Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Pharmacol Drug Dev ; 11(12): 1419-1429, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36062869

RESUMO

The capsid assembly modulator JNJ-56136379 (bersacapavir) disrupts hepatitis B virus replication. It is metabolized via cytochrome P450 (CYP) 3A, but little is known about the drug-drug interactions of JNJ-56136379 when combined with drugs that inhibit or are metabolized by CYP3A. In a phase 1, open-label trial (NCT03945539), healthy adults received 1 dose of JNJ-56136379 with and without 21 days of prior exposure to itraconazole 200 mg (CYP3A inhibitor). In a second phase 1, open-label trial (NCT03111511), healthy women received 1 dose of drospirenone/ethinyl estradiol and midazolam before and after 15 days of JNJ-56136379. Itraconazole increased the area under the plasma concentration-time curve (AUC) of JNJ-56136379 by 38%. JNJ-56136379 reduced the maximum observed concentration and AUC of midazolam (CYP3A substrate) by 42%-54%, increased AUC of ethinyl estradiol by 1.6-fold, but had no effect on drospirenone pharmacokinetics. Overall, these results demonstrated that a strong CYP3A inhibitor (itraconazole) modestly increased JNJ-56136379 exposure. Furthermore, JNJ-56136379 was a weak inducer of CYP3A (midazolam) and increased ethinyl estradiol exposure; coadministration of high-dose estrogen-based contraceptives and JNJ-56136379 is not recommended.


Assuntos
Inibidores do Citocromo P-450 CYP3A , Vírus da Hepatite B , Adulto , Feminino , Humanos , Antivirais/efeitos adversos , Capsídeo/metabolismo , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A/farmacologia , Interações Medicamentosas , Etinilestradiol/farmacologia , Vírus da Hepatite B/metabolismo , Itraconazol/farmacocinética , Midazolam/farmacocinética
2.
J Clin Pharmacol ; 61(10): 1344-1355, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33991350

RESUMO

Based on the in vitro profile of bedaquiline against mycobacterial species, it is being investigated for clinical efficacy against pulmonary nontuberculous mycobacteria (PNTM). Being a cytochrome P450 3A substrate, pharmacokinetic interactions of bedaquiline are anticipated with clarithromycin (a cytochrome P450 3A inhibitor), which is routinely used in pulmonary nontuberculous mycobacteria treatment. This phase 1, randomized, crossover study assessed the impact of steady-state clarithromycin (500 mg every 12 hours for 14 days) on the pharmacokinetics of bedaquiline and its metabolite (M2) after single-dose bedaquiline (100 mg; n = 16). Using these data, population pharmacokinetic modeling and simulation analyses were performed to determine the effect of clarithromycin on steady-state bedaquiline exposure. Although no effect was observed on maximum plasma concentration of bedaquiline and time to achieve maximum plasma concentration, its mean plasma exposure increased by 14% after 10 days of clarithromycin coadministration, with slower formation of M2. Simulations showed that bedaquiline plasma trough concentration at steady state was higher (up to 41% until week 48) with clarithromycin coadministration as compared to its monotherapy (400 mg once daily for 2 weeks, followed by 200 mg 3 times a week for 46 weeks; reference regimen). The overall exposure of a simulated bedaquiline regimen (400 mg once dialy for 2 weeks, followed by 200 mg twice a week for 46 weeks) with clarithromycin was comparable (<15% difference) to the monotherapy. Overall, combination of bedaquiline (400 mg once daily for 2 weeks, followed by 200 mg twice a week for 46 weeks) with clarithromycin seems a suitable regimen to be explored for efficacy and safety against pulmonary nontuberculous mycobacteria.


Assuntos
Antibacterianos/farmacologia , Antituberculosos/farmacocinética , Claritromicina/farmacologia , Diarilquinolinas/farmacocinética , Adulto , Antibacterianos/farmacocinética , Antibacterianos/uso terapêutico , Antituberculosos/uso terapêutico , Claritromicina/farmacocinética , Claritromicina/uso terapêutico , Estudos Cross-Over , Inibidores do Citocromo P-450 CYP3A/farmacologia , Inibidores do Citocromo P-450 CYP3A/uso terapêutico , Diarilquinolinas/uso terapêutico , Quimioterapia Combinada , Feminino , Humanos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Micobactérias não Tuberculosas/efeitos dos fármacos
3.
Adv Ther ; 37(4): 1703, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32133584

RESUMO

In the original article Ninth and Tenth authors were incorrectly omitted from the author group. The correct author group is Joris Vandenbossche, Wolfgang Jessner, Maarten van den Boer, Jeike Biewenga, Jan Martin Berke, Willem Talloen, Loeckie De Zwart, Jan Snoeys, Koen Vandyck, John Fry, Jeysen Yogaratnam.

4.
Adv Ther ; 36(9): 2450-2462, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31267367

RESUMO

INTRODUCTION: Hepatitis B viral capsid assembly is an attractive target for new antiviral treatments. JNJ-56136379 (JNJ-6379) is a potent capsid assembly modulator in vitro with a dual mode of action. In Part 1 of this first-in-human study in healthy adults, the pharmacokinetics (PK), safety and tolerability of JNJ-6379 were evaluated following single ascending and multiple oral doses. METHODS: This was a double-blind, randomized, placebo-controlled study in 30 healthy adults. Eighteen subjects were randomized to receive single doses of JNJ-6379 (25 to 600 mg) or placebo. Twelve subjects were randomized to receive 150 mg JNJ-6379 or placebo twice daily for 2 days, followed by 100 mg JNJ-6379 or placebo daily for 10 days. RESULTS: The maximum observed plasma concentration and the area under the curve increased dose proportionally from 25 to 300 mg JNJ-6379. Following multiple dosing, steady-state conditions were achieved on day 8. Steady-state clearance was similar following single and multiple dosing, suggesting time-linear PK. All adverse events (AEs) reported were mild to moderate in severity. There were no serious AEs or dose-limiting toxicities and no apparent relationship to dose for any AE. CONCLUSION: JNJ-6379 was well tolerated in this study. Based on the safety profile and plasma exposures of JNJ-6379 in healthy subjects, a dosing regimen was selected for Part 2 of this study in patients with chronic hepatitis B. This is anticipated to achieve trough plasma exposures of JNJ-6379 at steady state of more than three times the 90% effective concentration of viral replication determined in vitro. TRIAL REGISTRATION: Clinicaltrials.gov identifier, NCT02662712. FUNDING: Janssen Pharmaceutica.


Assuntos
Antivirais/administração & dosagem , Azepinas/farmacologia , Capsídeo/efeitos dos fármacos , Hepatite B Crônica/tratamento farmacológico , Piperidinas/farmacologia , Adulto , Área Sob a Curva , Azepinas/administração & dosagem , Azepinas/efeitos adversos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Vírus da Hepatite B/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Piperidinas/administração & dosagem , Piperidinas/efeitos adversos
5.
Biom J ; 61(5): 1104-1119, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30259557

RESUMO

The primary goal of "in vitro-in vivo correlation" (IVIVC) is the reliable prediction of the in vivo serum concentration-time course, based on the in vitro drug dissolution or release profiles. IVIVC methods are particularly appropriate for formulations that are released over an extended period of time or with a lag in absorption and may support approving a change in formulation of a drug without additional bioequivalence trials in human subjects. Most of the current IVIVC models are assessed using frequentist methods, such as linear regression, based on averaged data and entail complex and potentially unstable mathematical deconvolution. The proposed IVIVC approach includes (a) a nonlinear-mixed effects model for the in vitro release data; (b) a population pharmacokinetic (PK) compartment model for the in vivo immediate release (IR) data; and (c) a system of ordinal differential equations (ODEs), containing the submodels (a) and (b), which approximates and predicts the in vivo controlled release (CR) data. The innovation in this paper consists of splitting the parameter space between submodels (a) and (b) versus (c). Subsequently, the uncertainty on these parameters is accounted for using a Bayesian framework, that is estimates from the first two submodels serve as priors for the Bayesian hierarchical third submodel. As such, the Bayesian method explained ensures a natural integration and transfer of knowledge between various sources of information, balancing possible differences in sample size and parameter uncertainty of in vitro and in vivo studies. Consequently, it is a very flexible approach yielding results for a broad range of data situations. The application of the method is demonstrated for a transdermal patch (TD).


Assuntos
Biometria/métodos , Modelos Biológicos , Teorema de Bayes , Composição de Medicamentos , Permeabilidade , Soro/metabolismo , Pele/metabolismo
6.
J Psychopharmacol ; 32(12): 1341-1350, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30260294

RESUMO

BACKGROUND: Central nervous system-derived interleukin-1ß plays a role in mood disorders. P2X7 receptor activation by adenosine-triphosphate leads to the release of interleukin-1ß. AIMS: This first-in-human study evaluated safety, tolerability, pharmacokinetics and pharmacodynamics of a novel central nervous system-penetrant P2X7 receptor antagonist, JNJ-54175446, in healthy participants. METHODS: The study had three parts: an ascending-dose study in fasted participants (0.5-300 mg JNJ-54175446); an ascending-dose study in fed participants (50-600 mg); and a cerebrospinal fluid study (300 mg). Target plasma concentrations were based on estimated plasma effective concentration (EC)50 (105 ng/mL) and EC90 (900 ng/mL) values for central nervous system P2X7 receptor binding. RESULTS: Seventy-seven participants received a single oral dose of JNJ-54175446 ( n=59) or placebo ( n=18). Area under the curve of concentration time extrapolated to infinity (AUC∞) increased dose-proportionally; maximum concentration (Cmax) of plasma (Cmax,plasma) increased less than dose-proportionally following single doses of JNJ-54175446. Because food increases bioavailability of JNJ-54175446, higher doses were given with food to evaluate safety at higher exposures. The highest Cmax,plasma reached (600 mg, fed) was 1475±163 ng/mL. JNJ-54175446 Cmax in cerebrospinal fluid, a proxy for brain penetration, was seven times lower than in total plasma; unbound Cmax,plasma and Cmax,CSF were comparable (88.3±35.7 vs 114±39 ng/mL). JNJ-54175446 inhibited lipopolysaccharide/3'-O-(4-benzoylbenzoyl)-ATP-induced interleukin-1ß release from peripheral blood in a dose-dependent manner (inhibitory concentration (IC)50:82 ng/mL; 95% confidence interval: 48-94). Thirty-three of 59 (55.9%) participants reported at least one treatment-emergent adverse event; the most common adverse event being headache (11/59, 18.6%). CONCLUSION: Plasma exposure of JNJ-54175446 was dose-dependent. No serious adverse events occurred. Single-dose administration of JNJ-54175446>10 mg attenuated ex-vivo lipopolysaccharide-induced interleukin-1ß release in peripheral blood. Passive brain penetration of JNJ-54175446 was confirmed.


Assuntos
Encéfalo/metabolismo , Interleucina-1beta/metabolismo , Antagonistas do Receptor Purinérgico P2X/administração & dosagem , Piridinas/administração & dosagem , Triazóis/administração & dosagem , Administração Oral , Adolescente , Adulto , Idoso , Área Sob a Curva , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Interações Alimento-Droga , Humanos , Concentração Inibidora 50 , Lipopolissacarídeos/farmacologia , Masculino , Pessoa de Meia-Idade , Antagonistas do Receptor Purinérgico P2X/farmacocinética , Antagonistas do Receptor Purinérgico P2X/farmacologia , Piridinas/farmacocinética , Piridinas/farmacologia , Distribuição Tecidual , Triazóis/farmacocinética , Triazóis/farmacologia , Adulto Jovem
7.
Eur J Pharm Sci ; 96: 626-642, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27693299

RESUMO

Three Physiologically Based Pharmacokinetic software packages (GI-Sim, Simcyp® Simulator, and GastroPlus™) were evaluated as part of the Innovative Medicine Initiative Oral Biopharmaceutics Tools project (OrBiTo) during a blinded "bottom-up" anticipation of human pharmacokinetics. After data analysis of the predicted vs. measured pharmacokinetics parameters, it was found that oral bioavailability (Foral) was underpredicted for compounds with low permeability, suggesting improper estimates of intestinal surface area, colonic absorption and/or lack of intestinal transporter information. Foral was also underpredicted for acidic compounds, suggesting overestimation of impact of ionisation on permeation, lack of information on intestinal transporters, or underestimation of solubilisation of weak acids due to less than optimal intestinal model pH settings or underestimation of bile micelle contribution. Foral was overpredicted for weak bases, suggesting inadequate models for precipitation or lack of in vitro precipitation information to build informed models. Relative bioavailability was underpredicted for both high logP compounds as well as poorly water-soluble compounds, suggesting inadequate models for solubility/dissolution, underperforming bile enhancement models and/or lack of biorelevant solubility measurements. These results indicate areas for improvement in model software, modelling approaches, and generation of applicable input data. However, caution is required when interpreting the impact of drug-specific properties in this exercise, as the availability of input parameters was heterogeneous and highly variable, and the modellers generally used the data "as is" in this blinded bottom-up prediction approach.


Assuntos
Biofarmácia/métodos , Simulação por Computador , Modelos Biológicos , Preparações Farmacêuticas/classificação , Preparações Farmacêuticas/metabolismo , Administração Oral , Avaliação Pré-Clínica de Medicamentos/métodos , Previsões , Humanos , Absorção Intestinal/efeitos dos fármacos , Absorção Intestinal/fisiologia , Preparações Farmacêuticas/administração & dosagem
8.
Eur J Pharm Sci ; 96: 610-625, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27816631

RESUMO

Orally administered drugs are subject to a number of barriers impacting bioavailability (Foral), causing challenges during drug and formulation development. Physiologically-based pharmacokinetic (PBPK) modelling can help during drug and formulation development by providing quantitative predictions through a systems approach. The performance of three available PBPK software packages (GI-Sim, Simcyp®, and GastroPlus™) were evaluated by comparing simulated and observed pharmacokinetic (PK) parameters. Since the availability of input parameters was heterogeneous and highly variable, caution is required when interpreting the results of this exercise. Additionally, this prospective simulation exercise may not be representative of prospective modelling in industry, as API information was limited to sparse details. 43 active pharmaceutical ingredients (APIs) from the OrBiTo database were selected for the exercise. Over 4000 simulation output files were generated, representing over 2550 study arm-institution-software combinations and approximately 600 human clinical study arms simulated with overlap. 84% of the simulated study arms represented administration of immediate release formulations, 11% prolonged or delayed release, and 5% intravenous (i.v.). Higher percentages of i.v. predicted area under the curve (AUC) were within two-fold of observed (52.9%) compared to per oral (p.o.) (37.2%), however, Foral and relative AUC (Frel) between p.o. formulations and solutions were generally well predicted (64.7% and 75.0%). Predictive performance declined progressing from i.v. to solution and immediate release tablet, indicating the compounding error with each layer of complexity. Overall performance was comparable to previous large-scale evaluations. A general overprediction of AUC was observed with average fold error (AFE) of 1.56 over all simulations. AFE ranged from 0.0361 to 64.0 across the 43 APIs, with 25 showing overpredictions. Discrepancies between software packages were observed for a few APIs, the largest being 606, 171, and 81.7-fold differences in AFE between SimCYP and GI-Sim, however average performance was relatively consistent across the three software platforms.


Assuntos
Biofarmácia/métodos , Simulação por Computador , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Administração Oral , Avaliação Pré-Clínica de Medicamentos/métodos , Previsões , Humanos , Absorção Intestinal/efeitos dos fármacos , Absorção Intestinal/fisiologia , Preparações Farmacêuticas/administração & dosagem
9.
Clin Pharmacol Drug Dev ; 4(1): 41-48, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26097791

RESUMO

Domperidone effects on QTc duration were assessed in a single-center, double-blind, four-way crossover study of 44 healthy participants randomized to one of four treatment sequences consisting of four treatment periods separated by 4-9 days washout. On Day 1 of each 4-day period, participants began oral domperidone 10 or 20 mg q.i.d., matching placebo q.i.d., or single-dose moxifloxacin 400 mg (positive control)/placebo q.i.d. In each period, triplicate 12-lead electrocardiograms were recorded at baseline (30, 20, and 10 minutes predose), 8 timepoints after dosing on Days 1 and 4, and predose on Day 4. In mixed effects models, the largest difference for domperidone in least squares means for change from baseline QTcP versus placebo was 3.4 milliseconds (20 mg q.i.d., Day 4), 90% CI: 1.0-5.9, and <10 milliseconds at all timepoints for both domperidone dosages. Moxifloxacin response confirmed assay sensitivity. Participants achieved expected domperidone plasma exposures. No significant exposure-response relationship was found for QTc increase per ng/mL domperidone (90% CI of the slope estimate included zero at mean Cmax on Day 1 or Day 4). In summary, domperidone at doses up to 80 mg/day did not cause clinically relevant QTc interval prolongation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA