Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Drug Metab Dispos ; 51(12): 1607-1614, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37684056

ABSTRACT

Pomotrelvir is an orally bioavailable, target antiviral inhibitor of the main protease (Mpro) of coronaviruses, including severe acute respiratory syndrome coronavirus 2, the etiological agent of Coronavirus Disease 2019. The pharmacokinetics, metabolism and elimination of two [14C]-labeled microtracers of 5 µCi/700 mg pomotrelvir with separate labeling positions (isotopomers), [lactam carbonyl-14C-pomotelvir] and [benzene ring-U-14C-pomotrelvir], following a single oral dose in healthy adult males was evaluated in two separate cohorts. Pomotrelvir was rapidly absorbed and eliminated primarily through metabolism and subsequently excreted via urine and feces. There were no differences in pomotrelvir pharmacokinetics between the two cohorts. The mean total radioactive dose recovered was 93.8% (n = 8) in the lactam cohort (58% in urine and 36% in feces) and 94.2% (n = 8) in the benzene cohort (75% in urine and 19% in feces), with ≥80% of [14C] recovered within 96 hours after dosing. About 5% and 3% of the intact pomotrelvir was recovered in feces and urine, respectively. Eleven major metabolites were detected and characterized using liquid chromatography-accelerator mass spectrometry and liquid chromatography tandem mass spectrometry methods, with three and six different metabolites elucidated in the samples collected from lactam and benzene cohorts, respectively, and two metabolites observed in both cohorts. The major metabolism pathway of pomotrelvir is through hydrolysis of its peptide bonds followed by phase II conjugations. These results support that the application of two radiolabeled isotopomers provided a comprehensive metabolite profiling analysis and was a successful approach in identifying the major disposition pathways of pomotrelvir that has complex routes of metabolism. SIGNIFICANCE STATEMENT: An unconventional approach using two differentially labeled [14C] microtracers, [lactam carbonyl-14C-pomotrelvir] and [benzene ring-U-14C-pomotrelvir] evaluated the mass balance of orally administered pomotrelvir in healthy adult males in two separate cohorts. The radioactive dose recovered in excreta was about 94% for both cohorts. While the two isotopomers of the radiolabeled-pomotrelvir showed no major differences in pharmacokinetics overall, they allowed for differential detection of their radiolabeled metabolites and appropriate characterization of their plasma exposure and excretion in urine and feces.


Subject(s)
Benzene , Lactams , Adult , Humans , Male , Chromatography, High Pressure Liquid/methods , Benzene/analysis , Chromatography, Liquid , Biotransformation , Feces/chemistry , Lactams/analysis , Administration, Oral , Carbon Radioisotopes/analysis
2.
CPT Pharmacometrics Syst Pharmacol ; 12(10): 1553-1564, 2023 10.
Article in English | MEDLINE | ID: mdl-37614073

ABSTRACT

Pomotrelvir is a new chemical entity and potent direct-acting antiviral inhibitor of the main protease of coronaviruses. Here the cytochrome P450 (CYP)-mediated drug-drug interaction (DDI) potential of pomotrelvir was evaluated for major CYP isoforms, starting with in vitro assays followed by the basic static model assessment. The identified CYP3A4-mediated potential DDIs were evaluated clinically at a supratherapeutic dose of 1050 mg twice daily (b.i.d.) of pomotrelvir, including pomotrelvir coadministration with ritonavir (strong inhibitor of CYP3A4) or midazolam (sensitive substrate of CYP3A4). Furthermore, a physiologically-based pharmacokinetic (PBPK) model was developed within the Simcyp Population-based Simulator using in vitro and in vivo information and validated with available human pharmacokinetic (PK) data. The PBPK model was simulated to assess the DDI potential for CYP isoforms that pomotrelvir has shown a weak to moderate DDI in vitro and for CYP3A4 at the therapeutic dose of 700 mg b.i.d. To support the use of pomotrelvir in women of childbearing potential, the impact of pomotrelvir on the exposure of the representative oral hormonal contraceptive drugs ethinyl estradiol and levonorgestrel was assessed using the PBPK model. The overall assessment suggested weak inhibition of pomotrelvir on CYP3A4 and minimal impact of a strong CYP3A4 inducer or inhibitor on pomotrelvir PK. Therefore, pomotrelvir is not anticipated to have clinically meaningful DDIs at the clinical dose. These comprehensive in vitro, in clinic, and in silico efforts indicate that the DDI potential of pomotrelvir is minimal, so excluding patients on concomitant medicines in clinical studies would not be required.


Subject(s)
Cytochrome P-450 CYP3A , Hepatitis C, Chronic , Humans , Female , Antiviral Agents/pharmacology , Cytochrome P-450 Enzyme System , Drug Interactions , Protein Isoforms , Models, Biological , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Computer Simulation
3.
Br J Clin Pharmacol ; 88(7): 3211-3221, 2022 07.
Article in English | MEDLINE | ID: mdl-35072287

ABSTRACT

AIMS: Filgotinib is a potent, oral, JAK1-preferential inhibitor for the treatment of rheumatoid arthritis (RA). This report describes exposure-response (ER) analyses of filgotinib for dose confirmation based on three phase 3 and two phase 2 studies in moderate to severe RA patients. METHODS: The pharmacokinetic exposures used in ER analyses were derived from population pharmacokinetic analysis. The exposure-efficacy relationships were assessed for efficacy endpoints (ACR20/50/70 and DAS28) over effective area under curve (AUCeff ), the combined exposures of filgotinib and GS-829845 (major, active metabolite), with nonlinear logistic regression models developed. Also, a t-test was performed to compare the exposure between subjects who achieved response and those who did not. For the ER analyses of safety, exposures were examined between subjects who experienced and who did not experience the evaluated safety events, which was conducted separately for filgotinib and GS-829845. RESULTS: The nonlinear logistic regression showed increasing response with increasing exposure, with exposures at 200 mg dose primarily residing on the curve plateau. Also, AUCeff was significantly higher in the subjects who achieved responses compared to those who did not (10 900 vs 9900 h*ng/mL for ACR20, P value < .0001). For exposure-safety analyses, filgotinib and GS-829845 exposures were similar irrespective of the presence/absence of the evaluated safety endpoints, indicating no exposure-safety relationship for common treatment-emergent adverse events (TEAEs)/laboratory abnormalities and serious TEAEs/infections. CONCLUSIONS: ER analyses confirmed that filgotinib produced more robust therapeutic effects across the exposure range observed at 200 mg once daily compared to lower doses, and collectively with the lack of exposure-safety relationship, the 200 mg once daily dose was supported for commercialization.


Subject(s)
Arthritis, Rheumatoid , Pyridines , Triazoles , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/drug therapy , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Humans , Janus Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Treatment Outcome , Triazoles/adverse effects
4.
Clin Pharmacol Drug Dev ; 10(4): 376-383, 2021 04.
Article in English | MEDLINE | ID: mdl-32989920

ABSTRACT

Filgotinib (FIL) is a potent and selective JAK1 inhibitor in clinical development for treatment of severe inflammatory diseases. A drug-drug interaction study to evaluate the potential effect of FIL on the pharmacokinetics (PK) of the oral contraceptive levonorgestrel (LEVO)/ethinyl estradiol (EE) was conducted. This was a phase 1, open-label, randomized, crossover study in healthy female subjects (N = 24). Subjects received a single dose of LEVO (150 µg)/EE (30 µg) alone (reference), or in combination with multiple-dose FIL (200 mg once daily for 15 days; test). Intensive PK sampling was conducted, and safety was assessed throughout the study. PK interactions were evaluated using 90% confidence intervals of the geometric least squares mean ratios of the test versus reference treatments. All 24 subjects enrolled completed study treatments. Coadministration of FIL with the oral contraceptive did not alter the PK of LEVO and EE; the 90% confidence intervals of the geometric least squares mean ratios were contained within bioequivalence bounds (80%-125%). Exposures of FIL were consistent with observed clinical exposure data. Study treatments were generally well tolerated. All adverse events were mild. Coadministration with FIL did not alter the PK of LEVO/EE, and hormonal contraceptives can serve as an effective contraception method for subjects on FIL treatment.


Subject(s)
Contraceptives, Oral, Hormonal/pharmacokinetics , Ethinyl Estradiol/pharmacokinetics , Janus Kinase Inhibitors/pharmacology , Levonorgestrel/pharmacokinetics , Pyridines/pharmacology , Triazoles/pharmacology , Adult , Contraceptives, Oral, Hormonal/adverse effects , Cross-Over Studies , Drug Combinations , Drug Interactions , Ethinyl Estradiol/adverse effects , Female , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase Inhibitors/adverse effects , Levonorgestrel/adverse effects , Middle Aged , Pyridines/adverse effects , Triazoles/adverse effects , Young Adult
5.
Clin Pharmacol Ther ; 109(5): 1334-1341, 2021 05.
Article in English | MEDLINE | ID: mdl-33141923

ABSTRACT

Firsocostat (FIR: previously GS-0976), a highly sensitive OATP substrate, reduces hepatic de novo lipogenesis (DNL) by inhibiting acetyl-CoA carboxylases (ACC). Measuring the pharmacodynamic (PD) efficacy of FIR on DNL provides a unique opportunity to determine optimal dosing strategies for liver-targeted OATP substrates in settings of altered OATP function. A randomized, four-way crossover drug-drug interaction study was conducted. Hepatic DNL, a marker for ACC activity, was measured in 28 healthy volunteers after reference, single dose FIR 10 mg, FIR 10 mg plus the OATP inhibitor rifampin (RIF) 300 mg i.v., or RIF 300 mg i.v. (control for DNL effect of RIF), each separated by a 7-day washout. Samples were collected for pharmacokinetic (PK) and PD assessments through 24 hours after each treatment. Hepatic DNL and its inhibition by FIR were assessed. Twenty-four subjects completed the study. All adverse events were mild. RIF alone increased hepatic DNL area under the effect curve from time of administration up to the time of the last quantifiable concentration (AUEClast ; 35.7%). Despite a 5.2-fold increase in FIR plasma exposure (area under the concentration-time curve from zero to infinity (AUCinf )) when administered with RIF, FIR alone, and FIR + RIF had the same hepatic PD effect, 37.1% and 34.9% reduction in DNL AUEClast , respectively, compared with their respective controls. These findings indicate that large decreases in OATP activity do not alter hepatic intracellular exposure (as inferred by no change in PD) for drugs that are primarily eliminated hepatically and permeability rate-limited, such as FIR. These results support PK theory that has been difficult to test and provide practical guidance on administration of liver-targeted drugs in settings of reduced OATP function.


Subject(s)
Isobutyrates/pharmacokinetics , Liver/drug effects , Organic Anion Transporters/antagonists & inhibitors , Oxazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Adult , Drug Interactions , Female , Humans , Isobutyrates/administration & dosage , Isobutyrates/adverse effects , Isobutyrates/blood , Liver/metabolism , Male , Middle Aged , Oxazoles/administration & dosage , Oxazoles/adverse effects , Oxazoles/blood , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/blood , Rifampin/pharmacology
6.
Anal Biochem ; 593: 113611, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32035040

ABSTRACT

Tenofovir disoproxil fumarate (TDF) and tenofovir alafenamide (TAF) are both tenofovir (TFV) prodrugs, with the same active intracellular metabolite, TFV-diphosphate (TFV-DP). TAF delivers TFV-DP to target cells more efficiently and at lower doses than TDF, thereby substantially reducing systemic exposure to TFV, which results in improved bone and renal safety relative to TDF. As such, the method developed for the determination of TFV following TAF administration involved two key differences from determination of TFV following TDF administration. First, human plasma samples (500 µL) immediately upon collection were treated with 20% formic acid (40 µL) (plasma: formic acid ratio of 100:8) to minimize hydrolysis of TAF to TFV, and thereby avoided overestimation of TFV concentrations. Second, various TFV validation tests were conducted in the presence of TAF to mimic the high TAF:TFV ratios in clinical samples collected within ~2 h after dosing. The method for determination of TFV was developed and validated at a US lab and followed FDA and EMA guidelines. To support global clinical studies of TAF, the method was cross-validated (one-way) between the US lab and a China lab and was successfully used for TFV determination in plasma samples from a clinical study that involved healthy Chinese subjects.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacokinetics , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Tenofovir/blood , Adenine/pharmacokinetics , Alanine , Formates/chemistry , Humans , Prodrugs/pharmacokinetics
7.
Clin Pharmacol Drug Dev ; 9(1): 32-40, 2020 01.
Article in English | MEDLINE | ID: mdl-31797578

ABSTRACT

Filgotinib, a selective inhibitor of Janus kinase 1, is being developed for the treatment of chronic inflammatory diseases. Electrocardiograms evaluated the effect of filgotinib on the corrected QT (QTc) interval in 52 healthy subjects who received each of 4 treatments: filgotinib 200 mg (therapeutic dose), 450 mg (supratherapeutic dose), and placebo, each administered once daily for 7 days, and a single dose of moxifloxacin 400 mg (positive control). Plasma samples were collected for pharmacokinetic analysis. The QTc interval was calculated using Fridericia's correction factor (QTcF) or an individual correction factor (QTcI). The relationship between plasma concentrations of filgotinib and its major metabolite and time-matched, baseline-adjusted, placebo-corrected QTc (ΔΔQTc) was evaluated. Filgotinib did not prolong QTcF or QTcI and using an appropriate mixed-effect model, the upper limit of the 2-sided 90% confidence interval for ΔΔQTc for each filgotinib dose (200 and 450 mg) remained below 10 milliseconds at all postdose time points. There were no clinically relevant relationships between QTc interval and plasma concentrations of filgotinib or its major metabolite. Filgotinib, administered at 200 or 450 mg, was generally well tolerated. Results of this thorough QT study demonstrate that filgotinib and its major metabolite are not associated with QTc interval prolongation.


Subject(s)
Heart Rate/drug effects , Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Adult , Cross-Over Studies , Double-Blind Method , Electrocardiography/drug effects , Female , Healthy Volunteers , Humans , Long QT Syndrome , Male , Middle Aged , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/blood , Pyridines/pharmacokinetics , Triazoles/blood , Triazoles/pharmacokinetics
8.
Antivir Ther ; 24(8): 557-566, 2019.
Article in English | MEDLINE | ID: mdl-31933482

ABSTRACT

BACKGROUND: Cobicistat (COBI), a CYP3A inhibitor, is a pharmacokinetic enhancer that increases exposures of the HIV protease inhibitors (PIs) atazanavir (ATV) and darunavir (DRV). The potential drug interaction between COBI-boosted PIs and hormonal contraceptives, which are substrates of intestinal efflux transporters and extensively metabolized by CYP enzymes, glucuronidation and sulfation, was evaluated. METHODS: This was a Phase I, open-label, two cohort (n=18/cohort), fixed-sequence study in healthy females that evaluated the drug-drug interaction (DDI) between multiple-dose ATV+COBI or DRV+COBI and single-dose drospirenone/ethinyl estradiol (EE). DDIs were evaluated using 90% confidence intervals of the geometric least-squares mean ratios of the test (drospirenone/EE+boosted PI) versus reference (drospirenone/EE) using lack of DDI boundaries of 70-143%. Safety was assessed throughout the study. RESULTS: 29/36 participants completed the study. Relative to drospirenone/EE alone, drospirenone area under the plasma concentration versus time curve extrapolated to infinity (AUCinf) was 1.6-fold and 2.3-fold higher, and maximum observed plasma concentration (Cmax) was unaltered, upon coadministration with DRV+COBI and ATV+COBI, respectively. EE AUCinf decreased 30% with drospirenone/EE + DRV+COBI and was unchanged with ATV+COBI + drospirenone/EE, relative to drospirenone/EE alone. Study treatments were generally well tolerated. The majority of adverse events were mild and consistent with known safety profiles of the compounds. CONCLUSIONS: Consistent with COBI-mediated CYP3A inhibition, drospirenone exposure increased following coadministration with COBI-containing regimens, with a greater increase with ATV+COBI. Thus, clinical monitoring for drospirenone-associated hyperkalaemia is recommended with DRV+COBI and ATV+COBI should not be used with drospirenone. Lower EE exposure with DRV+COBI may be attributed to inductive effects of DRV on CYP enzymes and/or intestinal efflux transporters (that is, P-gp) involved in EE disposition.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Cobicistat/pharmacokinetics , Cobicistat/therapeutic use , Ethinyl Estradiol/pharmacokinetics , Adolescent , Adult , Androstenes/administration & dosage , Androstenes/pharmacokinetics , Area Under Curve , Atazanavir Sulfate/pharmacokinetics , Atazanavir Sulfate/therapeutic use , Cohort Studies , Contraceptives, Oral, Hormonal , Darunavir/pharmacokinetics , Darunavir/therapeutic use , Drug Interactions , Ethinyl Estradiol/pharmacology , Female , Half-Life , Humans , Mineralocorticoid Receptor Antagonists/administration & dosage , Mineralocorticoid Receptor Antagonists/pharmacokinetics , Young Adult
9.
J Acquir Immune Defic Syndr ; 78(4): 465-472, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29649076

ABSTRACT

BACKGROUND: Tenofovir alafenamide (TAF), a prodrug of the nucleotide analogue tenofovir (TFV), is an antiretroviral (ARV) agent approved either as a complete regimen [elvitegravir/cobicistat/emtricitabine (F)/TAF, rilpivirine/F/TAF, bictegravir/F/TAF], or for use with other ARVs (F/TAF), for treatment of HIV. TAF is a substrate of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) transporters. Disposition of TAF may be altered by comedications that can inhibit or induce P-gp or BCRP transporters. The effects of ARVs on the pharmacokinetics of TAF were evaluated in 3 studies. METHODS: Healthy participants received TAF administered alone or with rilpivirine in study 1, with dolutegravir, ritonavir-boosted atazanavir (ATV + RTV), lopinavir (LPV/RTV), or darunavir (DRV + RTV) in study 2, and with the pharmacokinetic enhancer cobicistat or efavirenz in study 3. RESULTS: Across the 3 studies, 98 participants received treatment with TAF and a coadministered agent (n = 10-34/cohort). All study treatments were well tolerated. TAF and TFV exposures were unaffected after co-administration with rilpivirine and dolutegravir. Coadministration with P-gp/BCRP inhibitors such as cobicistat or PI-based regimens (ATV + RTV, LPV/r, or DRV + RTV) resulted in a range of 6%-183% increases in TAF and 105%-316% increases in TFV exposure, whereas coadministration with a P-gp inducer, efavirenz, resulted in a 15%-24% decrease in TAF and TFV exposure. CONCLUSIONS: Evaluation of the drug interaction between TAF and other commonly prescribed boosted and unboosted ARVs provides characterization of the susceptibility of TAF and/or TFV pharmacokinetics to inhibitors or inducers of P-gp/BCRP transporters.


Subject(s)
Adenine/analogs & derivatives , Anti-Retroviral Agents/pharmacokinetics , Drug Interactions , Adenine/administration & dosage , Adenine/pharmacokinetics , Adolescent , Adult , Alanine , Anti-Retroviral Agents/administration & dosage , Drug Therapy, Combination/methods , Female , Healthy Volunteers , Humans , Male , Middle Aged , Tenofovir/analogs & derivatives , Young Adult
10.
Clin Pharmacokinet ; 57(11): 1369-1383, 2018 11.
Article in English | MEDLINE | ID: mdl-29644537

ABSTRACT

Ledipasvir/sofosbuvir (Harvoni®), a fixed-dose combination tablet of an NS5A inhibitor ledipasvir and an NS5B polymerase inhibitor sofosbuvir, is approved for the treatment of chronic hepatitis C virus infection. Ledipasvir/sofosbuvir exhibits a favorable drug-drug interaction profile and can be administered with various medications that may be used by hepatitis C virus-infected patients, including patients with comorbidities, such as co-infection with human immunodeficiency virus or immunosuppression following liver transplantation. Ledipasvir/sofosbuvir is not expected to act as a victim or perpetrator of cytochrome P450- or UDP-glucuronosyltransferase 1A1-mediated drug-drug interactions. With the exception of strong inducers of P-glycoprotein, such as rifampin, ledipasvir/sofosbuvir is not expected to act as a victim of clinically relevant drug-drug interactions. As a perpetrator of pharmacokinetic drug-drug interactions via P-glycoprotein/BCRP, ledipasvir/sofosbuvir should not be used with rosuvastatin and elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate, whereas its co-administration with amiodarone is not recommended because of a pharmacodynamic interaction. This review summarizes a number of drug interaction studies conducted in support of the clinical development of ledipasvir/sofosbuvir.


Subject(s)
Benzimidazoles/pharmacology , Benzimidazoles/pharmacokinetics , Drug Interactions , Fluorenes/pharmacology , Fluorenes/pharmacokinetics , Uridine Monophosphate/analogs & derivatives , Humans , Sofosbuvir , Uridine Monophosphate/pharmacokinetics , Uridine Monophosphate/pharmacology
11.
Clin Pharmacol Ther ; 104(6): 1182-1190, 2018 12.
Article in English | MEDLINE | ID: mdl-29569723

ABSTRACT

Drug transporter and cytochrome P450 expression is regulated by shared nuclear receptors and, hence, an inducer should induce both, although the magnitude may differ. The objective of this study was to establish relative induction relationships between CYP3A and drug transporters (P-glycoprotein (P-gp), organic anion transporting polypeptide (OATP), and breast cancer resistance protein (BCRP)) or other P450s (CYP2C9 and CYP1A2) using ascending doses of the prototypical pregnane xenobiotic receptor (PXR) agonist, rifampin, to elicit weak, moderate, and strong PXR agonism. Healthy subjects received dabigatran etexilate, pravastatin, rosuvastatin, and a midazolam/tolbutamide/caffeine cocktail before and after rifampin 2, 10, 75, or 600 mg q.d. Unlike CYP3A, only moderate induction of P-gp, OATP, and CYP2C9 was observed and dose-dependent induction of P-gp, OATP, and CYP2C9 was always one drug-drug interaction category lower than observed for CYP3A, even when correcting for probe drug sensitivity. Data from this study establish proof-of-concept that P450 induction data can be leveraged to inform on the effect on transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 Enzyme Inducers/administration & dosage , Membrane Transport Modulators/administration & dosage , Pregnane X Receptor/agonists , Rifampin/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Biotransformation , Computer Simulation , Cytochrome P-450 Enzyme Inducers/adverse effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction , Female , Healthy Volunteers , Humans , Male , Membrane Transport Modulators/adverse effects , Middle Aged , Models, Biological , Organic Anion Transporters/agonists , Organic Anion Transporters/metabolism , Pharmacokinetics , Pregnane X Receptor/metabolism , Rifampin/adverse effects , Risk Assessment , Substrate Specificity , Young Adult
12.
Clin Pharmacol Ther ; 104(6): 1191-1198, 2018 12.
Article in English | MEDLINE | ID: mdl-29569712

ABSTRACT

Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. The objective of this study was to determine if these relationships could be utilized to predict transporter induction by other CYP3A inducers (rifabutin and carbamazepine) and of another P-gp substrate, sofosbuvir. Healthy subjects received sofosbuvir and a six-probe drug cassette before and after 300 mg q.d. rifabutin or 300 mg b.i.d. carbamazepine. Induction of P-gp, CYP2C9, and decreased sofosbuvir exposure were successfully predicted by observed CYP3A induction. Carbamazepine induction of OATP was underpredicted, likely due to reported additional non-PXR agonism. The results demonstrate that the effect of a PXR agonist on CYP3A can be leveraged to inform on induction liability for other primarily PXR-regulated P450s/transporters, allowing for prioritization of targeted DDI assessments during new drug development.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Carbamazepine/administration & dosage , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 Enzyme Inducers/administration & dosage , Membrane Transport Modulators/administration & dosage , Pregnane X Receptor/agonists , Rifabutin/administration & dosage , Rifampin/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Biotransformation , Carbamazepine/adverse effects , Computer Simulation , Cytochrome P-450 CYP2C9/biosynthesis , Cytochrome P-450 Enzyme Inducers/adverse effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction , Female , Healthy Volunteers , Humans , Male , Membrane Transport Modulators/adverse effects , Middle Aged , Models, Biological , Organic Anion Transporters/agonists , Organic Anion Transporters/metabolism , Pregnane X Receptor/metabolism , Rifabutin/adverse effects , Rifampin/adverse effects , Risk Assessment , Sofosbuvir/metabolism , Substrate Specificity , Young Adult
13.
Clin Pharmacol Drug Dev ; 7(6): 641-651, 2018 08.
Article in English | MEDLINE | ID: mdl-29058821

ABSTRACT

This study evaluated the effect of supratherapeutic exposure of the anti-HCV drug ledipasvir on the QTc interval in healthy subjects. Sixty healthy volunteers were randomized to receive twice-daily blinded ledipasvir (120 mg) or placebo, administered for 10 days each, or single doses of open-label moxifloxacin (400 mg). Serial plasma samples for ledipasvir concentration analysis were collected after each treatment. Triplicate time-matched electrocardiograms were collected at baseline and after each treatment. Change from baseline in the QTc for ledipasvir or moxifloxacin versus placebo was determined using several correction formulas (primary: QTcF [Fridericia's]; secondary: QTcN [population] and QTcI [individual]). Pharmacokinetics and exposure-QTc relationships were evaluated. Ledipasvir AUC0-24 and Cmax achieved approximately 3.7-fold and 4.2-fold, respectively, above exposures observed following administration of ledipasvir/sofosbuvir (90/400 mg) to HCV-infected patients. There was a lack of effect of supratherapeutic ledipasvir on QTc intervals using all correction methods (upper bound of the 2-sided 90%CIs for the mean difference in time-matched baseline-corrected QTc between ledipasvir versus placebo < 10 milliseconds at all times). The lower bound of the 2-sided 96.67%CI for the mean difference in moxifloxacin versus placebo was >5 milliseconds, thereby establishing assay sensitivity. Categorical analyses did not demonstrate clinically relevant effects of ledipasvir on QTc intervals or other electrocardiogram parameters. No relationships between ledipasvir plasma concentration and QTc interval were observed. Ledipasvir does not prolong QTc interval. Based on these results and a previous TQT evaluation for sofosbuvir, the fixed-dose combination regimen of ledipasvir/sofosbuvir is not expected to prolong the QTc interval.


Subject(s)
Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Electrocardiography/drug effects , Fluorenes/administration & dosage , Fluorenes/adverse effects , Adolescent , Adult , Anti-HIV Agents/blood , Area Under Curve , Benzimidazoles/blood , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fluorenes/blood , Healthy Volunteers , Humans , Long QT Syndrome/chemically induced , Male , Middle Aged , Young Adult
14.
Article in English | MEDLINE | ID: mdl-28971607

ABSTRACT

The drug-drug interaction (DDI) potential between the fixed-dose combinations of ledipasvir/sofosbuvir 90/400 mg for hepatitis C virus and emtricitabine/rilpivirine/tenofovir alafenamide (TAF) 200/25/25 mg for HIV was evaluated in a randomized, open-label, single-center, multiple-dose, 3-way, 6-sequence, crossover Phase 1 study in 42 healthy subjects. Emtricitabine/rilpivirine/TAF had no relevant effect on the pharmacokinetic parameters of maximum concentration [Cmax ] and area under the concentration versus time curve over the dosing interval [AUCtau ] for ledipasvir, sofosbuvir, and the metabolites GS-566500 and GS-331007. Ledipasvir/sofosbuvir had no effect on the Cmax and AUCtau for rilpivirine and emtricitabine. The Cmax and AUCtau of tenofovir, the major metabolite of TAF, were increased by 62% and 75%, respectively. However, the resulting absolute tenofovir exposures were markedly lower than the historical tenofovir exposures following tenofovir disoproxil fumarate (TDF) and, as such, were not considered to be clinically relevant. In contrast, additional adverse effect monitoring is recommended upon coadministration of ledipasvir and TDF due to elevated tenofovir exposures resulting from the DDI. This difference is explained by the fact that TAF 25 mg results in markedly lower (~90%) plasma tenofovir exposure compared to TDF 300 mg. Ledipasvir/sofosbuvir and emtricitabine/rilpivirine/TAF were generally well tolerated when administered alone or in combination. HIV/hepatitis C virus-coinfected patients can coadminister ledipasvir/sofosbuvir and emtricitabine/rilpivirine/TAF without dosage adjustments.


Subject(s)
Adenine/analogs & derivatives , Benzimidazoles/pharmacokinetics , Emtricitabine/pharmacokinetics , Fluorenes/pharmacokinetics , Rilpivirine/pharmacokinetics , Uridine Monophosphate/analogs & derivatives , Adenine/administration & dosage , Adenine/pharmacokinetics , Alanine , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Benzimidazoles/administration & dosage , Cross-Over Studies , Drug Combinations , Drug Interactions , Emtricitabine/administration & dosage , Female , Fluorenes/administration & dosage , Healthy Volunteers , Humans , Male , Rilpivirine/administration & dosage , Sofosbuvir , Tenofovir/analogs & derivatives , Uridine Monophosphate/administration & dosage , Uridine Monophosphate/pharmacokinetics
15.
Article in English | MEDLINE | ID: mdl-28193657

ABSTRACT

Preclinical characterization of velpatasvir (VEL; GS-5816), an inhibitor of the hepatitis C virus (HCV) NS5A protein, demonstrated that it has favorable in vitro and in vivo properties, including potent antiviral activity against hepatitis C virus genotype 1 to 6 replicons, good metabolic stability, low systemic clearance, and adequate bioavailability and physicochemical properties, to warrant clinical evaluation. The phase 1 (first-in-human) study evaluated the safety, tolerability, and pharmacokinetics of VEL in healthy human subjects following administration of single and multiple (n = 7) once-daily ascending doses and of VEL in the presence and absence of food. Following administration of single and multiple doses, VEL was safe and well tolerated when administered at up to 450 mg and when administered with food. The pharmacokinetic behavior of VEL observed in humans was generally in agreement with that seen during preclinical characterization. Following administration of multiple doses, VEL trough concentrations were significantly greater than the protein-adjusted half-maximal (50%) effective concentration of VEL against HCV genotype 1 to 6 replicons at all evaluated doses greater than 5 mg. The pharmacokinetics of VEL were not significantly affected by administration with food. Collectively, the results of this study support the further clinical investigation of VEL administered once daily as part of a regimen with other pangenotypic direct-acting antivirals for the treatment of HCV infection.


Subject(s)
Antiviral Agents/therapeutic use , Carbamates/pharmacokinetics , Carbamates/therapeutic use , Hepacivirus/drug effects , Hepatitis C/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Viral Nonstructural Proteins/antagonists & inhibitors , Adult , Animals , Antiviral Agents/adverse effects , Carbamates/adverse effects , Dogs , Female , Healthy Volunteers , Hepatitis C/virology , Heterocyclic Compounds, 4 or More Rings/adverse effects , Humans , Macaca fascicularis , Male , Microsomes, Liver/metabolism , Middle Aged , Rats , Rats, Sprague-Dawley , Virus Replication/drug effects , Young Adult
16.
Clin Drug Investig ; 37(2): 195-205, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27785737

ABSTRACT

BACKGROUND AND OBJECTIVES: Entospletinib is a selective, reversible, adenosine triphosphate-competitive small-molecule spleen tyrosine kinase (SYK) inhibitor that blocks B cell receptor-mediated signaling and proliferation in B lymphocytes. This study evaluated the safety, pharmacokinetics, and pharmacodynamics of entospletinib in a double-blind, single/multiple ascending dose study in healthy volunteers. METHODS: In sequential cohorts, 120 subjects received entospletinib (25-1200 mg; fasted) as single or twice-daily oral doses for 7 days. Along with pharmacokinetics, the study assessed functional inhibition of ex vivo anti-immunoglobulin E-stimulated CD63 expression on basophils and pervanadate-evoked phosphorylated SYK (pSYK) Y525. Safety and tolerability were assessed throughout the study. RESULTS: Entospletinib was generally well-tolerated over a 48-fold dose range. Adverse events (AEs) were generally mild to moderate, with no AE-driven study drug discontinuations noted. Entospletinib displayed a median plasma half-life of 9-15 h; entospletinib exposures reached a plateau at ≥600 mg twice daily (likely due to solubility-limited absorption) and provided >90% CD63 inhibition at peak concentrations and >60% inhibition at trough concentrations (corresponding pSYK inhibition of >70 and >50%). CONCLUSION: The overall safety, pharmacokinetics, and pharmacodynamics profiles of entospletinib support further clinical evaluation.


Subject(s)
Syk Kinase/antagonists & inhibitors , Adolescent , Adult , B-Lymphocytes/drug effects , Basophils/drug effects , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Food-Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Tetraspanin 30/biosynthesis , Tetraspanin 30/drug effects , Vanadates/pharmacology , Young Adult
17.
Antimicrob Agents Chemother ; 60(9): 5135-40, 2016 09.
Article in English | MEDLINE | ID: mdl-27216057

ABSTRACT

Tenofovir alafenamide (TAF) is an oral prodrug of tenofovir (TFV) that has greater stability in plasma than TFV disoproxil fumarate (TDF) and circulates as intact TAF, resulting in the direct and higher lymphatic loading of and exposure to TFV diphosphate, the active moiety. Unlike TFV, TAF is minimally eliminated in urine. The pharmacokinetics (PK) of TAF and TFV in HIV-uninfected subjects with severe renal impairment and matched healthy controls were evaluated. Subjects with severe renal impairment (RI; estimated glomerular filtration rate [eGFR], 15 to 29 ml/min) and controls (eGFR, ≥90 ml/min) matched for age, gender, and body mass index received a single dose of TAF at 25 mg. Blood and urine samples for TAF and TFV PK determinations were collected over 7 days postdosing, and subjects were followed up at 14 days. A total of 14 renally impaired subjects and 13 control subjects enrolled and completed the study. The TAF maximum observed concentration in plasma (Cmax) and the area under the concentration-versus-time curve (AUC) extrapolated to infinite time (AUCinf) were 79% and 92% higher, respectively, in subjects with severe RI than the controls, primarily due to higher absorption. The TFV Cmax and AUCinf were 2.8-fold and 5.7-fold higher, respectively, in subjects with severe RI than the controls. In subjects with severe RI, TAF at 25 mg provided a TFV AUC 10 to 40% lower than that from historical TDF-based TFV exposures in subjects with normal renal function. There were no discontinuations due to adverse events. In subjects with severe RI receiving TAF at 25 mg, TAF exposures were higher than those for the controls; these differences are unlikely to be clinically meaningful. TFV exposures were higher than those for the controls but lower than the exposures in nonrenally impaired subjects on TDF-based regimens.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/blood , Renal Insufficiency, Chronic/blood , Adenine/blood , Adenine/pharmacokinetics , Aged , Alanine , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Case-Control Studies , Female , Glomerular Filtration Rate , HIV Infections , Humans , Male , Middle Aged , Renal Insufficiency, Chronic/physiopathology , Tenofovir/analogs & derivatives
18.
Clin Pharmacokinet ; 55(11): 1337-1351, 2016 11.
Article in English | MEDLINE | ID: mdl-27193156

ABSTRACT

Ledipasvir/sofosbuvir (Harvoni®), a fixed-dose combination tablet of an NS5A inhibitor ledipasvir and an NS5B polymerase inhibitor sofosbuvir, is approved in the US, European Union, Canada, and other regions for the treatment of chronic hepatitis C virus infection in adults. Following absorption, ledipasvir reaches maximum plasma concentrations (T max) 4-4.5 h post-dose and is eliminated with a terminal half-life (t 1/2) of 47 h. Sofosbuvir undergoes intracellular activation to an active triphosphate GS-461203 (not detected in plasma) and ultimately to GS-331007, a predominant circulating metabolite, which is the primary analyte of interest in clinical pharmacology studies. Sofosbuvir is rapidly absorbed and eliminated from plasma (T max: 0.8-1 h; t 1/2: 0.5 h). The peak plasma concentrations for GS-331007 are achieved between 3.5 and 4 h post-dose; the elimination t 1/2 for GS-331007 is 27 h. Ledipasvir/sofosbuvir exhibits a favorable clinical pharmacology profile; it can be administered once daily without regard to food and does not require dose modification in hepatitis C virus-infected patients with any degree of hepatic impairment or mild to moderate renal impairment. The pharmacokinetic profiles of ledipasvir, sofosbuvir, and GS-331007 (predominant circulating metabolite of sofosbuvir) are not significantly affected by demographic variables; pharmacokinetic/pharmacodynamic analyses reveal no exposure-response relationships for efficacy or safety. The review summarizes the clinical pharmacokinetics, pharmacodynamics, and pharmacokinetic/pharmacodynamic analyses for ledipasvir/sofosbuvir.


Subject(s)
Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Benzimidazoles/pharmacokinetics , Benzimidazoles/therapeutic use , Fluorenes/pharmacokinetics , Fluorenes/therapeutic use , Hepatitis C, Chronic/drug therapy , Uridine Monophosphate/analogs & derivatives , Animals , Antiviral Agents/pharmacology , Area Under Curve , Benzimidazoles/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Fluorenes/pharmacology , Half-Life , Humans , Hydrogen-Ion Concentration , Liver Failure/metabolism , Metabolic Clearance Rate , Renal Insufficiency/metabolism , Sofosbuvir , Sustained Virologic Response , Uridine Monophosphate/pharmacokinetics , Uridine Monophosphate/pharmacology , Uridine Monophosphate/therapeutic use
19.
J Acquir Immune Defic Syndr ; 72(3): 281-8, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26885802

ABSTRACT

BACKGROUND: Elvitegravir (EVG), a HIV integrase inhibitor, is metabolized primarily by CYP3A, and secondarily by UGT1A1/3; Efavirenz (EFV), a HIV non-nucleoside reverse transcriptase inhibitor, is metabolized by Cytochrome P450 (CYP) 2B6 and induces CYP3A and uridine diphosphate glucuronosyltransferase (UGT) with residual effects post discontinuation because of long T1/2 (40-55 hours). This study evaluated the pharmacokinetics after switching from efavirenz/emtricitabine/tenofovir disoproxil fumarate (EFV/FTC/TDF) to elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate (EVG/COBI/FTC/TDF). METHODS: Healthy subjects (n = 32 including n = 8 CYP2B6 poor metabolizers) received EVG/COBI/FTC/TDF (150/150/200/300 mg) on days 1-7, and after a washout, received EFV/FTC/TDF (600/200/300 mg) on days 15-28 and switched to EVG/COBI/FTC/TDF (150/150/200/300 mg) for 5 weeks (days 29-62). Pharmacokinetic assessments occurred on days 7, 28, 35, and 42; trough samples (Ctrough) were collected periodically until day 63. Safety was assessed throughout the study. RESULTS: Twenty-nine subjects completed with 3 adverse events leading to discontinuation; no grade ≥3 adverse events were reported. Post-EFV/FTC/TDF, mean EVG area under concentration (AUCtau) was 37% and 29% lower and mean Ctrough ∼3- and ∼5-fold above IC95, respectively, on days 35 and 42, and 7-8-fold above IC95 by 5 weeks. COBI AUCtau returned to normal by day 42. EVG glucuronide, GS-9200, AUCtau was higher (46% and 32% on days 35 and 42, respectively) postswitch. CYP2B6 poor metabolizers displayed higher EFV AUCtau and Cmax (125% and 91%, respectively) versus non-poor metabolizers, and lower EVG and COBI exposures. EFV Ctrough was >IC90 (10 ng/mL) in all subjects postswitch. FTC and tenofovir (TFV) exposures were unaffected. CONCLUSIONS: After EFV/FTC/TDF to EVG/COBI/FTC/TDF switch, EVG and/or EFV exposures were in an active range. These findings support further evaluation of switching regimens in HIV-1 patients.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Efavirenz, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/pharmacokinetics , Elvitegravir, Cobicistat, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/pharmacokinetics , Adolescent , Adult , Anti-HIV Agents/adverse effects , Drug Combinations , Drug-Related Side Effects and Adverse Reactions , Efavirenz, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/adverse effects , Elvitegravir, Cobicistat, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/adverse effects , Female , Healthy Volunteers , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
20.
Clin Pharmacokinet ; 55(5): 605-13, 2016 May.
Article in English | MEDLINE | ID: mdl-26519191

ABSTRACT

BACKGROUND AND OBJECTIVES: Velpatasvir (VEL; GS-5816) is a potent, pangenotypic hepatitis C virus (HCV), non-structural protein 5A inhibitor in clinical development for the treatment of chronic HCV infection. In vitro studies indicate that VEL may inhibit several drug transporters and be a substrate for enzyme/drug transport systems in vivo. The purpose of this study was to evaluate the potential of VEL as a perpetrator or victim of metabolic- and transporter-based drug-drug interactions using complementary probe drugs. METHODS: This Phase 1 study was a randomized, cross-over, open-label, single- and multiple-dose, five-cohort study. Serial blood samples were collected following oral administration of reference and test treatments. The primary pharmacokinetic parameters of each analyte were compared when administered alone or in combination. The 90% confidence intervals (CI) for the ratio of the geometric least-squares means of the test and reference treatments was calculated for each analyte and parameter of interest. RESULTS: This study demonstrated that VEL is a weak (P-gp, OATP) to moderate (breast cancer resistance protein) transport inhibitor. As a victim of interactions, VEL is moderately affected by potent inhibitors and to a greater extent, potent inducers of enzyme/drug transporter systems. CONCLUSIONS: The impact of specific transporters and overall contribution of drug transport vs. metabolizing enzymes on the disposition of VEL was characterized through the use of complementary probes, despite the lack of phenotypic specificity, and informs a broad range of drug-drug interaction recommendations.


Subject(s)
Antiviral Agents/pharmacokinetics , Carbamates/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Membrane Transport Proteins/metabolism , Adolescent , Adult , Antiviral Agents/adverse effects , Antiviral Agents/pharmacology , Carbamates/adverse effects , Carbamates/pharmacology , Cross-Over Studies , Drug Interactions , Female , Hepacivirus , Heterocyclic Compounds, 4 or More Rings/adverse effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...