Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Pharmacokinet ; 56(11): 1331-1342, 2017 11.
Article in English | MEDLINE | ID: mdl-28238203

ABSTRACT

BACKGROUND: JNJ-53718678 is a potent small-molecule inhibitor of the F-glycoprotein-mediated complex membrane fusion process of the respiratory syncytial virus. Here, we report the pharmacokinetics, the population pharmacokinetic modeling, and the safety and tolerability of JNJ-53718678 from the first-in-human, double-blind, randomized, placebo-controlled phase I study. METHODS: Healthy subjects were randomized (6:3) into five single-dose groups (25-1000 mg) or three multiple-dose groups [250 mg every 24 h (q24h), 500 mg q24h, 250 mg every 12 h; fed conditions for 8 days] to receive JNJ-53718678 or placebo. Blood and urine samples were collected at several timepoints up to 72 h after intake of JNJ-53718678 and analyzed using validated liquid chromatography-mass spectrometry methods. A population pharmacokinetic model was developed and validated. RESULTS: Peak plasma concentrations of JNJ-53718678 increased with increasing single (159 ± 54.9 to 6702 ± 1733 ng/mL) and multiple (on day 8, 1528 ± 256 to 2655 ± 591 ng/mL) doses. Steady-state conditions were reached on day 2 of the 8-day dosing regimen. Less than 4% of JNJ-53718678 was excreted in urine across all dose groups. Mean exposure of JNJ-53718678 was 7% lower in the fed state compared with the fasted state at the same dose. A two-compartment model with first-order absorption with parallel linear and non-linear elimination best described the pharmacokinetics of JNJ-53718678. No covariate effects were observed. CONCLUSIONS: A population pharmacokinetic model that describes the concentration data well with good precision of all parameter estimates was developed and validated. JNJ-53718678 was well tolerated at all single and multiple doses studied.


Subject(s)
Antiviral Agents/pharmacokinetics , Imidazolidines/pharmacokinetics , Indoles/pharmacokinetics , Models, Biological , Adolescent , Adult , Antiviral Agents/adverse effects , Antiviral Agents/blood , Antiviral Agents/urine , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Imidazolidines/adverse effects , Imidazolidines/blood , Imidazolidines/urine , Indoles/adverse effects , Indoles/blood , Indoles/urine , Male , Middle Aged , Respiratory Syncytial Viruses/drug effects , Young Adult
2.
Clin Pharmacokinet ; 56(5): 525-536, 2017 05.
Article in English | MEDLINE | ID: mdl-27665573

ABSTRACT

BACKGROUND: Etravirine is a non-nucleoside reverse transcriptase inhibitor indicated in combination with other antiretrovirals for treatment-experienced HIV patients ≥6 years of age. Etravirine is primarily metabolized by cytochrome P450 (CYP) 2C9, CYP2C19, and CYP3A. This analysis determined the impact of concomitant antiretrovirals and CYP2C9/CYP2C19 phenotype on the pharmacokinetics of etravirine. METHODS: We used 4728 plasma concentrations from 817 adult subjects collected from four clinical studies to develop the population pharmacokinetic model. The presence of atazanavir/ritonavir, lopinavir/ritonavir, darunavir/ritonavir, tenofovir disoproxil fumarate, or enfuvirtide together with the CYP2C9 and CYP2C19 phenotype and other demographics were evaluated. RESULTS: A one-compartment model with first-order input and a lag-time best described the data. Estimates of apparent total clearance (CL/F), apparent central volume of distribution (V c/F), first-order absorption rate constant (k a), and absorption lag-time were 41.7 L/h, 972 L, 1.16 h, and 1.32 h, respectively. Estimates of between-subject variability on CL/F, V c/F, and relative bioavailability (F) were 39.4 %CV (percentage coefficient of variation), 35.9 %CV and 35.5 %CV, respectively. Between-occasion variability on F was estimated to be 30.0 %CV. CL/F increased non-linearly with body weight and creatinine clearance (CLCR), and also varied based on CYP2C9/CYP2C19 phenotype. CONCLUSIONS: In this analysis, body weight, CLCR, and CYP2C9/CYP2C19 phenotype were found to describe some of the variability in CL/F. It was not possible to show an impact of concomitant antiretrovirals on the pharmacokinetics of etravirine for adults predominantly taking coadministered boosted protease inhibitors as a background antiretroviral regimen.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/pharmacokinetics , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C9/genetics , Pyridazines/administration & dosage , Pyridazines/pharmacokinetics , Adult , Clinical Trials as Topic/methods , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C9/metabolism , Drug Interactions/physiology , Drug Therapy, Combination , Female , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/genetics , HIV-1/drug effects , HIV-1/metabolism , Humans , Male , Nitriles , Polymorphism, Genetic/genetics , Pyrimidines , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics
3.
J Clin Pharmacol ; 56(11): 1395-1405, 2016 11.
Article in English | MEDLINE | ID: mdl-27060341

ABSTRACT

PIANO (NCT00665847) investigated etravirine pharmacokinetics, efficacy, and safety in children and adolescents. Treatment-experienced, HIV-1-infected patients (≥6 to <18 years) received etravirine 5.2 mg/kg twice daily (maximum 200 mg twice daily) plus background antiretrovirals. A population pharmacokinetic model was developed, and etravirine C0h and AUC0-12h were estimated. Relationships among intrinsic/extrinsic factors and etravirine pharmacokinetics and pharmacokinetics with pharmacodynamics were assessed. The best model describing etravirine pharmacokinetics consisted of a single compartment with sequential zero- and first-order absorption following a lag time. Interindividual variability terms were included on clearance (CL/F) and the first-order input rate constant (KA). The final model estimates (coefficient of variation, %) for CL/F and KA were 46.3 (11) L/h and 1.07 (34) h-1 , respectively. Overall, median (range) estimated etravirine C0h and AUC0-12h were 287 (2-2276) ng/mL and 4560 (62-28,865) ng · h/mL, respectively. Exposure was slightly lower in adolescents vs children. Sex and adherence did not affect etravirine pharmacokinetics. Factors significantly affecting etravirine exposure were body weight (higher with lower weight), race (lower in Asians than in white or black patients), and the use of certain HIV protease inhibitors. Virologic response (<50 copies/mL at week 48) was lower in the lowest etravirine AUC0-12h quartile vs the upper 3 quartiles (41% vs 67% to 76%). Rash occurred more frequently in the highest quartile than in the lower 3 quartiles (52% versus 8% to 20%). Etravirine 5.2 mg/kg twice daily in treatment-experienced, HIV-1-infected children and adolescents provides comparable exposure to that in adults receiving etravirine 200 mg twice daily and is the recommended dose for children and adolescents.


Subject(s)
HIV Infections/blood , HIV-1/drug effects , Pyridazines/administration & dosage , Pyridazines/blood , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/blood , Adolescent , Adult , Child , Drug Administration Schedule , Female , HIV Infections/drug therapy , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/pharmacokinetics , Humans , Male , Nitriles , Pyridazines/pharmacokinetics , Pyrimidines , Reverse Transcriptase Inhibitors/pharmacokinetics , Treatment Outcome
4.
Plant J ; 84(1): 1-19, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26296678

ABSTRACT

Next-generation sequencing technologies allow an almost exhaustive survey of the transcriptome, even in species with no available genome sequence. To produce a Unigene set representing most of the expressed genes of pea, 20 cDNA libraries produced from various plant tissues harvested at various developmental stages from plants grown under contrasting nitrogen conditions were sequenced. Around one billion reads and 100 Gb of sequence were de novo assembled. Following several steps of redundancy reduction, 46 099 contigs with N50 length of 1667 nt were identified. These constitute the 'Caméor' Unigene set. The high depth of sequencing allowed identification of rare transcripts and detected expression for approximately 80% of contigs in each library. The Unigene set is now available online (http://bios.dijon.inra.fr/FATAL/cgi/pscam.cgi), allowing (i) searches for pea orthologs of candidate genes based on gene sequences from other species, or based on annotation, (ii) determination of transcript expression patterns using various metrics, (iii) identification of uncharacterized genes with interesting patterns of expression, and (iv) comparison of gene ontology pathways between tissues. This resource has allowed identification of the pea orthologs of major nodulation genes characterized in recent years in model species, as a major step towards deciphering unresolved pea nodulation phenotypes. In addition to a remarkable conservation of the early transcriptome nodulation apparatus between pea and Medicago truncatula, some specific features were highlighted. The resource provides a reference for the pea exome, and will facilitate transcriptome and proteome approaches as well as SNP discovery in pea.


Subject(s)
Gene Expression Regulation, Plant , Pisum sativum/growth & development , Pisum sativum/genetics , Plant Root Nodulation/genetics , Plant Roots/growth & development , Plant Roots/genetics , RNA, Plant/genetics , High-Throughput Nucleotide Sequencing
5.
J Int AIDS Soc ; 17(4 Suppl 3): 19772, 2014.
Article in English | MEDLINE | ID: mdl-25397516

ABSTRACT

INTRODUCTION: COBI, a PK enhancer with no ARV activity is a more selective cytochrome P450 (CYP)3A inhibitor than ritonavir (RTV), does not induce CYP isozymes, and thus has less potential for drug-drug interactions. COBI boosts DRV PK as effectively as RTV in healthy volunteers. MATERIALS AND METHODS: This 48-week, phase IIIb, open-label, single-arm, US multicentre study (NCT01440569) included HIV-infected treatment-nave and experienced adults with no DRV RAMs, viral load (VL) ≥1000 c/mL, eGFR ≥80 mL/min and genotypic sensitivity to investigator-selected N[t]RTIs. Patients received DRV/COBI 800/150 mg qd (as single agents) plus two fully active N[t]RTIs. The primary endpoint was any treatment-emergent grade 3 or 4 AEs through Week 24. We report 48-week safety, efficacy and PK/PD results in treatment-nave patients. RESULTS: Of 313 ITT patients, 295 were treatment-nave (94%). In the treatment-nave cohort, 90% were male, 60% white and 294 (99.7%) received a TDF-containing regimen. Median baseline (BL) VL was 4.8 log10 c/mL and CD4(+) 370 cells/mm(3). Treatment-emergent grade 3 or 4 AEs regardless of causality were reported in 21 (7%) patients. AEs regardless of causality (any grade; ≥10% of patients) were: diarrhoea (27%), nausea (23%), URTI (15%) and headache (12%). Sixteen (5%) patients had AEs leading to study drug discontinuation, most frequently rash (three patients), hypersensitivity and nausea (two patients each). Consistent with the known inhibition of tubular creatinine secretion by COBI, there was a mean increase from BL in serum creatinine by week 2 (0.09 mg/dL), remaining stable through week 48 (mean 0.10 mg/dL increase from BL). At week 48, 83% of patients achieved VL<50 c/mL; FDA Snapshot); median increase in CD4(+) was 169 cells/mm(3). Eight patients met the criteria for resistance testing. M184V was detected in one pt receiving FTC. New primary RAMs were not detected in the other seven patients. The mean population PK-derived DRV AUC24h was 100,620 ng.h/mL and C0h 2,105 ng/mL (n=281). There were no clinically relevant relationships between DRV exposure and virologic response, AEs or laboratory parameters. CONCLUSIONS: The DRV PK of DRV/COBI was consistent with historical data for DRV/RTV. DRV/COBI 800/150 mg qd plus two N(t)RTIs had an 83% response and was well tolerated through Week 48. These results are similar to published data for DRV/RTV 800/100 mg qd, and support the use of DRV/COBI 800/150 mg qd in treatment-nave patients.

6.
J Antimicrob Chemother ; 69(10): 2591-605, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24951533

ABSTRACT

The ability to dose antiretroviral agents once daily simplifies the often complex therapeutic regimens required for the successful treatment of HIV infection. Thus, once-daily dosing can lead to improved patient adherence to medication and, consequently, sustained virological suppression and reduction in the risk of emergence of drug resistance. Several trials have evaluated once-daily darunavir/ritonavir in combination with other antiretrovirals (ARTEMIS and ODIN trials) or as monotherapy (MONET, MONOI and PROTEA trials) in HIV-1-infected adults. Data from ARTEMIS and ODIN demonstrate non-inferiority of once-daily darunavir/ritonavir against a comparator and, together with pharmacokinetic data, have established the suitability of once-daily darunavir/ritonavir for treatment-naive and treatment-experienced patients with no darunavir resistance-associated mutations. The findings of ARTEMIS and ODIN have led to recent updates to treatment guidelines, whereby once-daily darunavir/ritonavir, given with other antiretrovirals, is now a preferred treatment option for antiretroviral-naive adult patients and a simplified treatment option for antiretroviral-experienced adults who have no darunavir resistance-associated mutations. Once-daily dosing with darunavir/ritonavir is an option for treatment-naive and for treatment-experienced paediatric patients with no darunavir resistance-associated mutations based on the findings of the DIONE trial and ARIEL substudy. This article reviews the pharmacokinetics, efficacy, safety and tolerability of once-daily boosted darunavir. The feasibility of darunavir/ritonavir monotherapy as a treatment approach for some patients is also discussed. Finally, data on a fixed-dose combination of 800/150 mg of darunavir/cobicistat once daily are presented, showing comparable darunavir bioavailability to that obtained with 800/100 mg of darunavir/ritonavir once daily.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Age Factors , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Darunavir , Drug Interactions , Drug Therapy, Combination , Female , HIV Protease Inhibitors/adverse effects , HIV-1 , Humans , Male , Pregnancy , Sex Factors , Sulfonamides/adverse effects
7.
AIDS Res Ther ; 11: 39, 2014.
Article in English | MEDLINE | ID: mdl-25926858

ABSTRACT

BACKGROUND: Cobicistat is an alternative pharmacoenhancer to ritonavir. In healthy volunteers, darunavir exposure was comparable when darunavir 800 mg once daily was co-administered with cobicistat 150 mg once daily (as single agents or a fixed-dose combination) vs. with ritonavir 100 mg once daily. METHODS: This 48-week, Phase IIIb, single-arm, US multicenter study (NCT01440569) evaluated safety, efficacy and pharmacokinetics of darunavir/cobicistat 800/150 mg once daily (as single agents) plus two investigator-selected nucleoside/tide reverse transcriptase inhibitors (N[t]RTIs) in HIV-1-infected adults. Patients had no darunavir resistance-associated mutations (RAMs), plasma viral load (VL) ≥1000 HIV-1 RNA copies/ml, eGFR ≥80 ml/min and genotypic sensitivity to the two N[t]RTIs. The primary endpoint was any treatment-emergent grade 3 or 4 adverse events (AEs) through Week 24. RESULTS: The majority of the 313 intent-to-treat patients were treatment-naïve (295/313; 94%), male (89%), White (60%) and received a tenofovir-based regimen (99%). Median baseline VL and CD4(+) count overall were 4.8 log10 HIV-1 RNA copies/ml and 361 cells/mm(3), respectively. Overall, 86% of patients (268/313) completed the study. The majority of discontinuations were for AEs (15/313; 5%). The incidence of treatment-emergent grade 3 or 4 AEs regardless of causality was 6% through Week 24 and 8% through Week 48. Most common AEs through Week 48 were diarrhea (27%) and nausea (23%), which were grade 1 or 2 in severity. Week 48 virologic response rates (% with VL <50 HIV-1 RNA copies/ml; Snapshot analysis) were 81% overall and 83% in treatment-naïve patients; median increases in CD4(+) count at 48 weeks were 167 and 169 cells/mm(3), respectively. Of 15/313 patients who met the criteria for resistance analysis, one developed a darunavir RAM as a mixture with wild-type (I84I/V), without phenotypic resistance to darunavir. The mean population pharmacokinetic-derived darunavir areas under the plasma concentration-time curve were 102,000 overall and 100,620 ng•h/ml in treatment-naïve patients. No clinically relevant relationships were seen between darunavir exposure and virologic response, AEs or laboratory parameters. CONCLUSION: Darunavir/cobicistat 800/150 mg once daily was generally well tolerated through Week 48, with no new safety concerns. Pharmacokinetics, virologic and immunologic responses for darunavir/cobicistat were similar to previous data for darunavir/ritonavir 800/100 mg once daily.

8.
Basic Clin Pharmacol Toxicol ; 106(3): 256-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20102365

ABSTRACT

Animal-to-man extrapolation and therapeutic dose prediction are illustrated with two molecules designed to treat epilepsy. Synaptic vesicle protein 2A (SV2A) is the primary molecular target for their anticonvulsive effect, but additional mechanisms may also contribute. Brivaracetam (BRV), currently in phase 3 of clinical development, was used as the benchmark compound. A pharmacokinetic/pharmacodynamic model was built in NONMEM, relating the brain tissue concentrations of BRV in mice and the proportion of animals protected against convulsions in the pharmacological model of audiogenic seizures. Brain concentrations were linked with ex vivo binding to predict brain SV2A occupancy. A physiologically based pharmacokinetic model was developed for predicting BRV concentrations in human plasma and brain tissue. Predicted plasma profiles were in good agreement with observations. Predicted human brain concentrations of BRV and the mouse ex vivo binding pharmacokinetic/pharmacodynamic model were used to extrapolate brain SV2A occupancy at the human therapeutic dose. Secondly, for another compound also exhibiting selective affinity for the same target, similar pharmacokinetic/pharmacodynamic models were built from audiogenic seizure mouse data. Various dosing regimens of the new compound were simulated in order to reach the same brain SV2A occupancy as for the reference compound. These estimations support early development. Assumptions and limitations of the approach are discussed.


Subject(s)
Anticonvulsants , Disease Models, Animal , Epilepsy/drug therapy , Pharmacology , Physiology , Pyrrolidinones , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Epilepsy/blood , Epilepsy/metabolism , Humans , Logistic Models , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Predictive Value of Tests , Protein Binding , Pyrrolidinones/administration & dosage , Pyrrolidinones/pharmacokinetics , Pyrrolidinones/therapeutic use , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...