Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Pharmacol Drug Dev ; 11(10): 1165-1176, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35560823

RESUMEN

Ripretinib is a switch control KIT kinase inhibitor approved for treatment of adults with advanced gastrointestinal stromal tumors who received prior treatment with 3 or more kinase inhibitors, including imatinib. Ripretinib and its active metabolite (DP-5439) are cleared mainly via cytochrome P450 enzyme 3A4/5 (CYP3A4/5), and ripretinib solubility is pH-dependent, thus the drug-drug interaction potentials of ripretinib with itraconazole (strong CYP3A inhibitor), rifampin (strong CYP3A inducer), and pantoprazole (proton pump inhibitor) were each evaluated in open-label, fixed-sequence study designs. Overall, 20 participants received ripretinib 50 mg alone and with itraconazole 200 mg once daily, 24 participants received ripretinib 100 mg alone and with rifampin 600 mg once daily, and 25 participants received ripretinib 50 mg alone and with pantoprazole 40 mg once daily. Ripretinib exposure increased with concomitant itraconazole, with geometric least-squares (LS) mean ratios of ripretinib area under the concentration-time curve from 0 to ∞ (AUC0-∞ ) and maximum observed concentration (Cmax ) of 199% and 136%. Ripretinib exposure decreased with concomitant rifampin: geometric LS mean ratios for ripretinib AUC0-∞ and Cmax were 39% and 82%. Pantoprazole coadministration had no effect on ripretinib pharmacokinetics. No unexpected safety signals occurred. No dose adjustment is required for ripretinib coadministered with gastric acid reducers and strong CYP3A inhibitors; patients also receiving strong CYP3A inhibitors should be monitored more frequently for adverse reactions. Concomitant ripretinib use with strong CYP3A inducers should be avoided. Prescribers should refer to approved labeling for specific dose recommendations with concomitant use of strong and moderate CYP3A inducers.


Asunto(s)
Inductores del Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A , Inhibidores de Proteínas Quinasas , Adulto , Quimioterapia Combinada/efectos adversos , Ácido Gástrico/metabolismo , Humanos , Mesilato de Imatinib , Itraconazol/farmacología , Naftiridinas/farmacocinética , Pantoprazol , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de la Bomba de Protones , Rifampin , Urea/análogos & derivados
2.
Clin Pharmacol Ther ; 109(4): 867-891, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33555032

RESUMEN

Model-informed drug development (MIDD) has a long and rich history in infectious diseases. This review describes foundational principles of translational anti-infective pharmacology, including choice of appropriate measures of exposure and pharmacodynamic (PD) measures, patient subpopulations, and drug-drug interactions. Examples are presented for state-of-the-art, empiric, mechanistic, interdisciplinary, and real-world evidence MIDD applications in the development of antibacterials (review of minimum inhibitory concentration-based models, mechanism-based pharmacokinetic/PD (PK/PD) models, PK/PD models of resistance, and immune response), antifungals, antivirals, drugs for the treatment of global health infectious diseases, and medical countermeasures. The degree of adoption of MIDD practices across the infectious diseases field is also summarized. The future application of MIDD in infectious diseases will progress along two planes; "depth" and "breadth" of MIDD methods. "MIDD depth" refers to deeper incorporation of the specific pathogen biology and intrinsic and acquired-resistance mechanisms; host factors, such as immunologic response and infection site, to enable deeper interrogation of pharmacological impact on pathogen clearance; clinical outcome and emergence of resistance from a pathogen; and patient and population perspective. In particular, improved early assessment of the emergence of resistance potential will become a greater focus in MIDD, as this is poorly mitigated by current development approaches. "MIDD breadth" refers to greater adoption of model-centered approaches to anti-infective development. Specifically, this means how various MIDD approaches and translational tools can be integrated or connected in a systematic way that supports decision making by key stakeholders (sponsors, regulators, and payers) across the entire development pathway.


Asunto(s)
Antiinfecciosos/farmacología , Desarrollo de Medicamentos/organización & administración , Modelos Biológicos , United States Food and Drug Administration/organización & administración , Antibacterianos/farmacología , Antiinfecciosos/farmacocinética , Antifúngicos/farmacología , Antimaláricos/farmacología , Antituberculosos/farmacología , Antivirales/farmacología , Peso Corporal , Relación Dosis-Respuesta a Droga , Aprobación de Drogas/organización & administración , Descubrimiento de Drogas/organización & administración , Farmacorresistencia Microbiana/efectos de los fármacos , Farmacorresistencia Microbiana/fisiología , Humanos , Inmunidad/fisiología , Ivermectina/uso terapéutico , Pruebas de Función Renal , Pruebas de Función Hepática , Pruebas de Sensibilidad Microbiana , Oncocercosis Ocular/tratamiento farmacológico , Pediatría , Proyectos de Investigación , Estados Unidos , United States Food and Drug Administration/normas
3.
Clin Lung Cancer ; 16(2): 92-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25458558

RESUMEN

INTRODUCTION: New treatment options are needed for second-line therapy in patients with NSCLC. PATIENTS AND METHODS: This was a phase Ib/II study in patients with nonsquamous NSCLC in whom 1 previous platinum-based chemotherapy regimen had failed. Fifteen patients were enrolled in a dose escalation of eribulin mesylate in combination with pemetrexed (E+P). In phase II (n = 80), E+P at the maximum tolerated dose was compared with P. RESULTS: In phase Ib, the maximum tolerated dose of E+P was defined as eribulin 0.9 mg/m(2) with pemetrexed (500 mg/m(2)) each on day 1 of a 21-day cycle. In phase II, adverse events were comparable between groups. PFS and OS were similar between treatment groups. Median PFS was 21.4 weeks for E+P (n = 26; 95% confidence interval [CI], 12.7-39.6) and 23.4 weeks for P (n = 29; 95% CI, 17.1-29.9), with a hazard ratio of 1.0 (95% CI, 0.6-1.7). CONCLUSION: During phase Ib, E+P was tolerated only at a markedly lower dosing intensity relative to the eribulin monotherapy regimen approved for breast cancer and used in phase II studies of NSCLC. At the selected phase II dosing regimen, E+P was generally safe and well tolerated but provided no therapeutic advantage for the second-line treatment of locally advanced or metastatic nonsquamous NSCLC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/patología , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Furanos/administración & dosificación , Glutamatos/administración & dosificación , Guanina/administración & dosificación , Guanina/análogos & derivados , Humanos , Cetonas/administración & dosificación , Neoplasias Pulmonares/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Pemetrexed , Tasa de Supervivencia , Resultado del Tratamiento
4.
Antimicrob Agents Chemother ; 55(11): 5178-84, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21896909

RESUMEN

Zanamivir serum and pulmonary pharmacokinetics were characterized following intravenous (i.v.) or oral inhaled administration. I.v. zanamivir was given as intermittent doses of 100 mg, 200 mg, and 600 mg every 12 h (q12h) for two doses or as a continuous infusion (6-mg loading dose followed by 3 mg/h for 12 h). Oral inhaled zanamivir (two 5-mg inhalations q12h for two doses) was evaluated as well. Each zanamivir regimen was administered to six healthy subjects with serial pharmacokinetic sampling. In addition, a single bronchoalveolar lavage (BAL) fluid sample was collected at various time points and used to calculate epithelial lining fluid (ELF) drug concentrations for each subject. For intermittent i.v. administration of 100 mg, 200 mg, and 600 mg zanamivir, the median zanamivir concentrations in ELF collected 12 h after dosing were 74, 146, and 419 ng/ml, respectively, each higher than the historic mean 50% inhibitory concentrations for the neuraminidases of wild-type strains of influenza A and B viruses. Median ELF/serum zanamivir concentration ratios ranged from 55 to 79% for intermittent i.v. administration (when sampled 12 h after the last dose) and 43 to 45% for continuous infusion (when sampled 6 to 12 h after the start of the infusion). For oral inhaled zanamivir, the median zanamivir concentrations in ELF were 891 ng/ml for the first BAL fluid collection and 326 ng/ml for subsequent BAL fluid collections (when sampled 12 h after the last dose); corresponding serum drug concentrations were undetectable. This study demonstrates zanamivir's penetration into the human pulmonary compartment and supports the doses selected for the continuing development of i.v. zanamivir in clinical studies of influenza.


Asunto(s)
Pulmón/metabolismo , Zanamivir/administración & dosificación , Zanamivir/farmacocinética , Administración por Inhalación , Administración Oral , Adolescente , Adulto , Líquido del Lavado Bronquioalveolar/química , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Antimicrob Agents Chemother ; 52(2): 534-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18056271

RESUMEN

Rifabutin (RFB) is administered for treatment of tuberculosis and Mycobacterium avium complex infection, including use for patients coinfected with human immunodeficiency virus (HIV). Increased systemic exposure to RFB and its equipotent active metabolite, 25-O-desacetyl-RFB (dAc-RFB), has been reported during concomitant administration of CYP3A4 inhibitors, including ritonavir (RTV), lopinavir, and amprenavir (APV); therefore, a reduction in the RFB dosage is recommended when it is coadministered with these protease inhibitors. Fosamprenavir (FPV), the phosphate ester prodrug of the HIV type 1 protease inhibitor APV, is administered either with or without RTV. A randomized, open-label, two-period, two-sequence, balanced, crossover drug interaction study was conducted with 22 healthy adult subjects to compare steady-state plasma RFB pharmacokinetic parameters during concomitant administration of FPV-RTV (700/100 mg twice a day [BID]) with a 75%-reduced RFB dose (150 mg every other day [QOD]) to the standard RFB regimen (300 mg once per day [QD]) by geometric least-squares mean ratios. Relative to results with RFB (300 mg QD), coadministration of dose-adjusted RFB with FPV-RTV resulted in an unchanged RFB area under the concentration-time curve for 0 to 48 h (AUC(0-48)) and a 14% decrease in the maximum concentration of drug in plasma (C(max)), whereas the AUC(0-48) and C(max) of dAc-RFB were increased by 11- and 6-fold, respectively, resulting in a 64% increase in the total antimycobacterial AUC(0-48). Relative to historical controls, the plasma APV AUC from 0 h to the end of the dosing interval (AUC(0-tau)) and C(max) were increased approximately 35%, and the concentration at the end of the dosing interval at steady state was unchanged following coadministration of RFB with FPV-RTV. The safety profile of the combination of RFB and FPV-RTV was consistent with previously described events with RFB or FPV-RTV alone. Based on the results of this study, a reduction in the RFB dose by > or =75% (to 150 mg QOD or three times per week) is recommended when it is coadministered with FPV-RTV (700/100 mg BID).


Asunto(s)
Antituberculosos/farmacocinética , Carbamatos/farmacocinética , Organofosfatos/farmacocinética , Rifabutina/farmacocinética , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Adulto , Antituberculosos/efectos adversos , Antituberculosos/sangre , Área Bajo la Curva , Carbamatos/efectos adversos , Carbamatos/sangre , Estudios Cruzados , Esquema de Medicación , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Furanos , Humanos , Masculino , Persona de Mediana Edad , Organofosfatos/efectos adversos , Organofosfatos/sangre , Rifabutina/efectos adversos , Rifabutina/sangre , Ritonavir/efectos adversos , Ritonavir/sangre , Sulfonamidas/efectos adversos , Sulfonamidas/sangre
6.
Antimicrob Agents Chemother ; 51(8): 2982-4, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17517848

RESUMEN

Plasma ketoconazole (KETO), amprenavir (APV), and ritonavir (RTV) pharmacokinetics were evaluated in 15 healthy subjects after being treated with KETO at 200 mg once daily (QD), fosamprenavir (FPV)/RTV at 700/100 mg twice daily (BID), and then KETO at 200 mg QD plus FPV/RTV at 700/100 mg BID in this open-label study. The KETO area under the concentration-time curve at steady state was increased 2.69-fold with FPV/RTV. APV exposure was unchanged, and RTV exposure was slightly increased.


Asunto(s)
Antifúngicos/farmacocinética , Carbamatos/farmacocinética , Inhibidores de la Proteasa del VIH/farmacocinética , Cetoconazol/farmacocinética , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Adolescente , Adulto , Antifúngicos/administración & dosificación , Antifúngicos/sangre , Área Bajo la Curva , Carbamatos/administración & dosificación , Carbamatos/sangre , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Furanos , Inhibidores de la Proteasa del VIH/administración & dosificación , Inhibidores de la Proteasa del VIH/sangre , Humanos , Cetoconazol/administración & dosificación , Cetoconazol/sangre , Masculino , Persona de Mediana Edad , Organofosfatos/administración & dosificación , Organofosfatos/sangre , Organofosfatos/farmacocinética , Ritonavir/administración & dosificación , Ritonavir/sangre , Sulfonamidas/administración & dosificación , Sulfonamidas/sangre , Resultado del Tratamiento
7.
Antimicrob Agents Chemother ; 51(2): 560-5, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17088488

RESUMEN

Once-daily (QD) fosamprenavir (FPV) at 1,400 mg boosted with low-dose ritonavir (RTV) at 200 mg is effective when it is used in combination regimens for the initial treatment of human immunodeficiency virus infection. Whether a lower RTV boosting dose (i.e., 100 mg QD) could ensure sufficient amprenavir (APV) concentrations with improved safety/tolerability is unknown. This randomized, two 14-day-period, crossover pharmacokinetic study compared the steady-state plasma APV concentrations, safety, and tolerability of FPV at 1,400 mg QD boosted with either 100 mg or 200 mg of RTV QD in 36 healthy volunteers. Geometric least-square (GLS) mean ratios and the associated 90% confidence intervals (CIs) were estimated for plasma APV maximum plasma concentrations (Cmax), the area under the plasma concentration-time curve over the dosing period (AUC0-tau), and trough concentrations (Ctau) during each dosing period. Equivalence between regimens (90% CIs of GLS mean ratios, 0.80 to 1.25) was observed for the plasma APV AUC0-tau (GLS mean ratio, 0.90 [90% CI, 0.84 to 0.96]) and Cmax (0.97 [90% CI, 0.91 to 1.04]). The APV Ctau was 38% lower with RTV at 100 mg QD than with RTV at 200 mg QD (GLS mean ratio, 0.62 [90% CI, 0.55 to 0.69]) but remained sixfold higher than the protein-corrected 50% inhibitory concentration for wild-type virus, with the lowest APV Ctau observed during the 100-mg QD period being nearly threefold higher. The GLS mean APV Ctau was 2.5 times higher than the historical Ctau for unboosted FPV at 1,400 mg twice daily. Fewer clinical adverse drug events and smaller increases in triglyceride levels were observed with the RTV 100-mg QD regimen. Clinical trials evaluating the efficacy and safety of FPV at 1,400 mg QD boosted by RTV at 100 mg QD are now under way with antiretroviral therapy-naïve patients.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Carbamatos/farmacocinética , Organofosfatos/farmacocinética , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Adolescente , Adulto , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/efectos adversos , Fármacos Anti-VIH/sangre , Terapia Antirretroviral Altamente Activa , Carbamatos/administración & dosificación , Carbamatos/efectos adversos , Carbamatos/sangre , Femenino , Furanos , Infecciones por VIH/sangre , Infecciones por VIH/tratamiento farmacológico , Humanos , Masculino , Persona de Mediana Edad , Organofosfatos/administración & dosificación , Organofosfatos/efectos adversos , Profármacos/administración & dosificación , Profármacos/efectos adversos , Profármacos/farmacocinética , Ritonavir/administración & dosificación , Ritonavir/efectos adversos , Sulfonamidas/administración & dosificación , Sulfonamidas/efectos adversos , Sulfonamidas/sangre
8.
Antimicrob Agents Chemother ; 50(9): 3157-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16940117

RESUMEN

Fosamprenavir (FPV) with and without ritonavir (RTV) was added to the antiretroviral regimens of human immunodeficiency virus-infected subjects receiving nevirapine (NVP) to evaluate this drug interaction. Significant reductions in plasma amprenavir exposure (25 to 35%) were observed following coadministration of 1,400 mg of FPV twice a day (BID) and 200 mg of NVP BID. A regimen of 700 mg of FPV BID plus 100 mg of RTV BID may be coadministered with NVP without dose adjustment.


Asunto(s)
Fármacos Anti-VIH/farmacología , Carbamatos/farmacología , Infecciones por VIH/tratamiento farmacológico , VIH , Nevirapina/farmacología , Organofosfatos/farmacología , Ritonavir/farmacología , Sulfonamidas/farmacología , Adulto , Interacciones Farmacológicas , Femenino , Furanos , Inhibidores de la Proteasa del VIH/farmacología , Humanos , Masculino , Inhibidores de la Transcriptasa Inversa/farmacología
9.
J Acquir Immune Defic Syndr ; 42(1): 61-7, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16639341

RESUMEN

OBJECTIVES: To evaluate the drug interaction between fosamprenavir (FPV) and esomeprazole (ESO) after repeated doses in healthy adults. METHODS: Subjects received ESO 20 mg once daily (qd) for 7 days followed by either ESO 20 mg qd + FPV 1400 mg twice daily (bid) or ESO 20 mg qd + FPV 700 mg bid + ritonavir (RTV) 100 mg bid for 14 days in arms 1 and 2, respectively. After a 21- to 28-day washout, subjects received either FPV 1400 mg bid for 14 days (arm 1) or FPV 700 mg bid + RTV 100 mg bid for 14 days (arm 2). Pharmacokinetic sampling was conducted on the last day of each treatment. RESULTS: Simultaneous coadministration of ESO 20 mg qd with either FPV 1400 mg bid or FPV 700 mg bid + RTV 100 mg bid had no effect on steady-state amprenavir pharmacokinetics. The only effect on plasma ESO exposure was a 55% increase in area under the plasma concentration-time curve during a dosing interval, tau[AUC0-tau], after coadministration of ESO 20 mg qd with FPV 1400 mg bid. CONCLUSIONS: FPV 1400 mg bid or FPV 700 mg bid + RTV 100 mg bid may be coadministered simultaneously with ESO without dose adjustment. However, the impact of staggered administration of proton pump inhibitors (PPI) on plasma amprenavir exposure is unknown at present.


Asunto(s)
Carbamatos/administración & dosificación , Carbamatos/farmacocinética , Esomeprazol/administración & dosificación , Organofosfatos/administración & dosificación , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacocinética , Administración Oral , Adolescente , Adulto , Carbamatos/efectos adversos , Carbamatos/sangre , Diarrea/inducido químicamente , Esquema de Medicación , Combinación de Medicamentos , Esomeprazol/efectos adversos , Femenino , Furanos , Cefalea/inducido químicamente , Humanos , Masculino , Persona de Mediana Edad , Náusea/inducido químicamente , Organofosfatos/efectos adversos , Ritonavir/administración & dosificación , Ritonavir/efectos adversos , Sulfonamidas/efectos adversos , Sulfonamidas/sangre
10.
Antimicrob Agents Chemother ; 50(4): 1578-80, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16569890

RESUMEN

To compare the effect of ritonavir on plasma amprenavir pharmacokinetics, healthy adults received either fosamprenavir (700 mg twice a day [BID]) or amprenavir (600 mg BID) alone and in combination with ritonavir (100 mg BID). Ritonavir increased plasma amprenavir pharmacokinetic parameters to a similar extent when coadministered with either fosamprenavir or amprenavir.


Asunto(s)
Carbamatos/farmacología , Inhibidores de la Proteasa del VIH/farmacología , Organofosfatos/farmacocinética , Ritonavir/farmacología , Sulfonamidas/farmacología , Sulfonamidas/farmacocinética , Adulto , Área Bajo la Curva , Carbamatos/administración & dosificación , Estudios Cruzados , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Furanos , Humanos , Masculino , Persona de Mediana Edad , Organofosfatos/administración & dosificación , Ritonavir/administración & dosificación , Sulfonamidas/administración & dosificación
11.
Antimicrob Agents Chemother ; 50(3): 928-34, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16495253

RESUMEN

High-dose combinations of fosamprenavir (FPV) and ritonavir (RTV) were evaluated in healthy adult subjects in order to select doses for further study in multiple protease inhibitor (PI)-experienced patients infected with human immunodeficiency virus type 1. Two high-dose regimens, FPV 1,400 mg twice a day (BID) plus RTV 100 mg BID and FPV 1,400 mg BID plus RTV 200 mg BID, were planned to be compared to the approved regimen, FPV 700 mg BID plus RTV 100 mg BID, in a randomized three-period crossover study. Forty-two healthy adult subjects were enrolled, and 39 subjects completed period 1. Due to marked hepatic transaminase elevations, predominantly with FPV 1,400 mg BID plus RTV 200 mg BID, the study was terminated prematurely. For FPV 1,400 mg BID plus RTV 100 mg BID, the values for plasma amprenavir (APV) area under the concentration-time profile over the dosing interval (tau) at steady state [AUC(0-tau)], maximum concentration of drug in plasma (C(max)), and plasma concentration at the end of tau at steady state (C(tau)) were 54, 81, and 26% higher, respectively, and the values for plasma RTV AUC(0-tau), C(max), and C(tau) were 49% higher, 71% higher, and 11% lower, respectively, than those for FPV 700 mg BID plus RTV 100 mg BID. For FPV 1,400 mg BID plus RTV 200 mg BID, the values for plasma APV AUC(0-tau), C(max), and C(tau) were 26, 48, and 32% higher, respectively, and the values for plasma RTV AUC(0-tau), C(max), and C(tau) increased 4.15-fold, 4.17-fold, and 3.99-fold, respectively, compared to those for FPV 700 mg BID plus RTV 100 mg BID. FPV 1,400 mg BID plus RTV 200 mg BID is not recommended due to an increased rate of marked hepatic transaminase elevations and lack of pharmacokinetic advantage. FPV 1,400 mg BID plus RTV 100 mg BID is currently under clinical evaluation in multiple PI-experienced patients.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacocinética , Organofosfatos/farmacocinética , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Adolescente , Adulto , Carbamatos , Ensayos Clínicos como Asunto , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Furanos , Inhibidores de la Proteasa del VIH/efectos adversos , Inhibidores de la Proteasa del VIH/sangre , Seronegatividad para VIH , Humanos , Masculino , Persona de Mediana Edad , Organofosfatos/efectos adversos , Organofosfatos/sangre , Ritonavir/efectos adversos , Ritonavir/sangre , Sulfonamidas/efectos adversos , Sulfonamidas/sangre
12.
Clin Pharmacokinet ; 45(2): 137-68, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16485915

RESUMEN

Fosamprenavir is one of the most recently approved HIV-1 protease inhibitors (PIs) and offers reductions in pill number and pill size, and omits the need for food and fluid requirements associated with the earlier-approved HIV-1 PIs. Three fosamprenavir dosage regimens are approved by the US FDA for the treatment of HIV-1 PI-naive patients, including fosamprenavir 1,400 mg twice daily, fosamprenavir 1,400 mg once daily plus ritonavir 200mg once daily, and fosamprenavir 700 mg twice daily plus ritonavir 100mg twice daily. Coadministration of fosamprenavir with ritonavir significantly increases plasma amprenavir exposure. The fosamprenavir 700 mg twice daily plus ritonavir 100mg twice daily regimen maintains the highest plasma amprenavir concentrations throughout the dosing interval; this is the only approved regimen for the treatment of HIV-1 PI-experienced patients and is the only regimen approved in the European Union. Fosamprenavir is the phosphate ester prodrug of the HIV-1 PI amprenavir, and is rapidly and extensively converted to amprenavir after oral administration. Plasma amprenavir concentrations are quantifiable within 15 minutes of dosing and peak at 1.5-2 hours after fosamprenavir dosing. Food does not affect the absorption of amprenavir following administration of the fosamprenavir tablet formulation; therefore, fosamprenavir tablets may be administered without regard to food intake. Amprenavir has a large volume of distribution, is 90% bound to plasma proteins and is a substrate of P-glycoprotein. With <1% of a dose excreted in urine, the renal route is not an important elimination pathway, while the principal route of amprenavir elimination is hepatic metabolism by cytochrome P450 (CYP) 3A4. Amprenavir is also an inhibitor and inducer of CYP3A4. Furthermore, fosamprenavir is commonly administered in combination with low-dose ritonavir, which is also extensively metabolised by CYP3A4, and is a more potent CYP3A4 inhibitor than amprenavir. This potent CYP3A4 inhibition contraindicates the coadministration of certain CYP3A4 substrates and requires others to be co-administered with caution. However, fosamprenavir can be co-administered with many other antiretroviral agents, including drugs of the nucleoside/nucleotide reverse transcriptase inhibitor, non-nucleoside reverse transcriptase inhibitor and HIV entry inhibitor classes. Coadministration with other HIV-1 PIs continues to be studied.The extensive fosamprenavir and amprenavir clinical drug interaction information provides guidance on how to co-administer fosamprenavir and fosamprenavir plus ritonavir with many other commonly co-prescribed medications, such as gastric acid suppressants, HMG-CoA reductase inhibitors, antibacterials and antifungal agents.


Asunto(s)
Carbamatos/farmacocinética , Interacciones Farmacológicas , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/farmacocinética , VIH-1 , Organofosfatos/farmacocinética , Profármacos/farmacocinética , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Sulfonamidas/farmacocinética , Adulto , Carbamatos/sangre , Carbamatos/metabolismo , Furanos , Infecciones por VIH/metabolismo , Inhibidores de la Proteasa del VIH/efectos adversos , Inhibidores de la Proteasa del VIH/metabolismo , Semivida , Humanos , Absorción Intestinal , Organofosfatos/metabolismo , Organofosfatos/uso terapéutico , Profármacos/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacocinética , Inhibidores de la Transcriptasa Inversa/farmacología , Sulfonamidas/sangre , Sulfonamidas/metabolismo , Sulfonamidas/uso terapéutico , Distribución Tisular
13.
J Clin Pharmacol ; 44(3): 293-304, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14973306

RESUMEN

Twelve methadone-maintained HIV-negative subjects were given saquinavir/ritonavir (SQV/rtv) 1600 mg/100 mg once daily for 14 days. Pharmacokinetic evaluations of total and unbound methadone enantiomers (R and S) were conducted before and after SQV/rtv. SQV/rtv was well tolerated, with no ACTG Grade 3-4 adverse events, no evidence of sedation, and no changes in methadone dose. For R-methadone (active isomer), C(max), AUC(0-24 h), and C(min) were unchanged, but percent unbound 4 hours after dosing was reduced by 12%. For S-methadone, no differences in pharmacokinetic parameters of total drug were seen, but unbound concentrations were reduced by 15% and 21% at 4 and 24 hours after dosing, respectively. SQV trough concentrations exceeded the anticipated EC(50) (50 ng/mL) in 10/12 subjects, persisting for at least 6 hours after the final dose in 4/6 subjects. Once-daily SQV/rtv in methadone-maintained subjects is safe and not associated with any clinically significant interaction with methadone during 14 days of concomitant administration.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacología , Metadona/farmacocinética , Narcóticos/farmacocinética , Ritonavir/farmacología , Saquinavir/farmacología , Adulto , Cromatografía Liquida , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Interacciones Farmacológicas , Inhibidores de la Proteasa del VIH/sangre , Humanos , Masculino , Espectrometría de Masas , Tasa de Depuración Metabólica , Metadona/sangre , Metadona/química , Persona de Mediana Edad , Narcóticos/sangre , Narcóticos/química , Ritonavir/sangre , Saquinavir/sangre , Estereoisomerismo , Factores de Tiempo
14.
Pharmacotherapy ; 23(7): 835-42, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12885096

RESUMEN

STUDY OBJECTIVES: To determine the effects of concurrent, single doses of didanosine (both buffered and encapsulated enteric-coated bead formulations) on amprenavir steady-state pharmacokinetics, and to determine the effect of staggered dosing of the buffered formulation. DESIGN: Two-period, single-sequence, prospective, open-label drug interaction study with a 10-day washout interval. SETTING: Clinical research unit. SUBJECTS: Sixteen healthy volunteers without human immunodeficiency virus infection. INTERVENTION: Amprenavir 600 mg twice/day was given for the first 4 days of each treatment period, with 12-hour pharmacokinetic evaluations conducted on the last 2 days of each period. Amprenavir was administered according to the following sequential treatments (all fasting): amprenavir alone, concurrent with buffered didanosine, 1 hour before buffered didanosine, and concurrent with the encapsulated enteric-coated bead formulation of didanosine. MEASUREMENTS AND MAIN RESULTS: Plasma was collected 0, 1, 2, 3, 4, 6, 8, and 12 hours after dosing and assayed for amprenavir by using high-performance liquid chromatography. Noncompartmental pharmacokinetic parameters were determined. Geometric mean ratios for each treatment relative to amprenavir alone were determined and reported with 90% confidence intervals (CIs). No significant trends were noted in predose concentrations measured during either period. Area under the concentration-time curve during one 12-hour dosing interval (AUC12) was found to be bioequivalent for all treatments. Peak drug concentration (Cmax) was reduced by 15% on average with concurrent administration of buffered didanosine, and bioequivalence was not demonstrated for this parameter. For concurrent enteric-coated didanosine, geometric mean ratios for Cmax and AUC12 were 0.93 and 0.94, respectively. For buffered didanosine given 1 hour after amprenavir, geometric mean ratios were 1.06 and 1.10 for the same parameters, respectively. No differences were observed in 12-hour concentration (C12) with concurrent administration of buffered or enteric-coated didanosine. CONCLUSION: Amprenavir AUC12 and C12 are not significantly affected by concurrent administration of the buffered or enteric-coated formulations of didanosine. Therefore, amprenavir may be administered concurrently with either the buffered or the encapsulated enteric-coated bead formulation of didanosine in the fasting state.


Asunto(s)
Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/farmacocinética , Didanosina/farmacología , Inhibidores de la Transcriptasa Inversa/farmacología , Sulfonamidas/farmacocinética , Administración Oral , Adulto , Fármacos Anti-VIH/sangre , Área Bajo la Curva , Cápsulas , Carbamatos , Estudios Cruzados , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Furanos , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Sulfonamidas/sangre , Comprimidos Recubiertos , Factores de Tiempo
15.
Antimicrob Agents Chemother ; 47(5): 1694-9, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12709342

RESUMEN

To evaluate the pharmacokinetic effect of adding delavirdine mesylate to the antiretroviral regimens of human immunodeficiency virus (HIV)-infected patients stabilized on a full dosage of ritonavir (600 mg every 12 h), 12 HIV-1-infected subjects had delavirdine mesylate (400 mg every 8 h) added to their current antiretroviral regimens for 21 days. Ritonavir pharmacokinetics were evaluated before (day 7) and after (day 28) the addition of delavirdine, and delavirdine pharmacokinetics were evaluated on day 28. The mean values (+/- standard deviations) for the maximum concentration in serum (C(max)) of ritonavir, the area under the concentration-time curve from 0 to 12 h (AUC(0-12)), and the minimum concentration in serum (C(min)) of ritonavir before the addition of delavirdine were 14.8 +/- 6.7 micro M, 94 +/- 36 micro M. h, and 3.6 +/- 2.1 micro M, respectively. These same parameters were increased to 24.6 +/- 13.9 micro M, 154 +/- 83 micro M. h, and 6.52 +/- 4.85 micro M, respectively, after the addition of delavirdine (P is <0.05 for all comparisons). Delavirdine pharmacokinetic parameters in the presence of ritonavir included a C(max) of 23 +/- 16 micro M, an AUC(0-8) of 114 +/- 75 micro M. h, and a C(min) of 9.1 +/- 7.5 micro M. Therefore, delavirdine increases systemic exposure to ritonavir by 50 to 80% when the drugs are coadministered.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Delavirdina/farmacocinética , Infecciones por VIH/tratamiento farmacológico , Ritonavir/farmacocinética , Adulto , Área Bajo la Curva , Hidrocarburo de Aril Hidroxilasas/fisiología , Citocromo P-450 CYP2D6/fisiología , Citocromo P-450 CYP3A , Delavirdina/administración & dosificación , Quimioterapia Combinada , Femenino , Infecciones por VIH/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Oxidorreductasas N-Desmetilantes/fisiología , Ritonavir/administración & dosificación
16.
J Clin Pharmacol ; 43(2): 171-9, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12616670

RESUMEN

To determine the impact of gastric hypoacidity and acidic beverages on delavirdine mesylate pharmacokinetics in HIV-infected subjects, matched subjects with (n = 11) and without (n = 10) gastric hypoacidity received delavirdine 400 mg tid with either water or an acidic beverage (usually orange juice). The pharmacokinetics of delavirdine and its N-desalkyl metabolite were determined over 8 hours after 14 days of each treatment. Gastric pH was measured at baseline and during each pharmacokinetic evaluation. Delavirdine exposure (Cmax, AUC0-->8 h, and Cmin) was approximately 50% lower and the extent of delavirdine metabolism was higher in subjects with gastric hypoacidity. Orange juice produced a lower mean gastric pH compared to water and increased delavirdine absorption by 50% to 70% in subjects with gastric hypoacidity. However, orange juice had a marginal impact on delavirdine exposure in subjects without gastric hypoacidity. HIV-infected subjects with gastric hypoacidity significantly malabsorb delavirdine. Delavirdine administration with acidic beverages improves, but dose not normalize, absorption in these subjects.


Asunto(s)
Delavirdina/farmacocinética , Mucosa Gástrica/metabolismo , Infecciones por VIH/metabolismo , Adulto , Área Bajo la Curva , Bebidas , Citrus , Estudios Cruzados , Delavirdina/metabolismo , Femenino , Mucosa Gástrica/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Absorción Intestinal , Masculino
17.
Clin Drug Investig ; 23(4): 255-61, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-17535038

RESUMEN

OBJECTIVE: In a prior single-dose study that examined the effect of food on delavirdine pharmacokinetics in healthy volunteers, the absorption of delavirdine mesylate was delayed and the area under the curve was reduced by 26% in the presence of food. Since the complex, nonlinear pharmacokinetics of delavirdine do not permit a simple extrapolation of the results of a single-dose study to steady state, the present multiple-dose study was performed. PATIENTS AND STUDY DESIGN: Thirteen stable patients with HIV-1 infection (two females, 11 males; CD4 count range 124-588 cells/mm(3)) completed a randomised, crossover study in which subjects received two 14-day treatments with delavirdine mesylate 400mg every 8 hours. In treatment A, all delavirdine doses were administered on an empty stomach and in treatment B were taken with food. A pharmacokinetic evaluation was performed on day 14 of each treatment period. SETTING: An ambulatory AIDS research centre in an academic medical centre. INTERVENTIONS: Administration of delavirdine with and without food. MAIN OUTCOME MEASURES: Pharmacokinetic parameters for delavirdine. RESULTS: The maximum concentration (C(max)) [+/- standard deviation] in treatment A was 29.6 +/- 13.6muM and in treatment B it was 23.0 +/- 8.61muM (p = 0.037). The minimum concentrations (C(min)) were 9.45 +/- 6.7muM and 11.2 +/- 9.2muM, respectively (p > 0.05). The oral clearances (CL(oral)) were 17.8 +/- 41.6 L/h (treatment A) and 18.5 +/- 39.0 L/h (treatment B) [p > 0.05]. Similar patterns were observed for N-dealkylated delavirdine with a significant difference only in C(max) (4.13 vs 3.47muM [p = 0.022], treatment A vs B). CONCLUSIONS: These findings indicated that, in contrast to the increased CL(oral) noted in a prior single-dose study, food did not have a significant effect at steady state on the area under the plasma concentration-time curve or C(min). Although C(max) was significantly lower when the drug was taken taken with food, the clinical relevance of this parameter as compared with the trough concentration is unclear since the current focus for antiretrovirals is on maintaining trough concentrations in excess of in vitro inhibitory concentrations.

18.
Clin Drug Investig ; 23(5): 323-8, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-17535044

RESUMEN

BACKGROUND: Delavirdine is a non-nucleoside reverse transcriptase inhibitor with pH-dependent absorption characteristics that has received accelerated approval for the treatment of patients with HIV-1 infection. In a prior single-dose study concurrent administration of delavirdine mesylate and didanosine (buffered formulation) resulted in up to a 31% decrease in the area under the plasma delavirdine concentration versus time curve (AUC) compared with when both drugs were taken separately. OBJECTIVE: To evaluate the interaction of these two agents at steady state. STUDY DESIGN AND PATIENTS: A total of 11 HIV-infected subjects who were previously stabilised on didanosine were enrolled into a randomised, open-labelled crossover study. Nine subjects continued to receive their prescribed dose and schedule of didanosine, with each dose of didanosine taken either together with or 1 hour after delavirdine mesylate (400mg every 8 hours). Pharmacokinetic studies at baseline, day 14 and day 28 were conducted and the plasma concentrations of delavirdine and didanosine were determined. RESULTS: A lower delavirdine maximum plasma concentration (C(max)) [22.4 +/- 11 vs 35.5 +/- 17muM; p = 0.045] was noted when delavirdine and didanosine were taken together. However, no significant difference was noted for delavirdine AUC (114 +/- 56 muM.h compared with 153 +/- 79 muM.h [p = 0.181]). In addition, no differences were noted for didanosine pharmacokinetic parameters between treatments. CONCLUSION: These data indicate that patients receiving didanosine and delavirdine as part of a combination regimen during long-term therapy can be instructed to take them together in an attempt to enhance adherence to treatment with both antiretroviral agents.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...