Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Headache ; 63(5): 652-662, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37140071

RESUMO

OBJECTIVE: To assess the effect of single and multiple doses of rimegepant 75 mg dose on the pharmacokinetics of an oral contraceptive containing ethinyl estradiol (EE)/norgestimate (NGM) in healthy females of childbearing potential or non-menopausal females with tubal ligation. BACKGROUND: Females of childbearing age experience the highest prevalence of migraine and frequently inquire about the concomitant use of anti-migraine medications and contraceptives. Rimegepant, a calcitonin gene-related peptide receptor antagonist, demonstrated efficacy and safety for treating an acute migraine attack and preventing migraine. METHODS: This open-label, single-center, phase 1, drug-drug interaction study explored the effects of rimegepant 75 mg daily dose on the pharmacokinetics of an oral contraceptive containing EE/NGM 0.035 mg/0.25 mg in healthy females of childbearing potential or non-menopausal females with tubal ligation. During cycles 1 and 2, participants received EE/NGM once daily for 21 days followed by placebo tablets with inactive ingredients for 7 days. Rimegepant was administered during only cycle 2 for 8 days, from days 12 through 19. The primary endpoint was the effect of single and multiple doses of rimegepant on the pharmacokinetics of EE and norelgestromin (NGMN), an active metabolite of NGM, at steady state, including area under the concentration-time curve for 1 dosing interval (AUC0-τ,ss ) and maximum observed concentration (Css[max] ). RESULTS: The study enrolled 25 participants, with pharmacokinetic data assessed for 20 participants. A single 75 mg dose of rimegepant co-administered with EE/NGM increased exposures of EE and NGMN by ≤16% (geometric mean ratio [GMR], 1.03; 90% confidence interval [CI], 1.01-1.06; and GMR, 1.16; 90% CI, 1.13-1.20, respectively). After 8 days of co-administering EE/NGM with rimegepant, EE pharmacokinetic parameters, AUC0-τ,ss and Css(max) , increased by 20% (GMR, 1.20; 90% CI, 1.16-1.25) and 34% (GMR, 1.34; 90% CI, 1.23-1.46), respectively, and NGMN pharmacokinetic parameters increased by 46% (GMR, 1.46; 90% CI, 1.39-1.52) and 40% (GMR, 1.40; 90% CI, 1.30-1.51), respectively. CONCLUSIONS: The study identified modest elevations in overall EE and NGMN exposures after multiple doses of rimegepant, but these elevations are unlikely to be clinically relevant in healthy females with migraine.


Assuntos
Anticoncepcionais Orais Combinados , Etinilestradiol , Feminino , Humanos , Anticoncepcionais Orais Combinados/efeitos adversos , Anticoncepcionais Orais Combinados/farmacocinética , Etinilestradiol/farmacocinética , Piridinas
2.
J Neurotrauma ; 37(22): 2435-2444, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30816062

RESUMO

The purpose of this review is to highlight the pharmacological barrier to drug development for traumatic brain injury (TBI) and to discuss best practice strategies to overcome such barriers. Specifically, this article will review the pharmacological considerations of moving from the disease target "hit" to the "lead" compound with drug-like and central nervous system (CNS) penetrant properties. In vitro assessment of drug-like properties will be detailed, followed by pre-clinical studies to ensure adequate pharmacokinetic and pharmacodynamic characteristics of response. The importance of biomarker development and utilization in both pre-clinical and clinical studies will be detailed, along with the importance of identifying diagnostic, pharmacodynamic/response, and prognostic biomarkers of injury type or severity, drug target engagement, and disease progression. This review will detail the important considerations in determining in vivo pre-clinical dose selection, as well as cross-species and human equivalent dose selection. Specific use of allometric scaling, pharmacokinetic and pharmacodynamic criteria, as well as incorporation of biomarker assessments in human dose selection for clinical trial design will also be discussed. The overarching goal of this review is to detail the pharmacological considerations in the drug development process as a method to improve both pre-clinical and clinical study design as we evaluate novel therapies to improve outcomes in patients with TBI.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Desenvolvimento de Medicamentos/métodos , Fármacos Neuroprotetores/farmacologia , Animais , Biomarcadores/metabolismo , Humanos
3.
Clin Pharmacokinet ; 57(8): 911-928, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29353349

RESUMO

Daclatasvir is a first-in-class, highly selective, hepatitis C virus, non-structural protein 5a polymerase replication complex inhibitor with picomolar potency and broad genotypic coverage in vitro. Daclatasvir undergoes rapid absorption, with a time to reach maximum plasma concentration of 1-2 h and an elimination half-life of ~ 10 to 14 h observed in single-ascending dose studies. Steady state was achieved by day 4 in multiple-ascending dose studies. Daclatasvir can be administered without regard to food or pH modifiers. Daclatasvir exposure is similar between healthy subjects and subjects infected with hepatitis C virus. Intrinsic factors such as age, race, or sex do not impact daclatasvir exposure. No dose adjustment is necessary for patients with any degree of hepatic or renal impairment. Daclatasvir has low-to-moderate clearance with the predominant route of elimination via cytochrome P450 3A4-mediated metabolism and P-glycoprotein excretion and intestinal secretion. Renal clearance is a minor route of elimination for daclatasvir. As a result, the dose of daclatasvir should be reduced from 60 to 30 mg once daily when co-administered with strong inhibitors of cytochrome P450 3A4. No dose adjustment is required when daclatasvir is co-administered with moderate inhibitors of cytochrome P450 3A4. The dose of daclatasvir should be increased from 60 to 90 mg once daily when co-administered with moderate inducers of cytochrome P450 3A4. Co-administration of daclatasvir with strong inducers of cytochrome P450 3A4 is contraindicated. Concurrent medications with inhibitory effects on P-glycoprotein without concurrent inhibition of cytochrome P450 3A4 are unlikely to cause marked changes in daclatasvir exposure, as the clearance of daclatasvir is through both cytochrome P450 3A4 and P-glycoprotein. The potential for daclatasvir to affect the pharmacokinetics of concomitantly administered drugs that are substrates of the cytochrome P450 enzyme system is low. In vitro, daclatasvir is a weak-to-moderate inhibitor of transporters including organic cation transporter 1, P-glycoprotein, organic transporting polypeptide 1B1, organic transporting polypeptide 1B3, and breast cancer resistance protein, although in clinical studies, daclatasvir has not altered the pharmacokinetics of concomitantly administered drugs that are substrates of these transporters to an appreciable degree, except for rosuvastatin. In summary, daclatasvir is a hepatitis C virus, non-structural protein 5a-selective inhibitor with a well-characterized pharmacokinetic profile that forms part of potent and well-tolerated all-oral treatment regimens for chronic hepatitis C virus infection.


Assuntos
Antivirais/farmacocinética , Hepatite C Crônica/tratamento farmacológico , Imidazóis/farmacocinética , Absorção Fisiológica , Animais , Antivirais/administração & dosagem , Antivirais/sangue , Disponibilidade Biológica , Carbamatos , Ensaios Clínicos como Assunto , Citocromo P-450 CYP3A/metabolismo , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Meia-Vida , Hepatite C Crônica/sangue , Humanos , Imidazóis/administração & dosagem , Imidazóis/sangue , Pirrolidinas , Distribuição Tecidual , Valina/análogos & derivados , Proteínas não Estruturais Virais/antagonistas & inibidores
4.
Clin Pharmacol Drug Dev ; 6(2): 195-200, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28263460

RESUMO

Asunaprevir is an inhibitor of the hepatitis C virus (HCV) NS3/4A protease, demonstrating efficacy in clinical studies in patients infected with HCV genotype 1 or 4, with either peginterferon/ribavirin or combinations of direct-acting antivirals. Because of preferential distribution of asunaprevir to the liver via organic anion-transporting polypeptide (OATP)-mediated transport, asunaprevir demonstrates high apparent oral clearance and very low plasma concentrations. Asunaprevir plasma concentrations are markedly increased by single-dose rifampin (an OATP inhibitor) and in subjects with moderate to severe hepatic impairment. In addition, modestly higher plasma concentrations of asunaprevir have been noted in subjects infected with HCV relative to healthy subjects and in Asian subjects relative to whites. At the marketed dose, infrequent hepatic transaminase abnormalities were poorly predicted by plasma concentrations. For a compound with these characteristics, hepatic concentrations may have provided an improved understanding of the in vivo pharmacokinetic and pharmacodynamic data to support decision making during development.


Assuntos
Antivirais/farmacocinética , Isoquinolinas/farmacocinética , Fígado/metabolismo , Inibidores de Proteases/farmacocinética , Sulfonamidas/farmacocinética , Antivirais/efeitos adversos , Antivirais/sangue , Hepatite C/sangue , Hepatite C/metabolismo , Humanos , Isoquinolinas/efeitos adversos , Isoquinolinas/sangue , Inibidores de Proteases/efeitos adversos , Inibidores de Proteases/sangue , Sulfonamidas/efeitos adversos , Sulfonamidas/sangue
5.
J Clin Pharmacol ; 57(2): 235-246, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27432796

RESUMO

Voriconazole, a broad-spectrum triazole antifungal agent, is metabolized by cytochrome P450 (CYP) 2C19 and, to a lesser extent, by CYP3A. Genetic polymorphism of CYP2C19 not only plays a prominent role in its disposition but may also influence potential drug interactions with CYP450 modulators such as ritonavir. This study assessed 2-way drug interactions of voriconazole added on to ritonavir-boosted atazanavir in both CYP2C19 extensive-metabolizer (EM) and poor-metabolizer (PM) healthy subjects. Each subject received voriconazole alone on days 1-3, followed by a 7-day washout. Atazanavir/ritonavir 300/100 mg once daily was given on days 11-30 and voriconazole on days 21-30. Voriconazole doses were 200 mg (400 mg on days 1 and 21) twice daily and 50 mg (100 mg on days 1 and 21) twice daily for CYP2C19 EM and PM subjects, respectively. On coadministration, voriconazole AUC and Cmin decreased by 33% (90%CI, 22%-42%) and 39% (90%CI, 28%-49%), respectively, in CYP2C19 EMs, whereas voriconazole Cmax and AUC increased 4.4-fold (90%CI, 3.6-fold to 5.4-fold) and 5.6-fold (90%CI, 4.5-fold to 7.0-fold), respectively, in PMs. Adding voriconazole resulted in a 20%-30% decrease in atazanavir Cmin in both EMs and PMs. Ritonavir exposure was generally unchanged in either population. The safety and tolerability profiles of the combination were comparable with atazanavir/ritonavir and voriconazole administered alone. The most frequent adverse events with voriconazole were visual disturbance and headache. Coadministration of voriconazole and atazanavir/ritonavir is not recommended unless the benefit/risk to the patient justifies the use of the combination.


Assuntos
Antifúngicos/farmacocinética , Sulfato de Atazanavir/farmacocinética , Citocromo P-450 CYP2C19/genética , Inibidores da Protease de HIV/farmacocinética , Ritonavir/farmacocinética , Voriconazol/farmacocinética , Adolescente , Adulto , Antifúngicos/efeitos adversos , Área Sob a Curva , Sulfato de Atazanavir/efeitos adversos , Combinação de Medicamentos , Interações Medicamentosas , Feminino , Genótipo , Inibidores da Protease de HIV/efeitos adversos , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Ritonavir/efeitos adversos , Voriconazol/efeitos adversos , Adulto Jovem
6.
Antimicrob Agents Chemother ; 60(5): 2782-9, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26902761

RESUMO

BMS-663068 is an oral prodrug of the HIV-1 attachment inhibitor BMS-626529, which prevents viral attachment to host CD4(+) T cells by binding to HIV-1 gp120. To guide dose selection for the phase 3 program, pharmacokinetic/pharmacodynamic modeling was performed using data from two phase 2 studies with HIV-1-infected subjects (n = 244). BMS-626529 population pharmacokinetics were described by a two-compartment model with first-order elimination from the central compartment, zero-order release of prodrug from the extended-release formulation into a hypothetical absorption compartment, and first-order absorption into the central compartment. The covariates of BMS-663068 formulation type, lean body mass, baseline CD8(+) T-cell percentage, and ritonavir coadministration were found to be significant contributors to intersubject variability. Exposure-response analyses showed a relationship between the loge-transformed concentration at the end of a dosing interval (Ctau) normalized for the protein binding-adjusted BMS-626529 half-maximal (50%) inhibitory concentration (PBAIC50) and the change in the HIV-1 RNA level from the baseline level after 7 days of BMS-663068 monotherapy. The probability of achieving a decline in HIV-1 RNA level of >0.5 or >1.0 log10 copies/ml as a function of the loge-transformed PBAIC50-adjusted Ctau after 7 days of monotherapy was 99 to 100% and 57 to 73%, respectively, for proposed BMS-663068 doses of 400 mg twice daily (BID), 600 mg BID (not studied in the phase 2b study), 800 mg BID, 600 mg once daily (QD), and 1,200 mg QD. On the basis of a slight advantage in efficacy of BID dosing over QD dosing, similar responses for the 600- and 800-mg BID doses, and prior clinical observations, BMS-663068 at 600 mg BID was predicted to have the optimal benefit-risk profile and selected for further clinical investigation. (The phase 2a proof-of-concept study AI438006 and the phase 2b study AI438011 are registered at ClinicalTrials.gov under numbers NCT01009814 and NCT01384734, respectively.).


Assuntos
Fármacos Anti-HIV/uso terapêutico , Organofosfatos/uso terapêutico , Piperazinas/uso terapêutico , Triazóis/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Esquema de Medicação , Feminino , Infecções por HIV/tratamento farmacológico , Humanos , Concentração Inibidora 50 , Masculino , Pessoa de Meia-Idade , Organofosfatos/administração & dosagem , Organofosfatos/farmacocinética , Piperazinas/administração & dosagem , Piperazinas/farmacocinética , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Pró-Fármacos/uso terapêutico , Triazóis/administração & dosagem , Triazóis/farmacocinética
7.
Clin Pharmacokinet ; 54(12): 1205-22, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26177803

RESUMO

Asunaprevir is a tripeptidic acylsulfonamide inhibitor of the hepatitis C virus (HCV) NS3/4A protease. Asunaprevir undergoes rapid absorption, with a time to reach maximum plasma concentration (T max) of 2-4 h and an elimination half-life (t ½) of ≈15-20 h observed in single-ascending dose studies. Steady state was achieved by day 7 in multiple-ascending dose studies. The large food effect observed with earlier formulations was mitigated by the soft-gel capsule. Asunaprevir demonstrates high apparent oral clearance and minimal renal elimination, and is eliminated primarily via cytochrome P450 (CYP) 3A4-mediated hepatic oxidative metabolism and faecal excretion. Highly preferential distribution of asunaprevir to the liver occurs via organic anion-transporting polypeptide (OATP)-mediated transport and results in low plasma concentrations. The condition of the liver affects disposition, as higher asunaprevir plasma exposures are observed in patients infected with HCV and/or hepatic impairment. Japanese patients also have higher exposure relative to North American/European patients, but comparable safety at the registrational dose. Asunaprevir has a low potential to perpetrate drug-drug interactions via CYP3A4, P-glycoprotein and OATP, but is a moderate CYP2D6 inhibitor; concomitant drugs that are substrates of CYP2D6 or P-glycoprotein and have a narrow therapeutic index should be used with care. Asunaprevir plasma exposure is strongly affected by inhibitors of OATP transport. No clinically significant interactions were observed between asunaprevir and daclatasvir or daclatasvir/beclabuvir. Asunaprevir has a complex pharmacokinetic profile and forms part of potent and well-tolerated all-oral regimens for the treatment of chronic HCV infection.


Assuntos
Antivirais/farmacologia , Antivirais/farmacocinética , Isoquinolinas/farmacologia , Isoquinolinas/farmacocinética , Sulfonamidas/farmacologia , Sulfonamidas/farmacocinética , Animais , Antivirais/administração & dosagem , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/metabolismo , Humanos , Isoquinolinas/administração & dosagem , Inibidores de Proteases/administração & dosagem , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/farmacologia , Sulfonamidas/administração & dosagem , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/uso terapêutico
8.
Clin Drug Investig ; 35(5): 281-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25896946

RESUMO

BACKGROUND AND OBJECTIVE: Chronic hepatitis C virus (HCV) infection is a major cause of liver transplantation. Drug-drug interactions (DDIs) with cyclosporine and tacrolimus hindered the use of first-generation protease inhibitors in transplant recipients. The current study investigated DDIs between daclatasvir-a pan-genotypic HCV NS5A inhibitor with clinical efficacy in multiple regimens (including all-oral)-and cyclosporine or tacrolimus in healthy subjects. METHODS: Healthy fasted subjects (aged 18-49 years; body mass index 18-32 kg/m(2)) received single oral doses of cyclosporine 400 mg on days 1 and 9, and daclatasvir 60 mg once daily on days 4-11 (group 1, n = 14), or a single oral dose of tacrolimus 5 mg on days 1 and 13, and daclatasvir 60 mg once daily on days 8-19 (group 2, n = 14). Blood samples for pharmacokinetic analysis [by liquid chromatography with tandem mass spectrometry (LC-MS/MS)] were collected on days 1 and 9 for cyclosporine (72 h), on days 1 and 13 for tacrolimus (168 h) and on days 8 and 9 (group 1) or on days 12 and 13 (group 2) for daclatasvir (24 h). Plasma concentrations were determined by validated LC-MS/MS methods. RESULTS: Daclatasvir did not affect the pharmacokinetic parameters of cyclosporine or tacrolimus, and tacrolimus did not affect the pharmacokinetic parameters of daclatasvir. Co-administration of cyclosporine resulted in a 40 % increase in the area under the concentration-time curve of daclatasvir but did not affect its maximum observed concentration. CONCLUSION: On the basis of these observations in healthy subjects, no clinically relevant DDIs between daclatasvir and cyclosporine or tacrolimus are anticipated in liver transplant recipients infected with HCV; dose adjustments during co-administration are unlikely to be required.


Assuntos
Ciclosporina/administração & dosagem , Ciclosporina/farmacocinética , Imidazóis/administração & dosagem , Imidazóis/farmacocinética , Tacrolimo/administração & dosagem , Tacrolimo/farmacocinética , Adolescente , Adulto , Carbamatos , Ciclosporina/efeitos adversos , Ciclosporina/sangue , Interações Medicamentosas , Feminino , Voluntários Saudáveis , Humanos , Imidazóis/efeitos adversos , Imidazóis/sangue , Imunossupressores/administração & dosagem , Imunossupressores/efeitos adversos , Imunossupressores/farmacocinética , Masculino , Pessoa de Meia-Idade , Inibidores de Proteases/administração & dosagem , Inibidores de Proteases/efeitos adversos , Inibidores de Proteases/sangue , Inibidores de Proteases/farmacocinética , Pirrolidinas , Tacrolimo/efeitos adversos , Tacrolimo/sangue , Valina/análogos & derivados , Adulto Jovem
9.
Int J Clin Pharmacol Ther ; 53(4): 292-302, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25740262

RESUMO

OBJECTIVES: Assess the safety, tolerability, and pharmacokinetic (PK) profiles of daclatasvir (DCV) and asunaprevir (ASV) in healthy male Japanese subjects. METHODS: AI444-007 and AI447-005 were phase I, double-blind, placebo-controlled, sequential, single-ascending dose (SAD), and multiple-ascending dose (MAD) studies assessing DCV or ASV, respectively. Eight subjects per panel were randomized to study drug or placebo (3 : 1). In the SAD part of each study, subjects received single oral dose DCV 1/10/50/100/200 mg or ASV 200/400/600/900/1,200 mg. In MAD, subjects received 14-day oral multiple dose DCV 1/10/100 mg once-daily or ASV 200/400/600 mg every 12 hours. Serial PK blood sampling occurred from predose to 72-hours postdose or post-last-dose. Safety and tolerability was assessed throughout. RESULTS: 64 (SAD, n = 40; MAD, n = 24) and 65 (SAD, n = 40; MAD, n = 25) subjects were enrolled in AI444-007 and AI447-005, respectively. DCV and ASV were generally well tolerated, with no serious adverse events or clinicallyrelevant changes in vital signs or ECG parameters. Baseline demographic characteristics were comparable across treatment groups in both studies. DCV was readily absorbed, with median tmax of ~ 1 - 2 hours postdose and concentrations declining in a multi-phasic manner. Exposure generally increased dose-proportionally within dose-range studied. Steady-state was achieved between days 4 and 5 of multiple dosing. ASV was readily absorbed, with median tmax of ~ 2 - 4 hours postdose and concentrations declining in a biphasic manner. Exposure generally increased dose-proportionally within dose-range studied. Steady-state appeared to be achieved between days 3 - 5 of multiple dosing. CONCLUSIONS: Results suggest no clinically significant short-term safety signals with DCV and ASV at single or multiple doses in this population.


Assuntos
Antivirais/administração & dosagem , Antivirais/farmacocinética , Imidazóis/administração & dosagem , Imidazóis/farmacocinética , Isoquinolinas/administração & dosagem , Isoquinolinas/farmacocinética , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacocinética , Administração Oral , Adulto , Antivirais/efeitos adversos , Antivirais/sangue , Povo Asiático , Carbamatos , Método Duplo-Cego , Esquema de Medicação , Voluntários Saudáveis , Humanos , Imidazóis/efeitos adversos , Imidazóis/sangue , Isoquinolinas/efeitos adversos , Isoquinolinas/sangue , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Pirrolidinas , Sulfonamidas/efeitos adversos , Sulfonamidas/sangue , Valina/análogos & derivados , Adulto Jovem
10.
Antivir Ther ; 20(1): 29-37, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24704773

RESUMO

BACKGROUND: It is necessary to evaluate the impact of hepatic impairment on the pharmacokinetic profile of direct-acting antiviral agents for the treatment of HCV infection. METHODS: In this open-label, parallel group, multiple-dose study subjects (aged 18-70 years with a body mass index <35 kg/m(2)) with mild (n=6), moderate (n=6) and severe hepatic impairment (n=4) received asunaprevir 200 mg twice daily; healthy subjects (n=12) were matched (age, weight, gender) 1:1 to the first 4 subjects in each hepatic impairment group to act as controls. Pharmacokinetic sampling and analyses were performed on days 1 and 7 of dosing. Pharmacokinetic parameters were derived by non-compartmental methods. Geometric mean ratios (GMRs) and 90% CIs were used to assess the impact of hepatic impairment on the pharmacokinetics of asunaprevir, relative to healthy matched controls. RESULTS: Compared with healthy subjects, mild hepatic impairment did not result in meaningful alterations in asunaprevir exposure (day 7 maximal plasma concentration [Cmax] GMR: 0.58 [90% CI 0.35, 0.98]; area under the plasma concentration-time curve in one dosing interval [AUCtau] GMR: 0.79 [90% CI 0.55, 1.15]); clinically significant increases in asunaprevir exposure were observed in subjects with moderate (Cmax GMR: 5.03 [90% CI 2.99, 8.47]; AUCtau GMR: 9.83 [90% CI 6.76, 14.28]) and severe hepatic impairment (Cmax GMR: 22.92 [90% CI 12.57, 41.81]; AUCtau GMR: 32.08 [90% CI 20.84, 49.40]). Correlation between increased asunaprevir exposure and all individual components of the Child-Pugh classification system was observed in subjects with moderate and severe hepatic impairment. CONCLUSIONS: Mild hepatic impairment does not meaningfully affect the pharmacokinetic profile of asunaprevir. The dosing of asunaprevir in patients with moderate-to-severe hepatic impairment is not recommended. Clinicaltrials.gov identifier NCT01019070.


Assuntos
Antivirais/farmacocinética , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Isoquinolinas/farmacocinética , Inibidores de Proteases/farmacocinética , Sulfonamidas/farmacocinética , Adolescente , Adulto , Idoso , Antivirais/uso terapêutico , Área Sob a Curva , Índice de Massa Corporal , Estudos de Casos e Controles , Esquema de Medicação , Feminino , Hepacivirus/fisiologia , Hepatite C Crônica/patologia , Hepatite C Crônica/virologia , Humanos , Isoquinolinas/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/virologia , Masculino , Pessoa de Meia-Idade , Inibidores de Proteases/uso terapêutico , Índice de Gravidade de Doença , Sulfonamidas/uso terapêutico , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo
11.
Pharmacotherapy ; 33(3): 284-94, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23456732

RESUMO

STUDY OBJECTIVE: To investigate the pharmacokinetic and pharmacodynamic relationships of the human immunodeficiency virus (HIV)-protease inhibitor atazanavir (ATV) in the presence and absence of the pharmacokinetic booster ritonavir, utilizing ATV plasma trough concentrations (Ctrough ) and clinical biomarkers of antiviral efficacy and safety over 48 weeks. DESIGN: Randomized, open-label, multicenter, study designed to compare the efficacy and safety of ATV 300 mg plus ritonavir 100 mg (ATV300/r) with that of ATV 400 mg (ATV400). SETTING: Thirty clinic sites across 10 countries in Africa, Europe, North America, and South America. PATIENTS: Patients who were HIV-positive and treatment-naïve. INTERVENTIONS: Randomized to once-daily ATV400 (105 patients) or ATV300/r (95 patients) plus lamivudine and extended-release stavudine. MEASUREMENTS AND MAIN RESULTS: The Ctrough approximately 24 hours after the prior unobserved dose was measured through week 48. Composite Ctrough (i.e., the geometric mean of all trough concentrations over the 48 weeks), population inhibitory quotient ([IQ], i.e., Ctrough divided population estimated protein binding adjusted effective concentration at 90% [EC90 , 14 ng/ml]), composite population IQ (i.e., ATV composite trough divided by population estimated protein binding adjusted EC90 ), HIV RNA, CD4 cell counts, and metabolic and safety parameters were also assessed. For ATV400 and ATV300/r, respectively, geometric mean composite Ctrough (CV%) were 127 (106) ng/ml and 670 (63) ng/ml, geometric mean composite population IQ were 9 and 48, and composite Ctrough values of HIV EC90 or more were achieved in 98% and 100% of patients. High ATV Ctrough was associated with low HIV RNA at week 48; however, 88% of patients had HIV RNA less than 400 copies/ml in the lowest composite Ctrough quartile. There was no clear relationship between ATV Ctrough and changes in CD4 cell count. Increases in total bilirubin or jaundice were associated with higher Ctrough . Modest increases in triglycerides and cholesterol were associated with the addition of ritonavir. CONCLUSION: ATV-containing regimens with or without ritonavir achieved ATV exposures that provide robust antiretroviral efficacy and acceptable tolerability in treatment-naïve patients.


Assuntos
Fármacos Anti-HIV , Infecções por HIV/tratamento farmacológico , Oligopeptídeos , Piridinas , Ritonavir , Adulto , Idoso , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/uso terapêutico , Terapia Antirretroviral de Alta Atividade , Sulfato de Atazanavir , Contagem de Linfócito CD4 , Preparações de Ação Retardada , Relação Dose-Resposta a Droga , Esquema de Medicação , Farmacorresistência Viral , Feminino , Infecções por HIV/sangue , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/administração & dosagem , Oligopeptídeos/farmacocinética , Oligopeptídeos/uso terapêutico , Piridinas/administração & dosagem , Piridinas/farmacocinética , Piridinas/uso terapêutico , RNA Viral/sangue , Ritonavir/administração & dosagem , Ritonavir/farmacocinética , Ritonavir/uso terapêutico , Carga Viral/efeitos dos fármacos
12.
Clin Pharmacol Drug Dev ; 2(4): 316-27, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27121936

RESUMO

Asunaprevir (BMS-650032, ASV) is a potent, selective hepatitis C virus (HCV) NS3 protease inhibitor in clinical evaluation for chronic hepatitis C treatment. ASV pharmacokinetics were evaluated in four single- and multiple-ascending-dose studies in healthy subjects or subjects with HCV genotype 1 infection and in human mass balance and food-effect studies. Median Tmax was 2-4 hours. Although T½ was 14-23 hours, oral clearance was high (302-668 L/h at doses ≥100 mg). Steady state was achieved by Day 7. The ASV dose-exposure relationship was disproportionate at doses <200 mg but largely proportional for Cmax and AUC at clinically relevant doses of 200-600 mg (capsule). Following multiple doses, the accumulation index for AUC[TAU] and Cmin was lower at doses ≥200 mg, suggestive of possible auto-induction. ASV exposure increased when administered as a solution (vs. suspension) or with a high-fat meal; the effect was greater for Cmax than AUC, suggesting a saturable first-pass process, the mechanism of which remains to be defined. The apparent complexities of ASV pharmacokinetics will be further explored; nevertheless, data from these studies and antiviral activity in phase 2a/2b studies support further development. Clinical studies are ongoing with ASV in combination with other antivirals.

14.
J Acquir Immune Defic Syndr ; 36(5): 1034-40, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15247556

RESUMO

Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Phenytoin (PHT) is a CYP3A4 inducer and CYP2C9/CYP2C19 substrate. This study quantified the pharmacokinetic (PK) drug interaction between LPV/RTV and PHT. Open-label, randomized, multiple-dose, PK study in healthy volunteers. Subjects in arm A (n = 12) received LPV/RTV 400/100 mg twice daily (BID) (days 1-10), followed by LPV/RTV 400/100 mg BID + PHT 300 mg once daily (QD) (days 11-22). Arm B (n = 12) received PHT 300 mg QD (days 1-11), followed by PHT 300 mg QD + LPV/RTV 400/100 mg BID (days 12-23). Plasma samples were collected on day 11 and day 22; PK parameters were compared by geometric mean ratio (GMR, day 22:day 11). P values <0.05 were considered significant. Following PHT addition, LPV area under the concentration-time curve (AUC0-12h) decreased from 70.9 +/-37.0 to 49.6 +/- 25.1 microg.h/mL (GMR 0.67, P = 0.011) and C0h decreased from 6.0 +/- 3.2 to 3.6 +/- 2.3 microg/mL (GMR 0.54, P = 0.001). Following LPV/RTV addition, PHT AUC0-24h decreased from 191.0+/-89.2 to 147.8+/-104.5 microg.h/mL (GMR 0.69, P = 0.009) and C0h decreased from 7.0+/-4.0 to 5.3+/-4.1 microg/mL (GMR 0.66, P = 0.033). Concomitant LPV/RTV and PHT use results in a 2-way drug interaction. Phenytoin appears to increase LPV clearance via CYP3A4 induction, which is not offset by the presence of low-dose RTV. LPV/RTV may increase PHT clearance via CYP2C9 induction. Management should be individualized to each patient; dosage or medication adjustments may be necessary.


Assuntos
Anticonvulsivantes/administração & dosagem , Sistema Enzimático do Citocromo P-450/biossíntese , Inibidores da Protease de HIV/administração & dosagem , Fenitoína/administração & dosagem , Pirimidinonas/administração & dosagem , Ritonavir/administração & dosagem , Adulto , Anticonvulsivantes/efeitos adversos , Anticonvulsivantes/farmacocinética , Sistema Enzimático do Citocromo P-450/genética , Interações Medicamentosas , Feminino , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/efeitos adversos , Inibidores da Protease de HIV/farmacocinética , Humanos , Lopinavir , Masculino , Farmacogenética , Fenitoína/efeitos adversos , Fenitoína/farmacocinética , Pirimidinonas/efeitos adversos , Pirimidinonas/farmacocinética , Ritonavir/efeitos adversos , Ritonavir/farmacocinética , Convulsões/complicações , Convulsões/tratamento farmacológico
15.
Pharmacogenetics ; 14(5): 279-84, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15115913

RESUMO

This study utilized cytochrome P450 2D6 (CYP2D6) genotypes to explain variability of desipramine pharmacokinetics in a cohort of non-poor metabolizer individuals. In an interaction study utilizing desipramine as a probe, genotyping for the CYP2D6*3, *4, *5 and *6 alleles was used to screen out CYP2D6 poor metabolizers. Individuals were categorized according to these and additional alleles (CYP2D6*2, *9, *10, *17, *41 and x2). Genotypes of individuals heterozygous for two or three of *2, *17 and *41 alleles were confirmed by molecular haplotyping. Pharmacokinetic parameters of desipramine were analysed according to CYP2D6 category. Molecular haplotyping was necessary to definitively categorize four of 16 individuals. A subject who had unusually high plasma elimination half-time, exposure and metabolic ratios carried an intermediate metabolizer (IM) *9 allele in combination with a non-functional allele. This combination has a population frequency of less than 1 : 200. Individuals with *1/*1, *1/*2 and *2/*2 genotypes had lower than average plasma elimination half-time, exposure and metabolic ratios. For desipramine, additional genotyping of CYP2D6 IM alleles helped define subgroups of the CYP2D6-positive cohort. This suggests that genotyping for IM alleles will aid in interpretation of clinical trials involving CYP2D6 substrates. Due to the diversity of IM alleles, molecular haplotyping may be necessary to fully characterize CYP2D6 genotype-phenotype relationships.


Assuntos
Alelos , Antidepressivos Tricíclicos/farmacocinética , Citocromo P-450 CYP2D6/genética , Desipramina/farmacocinética , Estudos de Coortes , Feminino , Genótipo , Haplótipos , Humanos , Masculino , Fenótipo
16.
AIDS Res Hum Retroviruses ; 19(9): 825-35, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14585213

RESUMO

There is marked variability in the extent to which the three classes of antiretroviral (ARV) drugs bind to plasma proteins (<5 to >99%). Protease inhibitors (PIs), with the exception of indinavir, are more than 90% protein bound, mainly to alpha1-acid glycoprotein (AAG). Efavirenz, a nonnucleoside reverse transcriptase inhibitor (NNRTI), is more than 99% bound, mainly to albumin. Nucleoside reverse transcriptase inhibitors (NRTIs) are not highly protein bound. The pharmacological activity of ARV drugs is dependent on unbound drug entering cells that harbor the human immunodeficiency virus (HIV). There has been concern that changes in protein binding could impact on antiviral activity and management. However, for PIs and NNRTIs, and for many drugs given orally, altered plasma binding would not be expected to influence the average exposure to unbound (active) drug after chronic oral dosing. Nevertheless, there will be a change in the relationship between total and unbound concentrations that will be important if, as part of therapeutic drug monitoring, the total rather than the unbound drug is measured. Measuring drug concentrations that are needed to inhibit different HIV strains (wild type and drug resistant) in vitro could also cause confusion because most methods employ bovine serum in the assay medium, and unbound concentrations are not directly measured. Estimating unbound drug concentrations in human plasma and in incubation media can be highly method dependent and thus may affect the calculated IC50 (the concentration of drug that results in 50% inhibition of viral replication). Because inhibitory quotients (IQs = C(trough)/IC50) are becoming part of pharmacokinetic/pharmacodynamic (PK/PD) analyses of clinical trial data, the strengths and weaknesses of the methods used for the determination of unbound drug concentration in plasma and in vitro systems--ultracentrifugation, ultrafiltration, and equilibrium dialysis--need to be understood. Consensus on standard procedures must be reached. In June 2002, a panel of experts assembled by the Forum for Collaborative HIV Research met in Washington, DC, to review the basic principles of protein binding of ARV drugs, and to discuss the impact that changes in plasma protein binding may have on the PKs and activity of ARV drugs as well as on therapeutic drug monitoring. The purpose of the meeting was to discuss the following topics: (1) basic principles of protein binding and how changes in binding can impact on drug PKs and drug exposure in vivo, (2) variability in plasma protein binding among patients taking ARV drugs, (3) the impact of HIV infection and concomitant diseases on the extent of plasma protein binding, (4) the likelihood of clinically relevant drug interactions at the level of plasma protein binding, (5) the evidence that measuring unbound concentrations of ARV drugs in the plasma of patients gives more meaningful information than total drug concentration and, therefore, should be considered in routine therapeutic drug monitoring of ARV agents, (6) optimal method(s) for measuring the unbound concentration of drugs in vitro (for IC50 determination) and in vivo, and (7) future studies that need to be considered to fully understand the importance of plasma protein binding in therapeutic drug monitoring. This report summarizes the topics discussed at this meeting. It guides the reader through the discussions that allowed the panel to formulate a series of statements regarding the significance of plasma protein binding of ARV drugs when studied in vitro and in vivo. The roundtable participants also identified research priorities that are important for understanding the sources of inter- and intraindividual variability in protein binding in patients. These include obtaining data on unbound as well as on total concentrations in PK studies; looking at variants of AAG and whether they differ in binding affinity; and emphasizing the importance of developing a standard procedure for drug susceptibility assays used to determine IC50 values.


Assuntos
Antirretrovirais/farmacocinética , Animais , Antirretrovirais/farmacologia , Proteínas Sanguíneas/metabolismo , Interações Medicamentosas , Humanos , Taxa de Depuração Metabólica , Ligação Proteica , Distribuição Tecidual
17.
Pediatr Infect Dis J ; 22(3): 216-24, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12634581

RESUMO

BACKGROUND: Lopinavir/ritonavir has demonstrated antiviral activity in the HIV-infected adult. SUBJECTS: The objective of this study was to investigate a liquid coformulation of lopinavir/ritonavir, in combination with reverse transcriptase inhibitors, in HIV-infected children. METHODS: One hundred antiretroviral (ARV)-naive and ARV-experienced, nonnucleoside reverse transcriptase inhibitor-naive children between 6 months and 12 years of age participated in this Phase I/II, open label, multicenter trial. Subjects initially received either 230/57.5 mg/m(2) or 300/75 mg/m(2) lopinavir/ritonavir twice daily; ARV-naive subjects also received stavudine and lamivudine, whereas ARV-experienced subjects also received nevirapine and one or two nucleoside reverse transcriptase inhibitors. Lopinavir/ritonavir pharmacokinetics, safety and efficacy were evaluated. RESULTS: All subjects were escalated to the 300/75 mg/m(2) twice daily dose based on results from an interim pharmacokinetic and safety evaluation. The pharmacokinetics of lopinavir did not appear to be dependent on age when dosing was based on body surface area but were decreased on coadministration with nevirapine. Overall 79% of subjects had HIV RNA levels <400 copies/ml at Week 48 (intent-to-treat: missing = failure). Mean increases in absolute and relative (percent) CD4 counts from baseline to Week 48 were observed in both ARV-naive subjects (404 cells/mm(3); 10.3%) and ARV-experienced subjects (284 cells/mm(3); 5.9%). Only one subject prematurely discontinued the study because of a study drug-related adverse event. CONCLUSIONS: The liquid coformulation of lopinavir/ritonavir demonstrated durable antiviral activity and was safe and well-tolerated after 48 weeks of treatment in HIV-infected children.


Assuntos
Infecções por HIV/tratamento farmacológico , Dose Máxima Tolerável , Pirimidinonas/administração & dosagem , Inibidores da Transcriptase Reversa/administração & dosagem , Ritonavir/administração & dosagem , Administração Oral , Disponibilidade Biológica , Química Farmacêutica , Criança , Pré-Escolar , Intervalos de Confiança , Relação Dose-Resposta a Droga , Esquema de Medicação , Quimioterapia Combinada , Feminino , Seguimentos , Infecções por HIV/diagnóstico , Humanos , Lactente , Lopinavir , Masculino , Análise Multivariada , Pirimidinonas/farmacocinética , RNA Viral/efeitos dos fármacos , Inibidores da Transcriptase Reversa/farmacocinética , Ritonavir/farmacocinética , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento , Carga Viral
18.
Pharm Res ; 19(7): 1009-12, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12180532

RESUMO

PURPOSE: ABT-431 is a chemically stable, poorly soluble prodrug that rapidly converts in vivo to A-86929, a selective dopamine D-1 receptor agonist. This study was designed to evaluate the ability of the AERx pulmonary delivery system to deliver ABT-431 to the systemic circulation via the lung. METHODS: A 60% ethanol formulation of 50 mg/mL ABT-431 was used to prepare unit dosage forms containing 40 microL of formulation. The AERx system was used to generate a fine aerosol bolus from each unit dose that was collected either onto a filter assembly to chemically assay for the emitted dose or in an Andersen cascade impactor for particle size analysis. Plasma samples were obtained for pharmacokinetic analysis after pulmonary delivery and IV dosing of ABT-431 to nine healthy male volunteers. Doses from the AERx system were delivered as a bolus inhalation(s) (1, 2, 4, and 8 mg) and intravenous infusions were given over 1 hr (5 mg). Pharmacokinetic parameters of A-86929 were estimated using noncompartmental analysis. RESULTS: The emitted dose was 1.02 mg (%RSD = 11.0, n = 48). The mass median aerodynamic diameter of the aerosol was 2.9 +/- 0.1 microm with a geometric standard deviation of 1.3 +/- 0.1 (n = 15). Tmax (mean +/- SD) after inhalation ranged from 0.9 +/- 0.6 to 11.5 +/- 2.5. The mean absolute pulmonary bioavailibility (as A-86929) based on emitted dose ranged from 81.9% to 107.4%. CONCLUSIONS: This study demonstrated that the AERx pulmonary delivery system is capable of reproducibly generating fine nearly monodisperse aerosols of a small organic molecule. Aerosol inhalation utilizing the AERx pulmonary delivery system may be an efficient means for systemic delivery of small organic molecules such as ABT-431.


Assuntos
Agonistas de Dopamina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Inaladores Dosimetrados , Piridinas/administração & dosagem , Receptores de Dopamina D1/agonistas , Tetra-Hidronaftalenos/administração & dosagem , Administração por Inalação , Área Sob a Curva , Agonistas de Dopamina/sangue , Agonistas de Dopamina/química , Método Duplo-Cego , Sistemas de Liberação de Medicamentos/instrumentação , Humanos , Masculino , Inaladores Dosimetrados/estatística & dados numéricos , Tamanho da Partícula , Piridinas/sangue , Piridinas/química , Solubilidade , Tetra-Hidronaftalenos/sangue , Tetra-Hidronaftalenos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA