You are on page 1of 9

CLINICAL SCIENCE

Lopinavir/Ritonavir Induces the Hepatic Activity of


Cytochrome P450 Enzymes CYP2C9, CYP2C19, and
CYP1A2 But Inhibits the Hepatic and Intestinal Activity
of CYP3A as Measured by a Phenotyping Drug Cocktail
in Healthy Volunteers
Downloaded from https://journals.lww.com/jaids by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3/TxXCJJj86E4+NUV8xxlfwFYSWzFE+IuPoeELhGvCXQ= on 04/27/2020

Rosa F. Yeh, PharmD,* Vincent E. Gaver, PharmD,* Kristine B. Patterson, MD,Þ Naser L. Rezk, MS,*
Faustina Baxter-Meheux, BS,* Michael J. Blake, MD, PhD,þ Joseph J. Eron, Jr, MD,Þ
Cheri E. Klein, PhD,§ John C. Rublein, PharmD,§ and Angela D.M. Kashuba, PharmD*

ratios. Hepatic and intestinal + hepatic CYP3A activities were


Objective: The effect of lopinavir/ritonavir (LPV/r) administration
quantified by IV (CL) and PO (CL/F) MDZ clearance, respectively.
on cytochrome P450 (CYP) enzyme activity was quantified using a
Results: After LPV/r therapy, CYP2C9, CYP2C19, and CYP1A2 ac-
phenotyping biomarker cocktail. Changes in CYP2C9, CYP2C19,
tivity increased by 29%, 100%, and 43% (P = 0.001, 0.046, and 0.001),
CYP3A, CYP1A2, N-acetyltransferase-2 (NAT-2), and xanthine
respectively. No changes were seen in NAT-2 or XO activity. Hepatic
oxidase (XO) activities were evaluated using warfarin (WARF) +
and intestinal + hepatic CYP3A activity decreased by 77% (P G 0.001)
vitamin K, omeprazole (OMP), intravenous (IV) and oral (PO)
and 92% (P = 0.001), respectively.
midazolam (MDZ), and caffeine (CAF).
Conclusion: LPV/r therapy results in modest induction of
Design: Open-label, multiple-dose, pharmacokinetic study in
CYP1A2 and CYP2C9 and potent induction of CYP2C19 activity.
healthy volunteers.
Increasing doses of concomitant medications metabolized by these
Methods: Subjects (n = 14) simultaneously received PO WARF
enzymes may be necessary. LPV/r inhibited intestinal CYP3A
10 mg, vitamin K 10 mg, OMP 40 mg, CAF 2 mg/kg, and IV
to a greater extent than hepatic CYP3A activity. Doses of
MDZ 0.025 mg/kg on days (D) 1 and 14, and PO MDZ 5 mg on
concomitant CYP3A substrates should be reduced when combined
D2 and D15. LPV/r (400/100 mg twice daily) was administered on
with LPV/r, although intravenously administered compounds may
D4-17. CYP2C9 and CYP2C19 activities were quantified by S-WARF
require less of a relative dose reduction than orally administered
AUC0-inf and OMP/5-hydroxy OMP ratio, respectively. CYP1A2,
compounds.
NAT-2, and XO activities were quantified by urinary CAF metabolite
Key Words: lopinavir, ritonavir, pharmacokinetics, protease
inhibitors, cytochrome P450 enzyme system, induction, inhibition
Received for publication November 13, 2005; accepted February 1, 2006.
From the *School of Pharmacy, and †School of Medicine, University of
(J Acquir Immune Defic Syndr 2006;42:52Y60)
North Carolina at Chapel Hill, NC; ‡The Division of Pediatric
Pharmacology and Medical Toxicology, Pediatric Pharmacology Re-
search Unit, Children’s Mercy Hospital and Clinics, Kansas City, MO;
and §Abbott Laboratories, Clinical Pharmacokinetics and Antiviral
Global Team, Abbott Park, IL.
I n the treatment of HIV infection, drug interactions can be
detrimental to effective antiretroviral therapy as well as to
the treatment of other disease processes. HIV-infected
Funding source: Research grant support was provided by Abbott Laborato-
ries, the UNC Center for AIDS Research (P30A150410), the UNC patients usually take at least 3 antiretroviral agents and may
General Clinical Research Center (RR00046), the UNC Clinical Research receive other medications for supportive care, opportunistic
Nutrition Research Center (DK56350), AI54980 (A.D.M.K.), and the infections, and immunomodulation, in addition to using
Pediatric Pharmacology Research Unit, Children’s Mercy Hospitals and nutraceutical compounds for general well-being. Due to the
Clinics (grant 5 U10 HD01313-12, National Institute of Child Health and
Human Development, M.J.B.).
multiple effects of the nonnucleoside reverse transcriptase
Prior presentation: This research has been presented in part at the 5th inhibitors and protease inhibitors (PIs) on the cytochrome
International Workshop on Clinical Pharmacology of HIV Therapy, Rome, P450 (CYP) enzyme system, drug interactions are often
Italy, on April 1Y3, 2004, and the 6th International Workshop on Clinical unavoidable.1 The fixed-dose formulation of lopinavir (LPV)
Pharmacology of HIV Therapy, Quebec, Canada, on April 28Y30, 2005. and ritonavir (RTV) utilizes RTV’s potent inhibition of
Conflict of interest statement: Dr Eron is an ad hoc consultant to Abbott
Laboratories and receives support for conduct of clinical trials sponsored by CYP3A activity to enhance the systemic exposure of LPV.2
Abbott Laboratories through the University of North Carolina. Drs Klein CYP3A is the major enzyme subfamily responsible for the
and Rublein are employed by Abbott Laboratories. The remaining authors metabolism of most PIs and nonnucleoside reverse transcrip-
do not have conflicts of interest related to this research or manuscript. tase inhibitors. It is clear that lopinavir/ritonavir (LPV/r)
Reprints: Angela D.M. Kashuba, PharmD, 3318 Kerr Hall, CB#7360, School of
Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
affects CYP3A enzyme activity, as many drug interactions
27599 (e-mail: akashuba@unc.edu). have been noted between LPV/r and other CYP3A sub-
Copyright * 2006 by Lippincott Williams & Wilkins strates.3Y5 However, very little data are available on the effect

52 J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006 Lopinavir/Ritonavir and Cytochrome P450 Activity

of LPV/r on other CYP isoforms. In vitro experiments suggest Study Design


no significant inhibitory effects on other CYP isoenzymes,6 This was an open-label, pharmacokinetic (PK) study
but inductive effects have not been carefully studied. A consisting of two 48-hour inpatient visits at the Verne S.
number of clinical reports have shown decreased exposure to Caviness General Clinical Research Center (GCRC) at UNC
phenytoin,7 warfarin,8,9 methadone,10Y13 and ethinyl estradiol,10 Hospitals and 2 follow-up outpatient visits (days 4 and 17).
but the mechanisms for these are unclear. The duration of the study was 17 days. As previous
The primary objective of this study was to quantify the investigations have not noted significant menstrual cycle
influence of 10 days of LPV/r administration on the activity of effects on phenotyping biomarker data, this study did not
the 4 CYP isozymes responsible for metabolizing the majority control for menstrual cycle.17Y22
of drugs currently available. To accomplish this, we adminis- On days 1 and 14, all subjects were admitted to the
tered a modified BCooperstown 5 + 1 cocktail^ consisting of GCRC. After a peripheral IV line was placed for blood
caffeine (CAF, a substrate for CYP1A2, N-acetyltransferase-2 collection, patients emptied their bladder and were dosed
[NAT-2], and xanthine oxidase [XO]), warfarin (WARF, a with the phenotyping medications. On the first day of each PK
substrate for CYP2C9), omeprazole (OMP, a substrate for visit (days 1 and 14), each subject received simultaneous
CYP2C19), and intravenous (IV) and oral (PO) midazolam administration of 4 PO medications (CAF 2 mg/kg, WARF
(MDZ, a substrate for CYP3A, but not P-glycoprotein).14,15 10 mg, vitamin K 10 mg, and OMP 40 mg) and 1 IV medication
These data were generated to further understand currently (MDZ 0.025 mg/kg). CAF metabolites were continuously
documented drug interactions with LPV/r and to predict the collected in urine over 12 hours. Blood samples for WARF
extent of as-yet-undocumented drug interactions. were collected just before WARF administration and then at 3,
6, 9, 12, 24, 48, and 72 hours afterward. Blood samples for OMP
METHODS and 5-hydroxy metabolite (5-OH OMP) were collected 2 hours
after OMP administration. Blood samples for IV MDZ for the
Subjects first PK visit were collected just before MDZ administration
Sixteen volunteers were screened and assessed to be and at 5, 15, and 30 minutes and 1, 1.5, 2, 3, 4, 6, 8, 12, and
healthy by physical examination, medical history, and 24 hours afterward.
laboratory evaluations. Subjects weighed at least 50 kg, During inpatient visits, standardized research meals
were 18 to 45 years of age, and were HIV-1 seronegative. were served consisting of approximately 15% protein, 30%
Subjects were excluded if they had significant medical fat, and 55% carbohydrate for a total daily intake of 2000 to
conditions or laboratory abnormalities; had a history of 2500 calories. Meals were finished within 30 minutes and
allergy to any study medication; were 20% more or less than given at least 2 hours after administration of phenotyping
their ideal body weight; used any prescription or over-the- medications. Nutrient intake analysis was performed by a
counter medications, vitamin, or herbal supplements on a dietitian after each meal. No additional CAF was permitted.
regular basis; had a history of alcohol or drug abuse; were Twenty-four hours after the first phenotyping medica-
unable to abstain from alcohol or grapefruit juice for the tions were administered, on days 2 and 15, each subject
duration of the study; used tobacco products within 2 months received an PO dose of MDZ 5 mg. Blood samples for PO
before the first study visit; or had a baseline international MDZ were collected just before MDZ administration and at
normalized ratio (INR) greater than 1.2. Sexually active 5, 15, and 30 minutes and 1, 1.5, 2, 3, 4, 6, 8, 12, 18, and
women were required to have a negative urine pregnancy 24 hours afterward.
test and to use a barrier form of birth control for the duration Subjects were discharged from the GCRC approxi-
of the study. The University of North Carolina (UNC) mately 24 hours after the PO MDZ dose, on days 3 and 16.
institutional review board approved the study protocol, and Subjects returned 24 hours later, on days 4 and 17, for an
all subjects provided written informed consent before any outpatient visit to obtain a 72-hour WARF blood sample. At
study procedures. day 4 outpatient visit, subjects took their first dose of LPV/r
soft gelatin capsules, administered orally at the usual clinical
Materials dose of 400 LPV/100 mg RTV twice daily. Subjects were
LPV/r (Kaletra) soft gelatin capsules were obtained from advised to take LPV/r with food. Medication adherence for
Abbott Laboratories (Abbott Park, IL). CAF 2-mg/mL PO this 10-day period was assessed by pill counts performed by
solution (formulated from CAF citrate purified powder, the study coordinator on day 15 and dosing administration
Mallinckrodt, Phillipsburg, NJ), WARF 10-mg tablets (war- cards completed daily by the study subject. These data were
farin, Dupont Pharmaceuticals, Newark, DE), vitamin K 10-mg reviewed by the study coordinator before initiation of the
tablets (Mephyton, Merck and Co, Inc, Whitehouse Station, second inpatient study visit (day 14). Subjects with less than
NJ), OMP 40-mg capsules (Prilosec, AstraZeneca, Wilming- 95% adherence were removed from the study. In addition, to
ton, DE), MDZ 5-mg tablets (MDZ, Bedford Laboratories, further assess adherence, a blood sample for LPV and RTV
Bedford, OH), and MDZ 1-mg/mL IV solution (MDZ 2-mL concentrations was drawn upon GCRC admission, before
vials, Bedford Laboratories, Bedford, OH) were dispensed administration of the phenotyping medications. LPV/r was
from the UNC Hospitals Investigational Drug Service continued through day 17. During the second PK visit (PK 2,
pharmacy. Vitamin K was administered to prevent the phar- days 14Y17), phenotyping procedures were performed in the
macologic effect of WARF without altering the pharmacoki- same manner as during days 1 to 4 (PK 1). During PK 2,
netics of the WARF stereoisomers.16 LPV/r administration was directly observed and given with

* 2006 Lippincott Williams & Wilkins 53

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Yeh et al J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006

food, at least 2 hours after administration of the phenotyping validated reverse-phase HPLC method with ultraviolet
medications. detection.26 Briefly, 300 KL of plasma was combined with
Safety assessment at each PK visit included blood 3 KL of 100 Kg/mL racemic ethyl-warfarin internal
laboratory testing (complete blood count with differential, standard in nanopure water. Samples were extracted using
prothrombin time/INR, chemistry panel, liver function tests, and BOND ELUT C18 solid-phase extraction column (100 mg,
urine pregnancy test), physical examination by study physician, 10 mL; Varian, Harbor City, CA) and evaporated under
and adverse event assessment by the study coordinator. With nitrogen at 45-C. Samples were reconstituted in 150 KL of
MDZ, respiratory rate, oxygenation, and mean arterial pressure a 50% acetonitrile/50% glacial acetic acid (0.5%) solution
(MAP) were monitored every 15 minutes for 60 minutes after IV before injection onto a Agilent 1100 HPLC system. Isomer
administration and every 15 minutes for 120 minutes after PO separation was accomplished using a (R,R) Whelk-01 chiral
administration. Flumazenil was available at the bedside during column (5.0 Km, 250  4.6 mm; Regis Technologies,
the entire inpatient stay. Adverse events were assessed using the Morton Grove, IL) with a 5/100 Nitrile Guard Cartridge
Adult AIDS Clinical Trials Group grading scale23 at initiation (Regis Technologies) using gradient elution with a flow rate
and conclusion of both inpatient visits and outpatient visits. of 1.0 mL/min. Calibration standard curves ranged from 25
Subjects were contacted by the study coordinator during the first to 3000 ng/mL. Intraday and interday coefficients of
week of LPV/r administration to ensure medication tolerability variation were less than 10%.
and to assess any early adverse event occurrences.
Omeprazole
Analytical Methods Concentrations of OMP and its CYP2C19-mediated
Blood samples were collected in vacutainers containing 5-OH OMP were determined from blood plasma samples
8.55 mg K3EDTA (15% additive solution) as an anticoagulant using a modified, validated reverse-phase HPLC method
and kept on ice after collection for a maximum of 15 minutes. with ultraviolet detection.27 Briefly, 500 KL of plasma was
Blood plasma was separated by centrifugation at 2800 rpm for combined with 500 KL of 0.25 Kg/mL phenactin internal
15 minutes at 4-C. During the study visit, plasma was stored at standard in 100 mmol/L ammonium acetate buffer, pH 7.0.
the GCRC at j20-C. At the end of the study visit, frozen Samples were extracted using BOND ELUT-C8 columns
plasma samples were transferred to a temperature-monitored (100 mg, 1.0 mL; Varian). Samples were evaporated using a
freezer, j80-C, for storage until analysis. Turbo Vap LV Evaporator (Zymark Corp, Hopkinton, MA)
Urine containers were kept refrigerated at 4-C during the at 30-C for 30 minutes and reconstituted with 100 KL of
12-hour urine collection. After collection, total urine volume 15% acetonitrile/85% HPLC-grade water (pH = 12) before
was measured, and 15-mL aliquots of urine were combined injection onto an Agilent 1100 HPLC system. Samples were
with additional ascorbic acid to maintain a pH of less than 4. run on a XTerra MS-C8 column (3.5 Km, 3.9  100 mm;
These samples were frozen at j80-C until analysis. Waters Scientific, Franklin, MA) with a XTerra MS-C8 Guard
column (3.5 Km, 3  20 mm; Waters Scienific) using gradient
Caffeine elution with a flow rate of 800 KL/min. Calibration standard
Urinary concentrations of 5-acetylamino-6-formyl- curves ranged from 5 to 1000 ng/mL. Intraday and interday
amino-3-methyluracil (AFMU), 1-methylxanthine (1X), coefficients of variation across the range of concentrations
1-methyluric acid (1U), and 1,7-dimethyluric acid (17U) were less than 5% for both OMP and 5-OH OMP.
were determined by reverse-phase high-performance
liquid chromatography with diode-array ultraviolet detec- Midazolam
tion (Agilent 1100, Agilent Technologies, Palo Alto, Concentrations of MDZ were determined using a
CA).7,24,25 Urine was filtered through a 0.22-Km nylon modified, validated reverse-phase HPLC method with mass
centrifuge tube filter (Spin-X, Costar, Acton, MA) before spectroscopy detection.28 Briefly, 300 KL of plasma was added
analysis. Samples (20 KL) were injected onto the HPLC to 100 KL of 0.01 Kg/mL alprazolam internal standard in
system with analytes of interest eluted on a C18 column methanol. Samples were extracted with 1.5 mL tert-butyl
(4.6  250 mm, 5 Km; Aqua-C18, Phenomenex, Torrance, ether, evaporated to dryness using a Turbo Vap LV Evaporator
CA) using a mobile phase of methanol/0.1% acetic acid at 30-C for 10 minutes, and reconstituted with 100 KL of an
(85:15) at a flow rate of 1 mL/min. Eluates were monitored acetonitrile/water/acetic acid mixture before injection onto an
by UV detection at 275 and 290 nm with CAF and Agilent 1100 LCMSD system. Samples were run on a Luna C8
metabolite concentrations determined against calibration column (3.0 Km, 2  100 mm; Phenomenex, Torrance, CA)
standards. A 7-point standard curve using peak heights was using a gradient elution with a flow rate of 200 KL/min.
used to assess analyte concentrations. Standard curves for Calibration standard curves ranged from 0.1 to 500 ng/mL.
all metabolites were linear over a range of 6 to 200 mmol/L Intraday and interday coefficients of variation across the range
(r2 9 0.99). The coefficient of variation for all standards was of concentrations were less than 8%.
less than 10% except for the lower limit of quantitation
which was consistently less than 20%. Lopinavir and Ritonavir
LPV and RTV concentrations were measured individually
Warfarin using plasma obtained from the second inpatient visit using a
Concentrations of warfarin R- and S-isomers were validated HPLC method with ultraviolet detection.25,29 Briefly,
determined from blood plasma samples using a modified, 550 KL of plasma was combined with 550 KL of 1.0 mg/mL

54 * 2006 Lippincott Williams & Wilkins

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006 Lopinavir/Ritonavir and Cytochrome P450 Activity

TABLE 1. CYP Phenotyping Results Before and After LPV/r Administration


Results*
Enzyme Phenotyping Measure Before LPV/r (PK 1) After LPV/r (PK 2) GMR (95% CI)
j1
CYP2C9 S-warfarin AUC (Kg I h I mL ) 14.97 (11.22Y22.13) 10.65 (8.66Y14.58) 0.71 (0.65Y0.79)*
CYP2C19 (PM included) OMP/5-OH OMP 1.55 (0.50Y4.19) 0.57 (0.27Y1.22) 0.15 (0.17Y1.03)*
CYP2C19 (NO PM) OMP/5-OH OMP 1.31 (0.78Y2.38) 0.57 (0.31Y1.32) 0.47 (0.20Y0.98)*
CYP3A (hepatic) IV MDZ CL (mL I minj1 I kgj1) 3.66 (3.15Y4.51) 0.83 (0.71Y1.03) 0.23 (0.18Y0.31)*
CYP3A (intestinal and hepatic) PO MDZ CL/F (mL I minj1 I kgj1) 13.73 (10.94Y19.64) 1.10 (0.96Y1.31) 0.08 (0.07Y0.11)*
CYP1A2 (1X + 1U + AFMU)/17U 4.64 (4.06Y5.53) 6.61 (5.71Y8.12) 1.43 (1.34Y1.54)*
NAT-2 (PM included) AFMU/(1X + 1U + AFMU) 0.10 (0.13Y0.36) 0.09 (0.12Y0.34) 0.95 (0.09Y2.90)
NAT-2 (NO PM) AFMU/(1X + 1U + AFMU) 0.20 (0.15Y0.38) 0.19 (0.14Y0.36) 0.96 (0.002Y3.06)
XO 1U/(1X + 1U) 0.70 (0.68Y0.72) 0.72 (0.69Y0.76) 1.03 (0.98Y1.09)
Results are expressed as geometric mean (95% CI). PK indicates pharmacokinetic visit.
*P G 0.05.

MDZ internal standard. Samples were extracted using BOND as the geometric mean (95% confidence interval [CI]),
ELUT-C18 columns (1.0 mL, 100 mg; Varian), evaporated to unless otherwise stated.
dryness using a Turbo Vap LV Evaporator at 40-C, and
reconstituted in 100 KL of mobile phase before injection onto an
Agilent 1100 HPLC system. Samples were separated on a
RESULTS
Zorbax C18 analytical column (3.5 Km, 150  4.6 mm; Agilent,
Wilmington, DE) with a Zorbax C18 (3.5 Km, 12.5  4.6 mm; Baseline Characteristics
Agilent) guard column and facilitated via a gradient elution. Twenty-six subjects were screened, and 16 subjects
Calibration standard curves ranged from 25 to 5000 ng/mL. were enrolled into the study. One subject was withdrawn from
Intraday and interday coefficients of variation were less than 6% the study after 10 days of LPV/r due to the development of a
for all analytes. grade 2 rash. This resolved upon discontinuation of LPV/r
and administration of diphenhydramine and prednisone. An
additional subject was withdrawn due to poor LPV/r
Data Analysis and Statistical Methods adherence. Therefore, 14 subjects were used in the analysis
CYP1A2, NAT-2, and XO activities were defined by of CYP1A2, NAT-2, XO, CYP2C9, and CYP3A activities.
the urinary ratios of (1X + 1U + AFMU)/17U, AFMU/(1X + Of these subjects, 3 were white males, 4 were white females,
1U + AFMU), and 1U/(1X + 1U), respectively. CYP2C9 3 were African American males, and 4 were African
activity was quantified by S-WARF AUC 0-inf , and American females. Average age, weight, and height were 26.5
CYP2C19 activity was quantified by the OMP/5-OH OMP (23.9Y29.9) years, 70.3 (63.3Y80.0) kg, and 173.8 (168.1Y
ratio 2 hours postdose. Pharmacokinetic parameters for 180.1) cm, respectively. Because 3 subjects had no detectable
MDZ and WARF were derived from plasma concentrations serum OMP or 5-OH OMP at either the baseline PK visit or
using noncompartmental methods in WinNonLinPro V4.0.1 the treatment PK visit, 11 subjects were used to quantify
(Pharsight Corp, Mountain View, CA). Hepatic CYP3A CYP2C19 activity.
activity was quantified by IV MDZ clearance (CL) and All subjects were extensive metabolizers by phenotype
intestinal + hepatic CYP3A activity by the MDZ apparent for CYP1A2,17,30,31 XO,17,30,31 and CYP2C9.32 One white
oral clearance (CL/F). female was a CYP2C19 poor metabolizer (PM) by phenotype
A priori sample size calculations determined that 13 (having an OMP/5-OH OMP ratio 96),33,34 and 1 African-
evaluable study subjects would provide 80% power or more American female was an NAT-2 PM by phenotype [having an
to detect a 30% change in intraindividual phenotyping AFMU/(1X + 1U + AFMU) ratio 90.5].17,30,31
measures, assuming a type I error of 0.05. Statistical
calculations were performed using SAS JMP 5.0 (SAS,
Cary, NC) by either the paired Student t test or Wilcoxon Pharmacokinetic Analysis
rank sum test, depending on the distribution of the data. A summary of the phenotyping measures for each drug-
Geometric mean ratios (GMRs) were calculated to compare metabolizing enzyme before and after LPV/r administration
PK parameter changes within subjects between the 2 study and the GMRs are shown in Table 1. Figures 1A to C and
periods (PK 2/PK 1). Predictors of both baseline CYP Figures 2A and B illustrate individual subject changes in
activity and the magnitude of change in CYP activity with these markers upon exposure to LPV/r.
LPV/r exposure were evaluated using multivariate linear Upon LPV/r exposure, CYP1A2 metabolic ratios (Fig. 1A)
regression and age, sex, race, body mass index, and LPV increased significantly by 43% (34%Y54%, P = 0.001), from a
and RTV concentrations as covariates. A P value of less ratio of 4.64 (4.06Y5.53) to a ratio of 6.61 (5.71Y8.12). NAT-2
than 0.05 was considered significant. All data are presented and XO metabolic ratios did not change. NAT-2 metabolic

* 2006 Lippincott Williams & Wilkins 55

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Yeh et al J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006

FIGURE 1. A, CAF metabolic ratio (CYP1A2 activity): PK 1 vs.


PK 2. Individual volunteer data are represented by the dotted
lines, and geometric mean data are represented by the solid
line. B, S-WARF AUC0-inf (CYP2C9 activity): PK 1 vs. PK 2.
Individual volunteer data are represented by the dotted lines,
and geometric mean data are represented by the solid line.
C, OMP metabolic ratio: PK 1 vs. PK 2 (CYP2C19 activity).
Individual volunteer data are represented by the dotted lines,
and geometric mean data are represented by the solid line.
Geometric mean line does not include PM. PK indicates
pharmacokinetic visit.

ratios were 0.20 (0.15Y0.38) before LPV/r and 0.19 (0.14Y0.36) IV MDZ CL was used as a surrogate for hepatic
after LPV/r therapy (P = 0.34). XO metabolic ratios were 0.70 CYP3A activity, and the apparent oral clearance of MDZ
(0.68Y0.72) before LPV/r and 0.72 (0.69Y0.76) after LPV/r (CL/F) was used as a measure of combined intestinal and
therapy (P = 0.20). hepatic CYP3A activity. Upon exposure to LPV/r, IV MDZ
CYP2C9 activity was measured by S-WARF expo- CL and PO MDZ CL/F both decreased significantly,
sure (Fig. 1B). After LPV/r therapy, S-WARF AUC0-inf suggesting a potent inhibitory effect on CYP3A activity in
significantly declined by 29% (17%Y34%, P = 0.001), from both organs. IV MDZ CL declined by 77% (69%Y82%, P G
14.97 (11.22Y22.13) Kg I h I mLj1 to 10.65 (8.66Y14.58) 0.001), from 3.66 (3.15Y4.51) mL I minj1 I kgj1 to 0.83
Kg I h I mLj1. This represents a moderate increase in (0.71Y1.03) mL I minj1 I kgj1 (Fig. 2A). PO MDZ CL/F
CYP2C9 activity. Although not used as a CYP biomarker, declined by 92% (89%Y93%, P = 0.001), from 13.73 (10.94Y
R-WARF AUC0-inf was also calculated for each study visit. 19.64) mL I minj1 I kgj1 to 1.10 (0.96Y1.31) mL I minj1 I kgj1
R-WARF exposure significantly decreased by 37% (23%Y46%, (Fig. 2B). As seen in Figure 3, significant correlations were
P = 0.001), from 29.31 (22.17Y44.14) Kg I h I mLj1 to 18.39 noted between baseline hepatic CYP3A activity and the
(14.54Y27.86) Kg I h I mLj1. magnitude of LPV/r-induced hepatic inhibition (r2 = 0.44, P =
CYP2C19 activity was estimated using an OMP 0.01) as well as between baseline intestinal + hepatic CYP3A
metabolic ratio (Fig. 1C). Among CYP2C19 extensive activity and the magnitude of LPV/r-induced inhibition (r2 =
metabolizers, the OMP metabolic ratio decreased signifi- 0.59, P = 0.001).
cantly by 53% (12%Y80%, P = 0.046), from 1.31 (0.78Y2.38)
to 0.57 (0.31Y1.32). This represents a 100% increase in Medication Adherence
CYP2C19 activity. In the CYP2C19 PM, the OMP ratio also Based on dose administration cards and pill counts,
decreased from 10.0 to less than 1. These data suggest a medication adherence ranged from 95% to 100%. LPV
significant increase in CYP2C19 activity, regardless of and RTV concentrations were measured using serum
phenotype. samples collected upon admission for the PK 2 visit.

56 * 2006 Lippincott Williams & Wilkins

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006 Lopinavir/Ritonavir and Cytochrome P450 Activity

(86%) of 14 subjects; most by day 5 of LPV/r therapy. Two


subjects reported diarrhea throughout the study, but did not
report any diarrhea during their PK visits.
Both before and after LPV/r administration, no adverse
effects were associated with OMP, WARF, or vitamin K
administration. With MDZ administration, subjects displayed
either no or mild sedation. Respiratory rate decreased by a mean
of 2 (range, j6 to 0) breaths/min after the IV dose, and a mean
of 2 (range, j4 to 0) breaths/min after the PO dose. MAP
decreased by a mean of 4 (range, j20 to +5) mm Hg and
8 (range, j29 to 0) mm Hg after the IV and PO doses,
respectively. Subjects whose MAP decreased by 20 and 29 mm
Hg were otherwise asymptomatic of MDZ adverse events, and
the extent of MAP decrease was not dependent on LPV/r
administration. Flumazenil was not administered to any subject.

DISCUSSION
Treatment of HIV infection involves the use of multiple
antiretroviral medications. The number of drug interactions in
this population is unprecedented because many of these
compounds are metabolized by, and influence the activity of,
drug-metabolizing enzymes and transporters. Patients with
HIV infection often have concomitant disease states requiring
additional medication therapy which may also interact with
antiretroviral medications. Interactions are of particular con-
cern with drugs having narrow therapeutic indices, such as
anticonvulsants or anticoagulants. CYP phenotyping is a useful
tool for understanding and predicting these drug interactions.
Using phenotyping methods in healthy volunteers, we
found that 10 days of LPV/r therapy resulted in a 43% increase
in CYP1A2 activity, a 29% increase in CYP2C9 activity, and
an 100% increase in CYP2C19 activity. The degree of
CYP2C19 and CYP2C9 induction seen in this investigation is
consistent with in vitro data using prototypical inducers such
FIGURE 2. A, IV MDZ CL: PK 1 vs. PK 2 (hepatic CYP3A
activity). Individual volunteer data are represented by the
dotted lines, and geometric mean data are represented by the
solid line. B, PO MDZ CL/F: PK 1 vs. PK 2 (hepatic + intestinal
CYP3A activity). Individual volunteer data are represented by
the dotted lines, and geometric mean data are represented by
the solid line. PK indicates pharmacokinetic visit.

Subjects had samples obtained 8.4 T 0.6 hours (mean T


SD) after a self-recorded, nonobserved dose. LPV con-
centrations were 7.26 (5.86Y10.02) Kg/mL, and RTV
concentrations were 0.49 (0.36Y0.98) Kg/mL. These were
within the expected concentration range, as seen in
historical cohorts after observed dosing.4,7

Adverse Events
All study medications were generally well tolerated,
and all adverse effects were grade 2 or less, consistent with
the adverse reaction profile of LPV/r.4 The most common
FIGURE 3. Correlation between baseline MDZ clearance and
adverse event associated with LPV/r was diarrhea. All 14 the magnitude of change after 10 days of LPV/r. IV MDZ CL
subjects experienced diarrhea, classified as grade 1 in 11 (CYP3A hepatic activity) is represented by the open circles
subjects (79%) and as grade 2 in 3 subjects (21%). (y = 0.48 j 0.06x, r = 0.66, P = 0.01 [solid line]), and PO MDZ
Loperamide was required for symptom relief in 4 of 11 CL/F (CYP3A hepatic + intestinal activity) is represented by the
subjects with grade 1 diarrhea and 3 of 3 subjects with grade 2 closed circles (y = 0.15 j 0.004x, r = 0.77, P = 0.001 [dotted
diarrhea. Diarrhea resolved before the second PK visit in 12 line]). PK indicates pharmacokinetic visit.

* 2006 Lippincott Williams & Wilkins 57

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Yeh et al J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006

as rifampicin and phenobarbital, which show greater increases some in vitro data show that methadone is primarily
in CYP2C19 activity than increases in CYP2C9 activity.35 metabolized by CYP3A4 and, to a lesser extent, CYP2D6,44
Conversely, hepatic CYP3A activity declined by 77%, and other studies surmise that the decreased exposures may be due
intestinal + hepatic CYP3A activity decreased by 92%. No to induction of CYP2B6, CYP2C9, CYP2C19, and/or glucur-
effects were noted on NAT-2 or XO activities. Although there onidation pathways.13,42,43,45 Our data suggest that methadone
were no apparent differences in response associated with unlikely utilizes CYP3A4 as the primary pathway of metab-
weight, sex, race, age, and LPV or RTV concentrations (data olism (as LPV/r was found to be an overall inhibitor of
not shown), our sample size was not sufficient to conduct a CYP3A4) and provide support for the role of CYP2C induction
robust analysis. in decreasing methadone exposures. However, as with
To our knowledge, this is the first clinical study to show phenytoin, protein-binding interactions or stereoselective
induction of CYP1A2 by LPV/r. Commonly used CYP1A2 effects may also be involved.43,46
substrates include theophylline, antipsychotics (eg, clozapine, The results of this investigation also suggest that
haloperidol, and olanzapine), and tricyclic antidepressants concomitant LPV/r therapy may increase the risk of failure
(eg, amitriptyline). RTV at a dose of 500 mg twice daily has during treatment with drugs metabolized primarily by
been shown to decrease exposure to theophylline36 by 43% CYP2C19. The increase in CYP2C19 activity observed in the
and olanzapine37 by 53%. Although the RTV dose in these present study is consistent with other clinical data, such as the
previous studies was greater than what was used in the current LPV/r-mediated increase in AUC of the CYP2C19-generated
study (and in current clinical practice), the magnitude of nelfinavir M8 metabolite.4,47 Other commonly used CYP2C19
induction was similar. Antidepressant and antipsychotic substrates include diazepam,48 proton pump inhibitors (esome-
agents are commonly used in HIV-infected patients, and prazole, lansoprazole, and pantoprazole),49Y51 and antidepres-
induction of CYP1A2 by PI combinations which include sants (citalopram, sertraline, and amitriptyline).52,53 Interestingly,
RTV may result in decreased efficacy of these agents. the one subject phenotyped as a PM of CYP2C19 also exhibited a
As determined by S-WARF exposure, LPV/r modestly large degree of CYP2C19 induction. This finding is unusual, as
induced CYP2C9 activity. In vitro data from human liver PMs of other CYP isozymes are purported to have nonfunctional
microsomes suggest that LPV/r is a very weak inhibitor of alleles.54 However, 2 recent studies have found similar induction
CYP2C9 (Ki = 13.7Y23.0 Kmol/L [21.8Y36.6 Kg/mL]). potential in CYP2C19 PMs given the herbal product Ginkgo
However, the microsome system lacks the cellular machinery biloba.55,56 This phenomenon needs further investigation, but
necessary to evaluate induction potential.6 S-WARF accounts some hypotheses include induction of a non-CYP2C19 hydrox-
for approximately 70% of the overall anticoagulation response ylase or the presence of inducible, low-level CYP2C19 activity.56
with warfarin,38 and subtherapeutic INR values have been Consistent with in vitro and in vivo reports, LPV/r
reported in patients taking warfarin and at least 400 mg of RTV inhibited both hepatic and intestinal + hepatic CYP3A
twice daily.8,9 The clinical effect on INR could not be activity, increasing exposure to IV and PO MDZ. A greater
evaluated in this study, as vitamin K was given with WARF. degree of intestinal inhibition was seen in our study, likely
However, based on the changes in S- and R-WARF exposure, due to increased local concentrations of LPV and RTV during
it appears that low doses of RTV in combination with LPV absorption, and significant contribution of intestinal CYP3A
may also cause a reduction in INR response. activity to MDZ metabolism.57Y59 Although MDZ coadmin-
The results of this study are also helpful in understanding istration with LPV/r is currently contraindicated,4 these data
other unexpected drug interactions with LPV/r, such as that may support the cautious use of IV MDZ, initiated at 25% of
seen with phenytoin or methadone. We previously demonstrat- the dose and given at 3 times the regular dosing interval.
ed that LPV/r unexpectedly decreased phenytoin (PHT) Furthermore, for clinical studies which show overall induc-
exposure by 31%.7 It has been reported that, at therapeutic tion of CYP3A substrates by LPV/r,10Y13 the mechanism of
concentrations, PHT likely saturates CYP2C9, resulting in a induction is unlikely through CYP3A.
shift of PHT metabolism to the CYP2C19 pathway.39 It has In vitro, LPV/r inhibits CYP3A with an IC50 of 1.1 KM
also been reported that LPV/r has significant binding affinity (0.69 Kg/mL).6 The metabolism of drugs that are primarily
for CYP2C9 (IC50 = 13.7 T 4.0 Kmol/L [8.6 T 2.5 Kg/mL] at a CYP3A substrates such as atorvastatin,60 rifabutin,10 and
LPV/RTV concentration ratio of 3:1).6,40 Our data suggest that ketoconazole,61 when coadministered with LPV/r, is inhibited
LPV/r likely increased PHT metabolism via induction of both in vivo to a similar extent as that seen with MDZ. However,
CYP2C9 and CYP2C19, although the specific contribution of compounds that are also P-glycoprotein substrates (unlike MDZ)
each isozyme cannot be determined from this study. Because may be affected to a greater extent.62 For example, tacrolimus is
both PHT and LPV are highly protein bound (87%Y93%41 and both a CYP3A and P-glycoprotein substrate which requires a
98%Y99%,4 respectively), protein-binding interactions cannot 28- to 140-fold decrease in dose upon the addition of LPV/r.63
be discounted in contributing to altered PHT concentrations. Significant correlations were observed in this study
Total methadone exposures have been shown to decrease between the magnitude of decrease in CYP3A hepatic and
by 36% to 47% with concomitant LPV/r administration in both hepatic + intestinal clearances and initial CYP3A clearance.
HIV-infected and noninfected subjects,42,43 with inconsistent Subjects with the highest initial MDZ clearances (and,
reports of opioid withdrawal symptoms.10Y13 In 1 study, low- presumably, the highest CYP3A activity) exhibited the greatest
dose RTV (100 mg twice daily) was not associated with decline in activity with LPV/r treatment. This relationship has
changes in total methadone exposure; therefore, the reduction been noted for other CYP3A inhibitorYsubstrate interactions,
in methadone exposure was attributed to LPV.12 Although such as MDZ-ketoconazole,57 grapefruit juiceYfelodipine,58 and

58 * 2006 Lippincott Williams & Wilkins

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006 Lopinavir/Ritonavir and Cytochrome P450 Activity

MDZ-clarithromycin.59 This correlation is consistent with the 11. Clarke S, Mulcahy F, Bergin C, et al. Absence of opioid withdrawal
mechanism-based inhibition of CYP3A by RTV64 and, possibly, symptoms in patients receiving methadone and the protease inhibitor
lopinavir-ritonavir. Clin Infect Dis. 2002;34(8):1143Y1145.
LPV,65 where CYP3A is inactivated by the formation of a 12. McCance-Katz EF, Rainey PM, Friedland G, et al. The protease inhibitor
metabolic intermediate complex. Further characterization of the lopinavir-ritonavir may produce opiate withdrawal in methadone-
mechanism of the effects of LPV/r on CYP may improve in vivo maintained patients. Clin Infect Dis. 2003;37(4):476Y482.
prediction models. 13. Stevens RC, Rapaport S, Maroldo-Connelly L, et al. Lack of methadone
dose alterations or withdrawal symptoms during therapy with lopinavir/
Because this study was performed in healthy volunteers, ritonavir. J Acquir Immune Defic Syndr. 2003;33(5):650Y651.
it is possible that the magnitude of drug interactions between 14. Kim RB, Wandel C, Leake B, et al. Interrelationship between substrates
LPV/r and other CYP substrates may differ. However, LPV and inhibitors of human CYP3A and P-glycoprotein. Pharm Res. 1999;
and RTV exposures are similar in HIV-negative and HIV- 16(3):408Y414.
15. Chainuvati S, Nafziger AN, Leeder JS, et al. Combined phenotypic
positive individuals.4,7,66,67 Moreover, the magnitude of assessment of cytochrome P450 1A2, 2C9, 2C19, 2D6, and 3A,
interactions which have been performed in both HIV-negative N-acetyltransferase-2, and xanthine oxidase activities with the
and HIV-positive persons using LPV/r in combination with BCooperstown 5 + 1 cocktail^. Clin Pharmacol Ther. 2003;74(5):437Y447.
other drugs, such as efavirenz68,69 and saquinavir,70,71 is 16. Kim JS, Nafziger AN, Gaedigk A, et al. Effects of oral vitamin K on S- and
similar. At minimum, these data provide a signal to clinicians R-warfarin pharmacokinetics and pharmacodynamics: enhanced safety of
warfarin as a CYP2C9 probe. J Clin Pharmacol. 2001;41(7):715Y722.
for drug interaction potential with LPV/r and other CYP 17. Kashuba AD, Bertino JS Jr, Kearns GL, et al. Quantitation of three-
substrates, although the multiple medications taken by HIV- month intraindividual variability and influence of sex and menstrual
infected subjects could impact the magnitude of interaction. cycle phase on CYP1A2, N-acetyltransferase-2, and xanthine oxidase
In conclusion, our data have important implications for activity determined with caffeine phenotyping. Clin Pharmacol Ther.
the appropriate use of combination drug therapy with LPV/r. 1998;63(5):540Y551.
18. Kashuba AD, Bertino JS Jr, Rocci ML Jr, et al. Quantification of 3-month
Failure to consider CYP enzyme system pathways in intraindividual variability and the influence of sex and menstrual cycle
treatment regimens that include LPV/r may result in phase on CYP3A activity as measured by phenotyping with
subtherapeutic or supratherapeutic drug concentrations; the intravenous midazolam. Clin Pharmacol Ther. 1998;64(3):269Y277.
consequences of which might be treatment failure, the 19. Kim MJ, Bertino JS Jr, Gaedigk A, et al. Effect of sex and menstrual
cycle phase on cytochrome P450 2C19 activity with omeprazole used as
progression of underlying disease processes, or drug toxicity. a biomarker. Clin Pharmacol Ther. 2002;72(2):192Y199.
Phenotyping cocktails are an efficient and useful way to 20. Sconce EA, Khan TI, Wynne HA, et al. The impact of CYP2C9 and
clinically evaluate drug interaction potential and target select VKORC1 genetic polymorphism and patient characteristics upon
compounds for further evaluation. warfarin dose requirements: proposal for a new dosing regimen.
Blood. 2005;106(7):2329Y2333.
21. Smith HT, Jokubaitis LA, Troendle AJ, et al. Pharmacokinetics of
ACKNOWLEDGMENTS fluvastatin and specific drug interactions. Am J Hypertens. 1993;
6(11(pt 2)):375SY382S.
The authors thank Richard Bertz for scientific guidance 22. Vachharajani NN, Shyu WC, Smith RA, et al. The effects of age and
and support of this protocol, Nicole White for analytical gender on the pharmacokinetics of irbesartan. Br J Clin Pharmacol.
assistance, Chris Zurich for assistance with grants and 1998;46(6):611Y613.
contracts, the research volunteers, and the nurses and staff of 23. The Adult AIDS Clinical Trials Group Table for Grading Severity of
the UNC GCRC. Adult Adverse Experiences. Available at: http://aactg.s-3.com/members/
adulttox.html. Accessed September 8, 2004.
24. Kennedy MJ, Scripture CD, Kashuba AD, et al. Activities of cytochrome
REFERENCES P450 1A2, N-acetyltransferase 2, xanthine oxidase, and cytochrome
1. Piscitelli SC, Gallicano KD. Interactions among drugs for HIV and P450 2D6 are unaltered in children with cystic fibrosis. Clin Pharmacol
opportunistic infections. N Engl J Med. 2001;344(13):984Y996. Ther. 2004;75(3):163Y171.
2. Sham HL, Kempf DJ, Molla A, et al. ABT-378, a highly potent inhibitor 25. Grant DM, Tang BK, Kalow W. A simple test for acetylator phenotype
of the human immunodeficiency virus protease. Antimicrob Agents using caffeine (1984 Classical Article). Br J Clin Pharmacol.
Chemother. 1998;42(12):3218Y3224. 2004;58(7):S788YS793.
3. Cvetkovic RS, Goa KL. Lopinavir/ritonavir: a review of its use in the 26. Locatelli I, Kmetec V, Mrhar A, et al. Determination of warfarin
management of HIV infection. Drugs. 2003;63(8):769Y802. enantiomers and hydroxylated metabolites in human blood plasma by
4. Kaletra Product Information. Abbott Park, IL: Abbott Laboratories; liquid chromatography with achiral and chiral separation. J Chromatogr
February 2004. B Analyt Technol Biomed Life Sci. 2005;818(2):191Y198.
5. Corbett AH, Lim ML, Kashuba AD. Kaletra (lopinavir/ritonavir). 27. Kosten T, Oliveto A, Feingold A, et al. Desipramine and contingency
Ann Pharmacother. 2002;36(7Y8):1193Y1203. management for cocaine and opiate dependence in buprenorphine
6. Kumar GN, Dykstra J, Roberts EM, et al. Potent inhibition of the maintained patients. Drug Alcohol Depend. 2003;70(3):315Y325.
cytochrome P-450 3AYmediated human liver microsomal metabolism of 28. Kanazawa H, Okada A, Igarashi E, et al. Determination of midazolam
a novel HIV protease inhibitor by ritonavir: a positive drug-drug and its metabolite as a probe for cytochrome P450 3A4 phenotype by
interaction. Drug Metab Dispos. 1999;27(8):902Y908. liquid chromatographyYmass spectrometry. J Chromatogr A.
7. Lim ML, Min SS, Eron JJ, et al. Coadministration of lopinavir/ritonavir 2004;1031(1Y2):213Y218.
and phenytoin results in two-way drug interaction through cytochrome 29. Rezk NL, Tidwell RR, Kashuba AD. High-performance liquid chroma-
P-450 induction. J Acquir Immune Defic Syndr. 2004;36(5):1034Y1040. tography assay for the quantification of HIV protease inhibitors and
8. Knoell KR, Young TM, Cousins ES. Potential interaction involving non-nucleoside reverse transcriptase inhibitors in human plasma.
warfarin and ritonavir. Ann Pharmacother. 1998;32(12):1299Y1302. J Chromatogr B Analyt Technol Biomed Life Sci. 2004;805(2):241Y247.
9. Newshan G, Tsang P. Ritonavir and warfarin interaction. AIDS. 1999; 30. Saruwatari J, Nakagawa K, Shindo J, et al. A population phenotyping
13(13):1788Y1789. study of three drug-metabolizing enzymes in Kyushu, Japan, with use of
10. Bertz R, Hsu A, Lam W, et al. Pharmacokinetic interactions between the caffeine test. Clin Pharmacol Ther. 2002;72(2):200Y208.
Kaletra (lopinavir/ritonavir of ABT-378/r) and other non-HIV drugs 31. Fuhr U, Rost KL, Engelhardt R, et al. Evaluation of caffeine as a test drug
[Poster 438]. Paper presented at: 5th International Congress on Drug for CYP1A2, NAT2 and CYP2E1 phenotyping in man by in vivo versus
Therapy in HIV Infection; 2000; Glasgow, UK. in vitro correlations. Pharmacogenetics. 1996;6(2):159Y176.

* 2006 Lippincott Williams & Wilkins 59

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Yeh et al J Acquir Immune Defic Syndr & Volume 42, Number 1, May 2006

32. Lee CR, Goldstein JA, Pieper JA. Cytochrome P450 2C9 polymorphisms: 54. Tanaka E. Update: genetic polymorphism of drug metabolizing enzymes
a comprehensive review of the in-vitro and human data. Pharmacogenetics. in humans. J Clin Pharm Ther. 1999;24(5):323Y329.
2002;12(3):251Y263. 55. Yin OQ, Tomlinson B, Waye MM, et al. Pharmacogenetics and
33. Balian JD, Sukhova N, Harris JW, et al. The hydroxylation of herb-drug interactions: experience with Ginkgo biloba and omeprazole.
omeprazole correlates with S-mephenytoin metabolism: a population Pharmacogenetics. 2004;14(12):841Y850.
study. Clin Pharmacol Ther. 1995;57(6):662Y669. 56. Feng HJ, Huang SL, Wang W, et al. The induction effect of
34. Marinac JS, Balian JD, Foxworth JW, et al. Determination of CYP2C19 rifampicin on activity of mephenytoin 4-hydroxylase related to M1
phenotype in black Americans with omeprazole: correlation with mutation of CYP2C19 and gene dose. Br J Clin Pharmacol.
genotype. Clin Pharmacol Ther. 1996;60(2):138Y144. 1998;45(1):27Y29.
35. Madan A, Graham RA, Carroll KM, et al. Effects of prototypical 57. Tsunoda SM, Velez RL, von Moltke LL, et al. Differentiation of
microsomal enzyme inducers on cytochrome P450 expression in cultured intestinal and hepatic cytochrome P450 3A activity with use of
human hepatocytes. Drug Metab Dispos. 2003;31(4):421Y431. midazolam as an in vivo probe: effect of ketoconazole. Clin Pharmacol
36. Hsu A, Granneman GR, Witt G. Assessment of multiple doses of ritonavir Ther. 1999;66(5):461Y471.
on the pharmacokinetics of theophylline [abstract Mo.B.1200]. Presented 58. Lown KS, Bailey DG, Fontana RJ, et al. Grapefruit juice increases
at: XI International Conference on AIDS; 1996; Vancouver, Canada. felodipine oral availability in humans by decreasing intestinal CYP3A
37. Penzak SR, Hon YY, Lawhorn WD, et al. Influence of ritonavir on protein expression. J Clin Invest. 1997;99(10):2545Y2553.
olanzapine pharmacokinetics in healthy volunteers. J Clin 59. Gorski JC, Jones DR, Haehner-Daniels BD, et al. The contribution of
Psychopharmacol. 2002;22(4):366Y370. intestinal and hepatic CYP3A to the interaction between midazolam and
38. Takahashi H, Echizen H. Pharmacogenetics of warfarin elimination and clarithromycin. Clin Pharmacol Ther. 1998;64(2):133Y143.
its clinical implications. Clin Pharmacokinet. 2001;40(8):587Y603. 60. Carr R, Andre AK, Bertz RJ, et al. Concommitant administration of
39. Bajpai M, Roskos LK, Shen DD, et al. Roles of cytochrome P4502C9 ABT-378/ritonavir (ABT-378/r) results in a clinically important
and cytochrome P4502C19 in the stereoselective metabolism of pharmacokinetic (PK) interaction with atorvastatin (ATO) but not
phenytoin to its major metabolite. Drug Metab Dispos. 1996;24(12): pravastatin (PRA) [abstract 1644]. Presented at: 40th Interscience
1401Y1403. Conference on Antimicrobial Agents and Chemotherapy; 2000; Toronto,
40. Kumar GN, Rodrigues AD, Buko AM, et al. Cytochrome P450Ymediated Ontario, Canada.
metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in 61. Boffito M, Bonora S, Sales P, et al. Ketoconazole and lopinavir/ritonavir
human liver microsomes. J Pharmacol Exp Ther. 1996;277(1):423Y431. coadministration: boosting beyond boosting. AIDS Res Hum
41. Drug Facts and Comparisons, 58th ed. Wolters Kluwer Health, Inc.; 2004; Retroviruses. 2003;19(10):941Y942.
St. Louis, MO. 62. Vishnuvardhan D, Moltke LL, Richert C, et al. Lopinavir: acute exposure
42. Gerber JG, Rhodes RJ, Gal J. Stereoselective metabolism of methadone inhibits P-glycoprotein; extended exposure induces P-glycoprotein.
N-demethylation by cytochrome P4502B6 and 2C19. Chirality. 2004; AIDS. 2003;17(7):1092Y1094.
16(1):36Y44. 63. Jain AB, Venkataramanan R, Eghtesad B, et al. Effect of
coadministered lopinavir and ritonavir (Kaletra) on tacrolimus blood
43. Gerber JG, Rosenkranz S, Segal Y, et al. Effect of ritonavir/saquinavir on
concentration in liver transplantation patients. Liver Transpl.
stereoselective pharmacokinetics of methadone: results of AIDS Clinical
Trials Group (ACTG) 401. J Acquir Immune Defic Syndr. 2001;27(2): 2003;9(9):954Y960.
153Y160. 64. Ernest CS II, Hall SD, Jones DR. Mechanism-based inactivation of
44. Iribarne C, Dreano Y, Bardou LG, et al. Interaction of methadone with CYP3A by HIV protease inhibitors. J Pharmacol Exp Ther.
substrates of human hepatic cytochrome P450 3A4. Toxicology. 2005;312(2):583Y591.
65. Weemhoff JL, von Moltke LL, Richert C, et al. Apparent
1997;117(1):13Y23.
mechanism-based inhibition of human CYP3A in-vitro by lopinavir.
45. Foster DJ, Somogyi AA, Bochner F. Methadone N-demethylation in
J Pharm Pharmacol. 2003;55(3):381Y386.
human liver microsomes: lack of stereoselectivity and involvement of 66. Hsu A, Granneman GR, Witt G, et al. Multiple-dose pharmacokinetics
CYP3A4. Br J Clin Pharmacol. 1999;47(4):403Y412. of ritonavir in human immunodeficiency virusYinfected subjects.
46. Eap CB, Cuendet C, Baumann P. Binding of d-methadone, l-methadone, Antimicrob Agents Chemother. 1997;41(5):898Y905.
and dl-methadone to proteins in plasma of healthy volunteers: role of the 67. Kashuba AD, Tierney C, Downey GF, et al. Combining
variants of alpha 1-acid glycoprotein. Clin Pharmacol Ther. 1990;47(3): fosamprenavir with lopinavir/ritonavir substantially reduces
338Y346. amprenavir and lopinavir exposure: ACTG protocol A5143 results.
47. Khaliq Y, Gallicano K, Seguin I, et al. Single and multiple dose AIDS. 2005;19(2):145Y152.
pharmacokinetics of nelfinavir and CYP2C19 activity in human 68. Bertz R, Lam W, Hsu A, et al. Assessment of the pharmacokinetic
immunodeficiency virusYinfected patients with chronic liver disease. interaction between ABT-378/ritonavir (ABT-378/r) and efavirenz
Br J Clin Pharmacol. 2000;50(2):108Y115. (EFV) in healthy and in HIV+ subjects. Presented at: 40th Interscience
48. Bertilsson L, Henthorn TK, Sanz E, et al. Importance of genetic factors in Conference on Antimicrobial Agents and Chemotherapy; 2000;
the regulation of diazepam metabolism: relationship to S-mephenytoin, Toronto, Ontario, Canada.
but not debrisoquin, hydroxylation phenotype. Clin Pharmacol Ther. 69. Hsu A, Isaacson J, Brun S, et al. Pharmacokinetic-pharmacodynamic
1989;45(4):348Y355. analysis of lopinavir-ritonavir in combination with efavirenz and two
49. Sakai T, Aoyama N, Kita T, et al. CYP2C19 genotype and nucleoside reverse transcriptase inhibitors in extensively pretreated
pharmacokinetics of three proton pump inhibitors in healthy subjects. human immunodeficiency virusYinfected patients. Antimicrob Agents
Pharm Res. 2001;18(6):721Y727. Chemother. 2003;47(1):350Y359.
50. Kita T, Sakaeda T, Baba T, et al. Different contribution of CYP2C19 in 70. Bertz R, Ashbrenner E, Foit C, et al. Assessment of the steady-state
the in vitro metabolism of three proton pump inhibitors. Biol Pharm pharmacokinetics of three dosing regimens of saquinavir (400, 600,
Bull. 2003;26(3):386Y390. and 800 mg twice daily) administration as the hard gelatin
51. Furuta T, Shirai N, Sugimoto M, et al. Pharmacogenomics of proton capsule (HGC) in combination with lopinavir/ritonavir (LPV/r)
pump inhibitors. Pharmacogenomics. 2004;5(2):181Y202. 400/100 mg BID in HIV-infected adults. Presented at: 5th
52. Breyer-Pfaff U, Pfandl B, Nill K, et al. Enantioselective amitriptyline International Workshop on Clinical Pharmacology of HIV Therapy;
metabolism in patients phenotyped for two cytochrome P450 isozymes. 2004; Rome, Italy.
Clin Pharmacol Ther. 1992;52(4):350Y358. 71. Stephan C, Hentig N, Kourbeti I, et al. Saquinavir drug exposure is not
53. Breyer-Pfaff U. The metabolic fate of amitriptyline, nortriptyline and impaired by the boosted double protease inhibitor combination of
amitriptylinoxide in man. Drug Metab Rev. 2004;36(3-4):723Y746. lopinavir/ritonavir. AIDS. 2004;18(3):503Y508.

60 * 2006 Lippincott Williams & Wilkins

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like