Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 365(14): 1284-92, 2011 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-21991950

RESUMEN

BACKGROUND: Poor neurodevelopmental outcomes and recurrences of cutaneous lesions remain unacceptably frequent among survivors of neonatal herpes simplex virus (HSV) disease. METHODS: We enrolled neonates with HSV disease in two parallel, identical, double-blind, placebo-controlled studies. Neonates with central nervous system (CNS) involvement were enrolled in one study, and neonates with skin, eye, and mouth involvement only were enrolled in the other. After completing a regimen of 14 to 21 days of parenteral acyclovir, the infants were randomly assigned to immediate acyclovir suppression (300 mg per square meter of body-surface area per dose orally, three times daily for 6 months) or placebo. Cutaneous recurrences were treated with open-label episodic therapy. RESULTS: A total of 74 neonates were enrolled--45 with CNS involvement and 29 with skin, eye, and mouth disease. The Mental Development Index of the Bayley Scales of Infant Development (in which scores range from 50 to 150, with a mean of 100 and with higher scores indicating better neurodevelopmental outcomes) was assessed in 28 of the 45 infants with CNS involvement (62%) at 12 months of age. After adjustment for covariates, infants with CNS involvement who had been randomly assigned to acyclovir suppression had significantly higher mean Bayley mental-development scores at 12 months than did infants randomly assigned to placebo (88.24 vs. 68.12, P=0.046). Overall, there was a trend toward more neutropenia in the acyclovir group than in the placebo group (P=0.09). CONCLUSIONS: Infants surviving neonatal HSV disease with CNS involvement had improved neurodevelopmental outcomes when they received suppressive therapy with oral acyclovir for 6 months. (Funded by the National Institute of Allergy and Infectious Diseases; CASG 103 and CASG 104 ClinicalTrials.gov numbers, NCT00031460 and NCT00031447, respectively.).


Asunto(s)
Aciclovir/uso terapéutico , Antivirales/uso terapéutico , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Desarrollo Infantil/efectos de los fármacos , Herpes Simple/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Aciclovir/efectos adversos , Antivirales/efectos adversos , Enfermedades del Sistema Nervioso Central/prevención & control , Enfermedades del Sistema Nervioso Central/virología , Método Doble Ciego , Femenino , Herpes Simple/prevención & control , Humanos , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Prevención Secundaria
2.
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
3.
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
4.
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
5.
J Hepatol ; 52(4): 501-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20189674

RESUMEN

BACKGROUND & AIMS: Pegylated interferon (PEG-IFN) alfa-2b plus ribavirin (RBV) is the standard of care for adults with chronic hepatitis C but was not approved for the treatment of children at the time of this study. The aim of this study was to evaluate the efficacy and safety of PEG-IFN alfa-2b plus RBV in children. METHODS: Children and adolescents ages 3-17 years were treated with PEG-IFN alfa-2b (60microg/m(2)/week) plus RBV (15mg/kg/day). The duration of therapy was 24 weeks for genotype (G) 2 and G3 patients with low viral load (<600,000IU/ml) and 48 weeks for G1, G4, and G3 with high viral load (>or=600,000IU/ml). The primary end point was sustained virologic response (SVR), defined as undetectable hepatitis C virus (HCV) RNA 24 weeks after completion of therapy. RESULTS: SVR was attained by 70 (65%) children. Genotype was the main predictor of response: G1, 53%; G2/3, 93%; G4, 80%. SVRs were similar in younger and older children. Baseline viral load was the main predictor of response in the G1 cohort. No new safety signals were identified, and adverse events (AEs) were generally mild or moderate in severity. Dose was modified because of AEs in 25% of children; 1 child discontinued because of an AE (thrombocytopenia). No serious AEs related to study drugs were reported. CONCLUSION: Therapy with PEG-IFN alfa-2b plus RBV in children and adolescents with chronic hepatitis C offers favorable efficacy, reduced injection frequency, and an acceptable safety profile.


Asunto(s)
Antivirales/administración & dosificación , Hepacivirus/efectos de los fármacos , Hepatitis C Crónica/tratamiento farmacológico , Interferón-alfa/administración & dosificación , Polietilenglicoles/administración & dosificación , Ribavirina/administración & dosificación , Adolescente , Antivirales/efectos adversos , Antivirales/farmacocinética , Estatura , Peso Corporal , Niño , Desarrollo Infantil , Preescolar , Farmacorresistencia Viral/genética , Quimioterapia Combinada , Femenino , Genotipo , Hepacivirus/genética , Hepatitis C Crónica/virología , Humanos , Interferón alfa-2 , Interferón-alfa/efectos adversos , Interferón-alfa/farmacocinética , Masculino , Polietilenglicoles/efectos adversos , Polietilenglicoles/farmacocinética , Proteínas Recombinantes , Ribavirina/efectos adversos , Ribavirina/farmacocinética , Resultado del Tratamiento , Carga Viral/efectos de los fármacos
6.
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
7.
J Pediatric Infect Dis Soc ; 5(1): 53-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26407253

RESUMEN

BACKGROUND: Neonatal enterovirus sepsis has high mortality. Antiviral therapy is not available. METHODS: Neonates with suspected enterovirus sepsis (hepatitis, coagulopathy, and/or myocarditis) with onset at ≤15 days of life were randomized 2:1 to receive oral pleconaril or placebo for 7 days. Serial virologic (oropharynx, rectum, urine, serum), clinical, pharmacokinetic, and safety evaluations were performed. RESULTS: Sixty-one subjects were enrolled (43 treatment, 18 placebo), of whom 43 were confirmed enterovirus infected (31 treatment, 12 placebo). There was no difference in day 5 oropharyngeal culture positivity (primary endpoint; 0% in both groups). However, enterovirus-infected subjects in the treatment group became culture negative from all anatomic sites combined faster than placebo group subjects (median 4.0 versus 7.0 days, P = .08), and fewer subjects in the treatment group remained polymerase chain reaction (PCR)-positive from the oropharynx when last sampled (23% versus 58%, P = .02; median, 14.0 days). By intent to treat, 10/43 (23%) subjects in the treatment group and 8/18 (44%) in the placebo group died (P = .02 for 2-month survival difference); among enterovirus-confirmed subjects, 7/31 (23%) in the treatment group died versus 5/12 (42%) in the placebo group (P = .26). All pleconaril recipients attained concentrations greater than the IC90 after the first study day, but 38% were less than the IC90 during the first day of treatment. One subject in the treatment group and three in the placebo group had treatment-related adverse events. CONCLUSIONS: Shorter times to culture and PCR negativity and greater survival among pleconaril recipients support potential efficacy and warrant further evaluation.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Enterovirus/complicaciones , Infecciones por Enterovirus/tratamiento farmacológico , Enterovirus/efectos de los fármacos , Sepsis Neonatal/tratamiento farmacológico , Sepsis Neonatal/virología , Oxadiazoles/uso terapéutico , Antivirales/sangre , Antivirales/farmacocinética , Antivirales/orina , Método Doble Ciego , Enterovirus/genética , Enterovirus/aislamiento & purificación , Infecciones por Enterovirus/sangre , Infecciones por Enterovirus/orina , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Sepsis Neonatal/sangre , Sepsis Neonatal/orina , Orofaringe/virología , Oxadiazoles/sangre , Oxadiazoles/farmacocinética , Oxadiazoles/orina , Oxazoles , Recto/virología
8.
Clin Pharmacokinet ; 44(1): 99-109, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15634033

RESUMEN

OBJECTIVE: Delavirdine is a non-nucleoside reverse transcriptase inhibitor used in combination regimens for the treatment of HIV-1 infection. Our objective was to characterise the population pharmacokinetics of delavirdine in HIV-infected patients who participated in the adult AIDS Clinical Trials Group (ACTG) 260 and 261 studies. METHODS: ACTG 261 was a randomised, double-blind study of delavirdine 400mg three times daily, in various combination regimens; ACTG 260 was a concentration-targeted monotherapy study. Two hundred and thirty-four patients, and 1254 and 1251 plasma concentrations for delavirdine and N-delavirdine, respectively, were available for population pharmacokinetic analysis. The pharmacokinetic model (and initial parameters), based on previous studies, included two compartments for delavirdine (peripheral and central) and parallel clearance pathways (nonlinear conversion to N-delavirdine and first order clearance from the body). The model was one compartment for N-delavirdine with first order clearance. Diurnal variation of delavirdine and N-delavirdine oral clearance was modelled as a cosine function, with amplitude variation a fitted parameter. Pharmacokinetic parameter estimates were derived from iterative two-stage analysis; observed delavirdine and N-delavirdine concentrations fit with weighting by the inverse observation variance. Covariates were analysed by multiple general linear modelling. RESULTS: The mean (percent coefficient of variation [%CV]) CD4 count was 315 (109) cells/mm(3), weight 76.9 (14.7) kg, age 37 (8.5) years, and 15% of the population were women. Mean (%CV) population pharmacokinetic parameter estimates for delavirdine were: volume of distribution at steady state 67.6 (100) L, intrinsic oral clearance 19.8 (64) L/h, concentration at half the maximum velocity of metabolism (V(max)) 6.3 (69) micromol/L and first order oral clearance 0.57 (86) L/h. For N-delavirdine, the mean (%CV) apparent volume of distribution was 24.7 (75) L and apparent clearance 29.7 (42) L/h. The mean V(max) was 1376 (68) mg/day. The final model for average intrinsic clearance of delavirdine included race, sex, weight and age as significant covariates (p < 0.05); however, these covariates do not explain a significant proportion of the overall variability in the population. CONCLUSIONS: Delavirdine disposition exhibits nonlinear pharmacokinetics and large interpatient variability, and is significantly altered by time of day (impacting potential therapeutic drug monitoring and future pharmacokinetic study designs). Although race and sex appear to influence delavirdine pharmacokinetics, men and women and patients of different races should receive similar mg/kg dosage regimens. The presence of large interpatient variability supports the further investigation of the utility of therapeutic drug monitoring for delavirdine, if target drug concentrations can be better defined.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Carbamazepina/análogos & derivados , Delavirdina/farmacocinética , Infecciones por VIH/metabolismo , Adolescente , Adulto , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/uso terapéutico , Peso Corporal , Delavirdina/metabolismo , Delavirdina/uso terapéutico , Método Doble Ciego , Quimioterapia Combinada , Femenino , Infecciones por VIH/tratamiento farmacológico , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Grupos Raciales , Factores Sexuales
9.
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
10.
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
11.
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
12.
Pharmacotherapy ; 23(7): 866-70, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12885100

RESUMEN

STUDY OBJECTIVE: To determine if milk thistle (silymarin) alters the pharmacokinetics of indinavir. DESIGN: Sequential crossover trial. SETTING: General clinical research center. SUBJECTS: Ten healthy subjects. INTERVENTION: Indinavir 800 mg 3 times/day was given for four doses on days 1 and 2. Silymarin 160 mg 3 times/day was given on days 3-15. On day 16 and for one dose on day 17, both drugs were given at the same dosages. MEASUREMENTS AND MAIN RESULTS: Indinavir's pharmacokinetic parameters were evaluated at steady state both before and after administration of 14 days of silymarin. Blood samples were collected -0.25, 0.5, 1, 2, 3, 4, and 5 hours after indinavir dosing and assayed by high-performance liquid chromatography. The final pharmacokinetic model had first-order absorption after a lag time, and two compartments with first-order elimination from the central compartment. When given alone and combined with silymarin, respectively, the geometric mean (95% confidence interval [CI]) steady-state indinavir area under the plasma concentration-time curve was 20.7 hr x mg/L (15.3-28.2 hr x mg/L) and 19.4 hr x mg/L (15.8-23.6 hr x mg/L) and the trough plasma concentration was 0.340 mg/L (0.232-0.497 mg/L) and 0.232 mg/L (0.129-0.419 mg/L). CONCLUSION: Silymarin has no apparent effect on indinavir plasma concentrations.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacocinética , Indinavir/farmacocinética , Sustancias Protectoras/farmacología , Silimarina/farmacología , Administración Oral , Adolescente , Adulto , Área Bajo la Curva , Cromatografía Líquida de Alta Presión , Estudios Cruzados , Interacciones Farmacológicas , Femenino , Inhibidores de la Proteasa del VIH/sangre , Humanos , Indinavir/sangre , Masculino , Persona de Mediana Edad , Silybum marianum/química
13.
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
14.
J Econ Entomol ; 95(5): 1074-8, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12403437

RESUMEN

High carbon dioxide atmospheres combined with high temperature were effective for controlling Indian meal moth, Plodia interpunctella (Hübner) pupae. Pupae were exposed to atmospheres of 60, 80, or 98% carbon dioxide (CO2) in nitrogen (N2), or 60 or 80% CO2 in air at temperatures of 26.7 degrees C or 32.2 degrees C and 60% RH. Controlled atmosphere treatments at 32.2 degrees C controlled pupae faster than the same treatments at the lower temperature. At both temperatures high CO2 concentration treatments combined with nitrogen killed pupae faster than high CO2 concentration treatments combined with air. Exposure to 80% carbon dioxide mixed with nitrogen was the most effective treatment causing 100% mortality in 12 h at 32.2 degrees C and 93.3% mortality in 18 h at 26.6 degrees C. High-temperature controlled atmosphere treatments had no adverse effects on quality of two preserved floral products, Limonium sinuatum (L.) and Gypsophila elegans (Bieb.), tested for 12, 18, and 24 h according to industry standards.


Asunto(s)
Caryophyllaceae , Control de Insectos/métodos , Mariposas Nocturnas , Plumbaginaceae , Animales , Atmósfera , Dióxido de Carbono , Flores , Nitrógeno , Temperatura
15.
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.

16.
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.

17.
Clin Drug Investig ; 22(2): 125-34, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-23315400

RESUMEN

OBJECTIVE: To characterise the pharmacokinetics of indinavir during different phases of the menstrual cycle in HIV-infected women. DESIGN: Open-label study. SETTING: The immunodeficiency clinic at Erie County Medical Center, Buffalo, New York. PATIENTS: Ten HIV-infected women were enrolled in the study. Eligibility criteria included an acceptable medical history, chemistry profile, complete blood count with differential, lymphocyte profile, urinalysis and history of a regular menstrual cycle. PATIENTS had to be on a stable antiretroviral regimen that included indinavir 800mg taken every 8 hours. INTERVENTIONS: Blood sampling over an 8-hour period following an 800mg dose of indinavir during the menstrual, follicular and luteal phases of the menstrual cycle. MAIN OUTCOME MEASURES: Pharmacokinetic parameters in ten HIV-infected women adherent with indinavir 800mg every 8 hours during the menstrual, follicular and luteal phases of the menstrual cycle. Serum estradiol and progesterone levels were also obtained during each menstrual cycle phase. RESULTS: The peak plasma concentration, plasma concentration 8 hours after administration of a given dose of indinavir, elimination half-life and oral clearance of indinavir were not significantly different across the menstrual cycle phases. Indinavir exposure varied among the female patients with some individuals having similar areas under the concentration-time curve (AUCs) during the three phases while others had notable differences in AUC. Maximum plasma indinavir concentrations were highest during the follicular phase in four subjects, highest during the luteal phase in two individuals, and highest during the menstrual phase in three patients. CONCLUSIONS: No differences were found in indinavir pharmacokinetics during the menstrual cycle phases. Significant intra- and interpatient variability in indinavir pharmacokinetics were observed; however, indinavir exposure in women did not appear to be excessive compared with pharmacokinetic data obtained from prior studies conducted in men.

18.
Pediatr Infect Dis J ; 29(3): 195-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19949363

RESUMEN

BACKGROUND: When oseltamivir is administered in extremely high doses (500-1000 mg/kg) to young juvenile rats, central nervous system toxicity and death occurred in some animals. Mortality was not observed in older juvenile rats, suggesting a possible relationship between neurotoxicity and an immature blood-brain barrier. To assess potential neurologic adverse effects of oseltamivir use in infants, a retrospective chart review was performed in infants less than 12 months of age who received oseltamivir, amantadine, or rimantadine. METHODS: The primary objective was to describe the frequency of neurologic adverse events among children less than 12 months of age who received oseltamivir compared with those receiving adamantanes. Medical record databases, emergency department databases, and/or pharmacy records at 15 medical centers were searched to identify patients. RESULTS: Of the 180 infants identified as having received antiviral therapy, 115 (64%) received oseltamivir, 37 (20%) received amantadine, and 28 (16%) received rimantadine. The median dose of oseltamivir was 2.0 mg/kg/dose in 3- to 5-month-old and 2.2 mg/kg/dose in 9- to 12-month-old infants. The maximum dose administered was 7.0 mg/kg/dose. There were no statistically significant differences in the occurrence of adverse neurologic events during therapy among subjects treated with oseltamivir versus those treated with the adamantanes (P = 0.13). CONCLUSIONS: This is the largest report to date of oseltamivir use in children less than 12 months of age. Neurologic events were not more common with use of oseltamivir compared with that of the adamantanes. Dosing of oseltamivir was variable, illustrating the need for pharmacokinetic data in this younger population.


Asunto(s)
Adamantano/efectos adversos , Antivirales/efectos adversos , Gripe Humana/tratamiento farmacológico , Oseltamivir/efectos adversos , Rimantadina/efectos adversos , Adamantano/uso terapéutico , Antivirales/uso terapéutico , Sistema Nervioso Central/efectos de los fármacos , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Enfermedades del Sistema Nervioso/inducido químicamente , Oseltamivir/uso terapéutico , Estudios Retrospectivos , Rimantadina/uso terapéutico
19.
J Clin Virol ; 46 Suppl 4: S22-6, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19766534

RESUMEN

BACKGROUND: Ganciclovir protects against hearing deterioration in infants with symptomatic congenital cytomegalovirus (CMV) disease involving the central nervous system (CNS). OBJECTIVES: To assess the neurodevelopmental impact of ganciclovir therapy in this population. STUDY DESIGN: 100 neonates were enrolled into a controlled Phase III study of symptomatic congenital CMV involving the CNS, and were randomized to either 6 weeks of intravenous ganciclovir or no treatment. Denver developmental tests were performed at 6 weeks, 6 months, and 12 months. For each age, developmental milestones that > or =90% of normal children would be expected to have achieved were identified. The numbers of milestones not met ("delays") were determined for each subject. The average number of delays per subject was compared for each treatment group. RESULTS: At 6 months, the average number of delays was 4.46 and 7.51, respectively, for ganciclovir recipients and "no treatment" subjects (p=0.02). At 12 months, the average number of delays was 10.06 and 17.14, respectively (p=0.007). In a multivariate regression model, the effect of ganciclovir therapy remained statistically significant at 12 months (p=0.007). CONCLUSIONS: Infants with symptomatic congenital CMV involving the CNS receiving intravenous ganciclovir therapy have fewer developmental delays at 6 and 12 months compared with untreated infants. Based on these data as well as the previously published data regarding ganciclovir treatment and hearing outcomes, 6 weeks of intravenous ganciclovir therapy can be considered in the management of babies with symptomatic congenital CMV disease involving the CNS. If treatment is initiated, it should be started within the first month of life and patients should be monitored closely for toxicity, especially neutropenia. Since existing data only address the treatment of symptomatic congenital CMV disease involving the CNS, these data cannot be extrapolated to neonates with other manifestations of CMV disease, including asymptomatic babies and symptomatic babies who do not have CNS involvement.


Asunto(s)
Antivirales/uso terapéutico , Sistema Nervioso Central/virología , Infecciones por Citomegalovirus/tratamiento farmacológico , Discapacidades del Desarrollo/prevención & control , Ganciclovir/uso terapéutico , Antivirales/administración & dosificación , Sistema Nervioso Central/fisiopatología , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/congénito , Discapacidades del Desarrollo/virología , Femenino , Ganciclovir/administración & dosificación , Humanos , Lactante , Recién Nacido , Inyecciones Intravenosas , Masculino , Análisis Multivariante , Análisis de Regresión , Resultado del Tratamiento
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA