Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Clin Pharmacol ; 74(3): 365-371, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29198061

RESUMEN

PURPOSE: The aims of this study are to describe the development of PHARAO (Pharmacological Risk Assessment Online), a decision support system providing a risk profile for adverse events, associated with combined effects of multiple medicines, and to present data from a pilot study, testing the use, functionality, and acceptance of the PHARAO system in a clinical setting. METHODS: About 1400 substances were scored in relation to their risk to cause any of nine common and/or serious adverse effects. Algorithms for each adverse effect score were developed to create individual risk profiles from the patient's list of medication. The system was tested and integrated to the electronic medical record, during a 4-month period in two geriatric wards and three primary healthcare centers, and a questionnaire was answered by the users before and after the test period. RESULTS: A total of 732 substances were tagged with one or more of the nine risks, most commonly with the risk of sedation or seizures. During the pilot, the system was used 933 times in 871 patients. The most common signals generated by PHARAO in these patients were related to the risks of constipation, sedation, and bleeding. A majority of responders considered PHARAO easy to use and that it gives useful support in performing medication reviews. CONCLUSIONS: The PHARAO decision support system, designed as a complement to a database on drug-drug interactions used nationally, worked as intended and was appreciated by the users during a 4-month test period. Integration aspects need to be improved to minimize unnecessary signaling.


Asunto(s)
Enfermedad Crónica/tratamiento farmacológico , Sistemas de Apoyo a Decisiones Clínicas , Interacciones Farmacológicas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Geriatría/métodos , Polifarmacia , Atención Primaria de Salud/métodos , Anciano , Anciano de 80 o más Años , Actitud del Personal de Salud , Enfermedad Crónica/epidemiología , Comorbilidad , Revisión de la Utilización de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Finlandia/epidemiología , Evaluación Geriátrica , Humanos , Internet , Proyectos Piloto , Riesgo , Medición de Riesgo , Suecia/epidemiología , Recursos Humanos
2.
Eur J Clin Pharmacol ; 74(12): 1615-1622, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30167757

RESUMEN

PURPOSE: Buprenorphine has low oral bioavailability. Regardless of sublingual administration, a notable part of buprenorphine is exposed to extensive first-pass metabolism by the cytochrome P450 (CYP) 3A4. As drug interaction studies with buprenorphine are limited, we wanted to investigate the effect of voriconazole, a strong CYP3A4 inhibitor, on the pharmacokinetics and pharmacodynamics of oral buprenorphine. METHODS: Twelve healthy volunteers were given either placebo or voriconazole (orally, 400 mg twice on day 1 and 200 mg twice on days 2-5) for 5 days in a randomized, cross-over study. On day 5, they ingested 0.2 mg (3.6 mg during placebo phase) oral buprenorphine. We measured plasma and urine concentrations of buprenorphine and norbuprenorphine and monitored their pharmacological effects. Pharmacokinetic parameters were normalized for a buprenorphine dose of 1.0 mg. RESULTS: Voriconazole greatly increased the mean area under the plasma concentration-time curve (AUC0-18) of buprenorphine (4.3-fold, P < 0.001), its peak concentration (Cmax) (3.9-fold), half-life (P < 0.05), and excretion into urine (Ae; P < 0.001). Voriconazole also markedly enhanced the Cmax (P < 0.001), AUC0-18 (P < 0.001), and Ae (P < 0.05) of unconjugated norbuprenorphine but decreased its renal clearance (P < 0.001). Mild dizziness and nausea occurred during both study phases. CONCLUSIONS: Voriconazole greatly increases exposure to oral buprenorphine, mainly by inhibiting intestinal and liver CYP3A4. Effect on some transporters may explain elevated norbuprenorphine concentrations. Although oral buprenorphine is not commonly used, this interaction may become relevant in patients receiving sublingual buprenorphine together with voriconazole or other CYP3A4 or transporter inhibitors.


Asunto(s)
Analgésicos Opioides/farmacocinética , Antifúngicos/farmacología , Buprenorfina/farmacocinética , Voriconazol/farmacología , Adolescente , Adulto , Analgésicos Opioides/efectos adversos , Antifúngicos/efectos adversos , Área Bajo la Curva , Biotransformación , Buprenorfina/efectos adversos , Buprenorfina/análogos & derivados , Buprenorfina/metabolismo , Estudios Cruzados , Citocromo P-450 CYP3A/metabolismo , Mareo/inducido químicamente , Interacciones Farmacológicas , Femenino , Semivida , Voluntarios Sanos , Humanos , Masculino , Voriconazol/efectos adversos , Adulto Joven
3.
Eur J Clin Pharmacol ; 72(11): 1363-1371, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27510521

RESUMEN

PURPOSE: This study aimed to determine possible effects of voriconazole and posaconazole on the pharmacokinetics and pharmacological effects of sublingual buprenorphine. METHODS: We used a randomized, placebo-controlled crossover study design with 12 healthy male volunteers. Subjects were given a dose of 0.4 mg (0.6 mg during placebo phase) sublingual buprenorphine after a 5-day oral pretreatment with either (i) placebo, (ii) voriconazole 400 mg twice daily on the first day and 200 mg twice daily thereafter or (iii) posaconazole 400 mg twice daily. Plasma and urine concentrations of buprenorphine and its primary active metabolite norbuprenorphine were monitored over 18 h and pharmacological effects were measured. RESULTS: Compared to placebo, voriconazole increased the mean area under the plasma concentration-time curve (AUC0-∞) of buprenorphine 1.80-fold (90 % confidence interval 1.45-2.24; P < 0.001), its peak concentration (Cmax) 1.37-fold (P < 0.013) and half-life (t ½ ) 1.37-fold (P < 0.001). Posaconazole increased the AUC00-∞ of buprenorphine 1.25-fold (P < 0.001). Most of the plasma norbuprenorphine concentrations were below the limit of quantification (0.05 ng/ml). Voriconazole, unlike posaconazole, increased the urinary excretion of norbuprenorphine 1.58-fold (90 % confidence interval 1.18-2.12; P < 0.001) but there was no quantifiable parent buprenorphine in urine. Plasma buprenorphine concentrations correlated with the pharmacological effects, but the effects did not differ significantly between the phases. CONCLUSIONS: Voriconazole, and to a minor extent posaconazole, increase plasma exposure to sublingual buprenorphine, probably via inhibition of cytochrome P450 3 A and/or P-glycoprotein. Care should be exercised in the combined use of buprenorphine with triazole antimycotics, particularly with voriconazole, because their interaction can be of clinical importance.


Asunto(s)
Analgésicos Opioides/farmacocinética , Antifúngicos/farmacología , Buprenorfina/farmacocinética , Triazoles/farmacología , Voriconazol/farmacología , Administración Sublingual , Adulto , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Buprenorfina/efectos adversos , Buprenorfina/farmacología , Estudios Cruzados , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Voluntarios Sanos , Humanos , Masculino , Dolor/tratamiento farmacológico , Método Simple Ciego , Adulto Joven
4.
Eur J Clin Pharmacol ; 71(3): 321-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25560051

RESUMEN

BACKGROUND: Tramadol is widely used for acute, chronic, and neuropathic pain. Its primary active metabolite is O-desmethyltramadol (M1), which is mainly accountable for the µ-opioid receptor-related analgesic effect. Tramadol is metabolized to M1 mainly by cytochrome P450 (CYP)2D6 enzyme and to other metabolites by CYP3A4 and CYP2B6. We investigated the possible interaction of tramadol with the antifungal agents terbinafine (CYP2D6 inhibitor) and itraconazole (CYP3A4 inhibitor). METHODS: We used a randomized placebo-controlled crossover study design with 12 healthy subjects, of which 8 were extensive and 4 were ultrarapid CYP2D6 metabolizers. On the pretreatment day 4 with terbinafine (250 mg once daily), itraconazole (200 mg once daily) or placebo, subjects were given tramadol 50 mg orally. Plasma concentrations of tramadol and M1 were determined over 48 h and some pharmacodynamic effects over 12 h. Pharmacokinetic variables were calculated using standard non-compartmental methods. RESULTS: Terbinafine increased the area under plasma concentration-time curve (AUC0-∞) of tramadol by 115 % and decreased the AUC0-∞ of M1 by 64 % (P < 0.001). Terbinafine increased the peak concentration (C max) of tramadol by 53 % (P < 0.001) and decreased the C max of M1 by 79 % (P < 0.001). After terbinafine pretreatment the elimination half-life of tramadol and M1 were increased by 48 and 50 %, respectively (P < 0.001). Terbinafine reduced subjective drug effect of tramadol (P < 0.001). Itraconazole had minor effects on tramadol pharmacokinetics. CONCLUSIONS: Terbinafine may reduce the opioid effect of tramadol and increase the risk of its monoaminergic adverse effects. Itraconazole has no meaningful interaction with tramadol in subjects who have functional CYP2D6 enzyme.


Asunto(s)
Itraconazol/farmacología , Naftalenos/farmacología , Tramadol/administración & dosificación , Tramadol/farmacocinética , Administración Oral , Adulto , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Antifúngicos , Estudios Cruzados , Citocromo P-450 CYP2D6/genética , Inhibidores del Citocromo P-450 CYP2D6/farmacología , Inhibidores del Citocromo P-450 CYP3A/farmacología , Interacciones Farmacológicas , Femenino , Genotipo , Humanos , Masculino , Pruebas Neuropsicológicas , Terbinafina , Tramadol/sangre , Adulto Joven
5.
Eur J Clin Pharmacol ; 69(4): 867-75, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23099620

RESUMEN

PURPOSE: We assessed possible drug interactions of tramadol given concomitantly with the potent CYP2B6 inhibitor ticlopidine, alone or together with the potent CYP3A4 and P-glycoprotein inhibitor itraconazole. METHODS: In a randomized, placebo-controlled cross-over study, 12 healthy subjects ingested 50 mg of tramadol after 4 days of pretreatment with either placebo, ticlopidine (250 mg twice daily) or ticlopidine plus itraconazole (200 mg once daily). Plasma and urine concentrations of tramadol and its active metabolite O-desmethyltramadol (M1) were monitored over 48 h and 24 h, respectively. RESULTS: Ticlopidine increased the mean area under the plasma concentration-time curve (AUC0-∞) of tramadol by 2.0-fold (90 % confidence interval (CI) 1.6-2.4; p < 0.001) and Cmax by 1.4-fold (p < 0.001), and reduced its oral and renal clearance (p < 0.01). Ticlopidine reduced the AUC0-3 of M1 (p < 0.001) and the ratio of the AUC0-∞ of M1 to that of tramadol, but did not influence the AUC0-∞ of M1. Tramadol or M1 pharmacokinetics did not differ between the ticlopidine alone and ticlopidine plus itraconazole phases. CONCLUSIONS: Ticlopidine increased exposure to tramadol, reduced its renal clearance and inhibited the formation of M1, most likely via inhibition of CYP2B6 and/or CYP2D6. The addition of itraconazole to ticlopidine did not modify the outcome of the drug interaction. Concomitant clinical use of ticlopidine and tramadol may enhance the risk of serotonergic effects, especially when higher doses of tramadol are used.


Asunto(s)
Analgésicos Opioides/farmacocinética , Itraconazol/farmacología , Riñón/metabolismo , Ticlopidina/farmacología , Tramadol/análogos & derivados , Adulto , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/sangre , Analgésicos Opioides/orina , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Biotransformación , Estudios Cruzados , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Humanos , Itraconazol/administración & dosificación , Masculino , Tasa de Depuración Metabólica , Oxidorreductasas N-Desmetilantes/antagonistas & inhibidores , Ticlopidina/administración & dosificación , Tramadol/administración & dosificación , Tramadol/sangre , Tramadol/farmacocinética , Tramadol/orina , Adulto Joven
6.
Eur J Clin Pharmacol ; 69(6): 1293-301, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23242004

RESUMEN

PURPOSE: Tramadol is mainly metabolized by the cytochrome P450 (CYP) 2D6, CYP2B6 and CYP3A4 enzymes. The aim of this study was to evaluate the effect of enzyme induction with rifampicin on the pharmacokinetics and pharmacodynamics of oral and intravenous tramadol. METHODS: This was a randomized placebo-controlled crossover study design with 12 healthy subjects. After pretreatment for 5 days with rifampicin (600 mg once daily) or placebo, subjects were given tramadol either 50 mg intravenously or 100 mg orally. Plasma concentrations of tramadol and its active main metabolite O-desmethyltramadol (M1) were determined over 48 h. Analgesic and behavioral effects and whole blood 5-hydroxytryptamine (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA) concentrations were measured. RESULTS: Rifampicin reduced the mean area under the time-concentration curve (AUC0-∞) of intravenously administered tramadol by 43 % and that of M1 by 58 % (P < 0.001); it reduced the AUC0-∞ of oral tramadol by 59 % and that of M1 by 54 % (P < 0.001). Rifampicin increased the clearance of intravenous tramadol by 67 % (P < 0.001). Bioavailability of oral tramadol was reduced by rifampicin from 66 to 49 % (P = 0.002). The pharmacological effects of tramadol or whole blood serotonin concentrations were not influenced by pretreatment with rifampicin. CONCLUSIONS: Rifampicin markedly decreased the exposure to tramadol and M1 after both oral and intravenous administration. Therefore, rifampicin and other potent enzyme inducers may have a clinically important interaction with tramadol regardless of the route of its administration.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Sistema Enzimático del Citocromo P-450/biosíntesis , Rifampin/administración & dosificación , Tramadol/administración & dosificación , Tramadol/farmacocinética , Administración Oral , Analgésicos Opioides/sangre , Área Bajo la Curva , Disponibilidad Biológica , Biomarcadores/sangre , Biotransformación , Estudios Cruzados , Citocromo P-450 CYP2D6/genética , Sistema Enzimático del Citocromo P-450/genética , Esquema de Medicación , Interacciones Farmacológicas , Inducción Enzimática , Finlandia , Genotipo , Semivida , Humanos , Ácido Hidroxiindolacético/sangre , Inyecciones Intravenosas , Tasa de Depuración Metabólica , Metilación , Pruebas Neuropsicológicas , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Fenotipo , Desempeño Psicomotor/efectos de los fármacos , Serotonina/sangre , Tramadol/sangre
7.
Physiol Plant ; 145(4): 516-26, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22182287

RESUMEN

The aim of this study was to investigate plant frost hardiness responses to ultraviolet (UV) radiation, since the few results reported are largely contradictory. It was hypothesized that functional adaptation of life forms could explain these contradictions. Dwarf shrubs and tree seedlings, representing both evergreen and deciduous forms, were tested (Vaccinium vitis-idaea, Vaccinium myrtillus, Pinus sylvestris, Betula pubescens and its red form f. rubra). The research was performed in Sodankylä, Northern Finland (67°N), with enhanced UV-B- and UV-A-radiation treatments between 2002 and 2009. Plant frost hardiness was determined using the freeze-induced electrolyte leakage method in early autumn, during the onset of the frost hardening process. Additional physiological variables (malondialdehyde, glutathione, total phenols, C and N contents) were analyzed in V. vitis-idaea to explain the possible responses. These variables did not respond significantly to UV-radiation treatments, but explained the frost hardiness well (r² = 0.678). The main finding was that frost hardiness decreased in the evergreen shrub V. vitis-idaea, particularly with enhanced UV-A radiation. No significant responses were observed with the other plants. Therefore, this study does not support the idea that enhanced UV radiation could increase plant frost hardiness.


Asunto(s)
Congelación , Rayos Ultravioleta , Vaccinium vitis-Idaea/fisiología , Adaptación Fisiológica , Vaccinium vitis-Idaea/efectos de la radiación
8.
Eur J Clin Pharmacol ; 68(6): 979-86, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22286159

RESUMEN

PURPOSE: We examined the effect of grapefruit juice on the pharmacokinetics and pharmacodynamics of oral S-ketamine. METHODS: A randomized crossover open-label study design with two phases at an interval of 4 weeks was conducted in 12 healthy volunteers. Grapefruit juice or water was ingested 200 ml t.i.d. for 5 days. An oral dose of 0.2 mg/kg of S-ketamine was ingested on day 5 with 150 ml grapefruit juice or water. Plasma concentrations of ketamine and norketamine were determined for 24 h, and pharmacodynamic variables were recorded for 12 h. Noncompartmental methods were used to calculate pharmacokinetic parameters. RESULTS: Grapefruit juice increased the geometric mean value of the area under the plasma ketamine concentration-time curve(AUC0-∞) by 3.0-fold (range 2.4- to 3.6-fold; P<0.001), the peak plasma concentration (Cmax) by 2.1-fold (range 1.8- to 2.6-fold; P<0.001), and the elimination half-life by 24% (P<0.05) as compared to the water phase. The ratio of main metabolite norketamine to ketamine (AUCm/AUCp) was decreased by 57% (P<0.001) during the grapefruit phase.Self-rated relaxation was decreased (P<0.05) and the performance in the digit symbol substitution test was increased (P<0.05) after grapefruit juice, but other behavioral or analgesic effects were not affected. CONCLUSIONS: Grapefruit juice significantly increased the plasma concentrations of oral ketamine in healthy volunteers.Dose reductions of ketamine should be considered when using oral ketamine concomitantly with grapefruit juice.


Asunto(s)
Bebidas , Citrus paradisi , Interacciones Alimento-Droga , Ketamina/farmacocinética , Adulto , Anciano , Hidrocarburo de Aril Hidroxilasas/metabolismo , Estudios Cruzados , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP3A/metabolismo , Femenino , Humanos , Ketamina/administración & dosificación , Ketamina/análogos & derivados , Ketamina/sangre , Masculino , Persona de Mediana Edad , Oxidorreductasas N-Desmetilantes/metabolismo , Adulto Joven
9.
Antimicrob Agents Chemother ; 55(3): 1063-7, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21173180

RESUMEN

Our aim was to assess the effect of miconazole oral gel on the pharmacokinetics of oral oxycodone. In an open crossover study with two phases, 12 healthy volunteers took a single oral dose of 10 mg of immediate-release oxycodone with or without thrice-daily 85-mg miconazole oral gel treatment. The plasma concentrations of oxycodone and its oxidative metabolites were measured for 48 h. Pharmacological effects of oxycodone were recorded for 12 h. Pharmacokinetic parameters were compared by use of the geometric mean ratios (GMRs) and their 90% confidence interval (CIs). Pretreatment with miconazole oral gel caused a strong inhibition of the CYP2D6-dependent metabolism and moderate inhibition of the CYP3A4-dependent metabolism of oxycodone. The mean area under the concentration-time curve (AUC) from time zero to infinity (AUC(0-∞); GMR, 1.63; 90% CI, 1.48 to 1.79) and the peak concentration of oxycodone (GMR, 1.31; 90% CI, 1.19 to 1.44) were increased. The AUC of the CYP2D6-dependent metabolite oxymorphone was greatly decreased (GMR, 0.17; 90% CI, 0.09 to 0.31) by miconazole gel, whereas that of the CYP3A4-dependent metabolite noroxycodone was increased (GMR, 1.30; 90% CI, 1.15 to 1.47) by miconazole gel. Differences in the pharmacological response to oxycodone between phases were insignificant. Miconazole oral gel increases the exposure to oral oxycodone, but the clinical relevance of the interaction is moderate. Miconazole oral gel produces a rather strong inhibitory effect on CYP2D6, which deserves further study.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Miconazol/uso terapéutico , Oxicodona/uso terapéutico , Administración Oral , Adulto , Analgésicos Opioides/administración & dosificación , Inhibidores del Citocromo P-450 CYP2D6 , Inhibidores del Citocromo P-450 CYP3A , Femenino , Humanos , Masculino , Oxicodona/administración & dosificación , Dolor/tratamiento farmacológico , Adulto Joven
10.
Clin Drug Investig ; 31(3): 143-53, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21142269

RESUMEN

BACKGROUND AND OBJECTIVE: Oxycodone is a µ-opioid receptor agonist that is mainly metabolized by hepatic cytochrome P450 (CYP) enzymes. Because CYP enzymes can be inhibited by other drugs, the pharmacokinetics of oxycodone are prone to drug interactions. The aim of this study was to determine whether inhibition of CYP2D6 alone by paroxetine or inhibition of both CYP2D6 and CYP3A4 by a combination of paroxetine and itraconazole alters the pharmacokinetics of and pharmacological response to intravenous oxycodone. METHODS: We used a randomized, three-phase, crossover, placebo-controlled study design in 12 healthy subjects. The subjects were given 0.1 mg/kg of intravenous oxycodone after pre-treatments with placebo, paroxetine or a combination of paroxetine and itraconazole for 4 days. Plasma concentrations of oxycodone and its oxidative metabolites were measured over 48 hours, and pharmacokinetic and pharmacodynamic parameters subsequently evaluated. RESULTS: The effect of paroxetine on the plasma concentrations of oxycodone was negligible. The combination of paroxetine and itraconazole prolonged the mean elimination half-life of oxycodone from 3.8 to 6.6 hours (p < 0.001), and increased the exposure to oxycodone 2-fold (p < 0.001). However, these changes were not reflected in pharmacological response. CONCLUSION: The results of this study indicate that there are no clinically relevant drug interactions with intravenous oxycodone and inhibitors of CYP2D6. If both oxidative metabolic pathways via CYP3A4 and 2D6 are inhibited the exposure to intravenous oxycodone increases substantially.


Asunto(s)
Analgésicos Opioides/clasificación , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Oxicodona/farmacocinética , Adulto , Analgésicos Opioides/farmacología , Estudios Cruzados , Inhibidores del Citocromo P-450 CYP2D6 , Inhibidores del Citocromo P-450 CYP3A , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Femenino , Semivida , Humanos , Itraconazol/farmacología , Masculino , Oxicodona/farmacología , Paroxetina/farmacología , Adulto Joven
11.
Br J Clin Pharmacol ; 70(1): 78-87, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20642550

RESUMEN

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: Oxycodone is an opioid analgesic that is metabolized mainly in the liver by cytochrome P450 (CYP) 2D6 and 3A4 enzymes. So far, the effects of CYP2D6 or CYP3A4 inhibitors on the pharmacokinetics of oxycodone in humans have not been systematically studied. WHAT THIS STUDY ADDS: Drug interactions arising from CYP2D6 inhibition most likely have minor clinical importance for oral oxycodone. When both of CYP2D6 and CYP3A4 pathways are inhibited, the exposure to oral oxycodone is increased substantially. AIM: The aim of this study was to find out whether the inhibition of cytochrome P450 2D6 (CYP2D6) with paroxetine or concomitant inhibition of CYP2D6 and CYP3A4 with paroxetine and itraconazole, altered the pharmacokinetics and pharmacological response of orally administered oxycodone. METHODS: A randomized placebo-controlled cross-over study design with three phases was used. Eleven healthy subjects ingested 10 mg of oral immediate release oxycodone on the fourth day of pre-treatment with either placebo, paroxetine (20 mg once daily) or paroxetine (20 mg once daily) and itraconazole (200 mg once daily) for 5 days. The plasma concentrations of oxycodone and its oxidative metabolites were measured for 48 h, and pharmacological (analgesic and behavioural) effects were evaluated. RESULTS: Paroxetine alone reduced the area under concentration-time curve (AUC(0,0-48 h)) of the CYP2D6 dependent metabolite oxymorphone by 44% (P < 0.05), but had no significant effects on the plasma concentrations of oxycodone or its pharmacological effects when compared with the placebo phase. When both oxidative pathways of the metabolism of oxycodone were inhibited with paroxetine and itraconazole, the mean AUC(0,infinity) of oxycodone increased by 2.9-fold (P < 0.001), and its C(max) by 1.8-fold (P < 0.001). Visual analogue scores for subjective drug effects, drowsiness and deterioration of performance were slightly increased (P < 0.05) after paroxetine + itraconazole pre-treatment when compared with placebo. CONCLUSIONS: Drug interactions arising from CYP2D6 inhibition most likely have minor clinical importance for oral oxycodone if the function of the CYP3A4 pathway is normal. When both CYP2D6 and CYP3A4 pathways are inhibited, the exposure to oral oxycodone is increased substantially.


Asunto(s)
Inhibidores del Citocromo P-450 CYP2D6 , Adulto , Estudios Cruzados , Citocromo P-450 CYP3A , Interacciones Farmacológicas , Femenino , Humanos , Itraconazol/administración & dosificación , Itraconazol/farmacocinética , Itraconazol/farmacología , Masculino , Oxicodona/administración & dosificación , Oxicodona/farmacocinética , Oxicodona/farmacología , Paroxetina/administración & dosificación , Paroxetina/farmacocinética , Paroxetina/farmacología , Adulto Joven
12.
Physiol Plant ; 140(2): 128-40, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20497369

RESUMEN

Insulation provided by snow cover and tolerance of freezing by physiological acclimation allows Arctic plants to survive cold winter temperatures. However, both the protection mechanisms may be lost with winter climate change, especially during extreme winter warming events where loss of snow cover from snow melt results in exposure of plants to warm temperatures and then returning extreme cold in the absence of insulating snow. These events cause considerable damage to Arctic plants, but physiological responses behind such damage remain unknown. Here, we report simulations of extreme winter warming events using infrared heating lamps and soil warming cables in a sub-Arctic heathland. During these events, we measured maximum quantum yield of photosystem II (PSII), photosynthesis, respiration, bud swelling and associated bud carbohydrate changes and lipid peroxidation to identify physiological responses during and after the winter warming events in three dwarf shrub species: Empetrum hermaphroditum, Vaccinium vitis-idaea and Vaccinium myrtillus. Winter warming increased maximum quantum yield of PSII, and photosynthesis was initiated for E. hermaphroditum and V. vitis-idaea. Bud swelling, bud carbohydrate decreases and lipid peroxidation were largest for E. hermaphroditum, whereas V. myrtillus and V. vitis-idaea showed no or less strong responses. Increased physiological activity and bud swelling suggest that sub-Arctic plants can initiate spring-like development in response to a short winter warming event. Lipid peroxidation suggests that plants experience increased winter stress. The observed differences between species in physiological responses are broadly consistent with interspecific differences in damage seen in previous studies, with E. hermaphroditum and V. myrtillus tending to be most sensitive. This suggests that initiation of spring-like development may be a major driver in the damage caused by winter warming events that are predicted to become more frequent in some regions of the Arctic and that may ultimately drive plant community shifts.


Asunto(s)
Adaptación Fisiológica/fisiología , Ecosistema , Ericaceae/fisiología , Estaciones del Año , Regiones Árticas , Metabolismo de los Hidratos de Carbono , Carbohidratos/análisis , Ericaceae/clasificación , Ericaceae/metabolismo , Peroxidación de Lípido/fisiología , Meristema/metabolismo , Meristema/fisiología , Consumo de Oxígeno/fisiología , Fotosíntesis/fisiología , Complejo de Proteína del Fotosistema II/metabolismo , Hojas de la Planta/metabolismo , Hojas de la Planta/fisiología , Brotes de la Planta/metabolismo , Brotes de la Planta/fisiología , Nieve , Especificidad de la Especie , Temperatura , Vaccinium myrtillus/metabolismo , Vaccinium myrtillus/fisiología , Vaccinium vitis-Idaea/metabolismo , Vaccinium vitis-Idaea/fisiología
13.
Eur J Clin Pharmacol ; 66(10): 977-85, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20697700

RESUMEN

PURPOSE: this study aimed to investigate the effect of antivirals ritonavir and lopinavir/ritonavir on the pharmacokinetics and pharmacodynamics of oral oxycodone, a widely used opioid receptor agonist used in the treatment of moderate to severe pain. METHODS: a randomized crossover study design with three phases at intervals of 4 weeks was conducted in 12 healthy volunteers. Ritonavir 300 mg, lopinavir/ritonavir 400/100 mg, or placebo b.i.d. for 4 days was given to the subjects. On day 3, 10 mg oxycodone hydrochloride was administered orally. Plasma concentrations of oxycodone, noroxycodone, oxymorphone, and noroxymorphone were determined for 48 h. Pharmacokinetic parameters were calculated with standard noncompartmental methods. Behavioral effects and experimental cold pain analgesia were assessed for 12 h. ANOVA for repeated measures was used for statistical analysis. RESULTS: ritonavir and lopinavir/ritonavir increased the area under the plasma concentration-time curve of oral oxycodone by 3.0-fold (range 1.9- to 4.3-fold; P <0.001) and 2.6-fold (range 1.9- to 3.3-fold; P <0.001). The mean (± SD) elimination half-life increased after ritonavir and lopinavir/ritonavir from 3.6 ± 0.6 to 5.6 ± 0.9 h (P <0.001) and 5.7 ± 0.9 h (P <0.001), respectively. Both ritonavir (P <0.001) and lopinavir/ritonavir (P <0.05) increased the self-reported drug effect of oxycodone. CONCLUSIONS: ritonavir and lopinavir/ritonavir greatly increase the plasma concentrations of oral oxycodone in healthy volunteers and enhance its effect. When oxycodone is used clinically in patients during ritonavir and lopinavir/ritonavir treatment, reductions in oxycodone dose may be needed to avoid opioid-related adverse effects.


Asunto(s)
Analgésicos Opioides/farmacocinética , Inhibidores de la Proteasa del VIH/farmacología , Oxicodona/farmacocinética , Pirimidinonas/farmacología , Ritonavir/farmacología , Adulto , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/sangre , Analgésicos Opioides/farmacología , Área Bajo la Curva , Estudios Cruzados , Esquema de Medicación , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Finlandia , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/administración & dosificación , Inhibidores de la Proteasa del VIH/efectos adversos , Humanos , Modelos Lineales , Lopinavir , Masculino , Oxicodona/administración & dosificación , Oxicodona/sangre , Oxicodona/farmacología , Dolor/complicaciones , Dolor/tratamiento farmacológico , Pirimidinonas/administración & dosificación , Pirimidinonas/efectos adversos , Valores de Referencia , Ritonavir/administración & dosificación , Ritonavir/efectos adversos , Factores de Tiempo , Adulto Joven
14.
Eur J Clin Pharmacol ; 66(4): 387-97, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20076952

RESUMEN

BACKGROUND: The aim of this study was to investigate the effects of the cytochrome P450 3A4 (CYP34A) inhibitor itraconazole on the pharmacokinetics and pharmacodynamics of orally and intravenously administered oxycodone. METHODS: Twelve healthy subjects were administered 200 mg itraconazole or placebo orally for 5 days in a four-session paired cross-over study. On day 4, oxycodone was administered intravenously (0.1 mg/kg) in the first part of the study and orally (10 mg) in the second part. Plasma concentrations of oxycodone and its oxidative metabolites were measured for 48 h, and pharmacodynamic effects were evaluated. RESULTS: Itraconazole decreased plasma clearance (Cl) and increased the area under the plasma concentration-time curve (AUC0-infinity) of intravenous oxycodone by 32 and 51%, respectively (P<0.001) and increased the AUC(0-infinity) of orally administrated oxycodone by 144% (P<0.001). Most of the pharmacokinetic changes in oral oxycodone were seen in the elimination phase, with modest effects by itraconazole on its peak concentration, which was increased by 45% (P=0.009). The AUC(0-48) of noroxycodone was decreased by 49% (P<0.001) and that of oxymorphone was increased by 359% (P<0.001) after the administration of oral oxycodone. The pharmacologic effects of oxycodone were enhanced by itraconazole only modestly. CONCLUSIONS: Itraconazole increased the exposure to oxycodone by inhibiting its CYP3A4-mediated N-demethylation. The clinical use of itraconazole in patients receiving multiple doses of oxycodone for pain relief may increase the risk of opioid-associated adverse effects.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos Opioides/farmacocinética , Itraconazol/farmacología , Oxicodona/farmacología , Oxicodona/farmacocinética , Analgésicos Opioides/administración & dosificación , Área Bajo la Curva , Estudios Cruzados , Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A , Humanos , Inyecciones Intravenosas , Itraconazol/administración & dosificación , Itraconazol/farmacocinética , Morfinanos/farmacocinética , Morfinanos/farmacología , Oxidación-Reducción , Oxicodona/administración & dosificación , Oximorfona/farmacocinética , Oximorfona/farmacología
15.
Anesthesiology ; 110(6): 1371-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19417618

RESUMEN

BACKGROUND: Oxycodone is a mu-opioid receptor agonist that is metabolized mainly in the liver by cytochrome P450 3A and 2D6 enzymes. Rifampin is a strong inducer of several drug-metabolizing enzymes. The authors studied the interaction of rifampin with oxycodone. Their hypothesis was that rifampin enhances the CYP3A-mediated metabolism of oxycodone and attenuates its pharmacologic effect. METHODS: The protocol was a four-session, paired crossover. Twelve volunteers were given 600 mg oral rifampin or placebo once daily for 7 days. Oxycodone was given on day 6. In the first part of the study, 0.1 mg/kg oxycodone hydrochloride was given intravenously. In the second part of the study, 15 mg oxycodone hydrochloride was given orally. Concentrations of oxycodone and its metabolites noroxycodone, oxymorphone, and noroxymorphone were determined for 48 h. Psychomotor effects were characterized for 12 h by several visual analog scales. Analgesic effects were characterized by measuring the heat pain threshold and cold pain sensitivity. RESULTS: Rifampin decreased the area under the oxycodone concentration-time curve of intravenous and oral oxycodone by 53% and 86%, respectively (P < 0.001). Oral bioavailability of oxycodone was decreased from 69% to 21% (P < 0.001). Rifampin greatly increased the plasma metabolite-to-parent drug ratios for noroxycodone and noroxymorphone (P < 0.001). Pharmacologic effects of oral oxycodone were attenuated. CONCLUSIONS: Induction of cytochrome P450 3A by rifampin reduced the area under the oxycodone concentration-time curve of intravenous and oral oxycodone. The pharmacologic effects of oxycodone were modestly attenuated. To maintain adequate analgesia, dose adjustment of oxycodone may be necessary, when used concomitantly with rifampin.


Asunto(s)
Analgésicos Opioides/sangre , Antibacterianos/efectos adversos , Oxicodona/sangre , Rifampin/efectos adversos , Administración Oral , Adulto , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Disponibilidad Biológica , Estudios Cruzados , Citocromo P-450 CYP3A/biosíntesis , Citocromo P-450 CYP3A/metabolismo , Método Doble Ciego , Interacciones Farmacológicas , Inducción Enzimática/efectos de los fármacos , Femenino , Semivida , Humanos , Inyecciones Intravenosas , Masculino , Oxicodona/administración & dosificación , Oxicodona/farmacocinética , Adulto Joven
16.
Eur J Clin Pharmacol ; 65(1): 89-95, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18777173

RESUMEN

PURPOSE: The effect of topical miconazole oral gel and systemic oral voriconazole on the pharmacokinetics of oral etoricoxib was studied in 12 healthy volunteers. METHODS: Plasma concentrations of etoricoxib, miconazole, voriconazole, and thromboxane B(2) generation were followed after ingestion of 60 mg etoricoxib without pretreatment, after topical administration of miconazole oral gel (85 mg x 3, 3 days), or after oral voriconazole (400 mg x 2, 1st day, 200 mg x 2, 2nd day). RESULTS: Etoricoxib area under the plasma concentration-time curve (AUC(0-00)) and maximum plasma concentration (C(max)) geometric mean ratios (GMR) with/without miconazole were 1.69 {90% confidence interval (CI); 1.46-1.92} and 1.12 (90% CI; 0.99-1.25), respectively, and corresponding GMRs with/without voriconazole were 1.49 (90% CI; 1.37-1.61) and 1.19 (90% CI; 1.08-1.31), respectively. CONCLUSIONS: Miconazole oral gel and oral voriconazole produced comparable increase in the exposure to etoricoxib, presumably via CYP3A inhibition.


Asunto(s)
Antifúngicos/administración & dosificación , Inhibidores de la Ciclooxigenasa/farmacocinética , Inhibidores Enzimáticos/administración & dosificación , Miconazol/administración & dosificación , Piridinas/farmacocinética , Pirimidinas/administración & dosificación , Sulfonas/farmacocinética , Triazoles/administración & dosificación , Administración Oral , Administración Tópica , Adulto , Antifúngicos/sangre , Antifúngicos/farmacocinética , Biotransformación/efectos de los fármacos , Estudios Cruzados , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/sangre , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacocinética , Etoricoxib , Geles , Humanos , Masculino , Miconazol/sangre , Miconazol/farmacocinética , Piridinas/administración & dosificación , Piridinas/sangre , Pirimidinas/sangre , Pirimidinas/farmacocinética , Sulfonas/administración & dosificación , Sulfonas/sangre , Tromboxano B2/sangre , Triazoles/sangre , Triazoles/farmacocinética , Voriconazol , Adulto Joven
17.
Eur J Clin Pharmacol ; 65(3): 263-71, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18836708

RESUMEN

OBJECTIVE: We investigated the effect of voriconazole on the pharmacokinetics and pharmacodynamics of oxycodone. METHODS: Twelve healthy subjects ingested either voriconazole or placebo for 4 days in a randomized, cross-over study. On day 3, they ingested 10 mg oxycodone. Timed plasma samples were collected for the measurement of oxycodone, noroxycodone, oxymorphone, noroxymorphone and voriconazole up to 48 h, and pharmacodynamic effects were recorded. RESULTS: When voriconazole was taken at the same time as oxycodone, the mean area under the plasma concentration-time curve (AUC(0-infinity)) of oxycodone increased 3.6-fold (range 2.7- to 5.6-fold), peak plasma concentration 1.7-fold and elimination half-life 2.0-fold (p < 0.001) when compared to placebo. The AUC(0-infinity) ratio of noroxycodone to oxycodone was decreased by 92% (p < 0.001), and that of oxymorphone increased by 108% (p < 0.01). Pharmacodynamic effects of oxycodone were modestly increased by voriconazole. CONCLUSIONS: Voriconazole inhibits the CYP3A-mediated N-demethylation of oxycodone, drastically increasing exposure to oral oxycodone. Clinically, lower doses of oxycodone may be needed during voriconazole treatment to avoid opioid-related adverse effects especially after repeated dosing.


Asunto(s)
Analgésicos Opioides/farmacocinética , Antifúngicos/farmacología , Inhibidores Enzimáticos/farmacología , Oxicodona/farmacocinética , Pirimidinas/farmacología , Triazoles/farmacología , Administración Oral , Adulto , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/sangre , Antifúngicos/sangre , Antifúngicos/farmacocinética , Área Bajo la Curva , Estudios Cruzados , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A , Interacciones Farmacológicas , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacocinética , Femenino , Semivida , Humanos , Masculino , Oxicodona/administración & dosificación , Oxicodona/sangre , Pirimidinas/sangre , Pirimidinas/farmacocinética , Triazoles/sangre , Triazoles/farmacocinética , Voriconazol
18.
Eur J Clin Pharmacol ; 65(6): 627-33, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19205683

RESUMEN

OBJECTIVE: The aim was to develop a drug-drug interaction database (SFINX) to be integrated into decision support systems or to be used in website solutions for clinical evaluation of interactions. METHODS: Key elements such as substance properties and names, drug formulations, text structures and references were defined before development of the database. Standard operating procedures for literature searches, text writing rules and a classification system for clinical relevance and documentation level were determined. ATC codes, CAS numbers and country-specific codes for substances were identified and quality assured to ensure safe integration of SFINX into other data systems. Much effort was put into giving short and practical advice regarding clinically relevant drug-drug interactions. RESULTS: SFINX includes over 8,000 interaction pairs and is integrated into Swedish and Finnish computerised decision support systems. Over 31,000 physicians and pharmacists are receiving interaction alerts through SFINX. User feedback is collected for continuous improvement of the content. CONCLUSION: SFINX is a potentially valuable tool delivering instant information on drug interactions during prescribing and dispensing.


Asunto(s)
Sistemas de Administración de Bases de Datos , Sistemas de Apoyo a Decisiones Clínicas , Interacciones Farmacológicas
19.
J Steroid Biochem Mol Biol ; 110(1-2): 56-66, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18356043

RESUMEN

This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Screening with well-established chemical inhibitors showed that the formation of norethindrone was potently inhibited by CYP3A4 inhibitor ketoconazole (IC(50)=0.02 microM) and with CYP2C9 inhibitor sulphaphenazole (IC(50)=2.13 microM); the further biotransformation of norethindrone was strongly inhibited by ketoconazole (IC(50)=0.09 microM). Fluconazole modestly inhibited both lynestrenol bioactivation and norethindrone biotransformation. Lynestrenol bioactivation was mainly catalysed by recombinant human CYP2C9, CYP2C19 and CYP3A4; rCYP3A4 was responsible for the hydroxylation of norethindrone. A significant correlation was observed between norethindrone formation and tolbutamide hydroxylation, a CYP2C9-selective activity (r=0.63; p=0.01). Norethindrone hydroxylation correlated significantly with model reactions of CYP2C19 and CYP3A4. The greatest immunoinhibition of lynestrenol bioactivation was seen in incubations with CYP2C-Ab. The CYP3A4-Ab reduced norethindrone hydroxylation by 96%. Both lynestrenol and norethindrone were weak inhibitors of CYP2C9 (IC(50) of 32 microM and 46 microM for tolbutamide hydroxylation, respectively). In conclusion, CYP2C9, CYP2C19 and CYP3A4 are the primary cytochromes in the bioactivation of lynestrenol in vitro, while CYP3A4 catalyses the further metabolism of norethindrone.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Linestrenol/farmacocinética , Noretindrona/farmacocinética , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Biotransformación/efectos de los fármacos , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Fluconazol/farmacología , Humanos , Hidroxilación/efectos de los fármacos , Cetoconazol/farmacología , Linestrenol/química , Linestrenol/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Noretindrona/química , Noretindrona/metabolismo , Sulfafenazol/farmacología
20.
J Clin Pharmacol ; 48(8): 986-94, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18490497

RESUMEN

The effect of oral contraceptives (OCs) on melatonin metabolism was studied in 29 subjects genotyped for CYP1A2 SNP g.-163C>A polymorphism. Plasma melatonin and 6-OH-melatonin concentrations were measured after a 6-mg dose of melatonin using a validated liquid chromatography/mass spectrometry method. The mean melatonin AUC and C(max) values were 4- to 5-fold higher in OC users than in non-OC users (P < .0001), whereas the weight-adjusted clearance was significantly lower in OC users (P < .0001). No significant difference in melatonin pharmacokinetics between the genotypes and no additional effect by the genotype on the OC-induced increase in melatonin exposure were evident. Melatonin exposure had no significant effect on the subjects' state of alertness. In conclusion, a significant inhibitory effect of OCs on the CYP1A2-catalyzed melatonin metabolism was seen; thereby, OC use can alter CYP1A2-phenotyping results.


Asunto(s)
Anticonceptivos Hormonales Orales/efectos adversos , Inhibidores del Citocromo P-450 CYP1A2 , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Melatonina/farmacocinética , Polimorfismo Genético/genética , Adulto , Alelos , Área Bajo la Curva , Atención/efectos de los fármacos , Interacciones Farmacológicas , Femenino , Genotipo , Semivida , Humanos , Hidroxilación , Masculino , Melatonina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA