Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Article in English | MEDLINE | ID: mdl-30745392

ABSTRACT

The combination of the hepatitis C virus (HCV) nonstructural protein 5A (NS5A) inhibitor elbasvir and the NS3/4A protease inhibitor grazoprevir is a potent, once-daily therapy indicated for the treatment of chronic HCV infection in individuals coinfected with human immunodeficiency virus (HIV). We explored the pharmacokinetic interactions of elbasvir and grazoprevir with ritonavir and ritonavir-boosted HIV protease inhibitors in three phase 1 trials. Drug-drug interaction trials with healthy participants were conducted to evaluate the effect of ritonavir on the pharmacokinetics of grazoprevir (n = 10) and the potential two-way pharmacokinetic interactions of elbasvir (n = 30) or grazoprevir (n = 39) when coadministered with ritonavir-boosted atazanavir, lopinavir, or darunavir. Coadministration of ritonavir with grazoprevir increased grazoprevir exposure; the geometric mean ratio (GMR) for grazoprevir plus ritonavir versus grazoprevir alone area under the concentration-time curve from 0 to 24 h (AUC0-24) was 1.91 (90% confidence interval [CI]; 1.31 to 2.79). Grazoprevir exposure was markedly increased with coadministration of atazanavir-ritonavir, lopinavir-ritonavir, and darunavir-ritonavir, with GMRs for grazoprevir AUC0-24 of 10.58 (90% CI, 7.78 to 14.39), 12.86 (90% CI, 10.25 to 16.13), and 7.50 (90% CI, 5.92 to 9.51), respectively. Elbasvir exposure was increased with coadministration of atazanavir-ritonavir, lopinavir-ritonavir, and darunavir-ritonavir, with GMRs for elbasvir AUC0-24 of 4.76 (90% CI, 4.07 to 5.56), 3.71 (90% CI, 3.05 to 4.53), and 1.66 (90% CI, 1.35 to 2.05), respectively. Grazoprevir and elbasvir had little effect on atazanavir, lopinavir, and darunavir pharmacokinetics. Coadministration of elbasvir-grazoprevir with atazanavir-ritonavir, lopinavir-ritonavir, or darunavir-ritonavir is contraindicated, owing to an increase in grazoprevir exposure. Therefore, HIV treatment regimens without HIV protease inhibitors should be considered for HCV/HIV-coinfected individuals who are being treated with elbasvir-grazoprevir.


Subject(s)
Antiviral Agents/pharmacokinetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Hepatitis C/drug therapy , Adult , Amides , Antiviral Agents/pharmacology , Atazanavir Sulfate/pharmacokinetics , Atazanavir Sulfate/pharmacology , Benzofurans/pharmacokinetics , Benzofurans/pharmacology , Carbamates , Cyclopropanes , Darunavir/pharmacokinetics , Darunavir/pharmacology , Drug Interactions , Female , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Healthy Volunteers , Hepacivirus/drug effects , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lopinavir/pharmacokinetics , Lopinavir/pharmacology , Male , Middle Aged , Quinoxalines/pharmacokinetics , Quinoxalines/pharmacology , Ritonavir/pharmacokinetics , Ritonavir/pharmacology , Sulfonamides , Viral Nonstructural Proteins/antagonists & inhibitors , Young Adult
2.
Br J Clin Pharmacol ; 78(3): 587-98, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24617605

ABSTRACT

AIMS: Rosuvastatin and pitavastatin have been proposed as probe substrates for the organic anion-transporting polypeptide (OATP) 1B, but clinical data on their relative sensitivity and selectivity to OATP1B inhibitors are lacking. A clinical study was therefore conducted to determine their relative suitability as OATP1B probes using single oral (PO) and intravenous (IV) doses of the OATP1B inhibitor rifampicin, accompanied by a comprehensive in vitro assessment of rifampicin inhibitory potential on statin transporters. METHODS: The clinical study comprised of two separate panels of eight healthy subjects. In each panel, subjects were randomized to receive a single oral dose of rosuvastatin (5 mg) or pitavastatin (1 mg) administered alone, concomitantly with rifampicin (600 mg) PO or IV. The in vitro transporter studies were performed using hepatocytes and recombinant expression systems. RESULTS: Rifampicin markedly increased exposures of both statins, with greater differential increases after PO vs. IV rifampicin only for rosuvastatin. The magnitudes of the increases in area under the plasma concentration-time curve were 5.7- and 7.6-fold for pitavastatin and 4.4- and 3.3-fold for rosuvastatin, after PO and IV rifampicin, respectively. In vitro studies showed that rifampicin was an inhibitor of OATP1B1 and OATP1B3, breast cancer resistance protein and multidrug resistance protein 2, but not of organic anion transporter 3. CONCLUSIONS: The results indicate that pitavastatin is a more sensitive and selective and thus preferred clinical OATP1B probe substrate than rosuvastatin, and that a single IV dose of rifampicin is a more selective OATP1B inhibitor than a PO dose.


Subject(s)
Fluorobenzenes/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Quinolines/pharmacokinetics , Rifampin/pharmacology , Sulfonamides/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adult , Area Under Curve , Cross-Over Studies , Drug Interactions , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver-Specific Organic Anion Transporter 1 , Male , Middle Aged , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/metabolism , Rifampin/administration & dosage , Rosuvastatin Calcium , Solute Carrier Organic Anion Transporter Family Member 1B3 , Young Adult
3.
Clin Pharmacol Drug Dev ; 8(7): 952-961, 2019 10.
Article in English | MEDLINE | ID: mdl-31173673

ABSTRACT

Treatment of individuals coinfected with human immunodeficiency virus (HIV) and hepatitis C virus (HCV) requires careful consideration of potential drug-drug interactions. The pharmacokinetic interaction of the HCV fixed-dose combination treatment of elbasvir/grazoprevir (EBR/GZR) when coadministered with the fixed-dose combination HIV treatment of elvitegravir/cobicistat/tenofovir disoproxil fumarate/emtricitabine (EVG/COB/TDF/FTC) was evaluated in 22 healthy adults. In period 1, oral doses of EVG/COB/TDF/FTC (150 mg/150 mg/300 mg/200 mg) were administered once daily for 7 days. In period 2, oral doses of EBR/GZR (50 mg/100 mg) were administered once daily for 10 days. In period 3, oral doses of EVG/COB/TDF/FTC were coadministered with EBR/GZR once daily for 10 days. The pharmacokinetics of EVG/COB/TDF/FTC were not clinically meaningfully altered by concomitant EBR/GZR administration. Geometric mean ratios (90%CIs) for area under the plasma concentration-time curve from time 0 to 24 hours (AUC0-24 ) in the presence/absence of EBR/GZR were 1.1 (1.0, 1.2) for elvitegravir; 1.1 (1.0, 1.1) for emtricitabine; 1.2 (1.1, 1.2) for tenofovir; and 1.5 (1.4, 1.6) for cobicistat. In comparison, the AUC0-24 of elbasvir was ∼2 times higher and the AUC0-24 of grazoprevir was ∼5 times higher following concomitant administration of EVG/COB/TDF/FTC and EBR/GZR. Geometric mean ratios (90%CI) for AUC0-24 in the presence/absence of EVG/COB/TDF/FTC were 2.2 (2.0, 2.4) for elbasvir and 5.4 (4.5, 6.4) for grazoprevir. Coadministration of EVG/COB/TDF/FTC and EBR/GZR was generally well tolerated in healthy adults in this study. Nevertheless, because of the increased GZR exposure that occurs with coadministration of EVG/COB/TDF/FTC and EBR/GZR, coadministration of this combination is not recommended in those coinfected with HIV and HCV.


Subject(s)
Benzofurans/pharmacokinetics , Cobicistat/pharmacokinetics , Emtricitabine/pharmacokinetics , Imidazoles/pharmacokinetics , Quinolones/pharmacokinetics , Quinoxalines/pharmacokinetics , Tenofovir/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Benzofurans/administration & dosage , Cobicistat/administration & dosage , Drug Administration Schedule , Drug Combinations , Drug Interactions , Emtricitabine/administration & dosage , Female , Healthy Volunteers , Humans , Imidazoles/administration & dosage , Male , Middle Aged , Quinolones/administration & dosage , Quinoxalines/administration & dosage , Tenofovir/administration & dosage
4.
Clin Pharmacol Drug Dev ; 8(7): 962-970, 2019 10.
Article in English | MEDLINE | ID: mdl-31173674

ABSTRACT

Treatment of individuals coinfected with hepatitis C virus (HCV) and human immunodeficiency virus (HIV) requires careful consideration of potential drug-drug interactions. We evaluated the pharmacokinetic interaction of the direct-acting antiviral agents elbasvir and grazoprevir coadministered with the nucleotide reverse transcriptase inhibitor tenofovir disoproxil fumarate (TDF). Three open-label, multidose studies in healthy adults were conducted. In the first study (N = 10), participants received TDF 300 mg once daily, elbasvir 50 mg once daily, and elbasvir coadministered with TDF. In the second study (N = 12), participants received TDF 300 mg once daily, grazoprevir 200 mg once daily, and grazoprevir coadministered with TDF. In the third study (N = 14), participants received TDF 300 mg once daily and TDF 300 mg coadministered with coformulated elbasvir/grazoprevir 50 mg/100 mg once daily. Pharmacokinetics and safety were evaluated. Following coadministration, the tenofovir area under the plasma concentration-time curve to 24 hours and maximum plasma concentration geometric mean ratios (90% confidence intervals) for tenofovir and coadministered drug(s) versus tenofovir were 1.3 (1.2, 1.5) and 1.5 (1.3, 1.6), respectively, when coadministered with elbasvir; 1.2 (1.1, 1.3) and 1.1 (1.0, 1.2), respectively, when coadministered with grazoprevir; and 1.3 (1.2, 1.4) and 1.1 (1.0, 1.4), respectively, when coadministered with the elbasvir/grazoprevir coformulation. TDF had minimal effect on elbasvir and grazoprevir pharmacokinetics. Elbasvir and/or grazoprevir coadministered with TDF resulted in no clinically meaningful tenofovir exposure increases and was generally well tolerated, with no deaths, serious adverse events (AEs), discontinuations due to AEs, or laboratory AEs reported. No dose adjustments for elbasvir/grazoprevir or TDF are needed for coadministration in HCV/HIV-coinfected people.


Subject(s)
Antiviral Agents/pharmacokinetics , Benzofurans/pharmacokinetics , Imidazoles/pharmacokinetics , Quinoxalines/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Tenofovir/pharmacokinetics , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Area Under Curve , Benzofurans/administration & dosage , Benzofurans/adverse effects , Drug Administration Schedule , Drug Combinations , Drug Interactions , Female , HIV/drug effects , Healthy Volunteers , Hepacivirus/drug effects , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Male , Middle Aged , Quinoxalines/administration & dosage , Quinoxalines/adverse effects , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Tenofovir/administration & dosage , Tenofovir/adverse effects , Young Adult
5.
Clin Transl Sci ; 11(6): 553-561, 2018 11.
Article in English | MEDLINE | ID: mdl-30040872

ABSTRACT

We conducted two phase I trials to evaluate the pharmacokinetic interactions between elbasvir (EBR), grazoprevir (GZR), and methadone (MK-8742-P010 and MK-5172-P030) in non-hepatitis C virus (HCV)-infected participants on methadone maintenance therapy. Coadministration of EBR or GZR with methadone had no clinically meaningful effect on EBR, GZR, or methadone pharmacokinetics. The geometric mean ratios (GMRs) for R- and S-methadone AUC0-24 were 1.03 (90% confidence interval (CI), 0.92-1.15) and 1.09 (90% CI, 0.94-1.26) in the presence/absence of EBR; and 1.09 (90% CI, 1.02-1.17) and 1.23 (90% CI, 1.12-1.35) in the presence/absence of GZR. The GMRs for EBR and GZR AUC0-24 in participants receiving methadone relative to a healthy historical cohort not receiving methadone were 1.20 (90% CI, 0.94-1.53) and 1.03 (90% CI, 0.76-1.41), respectively. These results indicate that no dose adjustment is required for individuals with HCV infection receiving stable methadone therapy and the EBR/GZR fixed-dose regimen.


Subject(s)
Analgesics, Opioid/agonists , Antiviral Agents/pharmacokinetics , Benzofurans/pharmacokinetics , Imidazoles/pharmacokinetics , Methadone/pharmacokinetics , Quinoxalines/pharmacokinetics , Adult , Amides , Antiviral Agents/administration & dosage , Area Under Curve , Benzofurans/administration & dosage , Carbamates , Cyclopropanes , Drug Interactions , Drug Therapy, Combination/methods , Female , Healthy Volunteers , Hepatitis C/complications , Hepatitis C/drug therapy , Humans , Imidazoles/administration & dosage , Male , Methadone/administration & dosage , Middle Aged , Opiate Substitution Treatment/methods , Opioid-Related Disorders/complications , Opioid-Related Disorders/rehabilitation , Quinoxalines/administration & dosage , Sulfonamides , Young Adult
6.
Clin Transl Sci ; 11(6): 562-572, 2018 11.
Article in English | MEDLINE | ID: mdl-30040871

ABSTRACT

The aims of these phase I trials were to evaluate the pharmacokinetic interaction between elbasvir (EBR) or grazoprevir (GZR) and buprenorphine/naloxone (BUP/NAL). Trial 1 was a single-dose trial in healthy participants. Trial 2 was a multiple-dose trial in participants on BUP/NAL maintenance therapy. Coadministration of EBR or GZR with BUP/NAL had minimal effect on the pharmacokinetics of BUP/NAL, EBR, and GZR. The geometric mean ratios (GMRs (90% CI)) for BUP, norbuprenorphine, and NAL AUC0-∞ were 0.98 (0.89-1.08), 0.97 (0.86-1.09), and 0.88 (0.78-1.00) in the presence/absence of EBR; 0.98 (0.81-1.19), 1.13 (0.97-1.32), and 1.10 (0.82-1.47) in the presence/absence of GZR. The GMRs (90% CI) for EBR and GZR AUC0-∞ in the absence/presence of BUP/NAL were 1.22 (0.98-1.52) and 0.86 (0.63-1.18). In conclusion, no dose adjustment for BUP/NAL, EBR, or GZR is required for patients with HCV infection receiving EBR/GZR and BUP/NAL maintenance therapy.


Subject(s)
Analgesics, Opioid/agonists , Antiviral Agents/pharmacokinetics , Benzofurans/pharmacokinetics , Buprenorphine, Naloxone Drug Combination/pharmacokinetics , Imidazoles/pharmacokinetics , Quinoxalines/pharmacokinetics , Adult , Amides , Antiviral Agents/administration & dosage , Area Under Curve , Benzofurans/administration & dosage , Buprenorphine, Naloxone Drug Combination/administration & dosage , Carbamates , Cyclopropanes , Drug Interactions , Drug Therapy, Combination/methods , Female , Healthy Volunteers , Hepatitis C/complications , Hepatitis C/drug therapy , Humans , Imidazoles/administration & dosage , Male , Middle Aged , Opiate Substitution Treatment/methods , Opioid-Related Disorders/complications , Opioid-Related Disorders/rehabilitation , Quinoxalines/administration & dosage , Sulfonamides , Young Adult
7.
J Clin Pharmacol ; 44(9): 1054-62, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15317833

ABSTRACT

Simvastatin and fenofibrate are both commonly used lipid-regulating agents with distinct mechanisms of action, and their coadministration may be an attractive treatment for some patients with dyslipidemia. A 2-period, randomized, open-label, crossover study was conducted in 12 subjects to determine if fenofibrate and simvastatin are subject to a clinically relevant pharmacokinetic interaction at steady state. In treatment A, subjects received an 80-mg simvastatin tablet in the morning for 7 days. In treatment B, subjects received a 160-mg micronized fenofibrate capsule in the morning for 7 days, followed by a 160-mg micronized fenofibrate capsule dosed together with an 80-mg simvastatin tablet on days 8 to 14. Because food increases the bioavailability of fenofibrate, each dose was administered with food to maximize the exposure of fenofibric acid. The steady-state pharmacokinetics (AUC(0-24h), C(max), and t(max)) of active and total HMG-CoA reductase inhibitors, simvastatin acid, and simvastatin were determined following simvastatin administration with and without fenofibrate. Also, fenofibric acid steady-state pharmacokinetics were evaluated with and without simvastatin. The geometric mean ratios (GMRs) for AUC(0-24h) (80 mg simvastatin [SV] + 160 mg fenofibrate)/(80 mg simvastatin alone) and 90% confidence intervals (CIs) were 0.88 (0.80, 0.95) and 0.92 (0.82, 1.03) for active and total HMG-CoA reductase inhibitors. The GMRs and 90% CIs for fenofibric acid (80 mg SV + 160 mg fenofibrate/160 mg fenofibrate alone) AUC(0-24h) and C(max) were 0.95 (0.88, 1.04) and 0.89 (0.77, 1.02), respectively. Because both the active inhibitor and fenofibric acid AUC GMR 90% confidence intervals fell within the prespecified bounds of (0.70, 1.43), no clinically significant pharmacokinetic drug interaction between fenofibrate and simvastatin was concluded in humans. The coadministration of simvastatin and fenofibrate in this study was well tolerated.


Subject(s)
Fenofibrate/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypolipidemic Agents/pharmacokinetics , Simvastatin/pharmacokinetics , Adolescent , Adult , Area Under Curve , Biotransformation , Blood Cell Count , Cross-Over Studies , Drug Interactions , Electrocardiography/drug effects , Female , Fenofibrate/adverse effects , Half-Life , Humans , Hydroxymethylglutaryl CoA Reductases/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hypolipidemic Agents/adverse effects , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Middle Aged , Simvastatin/adverse effects
9.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(14-15): 1465-71, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19346171

ABSTRACT

Telcagepant (MK-0974) is a novel oral calcitonin gene-related peptide (CGRP) receptor antagonist and is currently under clinical development. Results from phases II and III clinical trials have suggested that telcagepant is effective for migraine treatment. A reliable and high throughput protein precipitation (PPT) method for determination of telcagepant in human plasma using liquid chromatography coupled with atmospheric pressure chemical ionization (APCI) tandem mass spectrometry has been developed. Clinical samples, internal standard (IS) and acetonitrile are transferred into 96-well plates using a robotic liquid handling system. An aliquot of 10 microL supernatant is directly injected into the LC-MS/MS system where separation is performed on a FluoPhase RP (150 x 2.1mm, 5 microm) column with an isocratic mobile phase (60% acetonitrile with 0.1% formic acid and 40% water with 0.1% formic acid) at 0.2 mL/min. The interfering 3S-diastereomer of telcagepant, which is observed in clinical samples, is chromatographically resolved from telcagepant. The PPT procedure significantly reduces the time required for sample processing and the assay is sufficiently sensitive for detection using both API 4000 and API 3000 mass spectrometers. The linear calibration range is 5-5000 nM using 200 microL of plasma. Assay intraday validation was conducted using six calibration curves derived from six lots of human control plasma. Calibration standard accuracy did not deviate by more than 3% and 6% of nominal values, and precision did not exceed 4% coefficient of variation (CV) and 10% CV, respectively on the API 4000 and API 3000. Several clinical phases IIb and III studies have been successfully supported with this assay.


Subject(s)
Azepines/blood , Chromatography, High Pressure Liquid/methods , Imidazoles/blood , Tandem Mass Spectrometry/methods , Humans , Migraine Disorders/blood , Migraine Disorders/drug therapy
10.
J Pharmacol Toxicol Methods ; 57(1): 61-9, 2008.
Article in English | MEDLINE | ID: mdl-17651990

ABSTRACT

INTRODUCTION: A Tecan-based enzyme inhibition assay has been developed for the determination of atorvastatin-derived 'active' and 'total' (active inhibitors plus atorvastatin lactone and other potential inhibitors following base hydrolysis) 3-hydroxy-3-methylglutaryl-Coenzyme A (HMG-CoA) reductase inhibitor concentrations in human plasma. Atorvastatin is an inhibitor of HMG-CoA reductase, which is a key rate-limiting enzyme in the cholesterol biosynthesis. Previously, atorvastatin-derived HMG-CoA reductase inhibitors were measured via enzyme inhibition assays by manual operation. METHODS: In this work, an enzyme assay procedure based on 8-tip Tecan robotics and set-up in a 96-well plate format with customized hardware is presented. Following protein precipitation of the plasma sample, an aliquot of the resulting supernatant is mixed with HMG-CoA reductase and (14)C-labeled HMG-CoA prior to incubation. The product, (14)C-mevalonic acid, is lactonized, separated from unreacted (14)C-substrate, and counted in a liquid scintillation counter. Plasma HMG-CoA reductase inhibitor concentrations are measured against atorvastatin as the standard. Tecan Genesis 150 and 200 robotic workstations were used for the protein precipitation, enzyme incubation, and product separation. RESULTS: The standard calibration range for the assay was 0.4-20 ng eq/mL. Intra-day precision (%CV) data for the calibration standard and quality control (QC) samples (n=5 replicates) were both

Subject(s)
Heptanoic Acids/analysis , Heptanoic Acids/blood , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Pyrroles/analysis , Pyrroles/blood , Atorvastatin , Heptanoic Acids/chemistry , Humans , Indicators and Reagents , Molecular Structure , Pyrroles/chemistry , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL