Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
BMC Public Health ; 24(1): 845, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38504193

RESUMO

PURPOSE: The significance of regular physical activity (PA) in reducing cardiovascular disease (CVD) risk is widely acknowledged. However, children in rural areas encounter specific barriers to PA compared to their urban counterparts. This study employs human-centered participatory co-design, involving community stakeholders in developing a multi-level PA intervention named Hoosier Sport. The primary hypothesis is the co-design sessions leading to the development of a testable intervention protocol. METHODS: Two co-design teams, each consisting of six children and six adults, were formed using human-centered participatory co-design facilitated by research faculty and graduate students. The process involved five co-design sessions addressing problem identification, solution generation, solution evaluation, operationalization, and prototype evaluation. Thematic analysis was employed to identify key themes and intervention components. RESULTS: Child co-designers (n = 6) ranged from 6th to 8th grade, averaging 12.6 years (SD = 1.8), while adult co-designers (n = 6) averaged 43.3 years (SD = 8.08). Thematic analysis revealed children emphasizing autonomy, the freedom to choose physical and non-physical activities, and the importance of building peer relationships during PA. Adult interviews echoed the importance of autonomy and choice in activities, with a focus on relatedness through positive role modeling. CONCLUSION: The prototype intervention and implementation strategies developed constitute a testable intervention aligned with Phase 1 of the ORBIT model. This testable prototype lays the groundwork for a collaborative campus-community partnership between the university and the local community, ensuring mutual benefits and sustainable impact.


Assuntos
Exercício Físico , Esportes , Criança , Adulto , Humanos , Instituições Acadêmicas , Estilo de Vida , Estudantes
2.
Br J Clin Pharmacol ; 88(1): 187-198, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34131942

RESUMO

AIMS: Omecamtiv mecarbil (OM) is a novel selective cardiac myosin activator under investigation for the treatment of heart failure. This study aimed to evaluate the effect of therapeutic concentrations of OM on electrocardiogram (ECG) parameters and exclude a clinically concerning effect on the rate-corrected QT (QTc) interval. METHODS: In part A, 70 healthy subjects received a 25 mg oral dose of OM, and pharmacokinetics were assessed. Only subjects with maximum observed plasma concentration ≤ 350 ng/mL (n = 60) were randomized into part B, where they received a single oral dose of placebo, 50 mg OM and 400 mg moxifloxacin in a 3-period, 3-treatment, 6-sequence crossover study with continuous ECG collection. RESULTS: After a 50-mg dose of OM, mean placebo-corrected change from baseline QTcF (∆∆QTcF; Fridericia correction) ranged from -6.7 ms at 1 hour postdose to -0.8 ms at 4 hours postdose. The highest upper bound of the 1-sided 95% confidence interval (CI) was 0.7 ms (4 h postdose). Moxifloxacin resulted in a clear increase in mean ∆∆QTcF, with a peak value of 13.1 ms (90% CI: 11.71-14.57) at 3 hours; lower bound of the 1-sided 95% CI was > 5 ms at all of the 3 prespecified time points. Based on a concentration-QTc analysis, an effect on ∆∆QTcF exceeding 10 ms can be excluded up to OM plasma concentrations of ~800 ng/mL. There were no serious or treatment-emergent adverse events leading to discontinuation from the study. CONCLUSION: OM does not have a clinically relevant effect on the studied ECG parameters.


Assuntos
Eletrocardiografia , Fluoroquinolonas , Estudos Cross-Over , Método Duplo-Cego , Fluoroquinolonas/efeitos adversos , Frequência Cardíaca , Humanos , Moxifloxacina/efeitos adversos , Ureia/análogos & derivados
3.
J Cardiovasc Pharmacol ; 79(4): 539-548, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34983909

RESUMO

ABSTRACT: Omecamtiv mecarbil is a small molecule that has been shown to improve cardiac function in patients with heart failure (HF) with reduced ejection fraction and is currently being developed as an oral modified release tablet for subjects with chronic HF. The objectives of this study were to analyze the pharmacokinetic (PK) properties of omecamtiv mecarbil and to investigate the effects of potential covariates on pertinent PK parameters using population PK modeling of data from 3 clinical trials in healthy subjects (n = 85) and 3 clinical trials in patients with HF (n = 4261). The population PK analysis was performed using a nonlinear mixed effects modeling approach. Omecamtiv mecarbil has a clearance of 11.7 L/h (0.701% relative standard error) and a central volume distribution of 275 L (2.12% relative standard error). The estimated half-life of omecamtiv mecarbil was 33 hours. Body weight and estimated glomerular filtration rate were significant covariates, but their effect on exposure was modest and lacked clinical relevance. Additional covariates, including sex, race, bilirubin, albumin, concomitant medications, New York Heart Association Functional Classification, N-terminal-pro hormone B-type natriuretic peptide, troponin I, creatine kinase MB, serum hemoglobin, tablet formulation, aspartate aminotransferase, and serum urea, were tested and found to have no impact on omecamtiv mecarbil exposures. The results of this integrated evaluation of the impact of covariates on the systemic exposure of omecamtiv mecarbil suggest that dose adjustment is not required for the studied subpopulations of patients with HF.


Assuntos
Insuficiência Cardíaca , Ureia , Voluntários Saudáveis , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Volume Sistólico , Ureia/análogos & derivados , Ureia/farmacocinética
4.
J Pharmacokinet Pharmacodyn ; 49(4): 455-469, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35870059

RESUMO

measures such as progression-free survival (PFS) and overall survival (OS) are commonly reported in literature for oncology trials, while time to progression (TTP) and post progression survival (PPS) are not usually reported. A time-variant transition hazard model was developed using an ordinary differential equation (ODE) model to estimate TTP and PPS from summary level PFS and OS. The model was applied to published data from immune checkpoint inhibitor trials for non-small cell lung cancer (NSCLC) in a meta-analysis framework. This model-based method was able to robustly estimate TTP and PPS from summary level OS and PFS data, provided a quantitative approach for understanding the patterns of disease progression across different treatments through the time-variant disease progression rate function, and provided a summary of how different treatments affect TTP and PPS. The proposed method can be generalized to characterize and quantify multiple time-to-event endpoints jointly in oncology trials and improve our understanding of disease prognostics for different treatments.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Progressão da Doença , Intervalo Livre de Doença , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Intervalo Livre de Progressão
5.
Drug Metab Dispos ; 49(8): 619-628, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34011533

RESUMO

Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that is currently in clinical development for the treatment of heart failure. The absorption and disposition of [14C]OM (60 µCi) were studied after a single intravenous infusion (35 mg over 1 hour) or oral solution dose (35 mg) in 14 healthy male subjects. Mean recovery of the administered [14C]OM dose was 85.1% and 86.5% over 336 hours for the intravenous and oral routes, respectively. After intravenous dosing, 47.8% and 37.3% of the dose was recovered in urine and feces, respectively; after oral dosing, 48.6% and 38.0% was recovered in urine and feces, respectively. Unchanged OM accounted for a minor percentage of radioactivity in urine (mean 7.7% of dose) and feces (mean 4.1% of dose) across all subjects. The major metabolites recovered in urine and feces were M3 (decarbamoylation product) and sequential metabolite M4 (lactam of M3), which accounted for means of 26.5% and 11.6% of the administered dose, respectively. The CYP4 family of enzymes was primarily responsible for the formation of M3 based on in vitro studies. Other metabolic pathways accounted for 14.9% of the administered dose. In pooled plasma, OM, M3, and M4 accounted for 83.8%, 6.0%, and 3.3% of the total [14C]OM-related materials. No other plasma metabolites constituted more than 3% of the administered dose. The bioavailability for OM solution was 93.5% after rapid and extensive absorption. SIGNIFICANCE STATEMENT: This study characterized the absorption and disposition of OM, a novel small molecule being developed for the treatment of heart failure. OM was primarily cleared through metabolism by the CYP4 family through oxidative cleavage of a terminal carbamate moiety that resembles hydrolysis.


Assuntos
Família 4 do Citocromo P450/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Eliminação Hepatobiliar/fisiologia , Absorção Intestinal/fisiologia , Eliminação Renal/fisiologia , Ureia/análogos & derivados , Administração Intravenosa , Administração Oral , Adulto , Disponibilidade Biológica , Biotransformação , Fármacos Cardiovasculares/administração & dosagem , Fármacos Cardiovasculares/farmacocinética , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Masculino , Ureia/administração & dosagem , Ureia/farmacocinética
6.
Biopharm Drug Dispos ; 42(7): 319-328, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34087948

RESUMO

Omecamtiv mecarbil (OM) is a cardiac myosin activator in clinical development for the treatment of heart failure. The effect of food on the pharmacokinetics (PK) of 25, 37.5, and 50 mg strength modified release (MR) tablets and the bioequivalence of two 25 mg tablets versus one 50 mg MR tablet were evaluated in two open-label, randomized, cross-over studies in healthy subjects. Subjects received two 25 mg tablets or one 50 mg OM MR tablet under fed or fasted states in Study 1 (n = 39), and single oral doses of 25 and 37.5 mg OM MR tablets and to assess its relative bioavailability to the 25 mg MR tablet, a 25 mg oral solution under fed or fasted states in Study 2 (n = 34). The area under the concentration-time curve (AUC) and the maximum observed concentration (Cmax ) of 25, 37.5, or 50 mg OM MR tablets were approximately 13%-22% higher and 31%-40% higher, respectively, when taken with food. The two 25 mg and one 50 mg OM MR tablets were bioequivalent (90% confidence intervals) of the geometric mean ratios for Cmax and AUC of OM were within 0.8-1.25 under the fasted or fed state. OM was well tolerated and all treatment-emergent events were mild in severity and resolved by the end of the study. In conclusion, these studies demonstrated that the effect of food on the PK of OM was minimal at all three studied strengths of the MR tablets, and two 25 mg MR tablets may be switched for one 50 mg MR tablet (EudraCT Number: 2019-003683-44).


Assuntos
Ureia/análogos & derivados , Administração Oral , Adulto , Área Sob a Curva , Miosinas Cardíacas , Estudos Cross-Over , Preparações de Ação Retardada , Substituição de Medicamentos , Jejum/metabolismo , Feminino , Interações Alimento-Droga , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Comprimidos , Equivalência Terapêutica , Ureia/administração & dosagem , Ureia/efeitos adversos , Ureia/sangue , Ureia/farmacocinética
7.
Br J Clin Pharmacol ; 83(3): 527-539, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27662429

RESUMO

AIM: The aim of the current study was to characterize the population pharmacokinetics of a triple direct-acting antiviral (DAA) regimen (3D) (ombitasvir, paritaprevir-ritonavir and dasabuvir) and adjunctive ribavirin, and estimate covariate effects in a broad spectrum of subjects with hepatitis C virus (HCV) genotype 1 infection. METHODS: Pharmacokinetic data from six phase III studies and one phase II study in subjects receiving the currently approved doses of the 3D ± ribavirin regimen for treating HCV genotype 1 infection for 12 weeks or 24 weeks were characterized using separate population pharmacokinetic models, built using each component of the regimen from nonlinear mixed-effects methodology in NONMEM 7.3. In the models, demographic and clinical covariates were tested. Models were assessed via goodness-of-fit plots, visual predictive checks and bootstrap evaluations. RESULTS: The population pharmacokinetic models for each component of the 3D ± ribavirin regimen (DAAs and ritonavir, n = 2348) and ribavirin (n = 1841) adequately described their respective plasma concentration-time data. Model parameter estimates were precise and robust, and all models showed good predictive ability. Significant covariate effects associated with apparent clearance and volume of distribution included age, body weight, gender, cirrhosis, HCV subtype, opioid or antidiabetic agent use, and creatinine clearance. CONCLUSION: The population pharmacokinetics of the 3D ± ribavirin regimen components in HCV-infected patients were characterized using phase II and III HCV clinical trial data. Although several statistically significant covariates were identified, their effects were modest and not clinically meaningful to necessitate dose adjustments for any component of the 3D regimen.


Assuntos
Anilidas/farmacocinética , Carbamatos/farmacocinética , Hepatite C/sangue , Compostos Macrocíclicos/farmacocinética , Ribavirina/farmacocinética , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Uracila/análogos & derivados , 2-Naftilamina , Adolescente , Adulto , Idoso , Anilidas/sangue , Antivirais/sangue , Antivirais/farmacocinética , Carbamatos/sangue , Ensaios Clínicos Fase II como Assunto/estatística & dados numéricos , Ensaios Clínicos Fase III como Assunto/estatística & dados numéricos , Ciclopropanos , Combinação de Medicamentos , Feminino , Humanos , Lactamas Macrocíclicas , Compostos Macrocíclicos/sangue , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Prolina/análogos & derivados , Ribavirina/sangue , Ritonavir/sangue , Sulfonamidas/sangue , Uracila/sangue , Uracila/farmacocinética , Valina , Adulto Jovem
8.
Clin Infect Dis ; 62(8): 972-9, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26740513

RESUMO

BACKGROUND: Guidelines for the treatment of human immunodeficiency virus (HIV) infection consistently recommend initiation of antiretroviral therapy in patients with hepatitis C virus (HCV)/HIV-1 coinfection. Therefore, potential drug interactions between antiretroviral drugs and HCV direct-acting antiviral agents (DAAs) must be carefully considered. The objective of this investigation was to evaluate the compatibility of a novel combination of DAAs (the 3D regimen) with commonly prescribed HIV-1 protease inhibitors (PIs). METHODS: Five phase 1, multiple-dose, open-label pharmacokinetic studies were performed in 144 healthy volunteers. Participants in each study were randomly assigned 1:1 into cohorts assessing the effects of the steady-state 3D regimen on steady-state HIV-1 PIs or vice versa. The 3D regimen comprised ombitasvir (25 mg once daily), paritaprevir/ritonavir (150/100 mg once daily), and dasabuvir (250 or 400 mg twice daily). The HIV-1 PIs assessed included atazanavir, darunavir, and lopinavir (administered with ritonavir). Safety, tolerability, and pharmacokinetic parameters were assessed to evaluate the compatibility of the drug regimens. RESULTS: Coadministration of the 3D regimen with the evaluated HIV-1 PIs was generally well tolerated in healthy volunteers. Morning administration of atazanavir (300 mg once daily) and darunavir regimens exhibited no clinically meaningful drug interactions with the 3D regimen. However, owing to higher paritaprevir and/or ritonavir exposures, evening administration of atazanavir (300 mg) plus ritonavir (100 mg) or lopinavir/ritonavir (800/200 mg) with the 3D regimen is not recommended. CONCLUSIONS: The 3D regimen can be coadministered with morning atazanavir and darunavir regimens. However, evening atazanavir plus ritonavir and lopinavir/ritonavir regimens are not recommended in combination with the 3D regimen.


Assuntos
Antivirais/administração & dosagem , Interações Medicamentosas , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/administração & dosagem , Hepatite C Crônica/tratamento farmacológico , 2-Naftilamina , Adolescente , Adulto , Anilidas/administração & dosagem , Anilidas/farmacocinética , Anilidas/uso terapêutico , Antivirais/farmacocinética , Antivirais/uso terapêutico , Carbamatos/administração & dosagem , Carbamatos/farmacocinética , Carbamatos/uso terapêutico , Coinfecção/tratamento farmacológico , Coinfecção/virologia , Ciclopropanos , Esquema de Medicação , Quimioterapia Combinada/efeitos adversos , Feminino , Infecções por HIV/complicações , Infecções por HIV/virologia , Inibidores da Protease de HIV/efeitos adversos , Inibidores da Protease de HIV/farmacocinética , HIV-1/efeitos dos fármacos , Voluntários Saudáveis , Hepacivirus/efeitos dos fármacos , Hepatite C/complicações , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Hepatite C Crônica/complicações , Hepatite C Crônica/virologia , Humanos , Lactamas Macrocíclicas , Compostos Macrocíclicos/administração & dosagem , Compostos Macrocíclicos/farmacocinética , Compostos Macrocíclicos/uso terapêutico , Masculino , Pessoa de Meia-Idade , Prolina/análogos & derivados , Ritonavir/administração & dosagem , Ritonavir/farmacocinética , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapêutico , Uracila/administração & dosagem , Uracila/análogos & derivados , Uracila/farmacocinética , Uracila/uso terapêutico , Valina , Adulto Jovem
9.
Antimicrob Agents Chemother ; 60(5): 2965-71, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26953200

RESUMO

The three direct-acting antiviral agent (3D) regimen is a novel combination of direct-acting antiviral agents (DAAs) that has proven effective for the treatment of hepatitis C virus (HCV) infection. Given the potential for coadministration in patients with human immunodeficiency virus infection, possible drug interactions with antiretroviral drugs must be carefully considered. Four phase 1, multiple-dose pharmacokinetic studies were conducted in healthy volunteers (n = 66). The 3D regimen of 150/100 mg daily paritaprevir/ritonavir, 25 mg daily ombitasvir, and 400 mg twice-daily dasabuvir was administered alone or in combination with 200 mg daily of emtricitabine and 300 mg daily of tenofovir disoproxil fumarate (tenofovir DF), 25 mg daily of rilpivirine, or 400 mg of raltegravir twice daily. A 2-DAA regimen of 150/100 mg daily paritaprevir/ritonavir and 400 mg of dasabuvir twice daily was also studied in combination with efavirenz/emtricitabine/tenofovir DF at 600/200/300 mg daily, respectively (Atripla; Bristol-Myers Squibb). Pharmacokinetic parameters were determined from plasma drug concentrations. No clinically significant drug interactions were observed (≤32% change in exposure) between the 3D regimen and that of emtricitabine plus tenofovir DF. Raltegravir exposure was increased up to 134% when the drug was coadministered with the 3D regimen. Although coadministration with rilpivirine was well tolerated in healthy volunteers, observed elevations in rilpivirine exposures may increase the potential for adverse drug reactions. Concomitant use of the 2-DAA regimen and efavirenz/emtricitabine/tenofovir DF was discontinued owing to poor tolerability and adverse events. No dose adjustment is required during coadministration of raltegravir, tenofovir DF, or emtricitabine with the 3D regimen. Rilpivirine is not recommended and efavirenz is contraindicated for coadministration with the 3D regimen.


Assuntos
Antirretrovirais/farmacologia , Antivirais/farmacologia , Benzoxazinas/farmacologia , Emtricitabina/farmacologia , Hepacivirus/efeitos dos fármacos , Raltegravir Potássico/farmacologia , Rilpivirina/farmacologia , Tenofovir/farmacologia , Alcinos , Ciclopropanos , Interações Medicamentosas , HIV-1/efeitos dos fármacos , Humanos
10.
Antimicrob Agents Chemother ; 60(2): 855-61, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26596948

RESUMO

The combination of ombitasvir (an NS5A inhibitor), paritaprevir (an NS3/4A inhibitor) coadministered with ritonavir (r), and dasabuvir (an NS5B nonnucleoside polymerase inhibitor), referred to as the 3D regimen, and the combination of ombitasvir-paritaprevir-r, referred to as the 2D regimen, have demonstrated high efficacy with and without ribavirin in hepatitis C virus (HCV)-infected subjects. These regimens have potential for coadministration with sofosbuvir (nucleoside NS5B inhibitor) in the treatment of HCV. This phase 1, drug-drug interaction, open-label, multiple-dose study enrolled 32 healthy subjects to receive the 3D or 2D regimen in combination with sofosbuvir. Doses of study drugs were as follows: ombitasvir-paritaprevir-r, 25/150/100 mg daily (QD); dasabuvir, 250 mg twice daily (BID); and sofosbuvir, 400 mg QD. Blood samples were collected on study days 7, 14, and 21 for evaluating drug interaction at steady state. The effect of the 3D and 2D regimens on the pharmacokinetics of sofosbuvir and its circulating metabolite GS-331007 and vice versa was assessed by a repeated-measures analysis. Exposures of the 3D and 2D regimens were similar (≤20% change) during coadministration with sofosbuvir and during administration alone. Sofosbuvir exposures were 61% to 112% higher with the 3D regimen and 64% to 93% higher with the 2D regimen than with sofosbuvir alone. GS-331007 total exposures were 27% and 32% higher with the 3D and 2D regimens, respectively, than with sofosbuvir alone. Increases in sofosbuvir and GS-331007 exposures likely resulted from breast cancer resistance protein (BCRP) and/or P glycoprotein (P-gp) transporter inhibition by paritaprevir and ritonavir. No subjects discontinued the study due to study drug-related adverse events. No dose adjustment is recommended for 3D, 2D, or sofosbuvir in clinical trials exploring the safety and efficacy of the combination. (This study has been registered at ClinicalTrials.gov under registration no. NCT02356562 and NCT02292719.).


Assuntos
Anilidas/uso terapêutico , Antivirais/uso terapêutico , Carbamatos/uso terapêutico , Interações Medicamentosas/fisiologia , Hepatite C Crônica/tratamento farmacológico , Compostos Macrocíclicos/uso terapêutico , Ritonavir/uso terapêutico , Sofosbuvir/uso terapêutico , Sulfonamidas/uso terapêutico , Uracila/análogos & derivados , 2-Naftilamina , Adulto , Anilidas/efeitos adversos , Anilidas/farmacocinética , Antivirais/efeitos adversos , Antivirais/farmacocinética , Carbamatos/efeitos adversos , Carbamatos/farmacocinética , Ciclopropanos , Quimioterapia Combinada , Feminino , Hepacivirus/efeitos dos fármacos , Humanos , Lactamas Macrocíclicas , Compostos Macrocíclicos/efeitos adversos , Compostos Macrocíclicos/farmacocinética , Masculino , Prolina/análogos & derivados , Ritonavir/efeitos adversos , Ritonavir/farmacocinética , Sofosbuvir/efeitos adversos , Sofosbuvir/farmacocinética , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética , Uracila/efeitos adversos , Uracila/farmacocinética , Uracila/uso terapêutico , Valina
11.
Antimicrob Agents Chemother ; 60(1): 105-14, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26459906

RESUMO

The two direct-acting antiviral (2D) regimen of ombitasvir and paritaprevir (administered with low-dose ritonavir) is being developed for treatment of genotype subtype 1b and genotypes 2 and 4 chronic hepatitis C virus (HCV) infection. Drug-drug interactions were evaluated in healthy volunteers to develop dosing recommendations for HCV-infected subjects. Mechanism-based interactions were evaluated for ketoconazole, pravastatin, rosuvastatin, digoxin, warfarin, and omeprazole. Interactions were also evaluated for duloxetine, escitalopram, methadone, and buprenorphine-naloxone. Ratios of geometric means with 90% confidence intervals for the maximum plasma concentration and the area under the plasma concentration-time curve were estimated to assess the magnitude of the interactions. For most medications, coadministration with the 2D regimen resulted in a <50% change in exposures. Ketoconazole, digoxin, pravastatin, and rosuvastatin exposures increased by up to 105%, 58%, 76%, and 161%, respectively, and omeprazole exposures decreased by approximately 50%. Clinically meaningful changes in ombitasvir, paritaprevir, or ritonavir exposures were not observed. In summary, all 11 medications evaluated can be coadministered with the 2D regimen, with most medications requiring no dose adjustment. Ketoconazole, digoxin, pravastatin, and rosuvastatin require lower doses, and omeprazole may require a higher dose. No dose adjustment is required for the 2D regimen.


Assuntos
Anilidas/farmacocinética , Antivirais/farmacocinética , Carbamatos/farmacocinética , Compostos Macrocíclicos/farmacocinética , Ritonavir/farmacocinética , Adulto , Anilidas/sangue , Antiácidos/sangue , Antiácidos/farmacocinética , Antiarrítmicos/sangue , Antiarrítmicos/farmacocinética , Anticoagulantes/sangue , Anticoagulantes/farmacocinética , Antidepressivos/sangue , Antidepressivos/farmacocinética , Antifúngicos/sangue , Antifúngicos/farmacocinética , Antivirais/sangue , Área Sob a Curva , Carbamatos/sangue , Ciclopropanos , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Voluntários Saudáveis , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/sangue , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacocinética , Lactamas Macrocíclicas , Compostos Macrocíclicos/sangue , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Antagonistas de Entorpecentes/sangue , Antagonistas de Entorpecentes/farmacocinética , Prolina/análogos & derivados , Ritonavir/sangue , Sulfonamidas , Valina
12.
Ther Drug Monit ; 38(5): 640-5, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27310199

RESUMO

BACKGROUND: Interactions between tacrolimus and cyclosporine (CSA) and the 3 direct-acting antiviral regimen (3D) of ombitasvir, paritaprevir/ritonavir, and dasabuvir necessitate a priori dose adjustments for the immunosuppressants to achieve desired levels. Modeling and simulations based on data in healthy subjects predicted that tacrolimus 0.5 mg every 7 days or 0.2 mg every 3 days, and CSA at one-fifth the total daily dose administered once daily, would achieve desired trough concentrations (Ctrough) during 3D treatment. The success of these dosing recommendations was evaluated by analyzing pharmacokinetic data from liver transplant recipients in the CORAL-I study. METHODS: A population pharmacokinetic model was developed using tacrolimus dosing and Ctrough data before and during 3D treatment (n = 29). The model was used to simulate various tacrolimus dosing regimens and predict tacrolimus concentration-time profiles during 3D treatment. CSA Ctrough data before and during 3D treatment (n = 5) were also summarized. RESULTS: A one-compartment model with first-order absorption adequately described tacrolimus pharmacokinetic profiles during the first 4 weeks of 3D treatment. Estimated tacrolimus Ctrough values (median; interquartile range) before and during 3D treatment were comparable (5.7 ng/mL; 4.9-6.5 ng/mL versus 5.2 ng/mL; 4.2-6.3 ng/mL, respectively). Based on simulations, in a patient with a starting Ctrough of 6 ng/mL, 0.5 mg tacrolimus every 7 or 14 days or 0.2 mg tacrolimus every 3 days will result in Ctrough levels of 6-9 ng/mL, 4-6 ng/mL, and 6-10 ng/mL, respectively, during 3D treatment. For CSA, Ctrough values (median; interquartile range) before and during 3D treatment were comparable (126 ng/mL; 94-140 ng/mL versus 104 ng/mL; 82-140 ng/mL). CONCLUSIONS: Observed data for tacrolimus and CSA in liver transplant recipients confirm that the recommended dosing strategies are valid and therapeutic levels of immunosuppression can be maintained during 3D treatment.


Assuntos
Anilidas/farmacologia , Carbamatos/farmacologia , Ciclosporina/farmacocinética , Compostos Macrocíclicos/farmacologia , Sulfonamidas/farmacologia , Tacrolimo/farmacocinética , Uracila/análogos & derivados , 2-Naftilamina , Adolescente , Adulto , Idoso , Antivirais/farmacologia , Ciclopropanos , Cálculos da Dosagem de Medicamento , Quimioterapia Combinada , Feminino , Hepatite C/tratamento farmacológico , Humanos , Imunossupressores/farmacocinética , Lactamas Macrocíclicas , Transplante de Fígado , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Prolina/análogos & derivados , Ritonavir/farmacologia , Uracila/farmacologia , Valina , Adulto Jovem
13.
J Hepatol ; 63(4): 805-12, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26070406

RESUMO

BACKGROUND & AIMS: Paritaprevir, ombitasvir, and dasabuvir are direct-acting antivirals for treatment of chronic hepatitis C virus (HCV) infection. The aim of this study was to characterize the effects of mild, moderate, and severe hepatic impairment on the pharmacokinetics of these drugs. METHODS: HCV-negative subjects with normal hepatic function (n=7) or mild (Child-Pugh A, n=6), moderate (Child-Pugh B, n=6), or severe (Child-Pugh C, n=5) hepatic impairment received a single-dose of the combination of paritaprevir plus ritonavir (paritaprevir/r, 200/100 mg), ombitasvir (25 mg), and dasabuvir (400 mg). Plasma samples were collected through 144 hours after administration for pharmacokinetic assessments. RESULTS: Paritaprevir, ombitasvir, dasabuvir, and ritonavir exposures (maximal plasma concentration, C(max), and area under the concentration-time curve, AUC) were minimally affected in subjects with mild or moderate hepatic impairment. Differences in exposures between healthy controls and subjects with mild or moderate hepatic impairment were less than 35%, except for 62% higher paritaprevir AUC in subjects with moderate hepatic impairment. Paritaprevir and dasabuvir AUC were significantly higher in subjects with severe hepatic impairment (950% and 325%, respectively). However, ombitasvir AUC was 54% lower and ritonavir AUC was comparable. Adverse events included eye stye, insomnia, and pain from an infiltrated intravenous line. CONCLUSIONS: The changes observed in paritaprevir, ritonavir, ombitasvir, and dasabuvir exposures in subjects with mild or moderate hepatic impairment do not necessitate dose adjustment. Subjects with severe hepatic impairment had substantially higher paritaprevir and dasabuvir exposures.


Assuntos
Anilidas/farmacocinética , Carbamatos/farmacocinética , Insuficiência Hepática/sangue , Hepatite C Crônica/tratamento farmacológico , Compostos Macrocíclicos/farmacocinética , Ribavirina/farmacocinética , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Uracila/análogos & derivados , 2-Naftilamina , Anilidas/administração & dosagem , Antivirais/administração & dosagem , Antivirais/farmacocinética , Carbamatos/administração & dosagem , Ciclopropanos , Inibidores do Citocromo P-450 CYP3A/administração & dosagem , Inibidores do Citocromo P-450 CYP3A/farmacocinética , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Insuficiência Hepática/etiologia , Hepatite C Crônica/sangue , Hepatite C Crônica/complicações , Humanos , Lactamas Macrocíclicas , Testes de Função Hepática , Compostos Macrocíclicos/administração & dosagem , Masculino , Pessoa de Meia-Idade , Prolina/análogos & derivados , Ribavirina/administração & dosagem , Ritonavir/administração & dosagem , Sulfonamidas/administração & dosagem , Resultado do Tratamento , Uracila/administração & dosagem , Uracila/farmacocinética , Valina
14.
J Hepatol ; 63(1): 20-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25646891

RESUMO

BACKGROUND & AIMS: Paritaprevir (administered with ritonavir, PTV/r), ombitasvir (OBV), and dasabuvir (DSV) are direct-acting antiviral agents (DAAs) for the treatment of chronic hepatitis C virus (HCV) infection. Thirteen studies were conducted to characterize drug-drug interactions for the 3D regimen of OBV, PTV/r, and DSV and various medications in healthy volunteers to inform dosing recommendations in HCV-infected patients. METHODS: Mechanism-based drug-drug interactions were evaluated for gemfibrozil, ketoconazole, carbamazepine, warfarin, omeprazole, digoxin, pravastatin, and rosuvastatin. Drug-drug interactions with medications commonly used in HCV-infected patients were evaluated for amlodipine, furosemide, alprazolam, zolpidem, duloxetine, escitalopram, methadone, buprenorphine/naloxone, and oral contraceptives. Ratios of geometric means with 90% confidence intervals for maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) were used to determine the magnitude of interaction. RESULTS: Coadministration with the 3D regimen of OBV, PTV/r, and DSV resulted in a <2-fold change in mean Cmax and AUC for most medications and the DAAs, indicating minimal to modest interactions. Carbamazepine decreased PTV, ritonavir, and DSV exposures substantially, while gemfibrozil increased DSV exposures substantially. Although coadministration with ethinyl estradiol-containing contraceptives resulted in elevated alanine aminotransferase levels, coadministration with a progestin-only contraceptive did not. CONCLUSIONS: The majority of medications can be coadministered with the 3D regimen of OBV, PTV/r, and DSV without dose adjustment, or with clinical monitoring or dose adjustment. Although no dose adjustment is necessary for the 3D regimen when coadministered with 17 of the 20 medications, coadministration with gemfibrozil, carbamazepine, or ethinyl estradiol-containing contraceptives is contraindicated.


Assuntos
Anilidas/administração & dosagem , Carbamatos/administração & dosagem , Hepatite C Crônica/tratamento farmacológico , Compostos Macrocíclicos/administração & dosagem , Ritonavir/administração & dosagem , Sulfonamidas/administração & dosagem , Uracila/análogos & derivados , 2-Naftilamina , Administração Oral , Adolescente , Adulto , Anilidas/farmacocinética , Antivirais/administração & dosagem , Antivirais/farmacocinética , Carbamatos/farmacocinética , Ciclopropanos , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Hepacivirus/imunologia , Anticorpos Anti-Hepatite C/imunologia , Hepatite C Crônica/sangue , Hepatite C Crônica/virologia , Humanos , Lactamas Macrocíclicas , Compostos Macrocíclicos/farmacocinética , Masculino , Pessoa de Meia-Idade , Prolina/análogos & derivados , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Uracila/administração & dosagem , Uracila/farmacocinética , Valina , Adulto Jovem
15.
Br J Clin Pharmacol ; 78(1): 94-105, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24433449

RESUMO

AIMS: Levodopa-carbidopa intestinal gel (LCIG) provides continuous levodopa-carbidopa delivery through intrajejunal infusion. This study characterized the population pharmacokinetics of levodopa following a 16 h jejunal infusion of LCIG or frequent oral administration of levodopa-carbidopa tablets (LC-oral) in subjects with advanced Parkinson's disease (PD). METHODS: A non-linear mixed-effects model of levodopa pharmacokinetics was developed using serial plasma concentrations from an LCIG phase 1 study and a phase 3 double-blind, double-dummy study of the efficacy and safety of LCIG compared with LC-oral in advanced PD patients (n = 68 for model development; 45 on LCIG and 23 on LC-oral). The final model was internally evaluated using stochastic simulations and bootstrap and externally evaluated using sparse pharmacokinetic data from 311 subjects treated in a long term safety study of LCIG. RESULTS: The final model was a two compartment model with a transit compartment for absorption, first order elimination, bioavailability for LCIG (97%; confidence interval = 95% to 98%) relative to LC-oral, different first order transit absorption rate constants (LCIG = 9.2 h(-1) vs. LC-oral = 2.4 h(-1) ; corresponding mean absorption time of 7 min for LCIG vs. 25 min for LC-oral) and different residual (intra-subject) variability for LCIG (15% proportional error, 0.3 µg ml(-1) additive error) vs. LC-oral (29% proportional error, 0.59 µg ml(-1) additive error). Estimated oral clearance and steady-state volume of distribution for levodopa were 24.8 l h(-1) and 131 l, respectively. CONCLUSIONS: LCIG administration results in faster absorption, comparable levodopa bioavailability and significantly reduced intra-subject variability in levodopa concentrations relative to LC-oral administration.


Assuntos
Carbidopa/administração & dosagem , Levodopa/administração & dosagem , Levodopa/farmacocinética , Modelos Biológicos , Doença de Parkinson/tratamento farmacológico , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Antiparkinsonianos/administração & dosagem , Antiparkinsonianos/sangue , Antiparkinsonianos/farmacocinética , Disponibilidade Biológica , Carbidopa/sangue , Carbidopa/farmacocinética , Método Duplo-Cego , Combinação de Medicamentos , Feminino , Géis , Humanos , Infusões Parenterais , Levodopa/sangue , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/sangue , Comprimidos
16.
Br J Clin Pharmacol ; 77(6): 965-74, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24215171

RESUMO

AIMS: ABT-288 is a potent and selective H3 receptor antagonist with procognitive effects in several preclinical models. In previous studies, 3 mg once daily was the maximal tolerated dose in healthy volunteers. This study characterized the safety, tolerability and pharmacokinetics of ABT-288 in stable subjects with schizophrenia. METHODS: This was a randomized, double-blind, placebo-controlled, dose-escalating study of ABT-288 (10 dose levels, from 1 to 60 mg once daily for 14 days) in stable subjects with schizophrenia treated with an atypical antipsychotic. In each dose group, five to seven and two to three participants were assigned to ABT-288 and placebo, respectively. RESULTS: Of the 67 participants enrolled, nine participants (on ABT-288) were prematurely discontinued, in seven of these due to adverse events. ABT-288 was generally safe and tolerated at doses up to 45 mg once daily. The most common adverse events, in decreasing frequency (from 31 to 5%), were abnormal dreams, headache, insomnia, dizziness, somnolence, dysgeusia, dry mouth, psychotic disorder, parosmia and tachycardia. Adverse events causing early termination were psychotic events (four) and increased creatine phosphokinase, pyrexia and insomnia (one each). The half-life of ABT-288 ranged from 28 to 51 h, and steady state was achieved by day 12 of dosing. At comparable multiple doses, ABT-288 exposure in subjects with schizophrenia was 45% lower than that previously observed in healthy subjects. At trough, ABT-288 cerebrospinal fluid concentrations were 40% of the total plasma concentrations. CONCLUSIONS: ABT-288 was tolerated at a 15-fold higher dose and 12-fold higher exposures in subjects with schizophrenia than previously observed in healthy volunteers. The greater ABT-288 tolerability was not due to limited brain uptake.


Assuntos
Antagonistas dos Receptores Histamínicos H3/efeitos adversos , Piridazinas/efeitos adversos , Pirróis/efeitos adversos , Esquizofrenia/tratamento farmacológico , Adulto , Área Sob a Curva , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Piridazinas/farmacocinética , Pirróis/farmacocinética
17.
Biopharm Drug Dispos ; 35(7): 417-29, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25041811

RESUMO

ABT-384 is a potent and selective inhibitor of 11ß-hydroxysteroid dehydrogenase type 1 (HSD-1). The pharmacokinetics of ABT-384 was evaluated in healthy volunteers in single-dose (1, 8, 20, 50, 120 and 240 mg) and multiple-dose studies (1, 2, 4, 8, 20, 30 and 100 mg once daily). Less than dose-proportional pharmacokinetics of ABT-384 was observed when ABT-384 was administered at single doses lower than 8 mg. This nonlinear phenomenon disappeared after repeated doses. The dose-normalized plasma concentration-time curves superposed across all dose groups on day 7, but not on day 1. This phenomenon cannot be explained by the half-life of ABT-384. Based on available data, the nonlinearity is likely due to binding of ABT-384 to a high-affinity-low-capacity site, such that this interaction was reflected in ABT-384 pharmacokinetics. To characterize the pharmacokinetics of ABT-384, a population pharmacokinetic model for ABT-384 was constructed. The model provided reasonable fitting for both single- and multiple-dose data. Further investigation is warranted to evaluate the disposition of ABT-384 at low doses using a larger number of subjects. The constructed model would be useful in predicting ABT-384 concentrations at different doses and guiding the selection of dosing regimens in further clinical trials.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/sangue , Adamantano/análogos & derivados , Piperazinas/administração & dosagem , Piperazinas/sangue , Adamantano/administração & dosagem , Adamantano/sangue , Adamantano/farmacocinética , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Piperazinas/farmacocinética , Adulto Jovem
18.
Artigo em Inglês | MEDLINE | ID: mdl-39016337

RESUMO

Sotorasib is a small molecule drug that specifically and irreversibly inhibits the KRAS p.G12C mutant protein. This analysis investigated the impact of a high-calorie high-fat meal on the pharmacokinetics, safety, and tolerability of sotorasib in both healthy volunteers and patients with KRAS G12C advanced solid tumors. Each subject received a single oral dose of 360 or 960 mg of sotorasib under fasted conditions or with a high-fat meal (fed conditions). The geometric least squares means (GLSM) ratios (fed/fasted) for 360 mg of sotorasib Cmax and AUCinf were 1.03 and 1.38, respectively, in healthy volunteers (N = 14). The GLSM ratios (fed/fasted) for Cmax and AUC0-24h were 1.38 and 1.75, respectively, with 360 mg of sotorasib in cancer patients (N = 2). The GLSM ratios (fed/fasted) for Cmax and AUC0-24h were 0.660 and 1.25, respectively, with 960 mg of sotorasib in cancer patients (N = 8). Sotorasib was well tolerated in fast and fed conditions. The impact of a high-fat meal on sotorasib exposure is less than a 2-fold increase or decrease in Cmax and AUCs.

19.
Clin Pharmacol Drug Dev ; 13(7): 810-818, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38421129

RESUMO

Sotorasib is a small molecule that irreversibly inhibits the Kirsten rat sarcoma viral oncogene homolog (KRAS) protein with a G12C amino acid substitution mutant protein. The impact of cytochrome P450 (CYP) 3A4 inhibition and induction on sotorasib pharmacokinetics (PKs) was evaluated in 2 separate studies in healthy volunteers (N = 14/study). The impact of CYP3A4 inhibition was interrogated utilizing repeat doses of 200 mg of itraconazole, a strong CYP3A4 inhibitor, on 360 mg of sotorasib PKs. The impact of CYP3A4 induction was interrogated utilizing multiple doses of 600 mg of rifampin, a strong CYP3A4 inducer. Additionally, the impact of organic anion transporting polypeptide (OATP) 1B1/3 inhibition on 960 mg of sotorasib PKs was interrogated after a single dose of 600 mg of rifampin. CYP3A4 inhibition did not significantly impact sotorasib Cmax but did lead to a 26% increase in sotorasib AUCinf. CYP3A4 induction decreased sotorasib Cmax by 35% and AUCinf by 51%. OATP1B1/3 inhibition decreased sotorasib Cmax and AUCinf by 16% and 23%, respectively. These results support that sotorasib can be given together with strong CYP3A4 and OATP1B1/3 inhibitors but the co-administration of sotorasib and strong CYP3A4 inducers should be avoided.


Assuntos
Indutores do Citocromo P-450 CYP3A , Inibidores do Citocromo P-450 CYP3A , Citocromo P-450 CYP3A , Interações Medicamentosas , Transportador 1 de Ânion Orgânico Específico do Fígado , Rifampina , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado/antagonistas & inibidores , Transportador 1 de Ânion Orgânico Específico do Fígado/metabolismo , Masculino , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/antagonistas & inibidores , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/metabolismo , Adulto , Inibidores do Citocromo P-450 CYP3A/farmacologia , Inibidores do Citocromo P-450 CYP3A/administração & dosagem , Citocromo P-450 CYP3A/metabolismo , Rifampina/farmacologia , Rifampina/administração & dosagem , Indutores do Citocromo P-450 CYP3A/farmacologia , Feminino , Adulto Jovem , Administração Oral , Pessoa de Meia-Idade , Voluntários Saudáveis , Área Sob a Curva , Itraconazol/farmacologia , Itraconazol/administração & dosagem , Itraconazol/farmacocinética , Compostos de Espiro/farmacocinética , Compostos de Espiro/administração & dosagem , Compostos de Espiro/farmacologia
20.
CPT Pharmacometrics Syst Pharmacol ; 13(1): 154-167, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37860956

RESUMO

A multistate platform model was developed to describe time-to-event (TTE) endpoints in an oncology trial through the following states: initial, tumor response (TR), progressive disease (PD), overall survival (OS) event (death), censor to the last evaluable tumor assessment (progression-free survival [PFS] censor), and censor to study end (OS censor), using an ordinary differential equation framework. Two types of piecewise functions were used to describe the hazards for different events. Piecewise surge functions were used for events that require tumor assessments at the scheduled study visit times (TR, PD, and PFS censor). Piecewise constant functions were used to describe hazards for events that occur evenly throughout the study (OS event and OS censor). The multistate TTE model was applied to describe TTE endpoints from a published phase III study. The piecewise surge functions well-described the observed surges of hazards/events for TR, PD, PFS, and OS occurring near scheduled tumor assessments and showed good agreement with all Kaplan-Meier curves. With the flexibility of piecewise hazard functions, the model was able to evaluate covariate effects in a time-variant fashion to better understand the temporal patterns of disease prognosis through different disease states. This model can be applied to advance the field of oncology trial design and optimization by: (1) enabling robust estimations of baseline hazards and covariate effects for multiple TTE endpoints, (2) providing a platform model for understanding the composition and correlations between different TTE endpoints, and (3) facilitating oncology trial design optimization through clinical trial simulations.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Prognóstico , Intervalo Livre de Progressão , Oncologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA