Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Publication year range
1.
Pharm Res ; 40(11): 2639-2651, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37561322

ABSTRACT

PURPOSE: Ritlecitinib, an inhibitor of Janus kinase 3 and tyrosine kinase expressed in hepatocellular carcinoma family kinases, is in development for inflammatory diseases. This study assessed the impact of ritlecitinib on drug transporters using a probe drug and endogenous biomarkers. METHODS: In vitro transporter-mediated substrate uptake and inhibition by ritlecitinib and its major metabolite were evaluated. Subsequently, a clinical drug interaction study was conducted in 12 healthy adult participants to assess the effect of ritlecitinib on pharmacokinetics of rosuvastatin, a substrate of breast cancer resistance protein (BCRP), organic anion transporting polypeptide 1B1 (OATP1B1), and organic anion transporter 3 (OAT3). Plasma concentrations of coproporphyrin I (CP-I) and pyridoxic acid (PDA) were assessed as endogenous biomarkers for OATP1B1 and OAT1/3 function, respectively. RESULTS: In vitro studies suggested that ritlecitinib can potentially inhibit BCRP, OATP1B1 and OAT1/3 based on regulatory cutoffs. In the subsequent clinical study, coadministration of ritlecitinib decreased rosuvastatin plasma exposure area under the curve from time 0 to infinity (AUCinf) by ~ 13% and maximum concentration (Cmax) by ~ 27% relative to rosuvastatin administered alone. Renal clearance was comparable in the absence and presence of ritlecitinib coadministration. PK parameters of AUCinf and Cmax for CP-I and PDA were also similar regardless of ritlecitinib coadministration. CONCLUSION: Ritlecitinib does not inhibit BCRP, OATP1B1, and OAT3 and is unlikely to cause a clinically relevant interaction through these transporters. Furthermore, our findings add to the body of evidence supporting the utility of CP-I and PDA as endogenous biomarkers for assessment of OATP1B1 and OAT1/3 transporter activity.


Subject(s)
Neoplasm Proteins , Organic Anion Transporters , Adult , Humans , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Biomarkers , Drug Interactions , Membrane Transport Proteins/metabolism , Neoplasm Proteins/metabolism , Organic Anion Transporters/metabolism , Rosuvastatin Calcium/metabolism , Rosuvastatin Calcium/pharmacokinetics , Rosuvastatin Calcium/pharmacology
2.
Anal Chem ; 92(14): 9745-9754, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32551505

ABSTRACT

There is a growing interest in using endogenous compounds as drug transporter biomarkers to facilitate drug-drug interaction (DDI) risk assessment in early phase I clinical trials. Compared to other drug transporters, however, no valid biomarker for hepatic organic cation transporter (OCT) 1 has been described to date. The present work represents the first report of an endogenous compound, isobutyryl-l-carnitine (IBC), as a potential clinical OCT1 biomarker for DDI assessment. A hydrophilic interaction chromatography (HILIC)-mass spectrometry/high resolution mass spectrometry (MS/HRMS) assay with a simple sample preparation method was developed. The assay is capable of simultaneously quantifying multiple endogenous compounds, including IBC, thiamine, N1-methylnicotinamide (1-NMN), creatinine, carnitine, and metformin, which is a probe for OCT1 and OCT2 and MATE1 and MATE2K (multidrug and toxin extrusion proteins) in clinical studies. The HRMS assay was fit-for-purpose validated in human plasma and demonstrated good linearity, accuracy, and precision for all analytes. It was further applied to two phase I clinical trials to evaluate potential biomarkers for OCT1 and additional cation transporters (renal OCT2, MATE1, and MATE2K). The clinical data demonstrated that plasma IBC changes correlated well with in vitro data and supported its use as a liver OCT1 biomarker. The described HILIC-MS/HRMS assay can be used as a "biomarker cocktail" to simultaneously assess clinical DDI risk for the inhibition of OCT1/2 and MATEs in clinical studies with new drug candidates.


Subject(s)
Biomarkers/chemistry , Carnitine/analogs & derivatives , Enzyme Inhibitors/pharmacokinetics , Organic Cation Transporter 1/metabolism , Carnitine/chemistry , Clinical Trials, Phase I as Topic , Drug Interactions , Enzyme Inhibitors/pharmacology , Humans , Metformin/pharmacokinetics , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/antagonists & inhibitors , Organic Cation Transporter 1/chemistry , Organic Cation Transporter 2/metabolism
3.
Drug Metab Dispos ; 47(5): 493-503, 2019 05.
Article in English | MEDLINE | ID: mdl-30862625

ABSTRACT

The aim of the present study was to quantitatively evaluate the drug-drug interactions (DDIs) of maraviroc (MVC) with various perpetrator drugs, including telaprevir (TVR), using an in vitro data-informed physiologically based pharmacokinetic (PBPK) model. MVC showed significant active uptake and biliary excretion in sandwich-cultured human hepatocytes, and biphasic organic anion transporting polypeptide (OATP)1B1-mediated uptake kinetics in transfected cells (high-affinity K m ∼5 µM). No measureable active uptake was noted in OATP1B3- and OATP2B1-transfceted cells. TVR inhibited OATP1B1-mediated MVC transport in vitro, and also exhibited CYP3A time-dependent inhibition in human hepatocytes (inactivation constant, K I = 2.24 µM, and maximum inactivation rate constant, k inact = 0.0112 minute-1). The inactivation efficiency (k inact/K I) was approximately 34-fold lower in human hepatocytes compared with liver microsomes. A PBPK model accounting for interactions involving CYP3A, P-glycoprotein (P-gp), and OATP1B1 was developed based on in vitro mechanistic data. MVC DDIs with ketoconazole (inhibition of CYP3A and P-gp), ritonavir (inhibition of CYP3A and P-gp), efavirenz (induction of CYP3A), rifampicin (induction of CYP3A and P-gp; inhibition of OATP1B1), and TVR (inhibition of CYP3A, P-gp, and OATP1B1) were well described by the PBPK model with optimized transporter K i values implying that OATP1B1-mediated uptake along with CYP3A metabolism determines the hepatic clearance of MVC, and P-gp-mediated efflux limits its intestinal absorption. In summary, MVC disposition involves intestinal P-gp/CYP3A and hepatic OATP1B1/CYP3A interplay, and TVR simultaneously inhibits these multiple mechanisms leading to a strong DDI-about 9.5-fold increase in MVC oral exposure.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP3A/metabolism , Drug Interactions/physiology , Hepatocytes/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , Liver/metabolism , Maraviroc/metabolism , Biological Transport/physiology , Cell Line , HEK293 Cells , Humans , Kinetics , Membrane Transport Proteins/metabolism , Microsomes, Liver/metabolism
4.
Drug Metab Rev ; 49(1): 18-34, 2017 02.
Article in English | MEDLINE | ID: mdl-27718639

ABSTRACT

A key goal in the clinical development of a new molecular entity is to quickly identify whether it has the potential for drug-drug interactions. In particular, confirmation of in vitro data in the early stage of clinical development would facilitate the decision making and inform future clinical pharmacology study designs. Plasma 4ß-hydroxycholesterol (4ß-HC) is considered as an emerging endogenous biomarker for cytochrome P450 3A (CYP3A), one of the major drug metabolizing enzymes. Although there are increasing reports of the use of 4ß-HC in academic- and industry-sponsored clinical studies, a thorough review, summary and consideration of the advantages and challenges of using 4ß-HC to evaluate changes in CYP3A activity has not been attempted. Herein, we review the biology of 4ß-HC, its response to treatment with CYP3A inducers, inhibitors and mixed inducer/inhibitors in healthy volunteers and patients, the association of 4ß-HC with other probes of CYP3A activity (e.g. midazolam, urinary cortisol ratios), and present predictive pharmacokinetic models. We provide recommendations for studying hepatic CYP3A activity in clinical pharmacology studies utilizing 4ß-HC at different stages of drug development.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hydrocortisone/urine , Hydroxycholesterols/blood , Midazolam/blood , Biomarkers/blood , Biomarkers/urine , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Discovery , Drug Interactions , Humans , Hydroxycholesterols/pharmacokinetics , Liver/drug effects , Liver/metabolism , Models, Biological
5.
Bioanalysis ; 16(6): 347-362, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38376139

ABSTRACT

Background: It has become common practice to assess solute carrier transporter (SLC)-mediated drug-drug interactions (DDIs) by quantitating various individual endogenous compounds as biomarkers in human plasma and urine. The goal of this work was to develop biomarker multiplex assays that could be utilized during first in human studies to support the simultaneous assessment of clinical DDI risk across various SLCs. Methodology: Hydrophilic interaction chromatography-MS/MS methods were developed, and validations were performed. Results: The multiplex assays were applied to a first in human study. Placebo/reference subject biomarker data were consistent with single assay in-house and published data. Conclusion: This work demonstrates the utility of these multiplex methods to support the concurrent evaluation of clinical DDI risk across various SLCs.


Subject(s)
Liquid Chromatography-Mass Spectrometry , Tandem Mass Spectrometry , Humans , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Biomarkers , Membrane Transport Proteins , Drug Interactions
6.
Clin Transl Sci ; 17(1): e13644, 2024 01.
Article in English | MEDLINE | ID: mdl-38108609

ABSTRACT

PF-06835919, a ketohexokinase inhibitor, presented as an inducer of cytochrome P450 3A4 (CYP3A4) in vitro (human primary hepatocytes), and static mechanistic modeling exercises predicted significant induction in vivo (oral midazolam area under the plasma concentration-time curve [AUC] ratio [AUCR] = 0.23-0.79). Therefore, a drug-drug interaction study was conducted to evaluate the effect of multiple doses of PF-06835919 (300 mg once daily × 10 days; N = 10 healthy participants) on the pharmacokinetics of a single oral midazolam 7.5 mg dose. The adjusted geometric means for midazolam AUC and its maximal plasma concentration were similar following co-administration with PF-06835919 (vs. midazolam administration alone), with ratios of the adjusted geometric means (90% confidence interval [CI]) of 97.6% (90% CI: 79.9%-119%) and 98.9% (90% CI: 76.4%-128%), respectively, suggesting there was minimal effect of PF-06835919 on midazolam pharmacokinetics. Lack of CYP3A4 induction was confirmed after the preparation of subject plasma-derived small extracellular vesicles (sEVs) and conducting proteomic and activity (midazolam 1'-hydroxylase) analysis. Consistent with the midazolam AUCR observed, the CYP3A4 protein expression fold-induction (geometric mean, 90% CI) was low in liver (0.9, 90% CI: 0.7-1.2) and non-liver (0.9, 90% CI: 0.7-1.2) sEVs (predicted AUCR = 1.0, 90% CI: 0.9-1.2). Likewise, minimal induction of CYP3A4 activity (geometric mean, 90% CI) in both liver (1.1, 90% CI: 0.9-1.3) and non-liver (0.9, 90% CI: 0.5-1.5) sEVs was evident (predicted AUCR = 0.9, 90% CI: 0.6-1.4). The results showcase the integrated use of an oral CYP3A probe (midazolam) and plasma-derived sEVs to assess a drug candidate as inducer.


Subject(s)
Cytochrome P-450 CYP3A , Midazolam , Humans , Midazolam/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Proteomics , Pharmaceutical Preparations , Liquid Biopsy , Drug Interactions , Administration, Oral
7.
Antimicrob Agents Chemother ; 57(12): 6158-64, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24080663

ABSTRACT

This open-label, fixed-sequence, phase 1 study evaluated the pharmacokinetic interaction between maraviroc (MVC) and ritonavir-boosted fosamprenavir (FPV/r) in healthy subjects. In period 1, subjects received 300 mg of MVC twice daily (BID; cohort 1) or once daily (QD; cohort 2) for 5 days. In period 2, cohort 1 subjects received 700/100 mg of FPV/r BID alone on days 1 to 10 and then FPV/r at 700/100 mg BID plus MVC at 300 mg BID on days 11 to 20; cohort 2 subjects received FPV/r at 1,400/100 mg QD alone on days 1 to 10 and then FPV/r at 1,400/100 mg QD plus MVC at 300 mg QD on days 11 to 20. Pharmacokinetic parameters, assessed on day 5 of period 1 and on days 10 and 20 of period 2, included the maximum plasma concentration (Cmax), the concentration at end of dosing interval (Cτ), and the area under the curve over dosing interval (AUCτ). Safety and tolerability were also assessed. MVC geometric mean AUCτ, Cmax, and Cτ were increased by 149, 52, and 374%, respectively, after BID dosing with FPV/r, and by 126, 45, and 80%, respectively, after QD dosing. Amprenavir (the active form of the prodrug fosamprenavir) and ritonavir exposures were decreased in the presence of MVC with amprenavir AUCτ, Cmax, and Cτ decreased by 34 to 36% in the presence of FPV/r plus maraviroc BID and by 15 to 30% with FPV/r plus MVC QD both compared to FPV/r alone. The overall all-causality adverse-event (AE) incidence rate was 96.4%; all AEs were of mild or moderate severity. Commonly reported treatment-related AEs (>20% of patients overall) included diarrhea, fatigue, abdominal discomfort, headache, and nausea. No serious AEs or deaths occurred. In summary, maraviroc exposure increased in the presence of FPV/r, whereas MVC coadministration decreased amprenavir and ritonavir exposures. MVC dosed at 300 mg BID with FPV/r is not recommended due to concerns of lower amprenavir exposures; however, no dose adjustment is warranted with MVC at 150 mg BID in combination with FPV/r based on the available clinical data. MVC plus FPV/r was generally well tolerated; no new safety signals were detected.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Carbamates/pharmacokinetics , Cyclohexanes/pharmacokinetics , Organophosphates/pharmacokinetics , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Triazoles/pharmacokinetics , Adult , Anti-HIV Agents/blood , Area Under Curve , Carbamates/blood , Cyclohexanes/blood , Drug Administration Schedule , Drug Combinations , Drug Dosage Calculations , Drug Interactions , Furans , Humans , Male , Maraviroc , Middle Aged , Organophosphates/blood , Ritonavir/blood , Sulfonamides/blood , Triazoles/blood
8.
HIV Clin Trials ; 14(3): 99-109, 2013.
Article in English | MEDLINE | ID: mdl-23835512

ABSTRACT

PURPOSE: This open-label, nonrandomized, parallel-group study was conducted to explore the pharmacokinetics, safety, and tolerability of maraviroc in renally impaired subjects. METHODS: Subjects with normal renal function; mild, moderate, or severe renal impairment; or end-stage renal disease (ESRD) (n = 6 per group) were enrolled. Subjects with normal function (period 1), severe impairment, and ESRD received a single 300 mg dose of maraviroc. Subjects with normal function (period 2), mild impairment, and moderate impairment received 150 mg for 7 days at adjusted intervals of twice daily, once daily, and every 48 hours, respectively, with saquinavir/ritonavir (SQV/r). Maraviroc was quantified in plasma, urine, and dialysate by tandem high-performance liquid chromatography-mass spectrometry. RESULTS: With SQV/r, geometric mean steady-state maraviroc area under the plasma concentration-time curve for the dosing interval (AUCtau) was 5,341 (coefficient of variation [CV], 27%), 8,119 (35%), and 6,193 (27%) h•ng/mL, in normal function, mild, and moderate impairment groups, respectively. Without SQV/r, 2% to 3% of the maraviroc dose was recovered in urine versus 15% to 25% of steady-state dose when given with SQV/r. Moderate to high intersubject variability in exposure was noted. AUC from zero to infinity (AUCinf) was similar to historical single-dose data in subjects with ESRD: low in those with normal function, and high in those with severe impairment. Dialysis did not influence maraviroc exposure. Maraviroc was well tolerated. CONCLUSIONS: The data suggest that no dosing interval adjustments are required in subjects with renal impairment when maraviroc is administered alone. However, maraviroc dosing interval adjustment is warranted in renally impaired patients receiving potent CYP3A4 inhibitors. Reference to local prescribing information is recommended, because dose recommendations in renally impaired patients may differ between regions.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Cyclohexanes/pharmacokinetics , Renal Insufficiency/metabolism , Triazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Aged , Anti-HIV Agents/adverse effects , Anti-HIV Agents/blood , Area Under Curve , Case-Control Studies , Cyclohexanes/adverse effects , Cyclohexanes/blood , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Female , Gene Expression Regulation , Half-Life , Humans , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Male , Maraviroc , Middle Aged , Renal Dialysis , Severity of Illness Index , Triazoles/adverse effects , Triazoles/blood
9.
Clin Pharmacol Ther ; 113(1): 182-195, 2023 01.
Article in English | MEDLINE | ID: mdl-36239478

ABSTRACT

Ceftazidime-avibactam is a novel ß-lactam/ß-lactamase inhibitor combination developed to treat serious Gram-negative bacterial infections; approved indications include complicated urinary tract infection, complicated intra-abdominal infection, and hospital-acquired pneumonia including ventilator-associated pneumonia in patients ≥ 3 months old. Because of the predominantly renal clearance of ceftazidime and avibactam, dose adjustments (reductions) are required for patients with estimated creatinine clearance (CrCL) ≤ 50 mL/min. We describe the application of combined adult and pediatric population pharmacokinetic models in developing ceftazidime-avibactam dose recommendations for pediatric patients ≥ 2 to < 18 years old with body surface area-normalized CrCL ≤ 50 mL/min/1.73 m2 , including moderate, severe, or very severe renal impairment, or end-stage renal disease requiring hemodialysis, and for patients ≥ 3 months to < 2 years old with mild, moderate, or severe renal impairment. Models included allometric scaling for all subjects and simulations (1,000 subjects per age group, renal function group, and indication) were performed nonparametrically using post hoc random effects. Doses were selected based on simulated pediatric patients achieving steady-state exposures similar to adults and high probability of target attainment (using a simultaneous joint target for both ceftazidime and avibactam). Because there were few children with renal impairment in the ceftazidime-avibactam clinical trials, selected pediatric doses were guided by extrapolation and matching of adult exposures associated with efficacy and within established safety margins. The recommended doses for pediatric patients with estimated CrCL ≤ 50 mL/min/1.73 m2 use equivalent adjustments in dose quantity and/or administration interval (vs. the corresponding age group with normal renal function) as those for adults.


Subject(s)
Ceftazidime , Renal Insufficiency , Adult , Humans , Child , Infant , Adolescent , Ceftazidime/pharmacokinetics , Anti-Bacterial Agents/adverse effects , Drug Combinations , beta-Lactamase Inhibitors/therapeutic use , Monobactams , Kidney/physiology
10.
Clin Pharmacol Ther ; 113(5): 1058-1069, 2023 05.
Article in English | MEDLINE | ID: mdl-36524426

ABSTRACT

Hepatic impairment (HI) is known to modulate drug disposition and may lead to elevated plasma exposure. The aim of this study was to quantitate the in vivo OATP1B-mediated hepatic uptake activity in populations with varying degrees of HI. First, we measured baseline levels of plasma coproporphyrin-I, an endogenous OATP1B biomarker, in an open-label, parallel cohort study in adult subjects with normal liver function and mild, moderate, and severe HI (n = 24, 6/cohort). The geometric mean plasma concentrations of coproporphyrin-I were 1.66-fold, 2.81-fold (P < 0.05), and 7.78-fold (P < 0.0001) higher in mild, moderate, and severe impairment than those healthy controls. Second, we developed a dataset of 21 OATP1B substrate drugs with HI data extracted from literature. Median disease-to-healthy plasma area under the curve (AUC) ratios for substrate drugs were ~ 1.4, 3.0, and 6.4 for mild, moderate, and severe HI, respectively. Additionally, significant linear relationship was noted between AUC ratios of substrate drugs without and with co-administration of rifampin, a prototypic OATP1B inhibitor, and AUC ratios in moderate (P < 0.01) and severe (P < 0.001) HI. Third, a physiologically-based pharmacokinetic model analysis was conducted with 10 substrate drugs following estimation of relative OATP1B functional activity in virtual disease population models using coproporphyrin-I data and verification of substrate models with rifampin drug-drug interaction data. This approach adequately predicted plasma AUC change particularly in moderate (9 of 10 within 2-fold) and severe (5 of 5 within 2-fold) HI. Collective findings indicate progressive reduction, by as much as 90-92%, in OATP1B activity in the HI population.


Subject(s)
Liver Diseases , Rifampin , Adult , Humans , Coproporphyrins , Pharmaceutical Preparations , Cohort Studies , Biomarkers , Drug Interactions , Area Under Curve
11.
J Clin Pharmacol ; 63(7): 784-797, 2023 07.
Article in English | MEDLINE | ID: mdl-36807251

ABSTRACT

Ritlecitinib, an inhibitor of Janus kinase 3 and hepatocellular carcinoma family kinases, is in development as potential treatment for several inflammatory diseases. In vitro studies presented ritlecitinib as an inhibitor of hepatic organic cation transporter (OCT) 1, renal transporters OCT2 and multidrug and toxin extrusion (MATE) proteins 1/2K using multiple substrates, and ritlecitinib's major inactive metabolite M2, as an inhibitor of OCT1. A clinical interaction study with an OCT1 drug probe (sumatriptan) and relevant probe biomarkers for OCT/MATE was conducted to assess the effect of ritlecitinib on these transporters in healthy adult participants. The selectivity of sumatriptan for OCT1 was confirmed through a series of in vitro uptake assays. A simple static model was used to help contextualize the observed changes in sumatriptan area under the plasma concentration-time curve (AUC). Coadministration of a single 400-mg dose of ritlecitinib increased sumatriptan AUC from time 0 to infinity (AUCinf ) by ≈30% relative to a single 25-mg sumatriptan administration alone. When administered 8 hours after a ritlecitinib dose, sumatriptan AUCinf increased by ≈50% relative to sumatriptan given alone. Consistent with OCT1 inhibition, the AUC from time 0 to 24 hours of isobutyryl-L-carnitine decreased by ≈15% after ritlecitinib. Based on the evaluation of the renal clearance of N1 -methylnicotinamide, ritlecitinib does not exert clinically meaningful inhibition on renal OCT2 or MATE1/2K. This study confirmed that ritlecitinib and M2 are inhibitors of OCT1 but not OCT2 or MATE1/2K in healthy adults.


Subject(s)
Organic Cation Transport Proteins , Sumatriptan , Adult , Humans , Organic Cation Transporter 1 , Biomarkers , Cations/metabolism , HEK293 Cells
12.
Antimicrob Agents Chemother ; 56(5): 2408-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22371898

ABSTRACT

The objective of this study was to investigate the effect of a supratherapeutic dose of lersivirine (LRV) on corrected QT (QTc) interval using Fridericia's equation (QTcF) in healthy subjects. In this randomized, single-dose, placebo- and active-controlled 3-way crossover study, healthy adult males (n = 48) were randomized to receive LRV (2,400 mg), moxifloxacin (400 mg), or placebo for each treatment period. Triplicate 12-lead electrocardiogram measurements were performed, PK samples were collected, and vital signs were measured. Adverse event monitoring and safety laboratory testing were performed. All subjects were white (mean age, 39 years; body mass index [BMI], 25.6 kg/m(2)) and completed the study. Following LRV administration, the upper bound of the 90% confidence interval (CI) for time-matched adjusted mean differences to placebo QTcF at each time point postdose was below the regulatory threshold of 10 ms, satisfying the criteria for a negative thorough QT/QTc study. The highest upper bound of QTcF 90% CI occurred at 6 h for LRV (3.32 ms; 90% CI, 1.47 to 5.17 ms). The study was deemed adequately sensitive as the lower bound of the 90% CI for the adjusted mean QTcF differences between moxifloxacin and placebo at the moxifloxacin historical T(max) of 3 h was >5 ms (15.29 ms; 90% CI, 13.44 to 17.14 ms). There was no statistically significant relationship between LRV exposure and placebo-adjusted change from baseline QTcF or clinically significant changes in QRS complex, pulse rate (PR) interval, heart rate, or blood pressure. LRV (2,400 mg) did not prolong the QTcF interval, and no clinically relevant electrocardiogram or vital sign changes were observed in healthy subjects.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Heart/drug effects , Nitriles/pharmacokinetics , Pyrazoles/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/blood , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/blood , Anti-Infective Agents/pharmacokinetics , Aza Compounds/administration & dosage , Aza Compounds/blood , Aza Compounds/pharmacokinetics , Blood Pressure/drug effects , Cross-Over Studies , Dose-Response Relationship, Drug , Electrocardiography , Fluoroquinolones , Heart/physiology , Heart Rate/drug effects , Humans , Male , Middle Aged , Moxifloxacin , Nitriles/administration & dosage , Nitriles/blood , Placebos , Pyrazoles/administration & dosage , Pyrazoles/blood , Quinolines/administration & dosage , Quinolines/blood , Quinolines/pharmacokinetics , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/blood
13.
Antimicrob Agents Chemother ; 56(2): 887-92, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22123705

ABSTRACT

Lersivirine (UK-453,061) is a new nonnucleoside reverse transcriptase inhibitor currently being developed as a treatment for human immunodeficiency virus type 1 infection. Lersivirine shows potent activity against wild-type and clinically relevant drug-resistant strains. Previous studies have demonstrated that lersivirine is metabolized by glucuronidation via UGT2B7 and by cytochrome P450 3A4 (CYP3A4). Lersivirine is also a weak inducer of the CYP3A4 enzyme. Therefore, coadministered lersivirine could potentially affect the pharmacokinetics of maraviroc, a CCR5 antagonist metabolized by CYP3A4, and raltegravir, an integrase inhibitor metabolized by glucuronidation. Two open-label studies assessed the pharmacokinetics of raltegravir and of maraviroc when they were coadministered with lersivirine and the pharmacokinetics of lersivirine when it was coadministered with raltegravir. Minor, clinically nonsignificant effects on the pharmacokinetics of raltegravir coadministered with lersivirine were observed at steady state for raltegravir, with estimated mean changes of -15%, -29%, and +25% in the area under the concentration-time profile from time zero to the end of the dosing interval (AUC(tau)), maximum plasma concentration (C(max)), and concentration observed 12 h postdose (C(12)), respectively. There were no clinically relevant effects of steady-state raltegravir on lersivirine AUC(tau), C(max), or concentration observed 24 h postdose (C(24)) (estimated mean changes of -2 to +5%). Coadministration of lersivirine at steady state with maraviroc resulted in no clinically relevant effects on maraviroc AUC(tau), C(max), or C(12) (estimated mean changes of +3.4 to +8.6%). Lersivirine appeared to be generally well tolerated in these studies and appears to be suitable for coadministration with raltegravir or maraviroc without the need for dose modification.


Subject(s)
Anti-HIV Agents , Cyclohexanes , Nitriles , Pyrazoles , Pyrrolidinones , Reverse Transcriptase Inhibitors , Triazoles , Adolescent , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Cross-Over Studies , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacokinetics , Drug Interactions , Drug Therapy, Combination , Female , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/pharmacokinetics , HIV Integrase Inhibitors/administration & dosage , HIV Integrase Inhibitors/pharmacokinetics , Humans , Male , Maraviroc , Middle Aged , Nitriles/administration & dosage , Nitriles/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Pyrrolidinones/administration & dosage , Pyrrolidinones/pharmacokinetics , Raltegravir Potassium , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Treatment Outcome , Triazoles/administration & dosage , Triazoles/pharmacokinetics , Young Adult
14.
Antimicrob Agents Chemother ; 56(8): 4303-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22644026

ABSTRACT

Lersivirine is a nonnucleoside reverse transcriptase inhibitor (NNRTI) with a unique resistance profile exhibiting potent antiviral activity against wild-type HIV and several clinically relevant NNRTI-resistant strains. Lersivirine, a weak inducer of the cytochrome P450 (CYP) enzyme CYP3A4, is metabolized by CYP3A4 and UDP glucuronosyltransferase 2B7 (UGT2B7). Two open, randomized, two-way (study 1; study A5271008) or three-way (study 2; study A5271043) crossover phase I studies were carried out under steady-state conditions in healthy subjects. Study 1 (n = 17) investigated the effect of oral rifampin on the pharmacokinetics (PKs) of lersivirine. Study 2 (n = 18) investigated the effect of oral rifabutin on the PKs of lersivirine and the effect of lersivirine on the PKs of rifabutin and its active metabolite, 25-O-desacetyl-rifabutin. Coadministration with rifampin decreased the profile of the lersivirine area under the plasma concentration-time curve from time zero to 24 h postdose (AUC(24)), maximum plasma concentration (C(max)), and plasma concentration observed at 24 h postdose (C(24)) by 85% (90% confidence interval [CI], 83, 87), 83% (90% CI, 79, 85), and 92% (90% CI, 89, 94), respectively, versus the values for lersivirine alone. Coadministration with rifabutin decreased the lersivirine AUC(24), C(max), and C(24) by 34% (90% CI, 29, 39), 25% (90% CI, 16, 33), and 58% (90% CI, 52, 64), respectively, compared with the values for lersivirine alone. Neither the rifabutin concentration profile nor overall exposure was affected following coadministration with lersivirine. Lersivirine and rifabutin reduced the 25-O-desacetyl-rifabutin AUC(24) by 27% (90% CI, 21, 32) and C(max) by 27% (90% CI, 19, 34). Lersivirine should not be coadministered with rifampin, which is a potent inducer of CYP3A4, UGT2B7, and P-glycoprotein activity and thus substantially lowers lersivirine exposure. No dose adjustment of rifabutin is necessary in the presence of lersivirine; an upward dose adjustment of lersivirine may be warranted when it is coadministered with rifabutin.


Subject(s)
Nitriles/pharmacology , Nitriles/pharmacokinetics , Pyrazoles/pharmacology , Pyrazoles/pharmacokinetics , Rifabutin/pharmacology , Rifabutin/pharmacokinetics , Rifampin/pharmacology , Adolescent , Adult , Cross-Over Studies , Cytochrome P-450 CYP3A/metabolism , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Female , Glucuronosyltransferase/metabolism , HIV/drug effects , HIV Infections/drug therapy , Humans , Male , Middle Aged , Nitriles/administration & dosage , Nitriles/adverse effects , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacology , Rifabutin/administration & dosage , Rifabutin/analogs & derivatives , Rifampin/administration & dosage , Young Adult
15.
Br J Clin Pharmacol ; 73(5): 768-75, 2012 May.
Article in English | MEDLINE | ID: mdl-22040521

ABSTRACT

AIMS: To investigate the effect of inhibitors of cytochrome P450 (CYP) 3A4 and glucuronidation (UGT2B7) on the pharmacokinetics of lersivirine (UK-453,061), a next generation non-nucleoside reverse transcriptase inhibitor with a unique resistance profile, and to investigate the safety and tolerability of co-administration of lersivirine with these inhibitors. METHODS: Two open-label, randomized, placebo-controlled, crossover studies were conducted in healthy subjects. Study 1 investigated the effect of ketoconazole (400 mg once daily) on the pharmacokinetics of lersivirine (250 mg once daily). Subjects received ketoconazole 400 mg once daily or placebo on days 1-2 and received lersivirine 250 mg once daily and ketoconazole 400 mg once daily or placebo on days 3-9. Study 2 investigated the effect of valproic acid (VPA, sodium valproate, 1000 mg once daily) on the PK of lersivirine (500 mg once daily). On days 1-7, subjects received lersivirine 500 mg once daily plus either VPA 1000 mg or placebo. RESULTS: Compared with lersivirine alone, co-administration with ketoconazole increased the lersivirine mean area under the curve (AUC(0,24 h)) and maximum plasma concentration (C(max) ) by 82% (90% CI 74%, 91%) and 61% (90% CI 41%, 83%), respectively. VPA increased the mean lersivirine AUC(0,24 h) by 25% (90% CI 16%, 35%), with little effect on C(max) (2.5%, 90% CI -9%, 16%). There were no serious adverse events and no treatment-related discontinuations from either study. CONCLUSIONS: Inhibition of CYP3A4 and UGT2B7 by ketoconazole increased lersivirine exposure. Inhibition of UGT2B7-mediated glucuronidation by VPA had a modest effect on lersivirine exposure. Co-administration of lersivirine with either ketoconazole or VPA appeared to be well tolerated.


Subject(s)
Antifungal Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ketoconazole/pharmacology , Nitriles/pharmacokinetics , Pyrazoles/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Valproic Acid/pharmacology , Adult , Area Under Curve , Asian People , Cross-Over Studies , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Glucuronosyltransferase/metabolism , Humans , Male , Middle Aged , Nitriles/administration & dosage , Pyrazoles/administration & dosage , Reverse Transcriptase Inhibitors/administration & dosage , Young Adult
16.
Eur J Clin Pharmacol ; 68(11): 1567-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22527351

ABSTRACT

PURPOSE: Lersivirine is a next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) with a unique resistance profile that exhibits potent antiretroviral activity against wild-type human immunodeficiency virus and clinically relevant NNRTI-resistant strains. Results from in vitro and in vivo investigations suggest that lersivirine is a cytochrome P450 (CYP3A4) inducer that is metabolized by CYP3A4 and uridine diphosphate glucuronosyltransferase (UGT) 2B7. In order to formally assess the effects of lersivirine on CYP3A4 metabolism and/or glucuronidation, we performed studies aimed at investigating the effects of lersivirine co-administration on the pharmacokinetics (PK) of midazolam, ethinylestradiol and levonorgestrel. METHODS: Two drug-drug interaction studies were performed. Healthy subjects were co-administered (1) single dose midazolam, a prototypical CYP3A4 substrate, followed by 14 days of lersivirine twice daily with single dose midazolam on the final day of lersivirine dosing or (2) 10 days of once-daily (QD) lersivirine and QD oral contraceptives (OCs; ethinylestradiol and levonorgestrel), substrates for CYP3A4, UGT2B7, and/or P-glycoprotein. The effects of co-administration on the PK parameters of midazolam and OCs were assessed. RESULTS: At clinically relevant lersivirine doses (500-1,000 mg total daily dose), the mean plasma exposure of midazolam was reduced in a dose-dependent manner by 20-36 %. Co-administration of lersivirine 1,000 mg QD with OCs had minor PK effects, increasing ethinylestradiol exposure by 10 % and reducing levonorgestrel exposure by 13 %. CONCLUSIONS: These data further support previous observations that lersivirine is a weak CYP3A4 inducer, a weak inhibitor of glucuronidation, and a P-glycoprotein inhibitor. In both studies, lersivirine appeared to have a good safety and tolerability profile.


Subject(s)
Contraceptives, Oral/pharmacokinetics , Ethinyl Estradiol/pharmacokinetics , Levonorgestrel/pharmacokinetics , Midazolam/pharmacokinetics , Nitriles/pharmacology , Pyrazoles/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Adolescent , Adult , Biotransformation/drug effects , Contraceptives, Oral/administration & dosage , Contraceptives, Oral/adverse effects , Contraceptives, Oral/blood , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Combinations , Drug Interactions , Ethinyl Estradiol/adverse effects , Ethinyl Estradiol/blood , Female , Half-Life , Humans , Levonorgestrel/administration & dosage , Levonorgestrel/adverse effects , Levonorgestrel/blood , Male , Metabolic Clearance Rate , Midazolam/adverse effects , Midazolam/blood , Middle Aged , Nitriles/adverse effects , Pyrazoles/adverse effects , Reverse Transcriptase Inhibitors/adverse effects , Young Adult
17.
Clin Transl Sci ; 15(7): 1561-1580, 2022 07.
Article in English | MEDLINE | ID: mdl-35470578

ABSTRACT

Although rifampin drug-drug interaction (DDI) studies are routinely conducted, there have been instances of liver function test (LFT) elevations, warranting further evaluation. A literature review was conducted to identify studies in which combination with rifampin resulted in hepatic events and evaluate any similarities. Over 600 abstracts and manuscripts describing rifampin DDI studies were first evaluated, of which 30 clinical studies reported LFT elevations. Out of these, 11 studies included ritonavir in combination with other drug(s) in the rifampin DDI study. The number of subjects that were discontinued from treatment on these studies ranged from 0 to 71 (0-100% of subjects in each study). The number of subjects hospitalized for adverse events in these studies ranged from 0 to 41 (0-83.67% of subjects in each study). LFT elevations in greater than 50% of subjects were noted during the concomitant administration of rifampin with ritonavir-boosted protease inhibitors and with lorlatinib; with labeled contraindication due to observed hepatotoxicity related safety findings only for saquinavir/ritonavir and lorlatinib. In the lorlatinib and ritonavir DDI studies, considerable LFT elevations were observed rapidly, typically within 24-72 h following co-administration. A possible sequence effect has been speculated, where rifampin induction prior to administration of the combination may be associated with increased severity of the LFT elevations. The potential role of rifampin in the metabolic activation of certain drugs into metabolites with hepatic effects needs to be taken into consideration when conducting rifampin DDI studies, particularly those for which the metabolic profiles are not fully elucidated.


Subject(s)
HIV Infections , HIV Protease Inhibitors , Drug Interactions , HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , Humans , Lactams, Macrocyclic/therapeutic use , Liver Function Tests , Rifampin/adverse effects , Ritonavir/adverse effects
18.
Clin Pharmacol Ther ; 111(2): 425-434, 2022 02.
Article in English | MEDLINE | ID: mdl-34623637

ABSTRACT

Preparations of plasma-derived small extracellular vesicles (sEVs) were deployed as liquid biopsy to study cytochrome P450 (CYP) 3A4 (CYP3A4) induction following modafinil 400 mg once daily × 14 days (young healthy volunteers, N = 10 subjects). Induction was confirmed using the 4ß-hydroxycholesterol-to-cholesterol (4ßHC/C) ratio, a plasma CYP3A4/5 biomarker, with a mean 2.1-fold increase (Day 15 vs. Day 1; 90% confidence interval (CI) = 1.8-2.3; P value = 0.0004). Proteomic analysis revealed the induction (mean Day 15 vs. Day 1 fold-increase (90% CI)) of both liver (1.3 (1.1-1.5), P value = 0.014) and nonliver (1.9 (1.6-2.2), P value = 0.04) sEV CYP3A4 protein expression. In CYP3A5 nonexpresser subjects, the baseline (pre-dose) 4ßHC/C plasma ratio was more highly correlated with liver sEVs (r = 0.937, P value = 0.001) than nonliver sEVs (r = 0.619, P value = 0.101) CYP3A4 protein expression. When CYP3A5 expressers (CYP3A5*1/*3) were included, the correlation with liver sEVs (r = 0.761, P value = 0.011) and nonliver sEVs (r = 0.391, P value = 0.264) CYP3A4 protein was weaker. Although modafinil-induced changes in plasma 4ßHC/C ratio did not correlate with sEVs CYP3A4 protein expression, the individual subject sEVs proteomic data were used successfully to predict victim drug (midazolam, triazolam, dextromethorphan, 17α-ethinylestradiol, and abemaciclib) area under the plasma concentration-time curve (AUC) ratios (AUCRs) following modafinil. Based on the AUCR values, modafinil was classified as a weak to moderate CYP3A4 inducer (vs. rifampicin). For the first time, it was possible to deploy plasma-derived sEVs to study CYP3A4 induction beyond rifampicin, a more potent CYP3A4 inducer.


Subject(s)
Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A/biosynthesis , Modafinil/administration & dosage , Biomarkers/blood , Cytochrome P-450 CYP3A/blood , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A Inducers/adverse effects , Drug Administration Schedule , Drug Interactions , Enzyme Induction , Extracellular Vesicles/drug effects , Extracellular Vesicles/enzymology , Healthy Volunteers , Humans , Hydroxycholesterols/blood , Liquid Biopsy , Liver/enzymology , Modafinil/adverse effects , Models, Biological , Plasma/enzymology , Proteomics , Rifampin/administration & dosage , Rifampin/adverse effects , Time Factors
19.
Bioanalysis ; 14(14): 971-984, 2022 Jul.
Article in English | MEDLINE | ID: mdl-36066071

ABSTRACT

Aim: Novel urinary biomarker evaluation approaches to support inhibition assessment for renal transporters (e.g., OCT2, multidrug and toxin extrusion proteins [MATEs]). Methods: Highly sensitive and robust hydrophilic interaction chromatography-MS/high-resolution MS assays, for urine and plasma, were developed and characterized to evaluate transporter biomarkers including N1-methyladenosine and N1-methylnicotinamide. Results: The assays were simple and reliable with good selectivity and sensitivity, and successfully supported a clinical drug-drug interaction study with a drug candidate that presented in vitro inhibition of OCT2 and MATEs. Conclusion: The multiplexed assays enable a performance comparison, including biomarker specificity and sensitivity, that should increase the confidence in early clinical OCT2/MATEs drug-drug interaction risk assessment.


Subject(s)
Organic Cation Transport Proteins , Tandem Mass Spectrometry , Biomarkers , Drug Interactions , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism
20.
Clin Pharmacol Ther ; 111(2): 404-415, 2022 02.
Article in English | MEDLINE | ID: mdl-34605015

ABSTRACT

Quantitative prediction of drug-drug interactions (DDIs) involving organic anion transporting polypeptide (OATP)1B1/1B3 inhibition is limited by uncertainty in the translatability of experimentally determined in vitro inhibition potency (half-maximal inhibitory concentration (IC50 )). This study used an OATP1B endogenous biomarker-informed physiologically-based pharmacokinetic (PBPK) modeling approach to predict the effect of inhibitor drugs on the pharmacokinetics (PKs) of OATP1B substrates. Initial static analysis with about 42 inhibitor drugs, using in vitro IC50 values and unbound liver inlet concentrations (Iin,max,u ), suggested in vivo OATP1B inhibition risk for drugs with R-value (1+ Iin,max,u /IC50 ) above 1.5. A full-PBPK model accounting for transporter-mediated hepatic disposition was developed for coproporphyrin I (CP-I), an endogenous OATP1B biomarker. For several inhibitors (cyclosporine, diltiazem, fenebrutinib, GDC-0810, itraconazole, probenecid, and rifampicin at 3 different doses), PBPK models were developed and verified against available CP-I plasma exposure data to obtain in vivo OATP1B inhibition potency-which tend to be lower than the experimentally measured in vitro IC50 by about 2-fold (probenecid and rifampicin) to 37-fold (GDC-0810). Models verified with CP-I data are subsequently used to predict DDIs with OATP1B probe drugs, rosuvastatin and pitavastatin. The predicted and observed area under the plasma concentration-time curve ratios are within 20% error in 55% cases, and within 30% error in 89% cases. Collectively, this comprehensive study illustrates the adequacy and utility of endogenous biomarker-informed PBPK modeling in mechanistic understanding and quantitative predictions of OATP1B-mediated DDIs in drug development.


Subject(s)
Atorvastatin/pharmacokinetics , Coproporphyrins/blood , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Liver/drug effects , Membrane Transport Modulators/pharmacology , Models, Biological , Rosuvastatin Calcium/pharmacokinetics , Biomarkers/blood , Computer Simulation , Drug Interactions , HEK293 Cells , Humans , Liver/metabolism , Liver-Specific Organic Anion Transporter 1/genetics , Liver-Specific Organic Anion Transporter 1/metabolism , Risk Assessment , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL