Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arthritis Res Ther ; 24(1): 208, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-36008814

RESUMEN

BACKGROUND: Publications suggest immunomodulation co-therapy improves responder rates in uncontrolled/refractory gout patients undergoing pegloticase treatment. The MIRROR open-label trial showed a 6-month pegloticase + methotrexate co-therapy responder rate of 79%, compared to an established 42% pegloticase monotherapy responder rate. Longer-term efficacy/safety data are presented here. METHODS: Uncontrolled gout patients (serum urate [SU] ≥ 6 mg/dL and SU ≥ 6 mg/dL despite urate-lowering therapy [ULT], ULT intolerance, or functionally-limiting tophi) were included. Patients with immunocompromised status, G6PD deficiency, severe kidney disease, or methotrexate contraindication were excluded. Oral methotrexate (15 mg/week) and folic acid (1 mg/day) were administered 4 weeks before and during pegloticase therapy. Twelve-month responder rate (SU < 6 mg/dL for ≥ 80% during month 12), 52-week change from baseline in SU, and extended safety were examined. Efficacy analyses were performed for patients receiving ≥ 1 pegloticase infusion. Pharmacokinetics (PK)/anti-drug antibodies (ADAs) were examined and related to efficacy/safety findings. RESULTS: Fourteen patients were included (all male, 49.3 ± 8.7 years, 13.8 ± 7.4-year gout history, pre-therapy SU 9.2 ± 2.5 mg/dL). Three patients were non-responders and discontinued study treatment before 24 weeks, one patient exited the study per protocol at 24 weeks (enrolled prior to treatment extension amendment), and 10 remained in the study through week 52. Of the 10, 8 completed 52 weeks of pegloticase + methotrexate and were 12-month responders. The remaining two discontinued pegloticase + methotrexate at week 24 (met treatment goals) and stayed in the study under observation (allopurinol prescribed at physicians' discretion); one remained a responder at 12 months. At 52 weeks, change from baseline in SU was - 8.2 ± 4.1 mg/dL (SU 1.1 ± 2.4 mg/dL, n = 10). Gout flares were common early in treatment but progressively decreased while on therapy (weeks 1-12, 13/14 [92.9%]; weeks 36-52, 2/8 [25.0%]). One patient recovered from sepsis (serious AE). Two non-responders developed high ADA titers; fewer patients had trough concentrations (Cmin) below the quantitation limit (BQL), and the median Cmin was higher (1.03 µg/mL vs. BQL) than pegloticase monotherapy trials. CONCLUSIONS: Pegloticase + methotrexate co-therapy was well-tolerated over 12 months, with sustained SU lowering, progressive gout flare reduction, and no new safety concerns. Antibody/PK findings suggest methotrexate attenuates ADA formation, coincident with higher treatment response rates. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03635957 . Registered on 17 August 2018.


Asunto(s)
Gota , Gota/tratamiento farmacológico , Supresores de la Gota/efectos adversos , Humanos , Masculino , Metotrexato/uso terapéutico , Polietilenglicoles/uso terapéutico , Brote de los Síntomas , Resultado del Tratamiento , Urato Oxidasa/efectos adversos , Ácido Úrico
2.
Clin Pharmacokinet ; 60(8): 1029-1040, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33768488

RESUMEN

BACKGROUND AND OBJECTIVE: Thyroid eye disease (TED) is characterized by inflammation/expansion of orbital tissues, proptosis, and diplopia. Teprotumumab is the first US Food and Drug Administration-approved therapy for TED, administered as an initial intravenous infusion of 10 mg/kg followed by 20 mg/kg every 3 weeks for an additional seven infusions. The objective of this article is to discuss the pharmacokinetics and exposure-response profile for teprotumumab in patients with TED. METHODS: A population pharmacokinetic analysis was performed to characterize pharmacokinetics and select dosing in patients with TED. Exposure-response was evaluated for efficacy (proptosis response, clinical activity score categorical response, and diplopia response) and safety (hyperglycemia, muscle spasms, and hearing impairment) parameters. RESULTS: Teprotumumab pharmacokinetics was linear in patients with TED, with low systemic clearance (0.334 L/day), low volume of distribution (3.9 and 4.2 L for the central and peripheral compartment, respectively), and a long elimination half-life (19.9 days). The approved dosing regimen provided > 20 µg/mL for > 90% insulin-like growth factor 1 receptor saturation throughout the dosing interval. Model-predicted mean (± standard deviation) steady-state area under the concentration-time curve, peak, and trough concentrations in patients with TED were 131 (± 30.9) mg∙h/mL, 643 (± 130) µg/mL, and 157 (± 50.6) µg/mL, respectively. Female patients had a 15% higher steady-state peak concentration but a similar steady-state area under the concentration-time curve vs male patients. No other covariates affected teprotumumab pharmacokinetics. No meaningful correlations between teprotumumab exposures and efficacy or safety parameters were observed. CONCLUSIONS: Teprotumumab pharmacokinetics was well characterized in patients with TED, and generally consistent with other IgG1 antibodies. Efficacy was consistent across the exposure range with a well-tolerated safety profile supporting the current dose regimen for patients with TED.


Asunto(s)
Oftalmopatía de Graves , Factor I del Crecimiento Similar a la Insulina , Anticuerpos Monoclonales Humanizados , Femenino , Oftalmopatía de Graves/tratamiento farmacológico , Humanos , Masculino
3.
Clin Drug Investig ; 37(2): 195-205, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27785737

RESUMEN

BACKGROUND AND OBJECTIVES: Entospletinib is a selective, reversible, adenosine triphosphate-competitive small-molecule spleen tyrosine kinase (SYK) inhibitor that blocks B cell receptor-mediated signaling and proliferation in B lymphocytes. This study evaluated the safety, pharmacokinetics, and pharmacodynamics of entospletinib in a double-blind, single/multiple ascending dose study in healthy volunteers. METHODS: In sequential cohorts, 120 subjects received entospletinib (25-1200 mg; fasted) as single or twice-daily oral doses for 7 days. Along with pharmacokinetics, the study assessed functional inhibition of ex vivo anti-immunoglobulin E-stimulated CD63 expression on basophils and pervanadate-evoked phosphorylated SYK (pSYK) Y525. Safety and tolerability were assessed throughout the study. RESULTS: Entospletinib was generally well-tolerated over a 48-fold dose range. Adverse events (AEs) were generally mild to moderate, with no AE-driven study drug discontinuations noted. Entospletinib displayed a median plasma half-life of 9-15 h; entospletinib exposures reached a plateau at ≥600 mg twice daily (likely due to solubility-limited absorption) and provided >90% CD63 inhibition at peak concentrations and >60% inhibition at trough concentrations (corresponding pSYK inhibition of >70 and >50%). CONCLUSION: The overall safety, pharmacokinetics, and pharmacodynamics profiles of entospletinib support further clinical evaluation.


Asunto(s)
Quinasa Syk/antagonistas & inhibidores , Adolescente , Adulto , Linfocitos B/efectos de los fármacos , Basófilos/efectos de los fármacos , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Interacciones Alimento-Droga , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Tetraspanina 30/biosíntesis , Tetraspanina 30/efectos de los fármacos , Vanadatos/farmacología , Adulto Joven
4.
J Acquir Immune Defic Syndr ; 72(3): 281-8, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26885802

RESUMEN

BACKGROUND: Elvitegravir (EVG), a HIV integrase inhibitor, is metabolized primarily by CYP3A, and secondarily by UGT1A1/3; Efavirenz (EFV), a HIV non-nucleoside reverse transcriptase inhibitor, is metabolized by Cytochrome P450 (CYP) 2B6 and induces CYP3A and uridine diphosphate glucuronosyltransferase (UGT) with residual effects post discontinuation because of long T1/2 (40-55 hours). This study evaluated the pharmacokinetics after switching from efavirenz/emtricitabine/tenofovir disoproxil fumarate (EFV/FTC/TDF) to elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate (EVG/COBI/FTC/TDF). METHODS: Healthy subjects (n = 32 including n = 8 CYP2B6 poor metabolizers) received EVG/COBI/FTC/TDF (150/150/200/300 mg) on days 1-7, and after a washout, received EFV/FTC/TDF (600/200/300 mg) on days 15-28 and switched to EVG/COBI/FTC/TDF (150/150/200/300 mg) for 5 weeks (days 29-62). Pharmacokinetic assessments occurred on days 7, 28, 35, and 42; trough samples (Ctrough) were collected periodically until day 63. Safety was assessed throughout the study. RESULTS: Twenty-nine subjects completed with 3 adverse events leading to discontinuation; no grade ≥3 adverse events were reported. Post-EFV/FTC/TDF, mean EVG area under concentration (AUCtau) was 37% and 29% lower and mean Ctrough ∼3- and ∼5-fold above IC95, respectively, on days 35 and 42, and 7-8-fold above IC95 by 5 weeks. COBI AUCtau returned to normal by day 42. EVG glucuronide, GS-9200, AUCtau was higher (46% and 32% on days 35 and 42, respectively) postswitch. CYP2B6 poor metabolizers displayed higher EFV AUCtau and Cmax (125% and 91%, respectively) versus non-poor metabolizers, and lower EVG and COBI exposures. EFV Ctrough was >IC90 (10 ng/mL) in all subjects postswitch. FTC and tenofovir (TFV) exposures were unaffected. CONCLUSIONS: After EFV/FTC/TDF to EVG/COBI/FTC/TDF switch, EVG and/or EFV exposures were in an active range. These findings support further evaluation of switching regimens in HIV-1 patients.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Combinación Efavirenz, Emtricitabina y Fumarato de Tenofovir Disoproxil/farmacocinética , Combinación Elvitegravir, Cobicistat, Emtricitabina y Fumarato de Tenofovir Disoproxil/farmacocinética , Adolescente , Adulto , Fármacos Anti-VIH/efectos adversos , Combinación de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Combinación Efavirenz, Emtricitabina y Fumarato de Tenofovir Disoproxil/efectos adversos , Combinación Elvitegravir, Cobicistat, Emtricitabina y Fumarato de Tenofovir Disoproxil/efectos adversos , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
5.
J Clin Pharmacol ; 56(6): 723-32, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26449283

RESUMEN

Elvitegravir (EVG) is an HIV strand transfer integrase inhibitor approved for the treatment of HIV infection as a part of antiretroviral regimens containing cobicistat (COBI) or ritonavir (RTV) as a booster. The population pharmacokinetics of EVG in treatment-naive and -experienced HIV patients was determined, and the effects of demographic, biometric, and formulation covariates on EVG pharmacokinetics (PK) were evaluated. Data from 31 clinical studies (25 in healthy subjects, 6 phase 1b to phase 3 in HIV-1-infected patients) with COBI-boosted EVG studies (as EVG/co or EVG/COBI/FTC/TDF single-tablet regimen) or RTV-boosted EVG studies (EVG/r) were analyzed using NONMEM. The effect of the covariates age, sex, race, health status (healthy volunteers vs HIV patients), weight, body mass index (BMI), body surface area (BSA), creatinine clearance (estimated GFR), and formulation were evaluated. EVG PK, with COBI or RTV, was described by a 2-compartment model, with first-order absorption and elimination and an absorption lag time. A statistically significant, but not clinically relevant, effect of BSA on EVG clearance (CL) was observed. Coadministration of atazanavir or lopinavir with EVG/r had an effect on EVG CL consistent with the known interaction with these agents. No other covariate had a meaningful effect on EVG PK. EVG PK was well described in a population PK model with HIV-infected patients, with low PK variability and no relevant effect of demographic or biometric covariates.


Asunto(s)
Infecciones por VIH/sangre , Infecciones por VIH/tratamiento farmacológico , Inhibidores de Integrasa VIH/sangre , Quinolonas/administración & dosificación , Quinolonas/sangre , Adolescente , Adulto , Anciano , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/sangre , Niño , Quimioterapia Combinada , Femenino , Integrasa de VIH/sangre , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
6.
Cancer Chemother Pharmacol ; 77(1): 89-98, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26645408

RESUMEN

PURPOSE: Idelalisib is a potent PI3Kδ inhibitor that was recently approved for treating hematologic malignancies. The objective of this analysis was to develop a population pharmacokinetic model for idelalisib and its inactive metabolite GS-563117 and to evaluate the impact of covariates on idelalisib/GS-563117 PK. METHODS: Data from 10 phase I or II studies in healthy volunteers or patients with hematologic malignancies (n = 736) were analyzed using NONMEM. Stepwise forward addition followed by backward elimination was implemented in the covariate (age, gender, race, body weight, baseline CLcr, AST, ALT, disease status, and type of cancer) model building process. Various model assessment methods were used to evaluate the models. RESULTS: Idelalisib plasma PK was best described by a two-compartment model with first-order absorption, first-order elimination from the central compartment, and a lag time. A nonlinear relationship between dose and relative bioavailability was included in the final model. Two statistically significant covariates were identified and incorporated into the final model: health status (healthy vs. patient) on CL/F and Q/F and body weight on CL/F. Despite being a statistically significant covariate, the effect of body weight on idelalisib exposures was weak, as evidenced by minor changes of steady-state exposure (C trough: 16%; AUC and C max: 10%) for a patient with extreme body weight (5th and 95th percentile) relative to the typical patient, and not considered to be clinically relevant. CONCLUSIONS: PopPK models were developed to adequately describe the plasma concentrations of idelalisib and GS-563117. There were no covariate that had a clinically meaningful impact on idelalisib or GS-563117 exposure.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias Hematológicas/tratamiento farmacológico , Modelos Biológicos , Purinas/farmacocinética , Quinazolinonas/farmacocinética , Antineoplásicos/uso terapéutico , Área Bajo la Curva , Estudios de Casos y Controles , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Femenino , Humanos , Masculino , Dinámicas no Lineales , Inhibidores de las Quinasa Fosfoinosítidos-3 , Purinas/uso terapéutico , Quinazolinonas/uso terapéutico
7.
J Clin Pharmacol ; 55(8): 944-52, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25821156

RESUMEN

Idelalisib, a phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitor, is metabolized primarily by aldehyde oxidase to form GS-563117, an inactive metabolite, and is metabolized to a lesser extent by cytochrome P450 3A and uridine 5'-diphospho-glucuronosyltransferase 1A4. In a mass balance study, the orally administered idelalisib dose was recovered mainly in feces (∼78%). This study evaluated the pharmacokinetics and safety of a single 150-mg dose of idelalisib in subjects with moderate or severe hepatic impairment and in age-, sex-, and weight-matched, healthy controls. The idelalisib maximum observed plasma concentration was generally comparable in subjects with moderate or severe hepatic impairment versus healthy controls, whereas the mean area under the curve was higher (58% to 59%). GS-563117 exposures were lower in impaired versus healthy control subjects, likely because of lower formation in the setting of liver impairment. Exploratory analyses indicated no relevant relationships between idelalisib or GS-563117 plasma exposures and Child-Pugh-Turcotte scores. Single oral doses of idelalisib 150 mg were well tolerated, with most treatment-emergent adverse events (AEs) and laboratory abnormalities being grades 1 or 2 in severity. As such, no dose adjustment was required when initiating idelalisib treatment in patients with mild or moderate hepatic impairment, although close monitoring for potential AEs is recommended.


Asunto(s)
Antineoplásicos/farmacocinética , Hepatopatías/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Purinas/farmacocinética , Quinazolinonas/farmacocinética , Adulto , Anciano , Antineoplásicos/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Unión Proteica , Inhibidores de Proteínas Quinasas/efectos adversos , Purinas/efectos adversos , Quinazolinonas/efectos adversos
8.
J Clin Pharmacol ; 55(8): 909-19, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25760671

RESUMEN

Idelalisib, a potent phosphatidylinositol-3-kinase delta (PI3Kδ) inhibitor, is metabolized primarily by aldehyde oxidase to form GS-563117 and to a lesser extent by cytochrome P450 (CYP) 3A and uridine 5'-diphospho-glucuronosyltransferase 1A4. In vitro, idelalisib inhibits P-glycoprotein (P-gp) and organic anion transporting polypeptides 1B1 and 1B3, and GS-563117 is a time-dependent CYP3A inhibitor. This study enrolled 24 healthy subjects and evaluated (1) the effect of idelalisib on the pharmacokinetics (PK) of digoxin, a P-gp probe substrate, rosuvastatin, a breast cancer resistance protein, and OATP1B1/OATP1B3 substrate, and midazolam, a CYP3A substrate; and (2) the effect of a strong inducer, rifampin, on idelalisib PK. On treatment, the most common clinical adverse events (AEs) were headache and pyrexia. Grade 3 transaminase increases were observed in 5 of 24 subjects and were reversible. Two subjects had serious AEs after treatment completion (grade 3 pyrexia and/or drug-induced liver injury). Idelalisib coadministration did not affect digoxin and rosuvastatin PK. Coadministration with idelalisib increased plasma exposures of midazolam (138% and 437% for maximum observed plasma concentration [Cmax ] and area under the plasma concentration-time curve from time 0 extrapolated to infinity [AUCinf ], respectively), consistent with the in vitro finding of CYP3A inhibition by GS-563117. Rifampin caused a substantial decrease in idelalisib (58% and 75%, Cmax and AUCinf , respectively) and GS-563117 exposures, indicating an enhanced contribution of CYP3A to idelalisib metabolism under a strongly induced state.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Purinas/farmacología , Purinas/farmacocinética , Quinazolinonas/farmacología , Quinazolinonas/farmacocinética , Adulto , Antineoplásicos/sangre , Digoxina/sangre , Digoxina/farmacocinética , Interacciones Farmacológicas , Femenino , Voluntarios Sanos , Humanos , Masculino , Midazolam/sangre , Midazolam/farmacocinética , Persona de Mediana Edad , Inhibidores de las Quinasa Fosfoinosítidos-3 , Purinas/sangre , Quinazolinonas/sangre , Rifampin/farmacología , Rosuvastatina Cálcica/sangre , Rosuvastatina Cálcica/farmacocinética , Adulto Joven
9.
N Engl J Med ; 371(8): 711-22, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25140957

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) is a common cause of infant hospitalizations and is increasingly recognized as a cause of considerable morbidity and mortality. No accepted antiviral treatment exists. METHODS: We conducted a double-blind, placebo-controlled study of GS-5806, an oral RSV-entry inhibitor, in healthy adults who received a clinical challenge strain of RSV intranasally. Participants were monitored for 12 days. At the time of a positive test for RSV infection or 5 days after inoculation, whichever occurred first, participants were randomly assigned to receive GS-5806 or placebo in one of seven sequential cohorts. Cohorts 1 to 4 received a first dose of 50 mg of GS-5806 and then 25 mg daily for the next 4 days, cohort 5 received a first dose of 50 mg and then 25 mg daily for the next 2 days, cohort 6 received one 100-mg dose, and cohort 7 received a first dose of 10 mg and then 5 mg daily for the next 4 days. Dose selection for cohorts 5, 6, and 7 occurred after an interim analysis of data for cohorts 1 to 4. The primary end point was the area under the curve (AUC) for the viral load, which was assessed after administration of the first dose through the 12th day after inoculation. Secondary end points were mucus weight and symptom scores. RESULTS: Among the 54 participants in cohorts 1 to 4 who were infected with RSV, active treatment was associated with a lower viral load (adjusted mean, 250.7 vs. 757.7 log10 plaque-forming-unit equivalents [PFUe] × hours per milliliter; P<0.001), lower total mucus weight (mean, 6.9 g vs. 15.1 g; P=0.03), and a lower AUC for the change from baseline in symptom scores (adjusted mean, -20.2 vs. 204.9 × hours; P=0.005). The results were similar in cohorts 5, 6, and 7. Adverse events, including low neutrophil counts and increased levels of alanine aminotransferase, were more common among participants receiving GS-5806. CONCLUSIONS: Treatment with GS-5806 reduced the viral load and the severity of clinical disease in a challenge study of healthy adults. (Funded by Gilead Sciences; ClinicalTrials.gov number, NCT01756482.).


Asunto(s)
Antivirales/uso terapéutico , Pirazoles/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios , Sulfonamidas/uso terapéutico , Administración Oral , Adolescente , Adulto , Antivirales/efectos adversos , Antivirales/farmacocinética , Área Bajo la Curva , Método Doble Ciego , Femenino , Humanos , Indazoles , Masculino , Pirazoles/efectos adversos , Pirazoles/farmacocinética , Infecciones por Virus Sincitial Respiratorio/virología , Índice de Severidad de la Enfermedad , Sulfonamidas/efectos adversos , Sulfonamidas/farmacocinética , Carga Viral , Adulto Joven
10.
HIV Clin Trials ; 14(5): 216-23, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24144898

RESUMEN

BACKGROUND: Due to ongoing neuropsychiatric adverse events in some efavirenz (EFV)-treated patients, a switch to an alternative non-nucleoside reverse transcriptase inhibitor may be considered. Rilpivirine (RPV) has been coformulated as a single-tablet regimen (STR) with emtricitabine/tenofovir disoproxil fumarate (FTC/TDF), and the components have demonstrated noninferior efficacy to EFV+FTC/TDF, good tolerability profile, and high adherence. After discontinuation, EFV has an extended inductive effect on cytochrome P450 (CYP) 3A4 that, after switching, may reduce RPV exposures and adversely impact clinical outcomes. OBJECTIVE: This study examines the clinical implications of reduced RPV exposures with concomitant FTC/TDF and declining EFV exposures when patients, intolerant to EFV, switch from EFV/FTC/TDF to RPV/FTC/TDF. METHODS: This 48-week, phase 2b, open-label, multicenter study evaluated the efficacy and safety of switching from EFV/FTC/TDF (≥3 months duration) to RPV/FTC/TDF. Virologic suppression (HIV-1 RNA <50 copies/mL), safety, and EFV and RPV pharmacokinetics were assessed. RESULTS: At weeks 12 and 24, all 49 dosed subjects remained suppressed on RPV/FTC/TDF. At week 48, 46 (93.9%) subjects remained suppressed and virologic failure occurred in 2/49 (4.1%) subjects with no emergence of resistance. EFV concentrations were above the 90th percentile for inhibitory concentration (IC90) for several weeks after EFV discontinuation, and RPV exposures were in the range observed in phase 3 studies by approximately 2 weeks post switch. No subjects discontinued the study due to an adverse event. CONCLUSIONS: Switching from EFV/FTC/TDF to RPV/FTC/ TDF was a safe, efficacious option for virologically suppressed HIV-infected patients with EFV intolerance wishing to remain on an STR.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Adenina/administración & dosificación , Adenina/efectos adversos , Adenina/análogos & derivados , Adenina/uso terapéutico , Adulto , Alquinos , Fármacos Anti-VIH/efectos adversos , Benzoxazinas/administración & dosificación , Benzoxazinas/efectos adversos , Benzoxazinas/uso terapéutico , Ciclopropanos , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Antagonismo de Drogas , Emtricitabina , Femenino , Infecciones por VIH/epidemiología , VIH-1/genética , VIH-1/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Nitrilos/administración & dosificación , Nitrilos/efectos adversos , Nitrilos/farmacocinética , Nitrilos/uso terapéutico , Organofosfonatos/administración & dosificación , Organofosfonatos/efectos adversos , Organofosfonatos/uso terapéutico , Pirimidinas/administración & dosificación , Pirimidinas/efectos adversos , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , ARN Viral , Rilpivirina , Tenofovir , Estados Unidos , Carga Viral , Adulto Joven
11.
J Acquir Immune Defic Syndr ; 63(4): 449-55, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23807155

RESUMEN

OBJECTIVE: To evaluate the antiviral activity, safety, pharmacokinetics, and pharmacokinetics/pharmacodynamics of short-term monotherapy with tenofovir alafenamide (TAF), a next-generation tenofovir (TFV) prodrug. DESIGN: A phase 1b, randomized, partially blinded, active- and placebo-controlled, dose-ranging study. METHODS: Treatment-naive and experienced HIV-1-positive adults currently off antiretroviral therapy were randomized to receive 8, 25, or 40 mg TAF, 300 mg tenofovir disoproxil fumarate (TDF), or placebo, each once daily for 10 days. RESULTS: Thirty-eight subjects were enrolled. Baseline characteristics were similar across dose groups. Significant reductions in plasma HIV-1 RNA from baseline to day 11 were observed for all TAF dose groups compared with placebo (P < 0.01), with a median decrease of 1.08-1.73 log10 copies per milliliter, including a dose-response relationship for viral load decrease up to 25 mg. At steady state, 8, 25, and 40 mg TAF yielded mean TFV plasma exposures [area under the plasma concentration-time curve (AUCtau)] of 97%, 86%, and 79% lower, respectively, as compared with the TFV exposures observed with 300 mg TDF. For 25 and 40 mg TAF, the mean intracellular peripheral blood mononuclear cell tenofovir diphosphate AUCtau was ∼7-fold and ∼25-fold higher, relative to 300 mg TDF. CONCLUSIONS: Compared with 300 mg TDF, TAF demonstrated more potent antiviral activity, higher peripheral blood mononuclear cell intracellular tenofovir diphosphate levels, and lower plasma TFV exposures, at approximately 1/10th of the dose. This may translate into greater antiviral efficacy, a higher barrier to resistance, and an improved safety profile relative to TDF, supporting further investigation of TAF dosed once daily in HIV-infected patients.


Asunto(s)
Adenina/análogos & derivados , Antirretrovirales/farmacología , Antirretrovirales/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , VIH-1 , ARN Viral/sangre , Adenina/efectos adversos , Adenina/sangre , Adenina/farmacocinética , Adenina/farmacología , Adenina/uso terapéutico , Adulto , Alanina , Antirretrovirales/farmacocinética , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Fatiga/inducido químicamente , Femenino , Infecciones por VIH/sangre , Humanos , Masculino , Persona de Mediana Edad , Náusea/inducido químicamente , Organofosfatos/sangre , Organofosfonatos/farmacocinética , Organofosfonatos/farmacología , Organofosfonatos/uso terapéutico , Método Simple Ciego , Estadísticas no Paramétricas , Tenofovir , Carga Viral , Adulto Joven
12.
Liver Int ; 29(8): 1178-83, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19486291

RESUMEN

BACKGROUND: Adefovir dipivoxil has activity against wild-type and lamivudine-resistant hepatitis B virus (HBV) and is frequently used to manage HBV infection in transplant recipients. Calcineurin inhibitors are a central component of immunosuppressive therapy. AIMS: Study GS-02-531 was an open-label, multicentre drug interaction trial to examine potential drug interactions between adefovir and tacrolimus in stable post-transplant recipients. MATERIALS AND METHODS: Sixteen non-HBV-infected post-transplant recipients with median age 45.5 years (69% male, 44% Caucasian, 50% Hispanic and 6% Black) and stable hepatic and renal function on a stable daily dose of tacrolimus (2-10 mg total daily dose) were studied before (tacrolimus alone) and after co-administration of adefovir 10 mg daily for 14 days (Days 1-14). Pharmacokinetic (PK) analyses utilized non-compartmental methods. RESULTS: The median elimination half-life of tacrolimus was 14.47 and 12.59 h for Day 0 and Day 14 respectively. The geometric mean ratios for tacrolimus on Day 14 vs Day 0 were 105.2% [90% confidence interval (90% CI): 89.8-123%] for C(max) and 106.4% (90% CI: 92.9-122%) for AUC(tau). Both 90% CIs for the ratios were contained within the predefined lack of interaction bounds of 80 and 125% (i.e. within the bounds for the equivalence assessment), indicating that these PK parameters of tacrolimus are not significantly altered by co-administration of adefovir. Similarly, the observed adefovir PK parameters after 14 days of co-administration with tacrolimus were comparable to historical data in non-transplant patients receiving adefovir alone. Serum creatinine values were stable during the study period. CONCLUSION: There is no significant PK interaction between tacrolimus and adefovir co-administered to liver transplant recipients for 14 days.


Asunto(s)
Adenina/análogos & derivados , Antivirales/uso terapéutico , Inmunosupresores/farmacocinética , Trasplante de Hígado , Organofosfonatos/uso terapéutico , Tacrolimus/farmacocinética , Adenina/uso terapéutico , Adolescente , Adulto , Anciano , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Semivida , Hepatitis B/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...