44:2672-2678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.King, J. R., H. Wynn, R. Brundage, and E. P. Acosta. 2004. Pharmacokinetic enhancement of protease inhibitor therapy. Clin. Pharmacokinet. 43:291-310. [DOI] [PubMed] [Google Scholar]
  • 17.Koudriakova, T., E. Iatsimirskaia, I. Utkin, E. Gangl, P. Vouros, E. Storozhuk, D. Orza, J. Marinina, and N. Gerber. 1998. Metabolism of the human immunodeficiency virus protease inhibitors indinavir and ritonavir by human intestinal microsomes and expressed cytochrome P4503A4/3A5: mechanism-based inactivation of cytochrome P4503A by ritonavir. Drug Metab. Dispos. 26:552-561. [PubMed] [Google Scholar]
  • 18.Little, S. J., S. Holte, J. P. Routy, E. S. Daar, M. Markowitz, A. C. Collier, R. A. Koup, J. W. Mellors, E. Connick, B. Conway, M. Kilby, L. Wang, J. M. Whitcomb, N. S. Hellmann, and D. D. Richman. 2002. Antiretroviral-drug resistance among patients recently infected with HIV. N. Engl. J. Med. 347:385-394. [DOI] [PubMed] [Google Scholar]
  • 19.Palella, F. J., Jr., K. M. Delaney, A. C. Moorman, M. O. Loveless, J. Fuhrer, G. A. Satten, D. J. Aschman, S. D. Holmberg, et al. 1998. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. N. Engl. J. Med. 338:853-860. [DOI] [PubMed] [Google Scholar]
  • 20.van Heeswijk, R. P., A. Veldkamp, J. W. Mulder, P. L. Meenhorst, J. M. Lange, J. H. Beijnen, and R. M. Hoetelmans. 2001. Combination of protease inhibitors for the treatment of HIV-1-infected patients: a review of pharmacokinetics and clinical experience. Antivir. Ther. 6:201-229. [PubMed] [Google Scholar]
  • 21.Veldkamp, A. I., G. J. Weverling, J. M. Lange, J. S. Montaner, P. Reiss, D. A. Cooper, S. Vella, D. Hall, J. H. Beijnen, and R. M. Hoetelmans. 2001. High exposure to nevirapine in plasma is associated with an improved virological response in HIV-1-infected individuals. AIDS 15:1089-1095. [DOI] [PubMed] [Google Scholar]
  • 22.Yoakim, C., P. R. Bonneau, R. Deziel, L. Doyon, J. Duan, I. Guse, S. Landry, E. Malenfant, J. Naud, W. W. Ogilvie, J. A. O'Meara, R. Plante, B. Simoneau, B. Thavonekham, M. Bos, and M. G. Cordingley. 2004. Novel nevirapine-like inhibitors with improved activity against NNRTI-resistant HIV: 8-heteroarylthiomethyldipyridodiazepinone derivatives. Bioorg. Med. Chem. Lett. 14:739-742. [DOI] [PubMed] [Google Scholar]

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES