Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Chem Toxicol ; 38(4): 460-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25609003

RESUMEN

CONTEXT: Bilastine is a new oral selective, non-sedating histamine H1 antagonist for the symptomatic treatment of allergic rhinoconjunctivitis and urticaria. The European Medicines Agency requires an Environmental Risk Assessment (ERA) for all novel medicines for human use. OBJECTIVE: To calculate the bilastine predicted environmental concentration in surface water (PECsw; phase I ERA), and to determine the effects of bilastine on aquatic systems (phase II [tier A]). MATERIALS AND METHODS: Bilastine PECsw was calculated using the maximum daily dosage (20 mg), assuming that all administered bilastine was released into the aquatic environment. A persistence, bioaccumulation and toxicity assessment was conducted using the log Kow from the molecular structure. In phase II (tier A), a ready biodegradability test was performed, and bilastine's potential toxicity to various aquatic and sediment-dwelling micro-organisms was evaluated. RESULTS: Bilastine PECSW was calculated as 0.1 µg L(-1), and the compound was not readily biodegradable. Bilastine had no significant effects on Chironomus riparius midges, or on the respiration rate of activated sludge. For green algae, the bilastine no observed effect concentration (NOEC) was 22 mg L(-1); bilastine had no effect on zebra fish development, or on the reproduction rate of daphnids. DISCUSSION: Bilastine NOEC values against zebra fish, algae, daphnids, and aerobic organisms in activated sludge were at least 130 000-fold greater than the calculated PECSW value. CONCLUSION: No environmental concerns exist from bilastine use in patients with allergic rhinoconjunctivitis or urticaria.


Asunto(s)
Bencimidazoles/toxicidad , Monitoreo del Ambiente/métodos , Antagonistas de los Receptores Histamínicos H1 no Sedantes/toxicidad , Piperidinas/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Bencimidazoles/administración & dosificación , Chironomidae/efectos de los fármacos , Chlorophyta/efectos de los fármacos , Daphnia/efectos de los fármacos , Femenino , Antagonistas de los Receptores Histamínicos H1 no Sedantes/administración & dosificación , Masculino , Nivel sin Efectos Adversos Observados , Piperidinas/administración & dosificación , Medición de Riesgo/métodos , Aguas del Alcantarillado/microbiología , Pez Cebra
2.
Eur J Drug Metab Pharmacokinet ; 39(1): 33-41, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23619917

RESUMEN

Modern pharmacometrics can integrate and leverage all prior proprietary and public knowledge. Such methods can be used to scale across species or comparators, perform clinical trial simulation across alternative designs, confirm hypothesis and potentially reduce development burden, time and costs. Crucial yet typically lacking in integration is the pre-clinical stage. Prediction of PK in man, using in vitro and in vivo studies in different animal species, is increasingly well theorized but could still find wider application in drug development. The aim of the present work was to explore methods for bridging pharmacokinetic knowledge from animal species (i.v. and p.o.) and man (p.o.) into i.v. in man using the antihistamine drug bilastine as example. A model, predictive of i.v. PK in man, was developed on data from two pre-clinical species (rat and dog) and p.o. in man bilastine trials performed earlier. In the knowledge application stage, two different approaches were used to predict human plasma concentration after i.v. of bilastine: allometry (several scaling methods) and a semi-physiological method. Both approaches led to successful predictions of key i.v. PK parameters of bilastine in man. The predictive i.v. PK model was validated using later data from a clinical study of i.v. bilastine. Introduction of such knowledge in development permits proper leveraging of all emergent knowledge as well as quantification-based exploration of PK scenario, e.g. in special populations (pediatrics, renal insufficiency, comedication). In addition, the methods permit reduction or elimination and certainly optimization of learning trials, particularly those concerning alternative off-label administration routes.


Asunto(s)
Bencimidazoles/farmacocinética , Antagonistas de los Receptores Histamínicos/farmacocinética , Modelos Biológicos , Piperidinas/farmacocinética , Administración Intravenosa , Administración Oral , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/sangre , Perros , Femenino , Antagonistas de los Receptores Histamínicos/administración & dosificación , Antagonistas de los Receptores Histamínicos/sangre , Humanos , Bases del Conocimiento , Masculino , Modelos Animales , Piperidinas/administración & dosificación , Piperidinas/sangre , Ratas , Ratas Wistar , Reproducibilidad de los Resultados , Especificidad de la Especie
3.
Drug Chem Toxicol ; 35 Suppl 1: 1-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22616810

RESUMEN

This study evaluated the tissue distribution of total radioactivity in male albino, male pigmented, and time-mated female albino rats after oral administration of a single dose of [¹4C]-bilastine (20 mg/kg). Although only 1 animal was analyzed at each time point, there were apparent differences in bilastine distribution. Radioactivity was distributed to only a few tissues at low levels in male rats, whereas distribution was more extensive and at higher levels in female rats. This may be a simple sex-related difference. In each group and at each time point, concentrations of radioactivity were high in the liver and kidney, reflecting the role of these organs in the elimination process. In male albino rats, no radioactivity was measurable by 72 hours postdose. In male pigmented rats, only the eye and uveal tract had measurable levels of radioactivity at 24 hours. Measureable levels of radioactivity were retained in these tissues at the final sampling time point (336 hours postdose), indicating a degree of melanin-associated binding. In time-mated female rats, but not in albino or pigmented male rats, there was evidence of low-level passage of radioactivity across the placental barrier into fetal tissues as well as low-level transfer of radioactivity into the brain.


Asunto(s)
Bencimidazoles/farmacocinética , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacocinética , Piperidinas/farmacocinética , Animales , Autorradiografía , Bencimidazoles/metabolismo , Ojo , Femenino , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Masculino , Intercambio Materno-Fetal , Melaninas/metabolismo , Pigmentación , Piperidinas/metabolismo , Embarazo , Trazadores Radiactivos , Ratas , Ratas Endogámicas , Ratas Sprague-Dawley , Caracteres Sexuales , Distribución Tisular , Imagen de Cuerpo Entero
4.
Drug Chem Toxicol ; 35 Suppl 1: 18-24, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22616812

RESUMEN

Knowledge of the biotransformation of oral H1 antihistamines is clinically important because it can define their pharmacokinetic profile through possible effects on absorption (i.e., first-pass metabolism) and elimination. Further, clinically significant interactions with inhibitors of cytochrome P450 (CYP) have previously been reported for drugs of this therapeutic group, such as terfenadine and astemizole, indicating the possibility of drug-drug interactions involving agents that share the same metabolic pathway. The aim of this article was to review the preclinical testing of a new antihistamine (i.e., bilastine) in terms of its biotransformation in various animal species, including humans, and to evaluate its potential for possible drug-drug interactions involving the CYP system. A wide array of preclinical experiments were reviewed, all of which demonstrated that bilastine undergoes minimal metabolism in all species tested to date, including humans. Further, bilastine did not interact significantly, either as an inhibitor or inducer, with the CYP enzyme system, suggesting a low propensity for involvement in drug-drug interactions. These characteristics demonstrate the potential for bilastine to be a good choice for allergic patients receiving treatment for other concomitant diseases, including those with renal or hepatic dysfunction.


Asunto(s)
Bencimidazoles/metabolismo , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Piperidinas/metabolismo , Animales , Bencimidazoles/farmacocinética , Bencimidazoles/farmacología , Biotransformación , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacocinética , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacología , Humanos , Piperidinas/farmacocinética , Piperidinas/farmacología , Especificidad de la Especie
5.
Drug Chem Toxicol ; 35 Suppl 1: 8-17, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22616811

RESUMEN

Membrane transporters play a significant role in facilitating transmembrane drug movement. For new pharmacological agents, it is important to evaluate potential interactions (e.g., substrate specificity and/or inhibition) with human transporters that may affect their pharmacokinetics, efficacy, or toxicity. Bilastine is a new nonsedating H1 antihistamine indicated for the treatment of allergic rhinoconjunctivitis and urticaria. The in vitro inhibitory effects of bilastine were assessed on 12 human transporters: four efflux [multidrug resistance protein 1 (MDR1) or P-glycoprotein, breast cancer resistance protein (BCRP), multidrug resistance associated protein 2 (MRP2), and bile salt export pump) and eight uptake transporters (sodium taurocholate cotransporting polypeptide, organic cation transporter (OCT)1, organic anion transporter (OAT)1, OAT3, OCT2, OATP2B1, OATP1B1, and OATP1B3). Only mild inhibition was found for MDR1-, OCT1-, and OATP2B1-mediated transport of probe substrates at the highest bilastine concentration assayed (300 µM; half-maximal inhibitory concentration: ≥300 µM). Bilastine transport by MDR1, BCRP, OAT1, OAT3, and OCT2 was also investigated in vitro. Only MDR1 active transport of bilastine was relevant, whereas it did not appear to be a substrate of OCT2, OAT1, or OAT3, nor was it transported substantially by BCRP. Drug-drug interactions resulting from bilastine inhibition of drug transporters that would be generally regarded as clinically relevant are unlikely. Additionally, bilastine did not appear to be a substrate of human BCRP, OAT1, OAT3, or OCT2 and thus is not a potential victim of inhibitors of these transporters. On the other hand, based on in vitro evaluation, clinically relevant interactions with MDR1 inhibitors are anticipated.


Asunto(s)
Bencimidazoles/farmacología , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacología , Moduladores del Transporte de Membrana/farmacología , Piperidinas/farmacología , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Bencimidazoles/efectos adversos , Bencimidazoles/metabolismo , Transporte Biológico , Células CHO , Células CACO-2 , Línea Celular , Sistema Libre de Células/metabolismo , Cricetinae , Cricetulus , Perros , Evaluación Preclínica de Medicamentos , Antagonistas de los Receptores Histamínicos H1 no Sedantes/efectos adversos , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Humanos , Moduladores del Transporte de Membrana/efectos adversos , Moduladores del Transporte de Membrana/metabolismo , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Concentración Osmolar , Piperidinas/efectos adversos , Piperidinas/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Spodoptera
6.
Drug Chem Toxicol ; 35 Suppl 1: 25-33, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22616813

RESUMEN

As part of the bilastine development program, and as mandated by regulatory authorities, several studies were performed with oral bilastine in different animal species to evaluate its toxicity profile. Toxicokinetic analyses conducted in tandem to evaluate systemic exposure, gender differences, and dose proportionality in the different animal species indicated that animals were systemically exposed to bilastine during treatment. Repeated-dose toxicity studies in beagle dogs (52 weeks) and in rats and mice (13 weeks) showed that bilastine at doses up to 2,000 mg/kg/day was not associated with any mortality, ocular effects, or nodules/masses. Likewise, no bilastine-associated neoplastic lesions were observed in rats and mice after 104 weeks of treatment with bilastine at doses up to 2,000 mg/kg/day. In general, bilastine-related clinical signs, body-weight changes, food consumption, clinical chemistry, haematology, and macro- and microscopic findings were of low order and reversible, with effects present only at the highest doses administered. Bilastine (up to 1,000 mg/kg/day) was well tolerated in pregnant/lactating rats and in their offspring and subsequent generations. With respect to effects on embryofoetal development in rabbits, bilastine at 400 mg/kg/day (the highest dose evaluated) was assessed to be the no observed adverse effects level. Overall, bilastine demonstrated a favorable toxicity profile in all animal models investigated and at higher doses than the corresponding recommended daily human dosage.


Asunto(s)
Bencimidazoles/efectos adversos , Antagonistas de los Receptores Histamínicos H1 no Sedantes/efectos adversos , Piperidinas/efectos adversos , Administración Oral , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacocinética , Evaluación Preclínica de Medicamentos , Femenino , Desarrollo Fetal/efectos de los fármacos , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacocinética , Masculino , Nivel sin Efectos Adversos Observados , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Embarazo , Caracteres Sexuales , Pruebas de Toxicidad
7.
Clin Pharmacokinet ; 48(8): 543-54, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19705924

RESUMEN

OBJECTIVE: To model the pharmacokinetic and pharmacodynamic relationship of bilastine, a new histamine H(1) receptor antagonist, from single- and multiple-dose studies in healthy adult subjects. METHODS: The pharmacokinetic model was developed from different single-dose and multiple-dose studies. In the single-dose studies, a total of 183 subjects received oral doses of bilastine 2.5, 5, 10, 20, 50, 100, 120, 160, 200 and 220 mg. In the multiple-dose studies, 127 healthy subjects received bilastine 10, 20, 40, 50, 80, 100, 140 or 200 mg/day as multiple doses during a 4-, 7- or 14-day period. The pharmacokinetic profile of bilastine was investigated using a simultaneous analysis of all concentration-time data by means of nonlinear mixed-effects modelling population pharmacokinetic software NONMEM version 6.1. Plasma concentrations were modelled according to a two-compartment open model with first-order absorption and elimination. For the pharmacodynamic analysis, the inhibitory effect of bilastine (inhibition of histamine-induced wheal and flare) was assessed on a preselected time schedule, and the predicted typical pharmacokinetic profile (based on the pharmacokinetic model previously developed) was used. An indirect response model was developed to describe the pharmacodynamic relationships between flare or wheal areas and bilastine plasma concentrations. Finally, once values of the concentration that produced 50% inhibition (IC(50)) had been estimated for wheal and flare effects, simulations were carried out to predict plasma concentrations for the doses of bilastine 5, 10 and 20 mg at steady state (72-96 hours). RESULTS: A non-compartmental analysis resulted in linear kinetics of bilastine in the dose range studied. Bilastine was characterized by two-compartmental kinetics with a rapid-absorption phase (first-order absorption rate constant = 1.50 h(-1)), plasma peak concentrations were observed at 1 hour following administration and the maximal response was observed at approximately 4 hours or later. Concerning the selected pharmacodynamic model to fit the data (type I indirect response model), this selection is attributable to the presence of inhibitory bilastine plasma concentrations that decrease the input response function, i.e. the production of the skin reaction. This model resulted in the best fit of wheal and flare data. The estimates (with relative standard errors expressed in percentages in parentheses) of the apparent zero-order rate constant for flare or wheal spontaneous appearance (k(in)), the first-order rate constant for flare or wheal disappearance (k(out)) and bilastine IC(50) values were 0.44 ng/mL/h (14.60%), 1.09 h(-1) (15.14%) and 5.15 ng/mL (16.16%), respectively, for wheal inhibition, and 11.10 ng/mL/h (8.48%), 1.03 h(-1) (8.35%) and 1.25 ng/mL (14.56%), respectively, for flare inhibition. The simulation results revealed that bilastine plasma concentrations do not remain over the IC(50) value throughout the inter-dose period for doses of 5 and 10 mg. However, with a dose of 20 mg of bilastine administered every 24 hours, plasma concentrations remained over the IC(50) value during the considered period for the flare effect, and up to 20 hours for the wheal effect. CONCLUSION: Pharmacokinetic and pharmacodynamic relationships of bilastine were reliably described with the use of an indirect response pharmacodynamic model; this led to an accurate prediction of the pharmacodynamic activity of bilastine.


Asunto(s)
Bencimidazoles/administración & dosificación , Bencimidazoles/farmacocinética , Antagonistas de los Receptores Histamínicos/administración & dosificación , Antagonistas de los Receptores Histamínicos/farmacocinética , Modelos Biológicos , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Adulto , Antialérgicos/administración & dosificación , Antialérgicos/farmacocinética , Antialérgicos/farmacología , Bencimidazoles/farmacología , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Antagonistas de los Receptores Histamínicos/farmacología , Humanos , Masculino , Piperidinas/farmacología , Método Simple Ciego
8.
Neuropharmacology ; 52(3): 975-84, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17141811

RESUMEN

Serotonin (5-HT) and 5-HT(1A) receptors have been suggested to play a pivotal role in the mechanism of action of antidepressant drugs, particularly in the case of selective serotonin reuptake inhibitors (SSRIs). In the rat learned helplessness (LH) paradigm, a valid animal model of human depression, repeated treatment with the 5-HT(1A) receptor agonist 8-OH-DPAT (0.125 and 0.5mg/kg) and several classes of antidepressants such as the tricyclic agent desipramine (30 and 60mg/kg), the monoamine oxidase inhibitor (MAOI) pargyline (60mg/kg) and the SSRIs fluoxetine (15 and 30mg/kg), paroxetine (15 and 30mg/kg) and sertraline (30mg/kg) improved behavioural deficit in helpless rats. The involvement of serotonergic mechanisms in the antidepressant-like effect of these agents was investigated using the selective 5-HT(1A) receptor antagonist WAY 100,635 and the 5-HT synthesis inhibitor p-chlorophenylalanine (PCPA). Pretreatment with WAY 100,635 blocked the 8-OH-DPAT-induced reduction in escape failures, but did not counteract the antidepressant effect of fluoxetine and paroxetine. PCPA given alone did not modify helpless behaviour nor did it affect the behavioural effect of 8-OH-DPAT, fluoxetine and paroxetine. Adaptive changes in 5-HT(1A) receptor function were studied by measuring 8-OH-DPAT-mediated hypothermia and lower lip retraction (LLR) in the animals 24h after LH test session. Fluoxetine and paroxetine treatments caused a marked reduction in agonist-induced responses, an effect completely prevented by WAY 100,635 and PCPA. In conclusion, whereas direct agonist activity at postsynaptic 5-HT(1A) receptors attenuated helpless behaviour, the antidepressant-like effect of SSRIs was found to be independent of their actions on either 5-HT(1A) receptor function or extracellular 5-HT.


Asunto(s)
Conducta Animal/efectos de los fármacos , Desamparo Adquirido , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de la radiación , Temperatura Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Electrochoque/efectos adversos , Masculino , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Estadísticas no Paramétricas , Factores de Tiempo
9.
Pharm Res ; 22(11): 1769-82, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16158214

RESUMEN

PURPOSE: The twofold aim of this study was to characterize in vivo in rats the pharmacokinetics (PK) and pharmacodynamics (PD) of L6-OH, a metabolite of lerisetron with in vitro pharmacological activity, and evaluate the extent to which L6-OH contributes to the overall effect. METHODS: The PK of L6-OH was determined directly postmetabolite i.v. dose (PK-1), and also simultaneously for L (lerisetron concentration) and for generated L6-OH after lerisetron dose (200 microg kg(-1), i.v.), using Nonlinear Mixed Effects Modeling with an integrated parent-metabolite PK model (PK-2). Surrogate effect was measured by inhibition of serotonin-induced bradycardia. Protein binding was assayed via ultrafiltration and all quantification was performed via liquid chromatography-electrospray ionization-mass spectrometry. RESULTS: L6-OH showed elevated plasma and renal clearances, and volume of distribution (PK-1). The in vivo potency (PD) of L6-OH was high (EC(50) = 0.098 ng mL(-1) and EC(50unbound) = 0.040 ng mL(-1)). Total clearance for L (PK-2) in the presence of generated L6-OH (CL(L) = CL(-->L6-OH) + CL(n)) was 0.0139 L min(-1). Most of this clearance was L6-OH formation (F(c) = 99.6%), but only an 8.6% fraction of L6-OH was released into the bloodstream. The remainder undergoes biliar and fecal elimination. The parameters estimated from PK-2 were used to predict concentrations of L6-OH (Cp(L6)) generated after a lerisetron therapeutic dose (10 microg kg(-1)) in the rat. These concentrations are needed for the PD model and are below the quantification limit. Cp(L6max) was less than the EC(50) of L6-OH. CONCLUSIONS: We conclude that after lerisetron administration, L6-OH is extensively formed in the rat but it is quickly eliminated; therefore, besides being equipotent with the parent drug, the L6-OH metabolite does not influence the effect of lerisetron.


Asunto(s)
Bencimidazoles/metabolismo , Piperidinas/metabolismo , Antagonistas de la Serotonina/metabolismo , Animales , Bencimidazoles/farmacología , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Hidroxilación , Masculino , Modelos Biológicos , Piperidinas/farmacología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...