ArticlePDF Available

Steady-State Disposition of the Nonpeptidic Protease Inhibitor Tipranavir when Coadministered with Ritonavir

American Society for Microbiology
Antimicrobial Agents and Chemotherapy
Authors:

Abstract and Figures

The pharmacokinetic and metabolite profiles of the antiretroviral agent tipranavir (TPV), administered with ritonavir (RTV), in nine healthy male volunteers were characterized. Subjects received 500-mg TPV capsules with 200-mg RTV capsules twice daily for 6 days. They then received a single oral dose of 551 mg of TPV containing 90 microCi of [(14)C]TPV with 200 mg of RTV on day 7, followed by twice-daily doses of unlabeled 500-mg TPV with 200 mg of RTV for up to 20 days. Blood, urine, and feces were collected for mass balance and metabolite profiling. Metabolite profiling and identification was performed using a flow scintillation analyzer in conjunction with liquid chromatography-tandem mass spectrometry. The median recovery of radioactivity was 87.1%, with 82.3% of the total recovered radioactivity excreted in the feces and less than 5% recovered from urine. Most radioactivity was excreted within 24 to 96 h after the dose of [(14)C]TPV. Radioactivity in blood was associated primarily with plasma rather than red blood cells. Unchanged TPV accounted for 98.4 to 99.7% of plasma radioactivity. Similarly, the most common form of radioactivity excreted in feces was unchanged TPV, accounting for a mean of 79.9% of fecal radioactivity. The most abundant metabolite in feces was a hydroxyl metabolite, H-1, which accounted for 4.9% of fecal radioactivity. TPV glucuronide metabolite H-3 was the most abundant of the drug-related components in urine, corresponding to 11% of urine radioactivity. In conclusion, after the coadministration of TPV and RTV, unchanged TPV represented the primary form of circulating and excreted TPV and the primary extraction route was via the feces.
Content may be subject to copyright.
ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, July 2007, p. 2436–2444 Vol. 51, No. 7
0066-4804/07/$08.000 doi:10.1128/AAC.01115-06
Copyright © 2007, American Society for Microbiology. All Rights Reserved.
Steady-State Disposition of the Nonpeptidic Protease Inhibitor
Tipranavir when Coadministered with Ritonavir
Linzhi Chen,
1
* John P. Sabo,
1
Elsy Philip,
1
Yanping Mao,
1
Stephen H. Norris,
1
Thomas R. MacGregor,
1
Jan M. Wruck,
3
Sandra Garfinkel,
2
Mark Castles,
1
Amy Brinkman,
4
and Hernan Valdez
2
Departments of Drug Metabolism and Pharmacokinetics,
1
Virology,
2
and Biometrics and Data Management,
3
Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, and
Covance Laboratories Inc., Madison, Winsconsin
4
Received 4 September 2006/Returned for modification 28 January 2007/Accepted 30 April 2007
The pharmacokinetic and metabolite profiles of the antiretroviral agent tipranavir (TPV), administered with
ritonavir (RTV), in nine healthy male volunteers were characterized. Subjects received 500-mg TPV capsules
with 200-mg RTV capsules twice daily for 6 days. They then received a single oral dose of 551 mg of TPV
containing 90 Ci of [
14
C]TPV with 200 mg of RTV on day 7, followed by twice-daily doses of unlabeled 500-mg
TPV with 200 mg of RTV for up to 20 days. Blood, urine, and feces were collected for mass balance and
metabolite profiling. Metabolite profiling and identification was performed using a flow scintillation analyzer
in conjunction with liquid chromatography-tandem mass spectrometry. The median recovery of radioactivity
was 87.1%, with 82.3% of the total recovered radioactivity excreted in the feces and less than 5% recovered from
urine. Most radioactivity was excreted within 24 to 96 h after the dose of [
14
C]TPV. Radioactivity in blood was
associated primarily with plasma rather than red blood cells. Unchanged TPV accounted for 98.4 to 99.7% of
plasma radioactivity. Similarly, the most common form of radioactivity excreted in feces was unchanged TPV,
accounting for a mean of 79.9% of fecal radioactivity. The most abundant metabolite in feces was a hydroxyl
metabolite, H-1, which accounted for 4.9% of fecal radioactivity. TPV glucuronide metabolite H-3 was the most
abundant of the drug-related components in urine, corresponding to 11% of urine radioactivity. In conclusion,
after the coadministration of TPV and RTV, unchanged TPV represented the primary form of circulating and
excreted TPV and the primary extraction route was via the feces.
Tipranavir (TPV) is a nonpeptidic (15, 16) sulfonamide-
substituted dihydropyrone (Fig. 1) protease inhibitor (PI) mar-
keted for the treatment of human immunodeficiency virus
(HIV)-infected, treatment-experienced patients under the
trade name Aptivus. Most viruses resistant to PIs retain sus-
ceptibility to TPV (1, 6, 12). Like other PIs, TPV binds directly
to HIV aspartyl protease, thereby disrupting the catalytic site
of the enzyme and preventing the protease-dependent cleav-
age of HIV gag and gag-pol polyproteins into smaller func-
tional proteins (10, 15, 16). Clinical studies have demonstrated
the significant activity of TPV against HIV type 1 in infected
patients receiving twice-daily doses ranging from 300 to 1,200
mg (2, 3, 4, 9).
In clinical studies with healthy volunteers and HIV-infected
patients, TPV pharmacokinetic parameters, including the
steady-state trough concentration, the area under the concen-
tration-time curve (AUC) for plasma, the maximum concen-
tration in plasma during the steady state (C
max
), and the ap-
parent terminal half-life (t
1/2
), were substantially improved
with the concomitant administration of ritonavir (RTV) (8, 9).
Since RTV strongly inhibits cytochrome P450 3A4 (CYP3A4)
(5), the boosted levels of TPV in plasma with the coadminis-
tration of TPV and RTV (TPV/r) indicate that the metabolism
of TPV occurs via the cytochrome P450 (CYP450) pathway (8,
9). Phase II studies have also demonstrated that TPV is an
inducer of CYP450 metabolism or intestinal P glycoprotein
efflux, thereby lowering systemic RTV concentrations when
administered with RTV (8, 11, 14). Clinical studies with
healthy volunteers and HIV-infected patients have evaluated
potential drug interactions of TPV/r and established that no
TPV dose adjustments are required when TPV/r is adminis-
tered in conjunction with other commonly employed antiret-
roviral agents (13; F. D. Goebel, J. P. Sabo, T. R. MacGregor,
D. L. Mayers, and S. McCallister, presented at the HIV DART
Conference, 2002).
The purpose of the present study was to characterize the
pharmacokinetic and metabolite profiles of TPV when admin-
istered with RTV.
MATERIALS AND METHODS
Materials. TPV, [
14
C]TPV, and TPV glucuronide were synthesized at Boehr-
inger Ingelheim Pharmaceuticals, Inc. (Ridgefield, CT). The position of the
14
C
label is indicated in Fig. 1. The chemical identities of the compounds were
established by high-performance liquid chromatography, mass spectrometry, and
nuclear magnetic resonance. The radiochemical purity of [
14
C]TPV was deter-
mined by liquid chromatography (LC)-radiochromatography to be 98%. Aqua-
sol-2, Ultima Gold XR, and Ultima FLO-M scintillation cocktails were pur-
chased from PerkinElmer Life and Analytical Sciences (Shelton, CT). All other
chemicals were provided by standard commercial sources and were of reagent
grade or better.
Subjects, dosing, and sample collection. This phase I, open-label, multiple-
oral-dose study was conducted to characterize the pharmacokinetics of TPV and
its metabolites, to elucidate the disposition of the parent compound and the total
radioactivity by excretion and mass balance during the steady state, and to
* Corresponding author. Mailing address: Research and Develop-
ment, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury
Rd., Ridgefield, CT 06877. Phone: (203) 778-7870. Fax: (203) 791-
6003. E-mail: lchen@rdg.boehringer-ingelheim.com.
Published ahead of print on 7 May 2007.
2436
identify and quantify major metabolites of TPV in plasma, urine, and feces. The
study was conducted in accordance with the Nuclear Regulatory Commission
regulations and the Declaration of Helsinki (1964 and subsequent revisions). Of
the 12 healthy male volunteers enrolled, 9 completed all phases of the study,
including the administration of [
14
C]TPV/r. Subjects were nonsmokers and re-
ceived standardized meals and the same supervised doses of TPV/r, i.e., 500 mg
of TPV plus 200 mg of RTV twice daily for 6 days. On day 7, participants
received a light snack at 6:00 am, 60 min prior to the administration of 500 mg
of [
14
C]TPV (90 Ci) plus 200 mg of RTV. Subjects fasted for an additional 4 h
after the dose administration. At 7:00 pm that evening, participants resumed
dosing with nonradioactive TPV at 500 mg and RTV at 200 mg twice daily and
continued on this dosing regimen up to day 20 of the study. Subjects did not
receive any medications known to affect CYP450 activity, and coffee, tea, cola,
chocolate, St. John’s wort, milk thistle, garlic supplements, Seville oranges,
grapefruit, grapefruit juice, and alcohol were avoided during the course of the
study.
Blood samples were collected daily, immediately prior to the evening doses of
TPV/r on study days 1 through 7 (5 ml) and prior to the morning doses on study
days 8 through 15 (10 ml). On day 7, serial 10-ml blood samples were obtained
10 min prior to the radioactive dose and at 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, and
24 h post-radioactive dose. Both whole blood and plasma were analyzed for
radioactivity. Additional 10-ml blood samples for metabolite profiling were col-
lected at 3, 8, and 12 h post-radioactive dose. The 3-h time point coincides with
the mean peak plasma TPV concentration (8), while the 8- and 12-h time points
were chosen for metabolite profiling as late time points near the end of the 12-h
dosing interval. Plasma was prepared immediately after sampling by centrifuga-
tion at 5°C for 10 min at 2,000 g.
After the administration of the radioactive dose, urine and feces were col-
lected until the end of the study. All samples were stored at approximately 20°C
until analyzed.
Radioactivity measurement. Feces and tissue samples were homogenized in
water and a mixture of ethanol and water (1:1), respectively, which were approx-
imately three to five times the sample weight. Whole-blood, fecal homogenate,
and tissue homogenate samples were combusted before radioactivity counting
with a Packard 307 sample oxidizer (Packard Instrument Co., Meriden, CT), and
the resulting
14
CO
2
was trapped in a mixture of Perma Fluor and Carbo-Sorb
(Packard Instrument Co., Meriden, CT). Aliquots of samples (0.2 ml of blood
and plasma, 0.5 ml of urine, and 0.5 g of fecal and tissue homogenates) were
mixed with 5 ml of Ultima Gold XR scintillation cocktail and evaluated for
radioactivity by using Packard 2900TR liquid scintillation counters (Packard
Instrument Co.). Scintillation counting data (expressed in counts per minute)
were automatically corrected for counting efficiency by using the external stan-
dardization technique and an instrument-stored quench curve generated from a
series of sealed quenched standards.
Quantitation of TPV in plasma. The concentrations of TPV in plasma were
determined using a validated LC-tandem mass spectrometry (MS-MS) method.
Plasma samples were prepared by a two-step liquid-liquid extraction procedure;
the use of an ethyl acetate-hexane mixture was followed by a hexane wash.
Chromatographic separation was achieved using a 2.0- by 30-mm, 4-m-particle-
size Synergi Polar-RP column (Phenomenex Inc., Torrance, CA). The mobile
phase consisted of 50% 20 mM formic acid–10 mM acetic acid and 50% aceto-
nitrile; the run time was 6 min. The detector used was a MicroMass QuattroLC
triple-quadrupole mass spectrometer (Waters, Milford, MA), which was
equipped with an electrospray ion source and operated in positive-selective-
reaction monitoring mode. Key MS-MS operating parameters included the fol-
lowing: capillary voltage, 1 kV; nebulizer gas, 110 liters/h; desolvation gas, 1,300
liters/h; desolvation temperature, 300°C; cone voltage, 30 V; and collision offset,
20 V. Of the two overlapping calibration curves used in this study, one had a
calibration range of 25.0 to 2,000 ng/ml and the other had a calibration range of
1,000 to 20,000 ng/ml. Two calibration curves with different calibration ranges
were used to minimize sample manipulation during bioanalysis. All of the sam-
ples were assayed with the higher-range curve first. For the samples that had a
concentration of less than 1,000 ng/ml (the lower limit of quantitation of the
higher-range curve), there was no reportable result, and these samples were
subsequently reassayed with the lower-range curve.
Metabolite sample extraction. The plasma samples collected from the nine
subjects at each of the three time points, 3, 8, and 12 h postdose, were pooled for
metabolite profiling and identification. A 20- to 25-ml aliquot of the plasma pool
was evaporated at room temperature under a nitrogen stream in a Zymark
TurboVap LV evaporator (Zymark, Hopkinton, MA), and the residue was then
extracted four to six times with 10 to 20 ml of methanol. For each extraction, the
mixture was sonicated for 5 min, and the supernatant was separated by centrif-
ugation. The extracts were combined, evaporated with the TurboVap, and sub-
jected to further cleanup via multiple evaporation-extraction cycles with meth-
anol and acetonitrile. The final sample was reconstituted in 0.30 to 0.43 ml of
methanol and transferred to an autosampler vial for LC-radiochromatography-
MS-MS analysis. The total extraction recovery of radioactivity from plasma
ranged from 87.8 to 100.8%.
The urine samples collected from each subject during the first 24 h after
[
14
C]TPV dosing were pooled. The majority (50.0 to 63.3%) of urine radioac-
tivity was found in the urine from the 0-to-24-h sample collection period, and the
remaining radioactivity was spread throughout large volumes of urine across
several days. Equal percentages (by volume) of the 0-to-24-h urine samples were
combined to make the pools used for metabolite profiling and identification. To
a 100-ml aliquot of pooled urine was added 6 ml of 28 to 30% ammonium
hydroxide. The urine samples were then extracted via solid-phase extraction with
35-cm
3
/6-g Oasis HLB solid-phase extraction cartridges (Waters Corporation,
Millford, MA). After loading, the solid-phase extraction cartridge was washed
with 25 to 35 ml of 2% ammonium hydroxide, and the sample was eluted with
methanol. The eluate was evaporated and further cleaned up via multiple evap-
oration-extraction cycles with acetonitrile. The final samples were reconstituted
in 0.4 to 0.5 ml of methanol and transferred to an autosampler vial for metabolite
profiling and identification. The total extraction recovery of radioactivity from
urine ranged from 84.1 to 101.7%.
A fecal pool for each subject for metabolite profiling and identification was
also prepared by combining aliquots of fecal homogenates from three to five
sampling time points at equal percentages (by volume). Depending on the sub-
ject, feces from 0 to 24 h up to 120 to 144 h were pooled. The pools corresponded
to 90% or more of the total radioactivity ultimately excreted in feces. An 18- to
30-ml aliquot of the pooled fecal homogenate was centrifuged, the supernatant
was discarded, and the pellet was extracted with acetonitrile, methanol, and
acidified methanol. The extracts were combined, evaporated, and further cleaned
up via multiple evaporation-extraction cycles with acetonitrile. The final samples
were reconstituted in 0.3 to 0.5 ml of methanol and then transferred to an
autosampler vial for metabolite profiling and identification. The total extraction
recovery of radioactivity from feces ranged from 81.2 to 106.1%.
Metabolite profiling and identification. Metabolite profiling and identification
was carried out with an LC-radiochromatography-MS-MS system, which con-
sisted of an Agilent 1100 series high-performance liquid chromatography system
(Agilent, Palo Alto, CA) with a Packard 525 Radiomatic flow scintillation ana-
lyzer (Packard Instrument Co.) and a Finnigan LCQ Deca XP
ion trap mass
spectrometer (Finnigan, San Jose, CA) as detectors. The chromatographic sep-
aration was achieved with an xTerra MS C
18
column, 250 by 4.6 mm, with a 5-m
particle size (Waters Corporation). The mobile phase A comprised 0.1% acetic
acid containing 5% acetonitrile, and the mobile phase B was acetonitrile con-
taining 0.1% acetic acid. The gradient conditions were 18 to 50% mobile phase
B over 50 min and then to 100% over 20 min at 1 ml/min. The post column flow
was split in a 1:20 ratio via an Acurate flow splitter (LC Packings, San Francisco,
CA); 20 parts were sent to the flow scintillation analyzer and 1 part was sent to
the ion trap mass spectrometer. The flow scintillation analyzer was equipped with
a 250-l flow cell, and the detection window was 4 to 100 keV. Packard Ultima
FLO-M was used as the scintillation cocktail, and the flow rate was 2.5 ml/min.
The LCQ mass spectrometer was equipped with an electrospray ion source and
operated in positive mode. Key operating parameters included a spray voltage of
5 kV, a sheath gas flow of 35 U, an auxiliary gas flow of 0 U, a capillary
temperature of 300°C, and an MS-MS collision energy of 40% with MS-MS wide
excitation.
Pharmacokinetic analysis. The pharmacokinetic profiles of TPV were deter-
mined using plasma and blood TPV concentrations in a noncompartmental
model (WinNonlin; Pharsight Corporation, Mountain View, CA). The following
pharmacokinetic parameters for TPV in plasma and blood were analyzed: morn-
ing and evening trough concentrations, the C
max
, the concentration in plasma
12 h after dosing (C
12h
), the AUC, the time to maximum concentration (T
max
),
and the t
1/2
of total
14
C radioactivity.
FIG. 1. Structure of the sulfonamide-substituted dihydropyrone PI
TPV. The asterisk denotes the
14
C label.
VOL. 51, 2007 TIPRANAVIR-RITONAVIR DISPOSITION 2437
RESULTS
Mass balance and excretion of radiolabel. A total of nine
male subjects (eight white, one black) from 22 to 53 years of
age with body mass indices of 21 to 27 kg/m
2
received radio-
labeled TPV. Approximately 75.3% 23.7% (mean stan-
dard deviation [SD]) of the radiolabeled dose was recovered
from feces, urine, and tissue (Table 1; Fig. 2, inset). Subject 113
was excluded from the mass balance analyses because diarrhea
prevented complete sample collection. Subject 109 had 21.3%
of the radiolabeled dose recovered, and this result was an
outlier for the study group as a whole. For the remaining seven
subjects, the total recovery of radioactivity was 83.0%
10.0%. Feces represented the primary route of excretion, with
70.4% 24.0% of the total radioactivity dose recovered via
this route. Urinary excretion accounted for 4.6% 0.6% of the
total administered radioactive dose.
Radioactivity and TPV concentrations in plasma and blood.
The pharmacokinetic parameters for TPV and for total radio-
activity in plasma and blood are shown in Tables 2 and 3. Plots
of mean concentrations of radioactivity in plasma and whole
blood versus time are shown in Fig. 2. For all subjects, con-
centrations of radioactivity in plasma and blood declined
steadily to below the limit of quantitation by 96 and 120 h
postdose, respectively. The plots of blood and plasma radioac-
tivity concentrations versus time were parallel, and the ratio of
the concentration of circulating radioactivity in plasma to that
in blood was 1.89 0.08 at each time point. Therefore, ap-
proximately 85% of the TPV in blood was distributed in the
plasma, with the remaining TPV in erythrocytes, and the dis-
tribution between plasma and blood cells was approximately
constant. The plasma radioactivity-time profile could also be
superposed on the plasma TPV concentrations obtained using
FIG. 2. Radioactivity in plasma (E) and blood (F) after a single oral dose of [
14
C]TPV administered during the steady state. Means SDs
are shown (n9). (Inset) Cumulative excretion of radioactivity in urine () and feces (E) and total radioactivity (F). Means SDs are shown
(n8).
TABLE 1. Summary of cumulative recovery of radioactivity from samples from human subjects after oral administration of [
14
C]TPV
Sample type Cumulative % of radioactive dose recovered from subject: Mean
a
SD
104 106 108 109 113
a
116 122 123 127
Urine 5.07 4.94 4.39 5.60 3.88 4.21 4.36 4.50 3.64 4.59 0.60
Feces 84.4 62.2 81.4 15.7 32.3 87.0 65.3 83.2 83.8 70.4 24.0
Toilet tissue specimen 0.79 0.30 0.71 0.01 0.07 0.04 0.07 0.77 0.23 0.37 0.34
Total 90.3 67.4 86.5 21.3 36.3 91.3 69.7 88.5 87.7 75.3 23.7
a
Subject 113 had an episode of explosive diarrhea on day 13, and the sample was lost. Thus, the results for subject 113 were excluded from the calculationsofall
the means involving excreta.
2438 CHEN ET AL. ANTIMICROB.AGENTS CHEMOTHER.
the LC-MS-MS assay, indicating that the abundance of TPV
relative to the total level of radioactivity in plasma remained
approximately constant.
Pharmacokinetics. The pharmacokinetic profiles of TPV
were characterized by using the concentration data for
plasma as measured before and after the administration of
the multiple-dose regimen and the single 551-mg oral dose
of [
14
C]TPV. Pharmacokinetic data derived prior to the
administration of radiolabeled TPV suggested that the
steady-state plasma TPV concentration was achieved by day
7 of the twice-daily administration of TPV/r at 500 and 200
mg, respectively (Fig. 3). The range of trough plasma TPV
TABLE 2. Pharmacokinetic parameters for TPV in plasma
Parameter
a
Mean SD Minimum Median Maximum Geometric mean
d
Total TPV dose (mg) 551 1 549 551 552
T
max
(h) 2.9 1.1 1.5 3.0 5.0 2.8
C
max
(M) observed
b
99.39 23.37 74.93 92.08 141.39 97.09
C
max
(M) normalized
c
90.23 21.30 67.87 83.56 128.31 88.13
C
12h
(M) observed
b
25.85 18.84 6.94 22.49 72.27 21.44
C
12h
(M) normalized
c
23.46 17.06 6.29 20.41 65.46 19.46
AUC
0–12
(h M) observed
b
657.0 170.9 452.5 601.2 922.9 638.4
AUC
0–12
(h M) normalized
c
596.5 155.7 409.9 545.5 840.6 579.4
z
(h
1
)0.1631 0.0499 0.0952 0.1713 0.2495 0.1561
t
1/2
(h) 4.6 1.5 2.8 4.0 7.3
CL (liters/h) 1.47 0.36 0.99 1.52 2.02 1.43
V(liters) 9.6 3.2 4.9 9.2 16.3 9.2
a
Results from nine subjects were included in the calculations. The total TPV dose includes unlabeled TPV and [
14
C]TPV.
z
, first-order rate constant associated
with the terminal (log-linear) elimination phase; CL, clearance; V, volume of distribution.
b
Based on noncompartmental pharmacokinetic analysis using reported TPV concentrations.
c
Based on noncompartmental pharmacokinetic results corrected for the TPV dose, as follows: (observed pharmacokinetic metric/actual TPV dose) nominal TPV
dose, where the actual TPV dose is the dose administered to each subject and the nominal TPV dose is 500 mg.
d
The harmonic mean t
1/2
was 4.2 h.
TABLE 3. Pharmacokinetic parameters for radioactivity in plasma and blood
a
Biomatrix and
time period Value T
max
(h)
T
last
(h)
C
max
(M eq)
C
last
(M eq)
C
12h
(M eq)
z
(h
1
)
t
1/2
(h)
AUC
(h M eq)
CL
(liters/h)
V
(liters)
MRT
(h)
Blood, 0–12 h Mean 2.61 49.45 10.26 0.1694 4.20 297.35 2.64 15.68
SD 0.60 11.19 3.34 0.0296 0.74 61.52 0.56 2.65
Min 1.50 35.92 4.96 0.1210 3.05 233.85 1.78 10.25
Median 3.00 47.13 9.58 0.1653 4.19 277.18 2.69 15.89
Max 3.00 74.99 15.33 0.2272 5.73 429.04 3.48 19.87
gMean 2.54 48.47 9.75 0.1672 4.15 292.27 2.59 15.46
hMean 4.09
Plasma, 0–12 h Mean 2.56 90.55 18.50 0.1761 4.07 546.97 1.45 8.32
SD 0.53 15.98 6.54 0.0374 0.74 111.16 0.30 1.57
Min 2.00 70.99 7.30 0.1348 2.63 439.66 0.96 5.81
Median 3.00 90.57 18.39 0.1752 3.96 531.54 1.44 8.12
Max 3.00 122.91 28.77 0.2640 5.14 779.57 1.88 10.75
gMean 2.51 89.37 17.33 0.1730 4.01 537.81 1.42 8.18
hMean 3.94
Blood, 0–Mean 64.00 0.93 0.0482 16.16 492.45 1.98 44.23 16.56
SD 16.97 0.31 0.0236 4.27 127.75 0.56 12.75 4.15
Min 24.00 0.62 0.0358 6.36 288.85 1.29 29.09 7.18
Median 72.00 0.91 0.0399 17.37 449.10 2.03 42.79 16.96
Max 72.00 1.44 0.1090 19.38 709.82 3.17 63.41 22.64
gMean 60.92 0.89 0.0449 15.43 477.46 1.91 42.61 15.94
hMean 14.37
Plasma, 0–Mean 90.67 0.82 0.0334 21.32 910.92 1.06 32.47 18.29
SD 10.58 0.20 0.0058 3.55 220.61 0.25 9.61 2.66
Min 72.00 0.60 0.0268 16.58 600.71 0.70 18.98 13.48
Median 96.00 0.74 0.0327 21.22 891.71 1.03 30.93 18.40
Max 96.00 1.13 0.0418 25.87 1310.61 1.52 51.67 23.63
gMean 90.05 0.80 0.0329 21.05 887.71 1.03 31.27 18.11
hMean 20.78
a
Results for nine subjects were included in the calculations. Min, minimum; max, maximum; gMean, geometric mean; hMean, harmonic mean; T
last
, time of last
measurable concentration; C
last
, last measurable concentration;
z
, first-order rate constant associated with the terminal (log-linear) elimination phase; CL, clearance;
V, volume of distribution; MRT, mean residence time.
VOL. 51, 2007 TIPRANAVIR-RITONAVIR DISPOSITION 2439
concentrations for all subjects following the first two doses
of TPV/r was 30.6 to 98.4 M. TPV trough concentrations
increased through three additional dose administrations
with continued twice-daily dosing of TPV/r to a geometric
mean of 70.0 M on the evening of day 2, which was reflec-
tive of CYP3A inhibition by RTV. Mean evening trough
concentrations decreased by 6 M per day for the next 4
days, and by day 7 the trough TPV concentration was 21.9
M. This level was indicative of the establishment of equi-
librium between CYP3A inhibition by RTV and induction
by TPV. A steady state was established by day 7, as the
geometric mean ratio of the day 7 evening measurement to
the day 8 morning measurement was 1.07. Geometric mean
morning trough TPV concentrations from day 8 through day
15 averaged 18.2 M (range, 16.6 to 21.9 M).
The calculated pharmacokinetic parameters for TPV are
shown in Table 2, and those for total radioactivity are pre-
sented in Table 3. Using noncompartmental analyses, the me-
dian T
max
was calculated as 3.0 h (range, 1.5 to 5.0 h), with a
median normalized C
max
of 83.56 M (range, 67.87 to 128.31
M) and a median normalized C
12h
of 20.41 M (range, 6.29
to 65.46 M). The values were normalized for the actual ra-
dioactive dose (551 mg) given to the subjects as opposed to the
nominal 500-mg dose given on the other days. The median
elimination t
1/2
of TPV in plasma of 4.0 h (range, 2.8 to 7.3 h)
closely approximated the median elimination t
1/2
of radioac-
tivity in plasma of 3.96 h (range, 2.63 to 5.14 h) and that in
whole blood of 4.19 h (range, 3.05 to 5.73 h). For this group of
subjects, the median AUC from 0 to 12 h (AUC
0–12
) following
TPV/r administration at 500 and 200 mg was 545.5 h M
(range, 409.9 to 840.6 h M).
TPV metabolites. Unchanged TPV was the predominant
circulating component in the pooled plasma samples at 3, 8,
and 12 h postdose and accounted for 98.4 to 99.7% of the
plasma radioactivity (Table 4; Fig. 4a). Only a few trace me-
tabolites, including a TPV glucuronidation conjugate (H-3)
and an oxidation metabolite (H-1), were observed in radio-
chromatograms, and each metabolite accounted for 0.2%
of the plasma radioactivity. Based on this observation of
trace metabolites, it can be surmised that the plasma sam-
ples from all the subjects contained predominantly un-
changed drug and that pooling was necessary for metabolite
identification. Unchanged TPV also dominated the radio-
chromatograms for feces (Fig. 4c), accounting for 79.9% of
fecal radioactivity or, on average, 53.5% of the dose in the
nine subjects. Several metabolites were found in feces. Me-
tabolite H-1, the most abundant, was responsible for 3.2%
of the dose, while another oxidation metabolite, H-2, ob-
served in the feces from most of the subjects, represented
0.4% of the dose. The remaining metabolites in feces each
were less than 1.3% of the dose. In contrast, TPV was much
less abundant in urine and represented only 0.5% of urine
radioactivity or 0.1% of the dose (Fig. 4b). The majority of
radioactivity in urine was accounted for by many metabo-
lites. TPV glucuronide conjugate (H-3), accounting for 0.5%
of the dose, was the most abundant urinary metabolite; the
remaining urinary metabolites were each less than 0.4% of
the dose.
The TPV glucuronide conjugate metabolite (H-3) corre-
sponded to a radiochromatographic peak at approximately
52.5 min and a protonated molecular ion at m/z 779, 176
daltons higher than that corresponding to TPV. Upon collision
activation, the molecular ion gave protonated TPV via the loss
of glucuronide. The identification of H-3 was confirmed by a
comparison of the retention time and the MS-MS pattern to
the authentic standard.
Metabolite H-1 had a protonated molecular ion at m/z 619,
16 daltons higher than that of TPV (Fig. 5). MS-MS analysis of
H-1 produced daughter ions at m/z 575 (loss of CO
2
) and 411
FIG. 3. Steady-state concentrations of TPV in plasma (Cp). Solid
line, geometric mean; dashed line, median; day 1 to day 6, evening
trough concentrations; day 7, morning and evening trough concentra-
tions (evening concentrations were not normalized to the TPV dose of
approximately 551 mg per subject); day 8 to day 15, morning trough
concentrations (n7).
TABLE 4. Summary of abundance of radioactive metabolites in plasma, urine, and feces
Metabolite % of radioactivity (concn)
a
in plasma at: % of radioactivity
(% of radioactive dose) in excreta Total % of radioactive
dose in excreta
3 h 8 h 12 h Urine Feces
H-1 0.2 (0.10) ND ND 0.1 (0.1) 4.9 (3.2) 3.2
H-2 ND
b
ND ND ND 0.8 (0.4) 0.4
H-3 0.1 (0.05) ND ND 11.0 (0.5) ND 0.5
TPV 98.9 (48.6) 98.4 (21.0) 99.7 (10.7) 0.5 (0.1) 79.9 (53.5) 53.5
Total 99.2 (48.8) 98.4 (21.0) 99.7 (10.7) 11.5 (0.5) 85.6 (57.1) 57.6
a
Concentrations of metabolites are expressed as microgram equivalents of [
14
C]TPV per milliliter.
b
ND, not detected.
2440 CHEN ET AL. ANTIMICROB.AGENTS CHEMOTHER.
via the same fragmentation patterns as TPV. In addition, new
fragments were observed at m/z 557 (loss of CO
2
and loss of
H
2
O) and 495 (loss of C
7
H
6
O and loss of H
2
O). The mass
spectral analysis suggests that H-1 was likely a hydroxyl me-
tabolite. Similarly, H-2 was tentatively identified as another
oxidation metabolite, as illustrated in Fig. 5.
Safety. Nausea and loose stools were the most common
adverse events associated with TPV/r administration. There
were no serious adverse events, and only two subjects prema-
turely discontinued the use of the study drug, one due to nausea
and alanine aminotransferase elevation and the other due to
alanine aminotransferase elevation. The most common labora-
tory abnormalities were increases in transaminases (greater than
or equal to Division of AIDS grade 3 in two subjects, or 17%) and
mild increases in cholesterol and triglycerides.
DISCUSSION
In this study conducted with healthy male volunteers dosed
with twice-daily TPV/r at 500 and 200 mg, steady-state con-
centrations in plasma were achieved by day 7. Upon initial
dosing, trough TPV concentrations were higher than those
during the steady state, which is consistent with immediate
CYP3A inhibition by RTV (5, 8). Trough TPV concentrations
steadily decreased with twice-daily dosing until day 7 and
thereafter remained relatively constant, reflecting steady-state
attainment, through the remainder of the study (day 15). The
progressive decrease in the trough TPV concentration and
subsequent plateau suggest either CYP3A induction by TPV
and RTV or a decreased bioavailability of TPV and RTV,
followed by a balance of the discordant effects of TPV and
RTV on metabolism and absorption. The trough TPV concen-
trations and other pharmacokinetic characteristics observed in
this trial are consistent with those characterized in other clin-
ical studies of TPV at similar TPV/r doses (8, 9).
After the administration of the [
14
C]TPV/r dose on day 7
(steady state), TPV was rapidly absorbed, and concentrations
of both TPV and the radioactivity in plasma peaked at around
3 h postdose. TPV was found to be the predominant circulating
component in plasma, accounting for more than 98.4% of
plasma radioactivity at 3, 8, and 12 h postdose.
FIG. 4. Representative radiochromatograms for the 3-h plasma sample pooled from all nine subjects (a) and the urine (b) and feces (c) samples
from subject 106 after the administration of the [
14
C]TPV dose.
VOL. 51, 2007 TIPRANAVIR-RITONAVIR DISPOSITION 2441
Approximately 75.3% of the radiolabeled dose was recov-
ered from feces, urine, and tissue combined, and most was
excreted by 96 h after dosing. In general, the level of recovery
was lower than expected, perhaps as a result of incomplete
sample collection by subjects. Fecal excretion, which accounted
for approximately 70.4% of the radioactive dose, was the pri-
mary elimination route for TPV. On the other hand, urinary
excretion was responsible for 4.6% of the radioactive dose and
represented only a minor route of elimination. The absolute
bioavailability of TPV, in the presence of RTV, is not known.
The predominant recovery from feces in this study may indi-
cate extensive biliary excretion or pronounced P glycoprotein
efflux during the steady state. Although several metabolites
existed in feces, each represented 3.2% of the dose. Addi-
tionally, all metabolites in urine were minor, with each ac-
counting for 0.5% of the dose. The proposed metabolic path-
ways of TPV, in the presence of RTV, are shown in Fig. 6. The
minor contribution of the kidneys to the elimination of TPV
suggests that adjustments of the TPV/r dosage would be un-
likely to be necessary for renally impaired patients.
The lack of significant TPV metabolism during the steady
state in the presence of RTV coadministration and the signif-
icant decline in trough TPV concentrations from the initial
dosing until the steady state was attained on day 7 suggest that
the fractions of TPV and RTV absorbed are lower during the
steady state than after a single dose. TPV (11) and, to a lesser
extent, RTV (7) are substrates for P glycoprotein efflux trans-
port. In a drug interaction study with the P glycoprotein sub-
strate loperamide, TPV exhibited P glycoprotein self-induction
capability (11). In long-term clinical trials with TPV and RTV,
TPV has been shown to significantly reduce systemic RTV
exposure (14), presumably due to P glycoprotein efflux trans-
port.
This study demonstrated that the vast majority of the ad-
ministered dose of TPV remains unchanged in plasma and
excrements. The lack of TPV metabolism after the adminis-
tration of TPV with RTV would be expected because of the
profound inhibition of CYP3A by RTV (5). Previous studies
have suggested that the first-pass metabolism of TPV, without
RTV coadministration, occurs via the CYP3A pathway (8, 9),
producing a maximum exposure of plasma to TPV of less than
1M. In addition, this study did not identify any major, un-
expected TPV metabolites from an alternative pathway that
might potentially have altered the pharmacokinetics, efficacy,
and safety of TPV.
The major findings of this study, the unchanged nature of
the majority of the administered dose in the plasma and the
lack of any major, unexpected TPV metabolites, indicate that
the structure of TPV remains intact and available to bind to
HIV protease. The favorable TPV pharmacokinetic and me-
FIG. 5. Comparison of MS-MS results for the TPV standard (a) and metabolites H-1 (b) and H-2 (c).
2442 CHEN ET AL. ANTIMICROB.AGENTS CHEMOTHER.
tabolite profiles demonstrated in this study, along with the
distinct structure and resistance profile of TPV, support TPV/r
as an important treatment option for HIV-infected patients
with resistance to multiple PIs.
REFERENCES
1. Back, N. K. T., A. van Wijk, D. Remmerswaal, M. van Monfort, M. Nijhuis,
R. Schuurman, and C. A. B. Boucher. 2000. In-vitro tipranavir susceptibility
of HIV-1 isolates with reduced susceptibility to other protease inhibitors.
AIDS 14:101–102.
2. Cahn, P., J. Villacian, A. Lazzarin, C. Katlama, B. Grinsztejn, K. Arasteh, P.
Lopez, N. Clumeck, J. Gerstoft, N. Stavrianeas, S. Moreno, F. Antunes, D.
Neubacher, and D. Mayers. 2006. Ritonavir-boosted tipranavir demonstrates
superior efficacy to ritonavir-boosted protease inhibitors in treatment-expe-
rienced HIV-infected patients: 24-week results of the RESIST-2 trial. Clin.
Infect. Dis. 43:1347–1356.
3. Gathe, J., D. A. Cooper, C. Farthing, D. Jayaweera, D. Norris, G. Pierone,
C. R. Steinhart, B. Trottier, S. L. Walmsley, C. Workman, G. Mukwaya, V.
Kohlbrenner, C. Dohnanyi, S. McCallister, and D. Mayers for the RESIST-1
Study Team. 2006. Efficacy of the protease-inhibitors tipranavir plus ritona-
vir in treatment-experienced patients: 24-week analysis from the RESIST-1
trial. Clin. Infect. Dis. 43:1337–1346.
4. Gathe, J. C., G. Pierone, P. Piliero, K. Arasteh, R. Rubio, R. G. LaLonde, D.
Cooper, A. Lazzarin, V. M. Kohlbrenner, C. Dohnanyi, J. Sabo, and D.
Mayers. 2007. Efficacy and safety of three doses of tipranavir boosted with
ritonavir in treatment-experienced HIV type-1-infected patients. AIDS Res.
Hum. Retrovir. 23:216–223.
5. 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 P450 3A4/3A5:
mechanism-based inactivation of cytochrome P450 3A by ritonavir. Drug
Metab. Dispos. 26:552–561.
6. Larder, B. A., K. Hertogs, S. Bloor, C. van den Eynde, W. DeCian, Y. Wang,
W. W. Freimuth, and G. Tarpley. 2000. Tipranavir inhibits broadly protease
inhibitor-resistant HIV-1 clinical samples. AIDS 14:1943–1948.
7. Lee, C. G. L., M. M. Gottesman, C. O. Cardarelli, M. Ramachandra, K.-T.
Jeang, S. V. Ambudkar, I. Pastan, and S. Dey. 1998. HIV-1 protease inhib-
itors are substrates for the MDR1 multidrug transporter. Biochemistry 37:
3594–3601.
8. MacGregor, T. R., J. P. Sabo, S. H. Norris, P. Johnson, L. Galitz, and S.
McCallister. 2004. Pharmacokinetic characterization of different dose com-
binations of coadministered tipranavir and ritonavir in healthy volunteers.
HIV Clin. Trials 5:371–382.
9. McCallister, S., H. Valdez, K. Curry, T. MacGregor, M. Borin, W. Freimuth,
Y. Wang, and D. L. Mayers. 2004. 14-day dose-response study of the efficacy,
safety, and pharmacokinetics of the nonpeptidic protease inhibitor tipranavir
in treatment-naive HIV-1-infected patients. J. Acquir. Immune Defic. Syndr.
35:376–382.
10. Monini, P., C. Sgadari, G. Barillari, and B. Ensoli. 2003. HIV protease
inhibitors: antiretroviral agents with anti-inflammatory, anti-angiogenic and
anti-tumour activity. J. Antimicrob. Chemother. 51:207–211.
11. Mukwaya, G., T. R. MacGregor, D. Hoelscher, T. Heming, D. Legg, K.
Kavanaugh, P. Johnson, J. P. Sabo, and S. McCallister. 2005. Interaction of
ritonavir-boosted tipranavir with loperamide does not result in loperamide-
associated neurologic side effects in healthy volunteers. Antimicrob. Agents
Chemother. 49:4903–4910.
12. Poppe, S. M., D. E. Slade, K. T. Chong, R. R. Hinshaw, P. J. Pagano, M.
Markowitz, D. D. Ho, H. Mo, R. R. Gorman III, T. J. Dueweke, S. Thais-
rivongs, and W. G. Tarpley. 1997. Antiviral activity of the dihydropyrone
PNU-140690, a new nonpeptidic human immunodeficiency virus protease
inhibitor. Antimicrob. Agents Chemother. 41:1058–1063.
13. Roszko, P. J., K. Curry, B. Brazina, A. Cohen, E. L. Turkie, J. P. Sabo, T. R.
FIG. 6. Proposed metabolic pathways of TPV administered with RTV. Gluc, glucuronide.
VOL. 51, 2007 TIPRANAVIR-RITONAVIR DISPOSITION 2443
MacGregor, and S. McCallister. 2003. Standard doses of efavirenz (EFV),
zidovudine (ZDV), tenofovir (TDF), and didanosine (ddI) may be given with
tipranavir/ritonavir (TPV/r). Antivir. Ther. 8:S428.
14. Sabo, J. P., P. J. Piliero, A. Lawton, T. R. MacGregor, and J. Leith. 2006. A
comparison of steady-state trough plasma ritonavir concentrations for HIV
patients receiving an optimized background regimen and ritonavir-boosted
tipranavir (TPV/r), lopinavir (LPV/r), saquinavir (SQV/r) or amprenavir
(APV/r), poster 43. In Proceedings of the 7th International Workshop on
Clinical Pharmacology of HIV Therapy (Lisbon). Virology Education, Utre-
cht, The Netherlands.
15. Thaisrivongs, S., and J. W. Strohbach. 1999. Structure-based discovery of
tipranavir disodium (PNU-140690E): a potent, orally bioavailable, nonpep-
tidic HIV protease inhibitor. Biopolymers 51:51–58.
16. Turner, S. R., J. W. Strohbach, R. A. Tommasi, P. A. Aristoff, P. D.
Johnson, H. I. Skulnick, L. A. Dolak, E. P. Seost, P. K. Tomich, M. J.
Bohanon, M. M. Horng, J. C. Lynn, K. T. Chong, R. R. Hinshaw, K. D.
Watenpaugh, M. N. Janakiraman, and S. Thaisrivongs. 1998. Tipranavir
(PNU-140690): a potent, orally bioavailable nonpeptidic HIV protease
inhibitor of the 5,6-dihydro-4-hydroxy-2-pyrone sulfonamide class. J. Med.
Chem. 41:3467–3476.
2444 CHEN ET AL. ANTIMICROB.AGENTS CHEMOTHER.
... There is little clinical evidence about what should be the regimen of choice and its adequate dosage in CKD patients 7,8,28,252,253,264,274,276,281,[292][293][294][295][296] . Nevertheless, a series of general recommendations are summarized in Table 20. ...
... NNRTIs, PIs, INSTI and fusion inhibitors do not require a dose adjustment in patients with renal function deterioration 7,8,28,252,253,264,274,276,281,[292][293][294][295][296] . In these patients, fixed-dose drug combinations must be avoided due to drug dosage difficulties. ...
... The α-pyrones are six-membered cyclic unsaturated esters that share chemical and physical properties reminiscent to alkene and aromatic compound. These compounds occur abundantly in naturally occurring molecules and are responsible for vast range of biological activities including antibacterial, antifungal, neurotoxic, and phytotoxic properties (Dickinson, 1993;McGlacken and Fairlamb, 2005) and plant growth-regulating (Kobayashi et al., 1994;Tsuchiya et al., 1997) antitumor (Suzuki et al., 1997;Kondoh et al., 1999), and HIV protease-inhibiting activities (Thaisrivongs et al., 1996;Turner et al., 1998;Chen et al., 2007). ...
... However, on chronic administration of the PIs, which are now almost always co-administered with ritonavir (RTV), the role of biliary excretion of the PIs may be even greater due to CYP3A inactivation [12]. Indeed, this is observed with tipranavir (TPV) after chronic administration of TPV/RTV [13]. Therefore, the role of hepatic transporters could become more significant in the hepatic disposition of PIs under chronic administration of PIs and/or RTV-boosted PI regimen. ...
Article
Full-text available
Although HIV protease inhibitors (PIs) produce profound metabolic interactions through inactivation/inhibition of CYP3A enzymes, their role as victims of transporter-based drug-drug interactions (DDIs) is less well understood. Therefore, we investigated if the PIs, nelfinavir (NFV), ritonavir (RTV), lopinavir (LPV), or amprenavir (APV) were transported into sandwich-cultured human hepatocytes (SCHH), and whether OATPs contributed to this transport. Our findings showed that except for (3) H-APV, no significant decrease in the total hepatocyte accumulation of the (3) H-PIs was detected in the presence of the corresponding unlabeled PI, indicating that the uptake of the other PIs was not mediated. Further, hepatocyte biliary efflux studies using (3) H-APV and unlabeled APV confirmed this decrease to be due to inhibition of sinusoidal influx transporter(s) and not the canalicular efflux transporters. Moreover, this sinusoidal transport of APV was not OATP-mediated. Our results indicate the hepatic uptake of NFV, RTV, or LPV was primarily mediated by passive diffusion. APV's hepatic uptake was mediated by an unidentified sinusoidal transporter(s). Therefore, NFV, RTV or LPV will not be victims of DDIs involving inhibition of hepatic influx transporters; however, the disposition of APV may be affected if its sinusoidal transport is inhibited. Copyright © 2012 John Wiley & Sons, Ltd.
Article
Three novel SEK15-derived polyketides, strepolyketides A-C (1-3), and the known SEK15 (4) were identified from Streptomyces sp. HN2A53. Their structures were established by comprehensive 1D and 2D NMR spectroscopic analysis and HRESIMS data. Compounds 2-3 were the unique dimers of SEK15. Biological evaluation of these compounds showed that compounds 2-4 showed moderate inhibitory activity against influenza virus neuraminidase (NA) in vitro
Article
Eight new α-pyrones 1-8 and three known α-pyrones 9-11 were isolated from three marine-derived Nocardiopsis strains SCSIO 10419, SCSIO 04583, and SCSIO KS107. The structures of compounds 1-8 were elucidated by comprehensive spectral analyses. The absolute configurations of 4-deoxyphomapyrone C (1), 4-deoxy-11-hydroxyphomapyrone C (3), 4-deoxy-7R-hydroxyphomapyrone C (5), and phomapyrone C (11) were determined by TDDFT-ECD calculations for the solution conformers, which revealed that the conformation of the side chain was decisive for the sign of the characteristic high-wavelength ECD transition. (-)-4-Deoxy-8-hydroxyphomapyrone C (4) was isolated from SCSIO 10419 and was deduced as a diastereomeric mixture containing (8S)- and (8R)-4-deoxy-8-hydroxyphomapyrone C in a ratio of 2.6:1 (8R:8S), by chiral-phase HPLC analysis and Mosher's ester analysis. Interestingly, 7-hydroxymucidone (9) was isolated from both SCSIO 04583 and SCSIO KS107, as an enantiomeric mixture containing (7S)-hydroxymucidone (major in 9 from SCSIO 04583) and (7R)-hydroxymucidone (major in 9 from SCSIO KS107). α-Pyrones 3-5 were identified as three isomers of phomapyrone C (11) with diverse hydroxy substitutions. α-Pyrones 10-hydroxymucidone (6), 4-hydroxymucidone (8), and 9, differed in the position of the hydroxy group. Several α-pyrones exhibited moderate growth inhibitory activity against Micrococcus luteus and Bacillus subtilis.
Article
In this update, antiretroviral therapy (ART) is recommended for all patients infected by type 1 human immunodeficiency virus (HIV-1). The strength and grade of the recommendation vary depending on the CD4+ T-lymphocyte count, the presence of opportunistic infections or comorbid conditions, age, and the efforts to prevent the transmission of HIV. The objective of ART is to achieve an undetectable plasma viral load (PVL). Initial ART should comprise three drugs, namely, two nucleoside reverse transcriptase inhibitors (NRTI) and one drug from another family. Three of the recommended regimens, all of which have an integrase strand transfer inhibitor (INSTI) as the third drug, are considered a preferred regimen; a further seven regimens, which are based on an INSTI, an non-nucleoside reverse transcriptase inhibitor (NNRTI), or a protease inhibitor boosted with ritonavir (PI/r), are considered alternatives. The reasons and criteria for switching ART are presented both for patients with an undetectable PVL and for patients who experience virological failure, in which case the rescue regimen should include three (or at least two) drugs that are fully active against HIV. The specific criteria for ART in special situations (acute infection, HIV-2 infection, pregnancy) and comorbid conditions (tuberculosis and other opportunistic infections, kidney disease, liver disease, and cancer) are updated. © 2015 Elsevier España, S.L.U. y Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica.
Chapter
IntroductionDiscoverySynthesisPreclinical DevelopmentClinical DevelopmentConclusions References
Article
Tipranavir (Aptivus®) is a selective nonpeptidic HIV-1 protease inhibitor (PI) that is used in the treatment of treatment-experienced adults with HIV-1 infection. Tipranavir is administered orally twice daily and must be given in combination with low-dose ritonavir, which is used to boost its bioavailability. The durable efficacy of tipranavir, in combination with low-dose ritonavir (tipranavir/ritonavir 500 mg/200 mg twice daily), has been demonstrated in well designed trials in treatment-experienced adults infected with multidrug-resistant strains of HIV-1. In treatment-experienced adults with HIV-1 infection receiving an optimized background regimen, viral suppression was greater and immunological responses were better with regimens containing tipranavir/ritonavir than with comparator ritonavir-boosted PI-containing regimens. The efficacy benefit appeared to be more marked in patients receiving two fully active drugs in the regimen, with the combination of tipranavir/ritonavir and enfuvirtide (for the first time) appearing to be the most successful. Although tipranavir is generally well tolerated, clinical hepatitis and hepatic decompensation, and intracranial haemorrhage have been associated with the drug. Tipranavir also has a complex drug-interaction profile. Thus, tipranavir, administered with ritonavir, is an effective treatment option for use in the combination therapy of adults with HIV-1 infection who have been previously treated with other antiretroviral drugs. Pharmacological Properties Tipranavir is a highly potent and selective nonpeptidic sulfonamide-containing dihydropyrone HIV-1 PI with good in vitro activity against a broad range of laboratory strains and clinical isolates of HIV-1. In vitro, resistance to tipranavir is slow to develop and the process is complex. On-treatment genotyping of viral isolates from treatment-experienced patients in clinical trials indicate that the mutations predominantly emerging with tipranavir treatment are L33F/I/V, V82T/ L and I84V. In vitro, mutations of the HIV-1 wild-type virus that confer resistance to tipranavir also confer resistance to most other commonly used PIs, with the exception of saquinavir and darunavir. The activity of tipranavir is reduced in the presence of human plasma proteins. Synergistic, additive and antagonistic effects have been observed with tipranavir and various other antiretroviral drugs in vitro. Tipranavir is administered in combination with low-dose ritonavir (tipranavir/ ritonavir), which boosts its bioavailability resulting in the achievement of effective plasma/serum concentrations of tipranavir with a twice-daily regimen. Depending on the dose of tipranavir/ritonavir administered, maximum plasma tipranavir concentrations are reached within 1–5 hours and, in the majority of individuals, steady state is reached after approximately 7 days of twice-daily administration. At steady state, tipranavir pharmacokinetics are linear when tipranavir is boosted by low-dose ritonavir. The effect of tipranavir/ritonavir on cytochrome P450 (CYP) isoenzymes, particularly CYP3A, and on P-glycoprotein (P-gp) results in numerous pharmacokinetic interactions between tipranavir/ ritonavir and other drugs. At steady state, tipranavir/ritonavir shows moderate inhibitory effects on hepatic CYP3A4/5, strong inhibitory effects on intestinal CYP3A4/5 and a minimal effect on the activity of P-gp, according to preliminary results of a well designed drug interaction study in healthy volunteers. Therapeutic Efficacy The therapeutic efficacy of oral tipranavir/ritonavir 500 mg/200 mg twice daily versus that of a ritonavir-boosted comparator PI (CPI) [lopinavir, indinavir, saquinavir or amprenavir] has been evaluated in the similarly designed randomized, nonblind, multicentre RESIST (Randomized Evaluation of Strategic Intervention in multidrug reSistant patients with Tipranavir)-1 and RESIST-2 trials in highly antiretroviral-experienced, HIV-1-infected adult patients. All patients also received concurrent treatment with an optimized background regimen (with or without enfuvirtide). Study participants were HIV-1-infected adult patients who had received ≥3 months of previous treatment with a nucleoside reverse transcriptase inhibitor, a non-nucleoside reverse transcriptase inhibitor and a PI. Included patients had also previously received at least two PI-based regimens. At baseline, all patients had a plasma HIV-1 RNA level of ≥1000 copies/mL. Recipients of tipranavir/ritonavir experienced a sustained response to treatment, with a significantly larger proportion of patients achieving a confirmed viral load reduction of ≥1 log10 copies/mL at 48 weeks than recipients of CPIs (primary endpoint; pooled data). In addition, the tipranavir/ritonavir treatment group had a longer time to treatment failure than did patients treated with the CPIs (primary endpoint). Immunological responses, a secondary efficacy measure, were also significantly better in the tipranavir/ritonavir group than in the recipients of CPIs. Preliminary data indicate that the efficacy of tipranavir/ritonavir is sustained for up to 156 weeks. Tolerability In the RESIST trials, adverse events with tipranavir/ritonavir were typical of those seen with the ritonavir-boosted CPIs, despite the higher dosage of ritonavir received by patients in the tipranavir/ritonavir group. Gastrointestinal adverse events were the most common treatment-emergent events with tipranavir/ritonavir and were usually mild. The adverse event rate per 100 patient-exposure years of any adverse event was similar among patients receiving tipranavir/ritonavir and those receiving a CPI/ritonavir. Exposure-adjusted total rates of grade 3 or 4 laboratory abnormalities were also similar in the the two treatment groups. Grade 3 or 4 elevations in triglyceride levels, cholesterol levels and liver enzymes occurred significantly more frequently among patients receiving tipranavir/ritonavir than in the CPI/ritonavir recipients. However, the most highly treatment-experienced patients did not experience grade 3 or 4 AST or ALT elevations during 96 weeks of treatment with tipranavir/ritonavir in five phase IIb/III trials. When reported, grade 3 or 4 AST or ALT elevations were were asymptomatic in the majority of patients. Reports of clinical hepatitis and hepatic decompensation, including some fatalities, have been associated with tipranavir/ritonavir treatment, usually in patients receiving multiple medications and with advanced HIV-1-infection. Caution is recommended and increased liver enzyme monitoring should be considered in patients with liver enzyme abnormalities or hepatitis receiving tipranavir. In addition, tipranavir has been associated with fatal and nonfatal intracranial haemorrhage and should be administered with caution in patients with an increased bleeding risk (see boxed warning in the manufacturer’s prescribing information). However, rates of intracranial haemorrage in a large cohort of US Medi-Cal recipients not taking tipranavir were similar to those observed in premarketing trials of tipranavir. Rashes have been reported in patients receiving tipranavir/ritonavir.
Article
Full-text available
Objective: To update the 2010 recommendations on the evaluation and management of renal disease in HIV-infected patients. Methods: This document was approved by a panel of experts from the AIDS Working Group (GESIDA) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), the Spanish Society of Nephrology (S.E.N.), and the Spanish Society of Clinical Chemistry and Molecular Pathology (SEQC). The quality of evidence and the level of recommendation were evaluated using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system. Results: The basic renal work-up should include measurements of serum creatinine, estimated glomerular filtration rate by CKD-EPI, Urine protein-to-creatinine ratio, and urinary sediment. Tubular function tests should include determination of serum phosphate levels and urine dipstick for glucosuria. In the absence of abnormal values, renal screening should be performed annually. In patients treated with tenofovir or with risk factors for chronic kidney disease (CKD), more frequent renal screening is recommended. In order to prevent disease progression, potentially nephrotoxic antiretroviral drugs are not recommended in patients with CKD or risk factors for CKD. The document advises on the optimal time for referral of a patient to the nephrologist and provides indications for renal biopsy. The indications for and evaluation and management of dialysis and renal transplantation are also addressed. Conclusions: Renal function should be monitored in all HIV-infected patients. The information provided in this document should enable clinicians to optimize the evaluation and management of HIV-infected patients with renal disease.
Article
Full-text available
PNU-140690 is a member of a new class of nonpeptidic human immunodeficiency virus (HIV) protease inhibitors (sulfonamide-containing 5,6-dihydro-4-hydroxy-2-pyrones) discovered by structure-based design. PNU-140690 has excellent potency against a variety of HIV type 1 (HIV-1) laboratory strains and clinical isolates, including those resistant to the reverse transcriptase inhibitors zidovudine or delavirdine. When combined with either zidovudine or delavirdine, PNU-140690 contributes to synergistic antiviral activity. PNU-140690 is also highly active against HIV-1 variants resistant to peptidomimetic protease inhibitors, underscoring the structural distinctions between PNU-140690 and substrate analog protease inhibitors. PNU-140690 retains good antiviral activity in vitro in the presence of human plasma proteins, and preclinical pharmacokinetic studies revealed good oral bioavailability. Accordingly, PNU-140690 is a candidate for clinical evaluation.
Article
Full-text available
A broad screening program previously identified phenprocoumon (1) as a small molecule template for inhibition of HIV protease. Subsequent modification of this lead through iterative cycles of structure-based design led to the activity enhancements of pyrone and dihydropyrone ring systems (II and V) and amide-based substitution (III). Incorporation of sulfonamide substitution within the dihydropyrone template provided a series of highly potent HIV protease inhibitors, with structure-activity relationships described in this paper. Crystallographic studies provided further information on important binding interactions responsible for high enzymatic binding. These studies culminated in compound VI, which inhibits HIV protease with a Ki value of 8 pM and shows an IC90 value of 100 nM in antiviral cell culture. Clinical trials of this compound (PNU-140690, Tipranavir) for treatment of HIV infection are currently underway.
Article
Tipranavir (TPV), a novel nonpeptidic protease inhibitor (NPPI), was administered to treatment-naive HIV-1-infected patients over 14 days in a randomized, multicenter, open-label, parallel-group trial to evaluate the efficacy and tolerability of a self-emulsifying drug delivery system (SEDDS) formulation, in combination with ritonavir (RTV). Of the 31 patients enrolled, 10 were randomized to receive TPV 1200 mg twice daily (TPV 1200), 10 patients received TPV 300 mg + RTV 200 mg twice daily (TPV/r 300/200), and 11 patients received TPV 1200 mg + RTV 200 mg twice daily (TPV/r 1200/200). The median baseline viral load and CD4 cell count were 4.96 log(10) copies/mL and 244 cells/mm(3), respectively. After 14 days, the median decrease in viral load was -0.77 log(10) in the TPV 1200 group, -1.43 log(10) in the TPV/r 300/200 group, and -1.64 log(10) in the TPV/r 1200/200 group. TPV exposure was increased by 24- and 70-fold in the TPV/r 300/200 and 1200/200 groups, respectively, compared with TPV 1200 alone. There were no significant differences across treatment arms with regard to drug-related adverse events. TPV/r appeared to be safe, effective, and well tolerated during 14 days of treatment.
Article
The FDA approved HIV-1 protease inhibitors, ritonavir, saquinavir, and indinavir, are very effective in inhibiting HIV-1 replication, but their long-term efficacy is unknown. Since in vivo efficacy depends on access of these drugs to intracellular sites where HIV-1 replicates, we determined whether these protease inhibitors are recognized by the MDR1 multidrug transporter (P-glycoprotein, or P-gp), thereby reducing their intracellular accumulation. In vitro studies in isolated membrane preparations from insect cells infected with MDR1-expressing recombinant baculovirus showed that these inhibitors significantly stimulated P-gp-specific ATPase activity and that this stimulation was inhibited by SDZ PSC 833, a potent inhibitor of P-gp. Furthermore, photoaffinity labeling of P-gp with the substrate analogue [125I]iodoarylazidoprazosin (IAAP) was inhibited by all three inhibitors. Cell-based approaches to evaluate the ability of these protease inhibitors to compete for transport of known P-gp substrates showed that all three HIV-1 protease inhibitors were capable of inhibiting the transport of some of the known P-gp substrates but their effects were generally weaker than other documented P-gp modulators such as verapamil or cyclosporin A. Inhibition of HIV-1 replication by all three protease inhibitors was reduced but could be restored by MDR1 inhibitors in cells expressing MDR1. These results indicate that the HIV-1 protease inhibitors are substrates of the human multidrug transporter, suggesting that cells in patients that express the MDR1 transporter will be relatively resistant to the anti-viral effects of the HIV-1 protease inhibitors, and that absorption, excretion, and distribution of these inhibitors in the body may be affected by the multidrug transporter.
Article
Both ritonavir and indinavir were readily metabolized by human intestinal microsomes. Comparison of the patterns of metabolites in incubations with enterocyte microsomes and expressed cytochrome P450 (CYP) isozymes and immunoinhibition and chemical inhibition studies showed the essential role of the CYP3A subfamily in the metabolism of both protease inhibitors by the small intestine. Ritonavir was similarly biotransformed by microsomes containing expressed CYP3A4 or CYP3A5 isozymes (KM = 0.05-0.07 microM, Vmax = 1-1.4 nmol/min/nmol CYP). In contrast, both the patterns of metabolites and the enzyme kinetic parameters for the metabolism of indinavir by expressed CYP3A5 (KM = 0.21 microM, Vmax = 0.24 nmol/min/nmol CYP) and CYP3A4 (KM = 0.04 microM, Vmax = 0.68 nmol/min/nmol CYP) were different. The biotransformation of both indinavir and ritonavir in human enterocyte microsomes was characterized by very low KM values (0.2-0.4 microM for indinavir and <0.1 microM for ritonavir). The Vmax for indinavir metabolism was greater in enterocyte (163 +/- 35 pmol/min/mg protein) than in liver (68 +/- 44 pmol/min/mg protein) microsomes. The metabolism of ritonavir in liver and enterocyte microsomes was associated with inactivation of CYP3A. The initial Vmax for ritonavir metabolism by enterocyte microsomes was 89 +/- 59 pmol/min/mg protein. The apparent inactivation rate constants for intestinal CYP3A and expressed CYP3A4 were 0.078 and 0.135 min-1, respectively. Metabolic inactivation of CYP3A by ritonavir explains the improved bioavailability and pharmacokinetics of ritonavir and the sustained elevation of blood levels of other, concomitantly administered, substrates of CYP3A.
Article
Efforts to develop therapeutically relevant HIV protease inhibitors as medicinal agents in confronting the AIDS crisis have been aided by the wealth of fundamental information acquired during related drug discovery campaigns against other aspartyl proteases. This knowledge base was brought to full force with the broad screening identification of small, nonpeptidic, inhibitory molecules as templates for chemical elaboration. Significantly, the ability to collect crystallographic data on the inhibitor-enzyme complexes in a rapid fashion afforded the opportunity for a structure-based approach to drug discovery. Iterative cycles of synthesis, biological testing, and structural information gathering followed by prudent design modifications afforded compounds suitable for clinical evaluation. Displaying high enzymatic inhibition (Ki = 8 pM), potent in vitro antiviral cell culture activity (IC90 = 100 nM), and a useful pharmacokinetic profile, PNU-140690E (Tipranavir disodium) has entered into clinical studies. Promising results from these early trials supported further evaluation of this compound in HIV-infected individuals. PNU-140690E is currently under extensive clinical study.
Article
Although the use of HIV-1 protease inhibitors (PI) has substantially benefited HIV-1-infected individuals, new PI are urgently needed, as broad PI resistance and therapy failure is common. The antiviral activity of tipranavir (TPV), a non-peptidic PI, was assessed in in vitro culture for 134 clinical isolates with a wide range of resistance to currently available peptidomimetic PI. The susceptibility of all 134 variants was then re-tested with the four PI simultaneously with TPV, using the Antivirogram assay. Of 105 viruses with more than tenfold resistance to three or four PI and an average of 6.1 PI mutations per sample, 95 (90%) were susceptible to TPV; eight (8%) had four- to tenfold resistance to TPV and only two (2%) had more than tenfold resistance. The substantial lack of PI cross-resistance to TPV shown by highly PI-resistant clinical isolates makes TPV an attractive new-generation HIV inhibitor.
Article
Tipranavir (TPV), a novel nonpeptidic protease inhibitor (NPPI), was administered to treatment-naive HIV-1-infected patients over 14 days in a randomized, multicenter, open-label, parallel-group trial to evaluate the efficacy and tolerability of a self-emulsifying drug delivery system (SEDDS) formulation, in combination with ritonavir (RTV). Of the 31 patients enrolled, 10 were randomized to receive TPV 1200 mg twice daily (TPV 1200), 10 patients received TPV 300 mg + RTV 200 mg twice daily (TPV/r 300/200), and 11 patients received TPV 1200 mg + RTV 200 mg twice daily (TPV/r 1200/200). The median baseline viral load and CD4 cell count were 4.96 log10 copies/mL and 244 cells/mm, respectively. After 14 days, the median decrease in viral load was -0.77 log10 in the TPV 1200 group, -1.43 log10 in the TPV/r 300/200 group, and -1.64 log10 in the TPV/r 1200/200 group. TPV exposure was increased by 24- and 70-fold in the TPV/r 300/200 and 1200/200 groups, respectively, compared with TPV 1200 alone. There were no significant differences across treatment arms with regard to drug-related adverse events. TPV/r appeared to be safe, effective, and well tolerated during 14 days of treatment.