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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Drug Metab Dispos ; 50(1): 65-75, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34620695

RESUMEN

Small molecules that present complex absorption, distribution, metabolism, and elimination (ADME) properties can be challenging to investigate as potential therapeutics. Acquiring data through standard methods can yield results that are insufficient to describe the in vivo situation, which can affect downstream development decisions. Implementing in vitro-in vivo-in silico strategies throughout the drug development process is effective in identifying and mitigating risks while speeding up their development. Risdiplam (Evrysdi)-an orally bioavailable, small molecule approved by the US Food and Drug Administration and more recently by the European Medicines Agency for the treatment of patients ≥2 months of age with spinal muscular atrophy-is presented here as a case study. Risdiplam is a low-turnover compound whose metabolism is mediated through a non-cytochrome P450 enzymatic pathway. Four main challenges of risdiplam are discussed: predicting in vivo hepatic clearance, determining in vitro metabolites with regard to metabolites in safety testing guidelines, elucidating enzymes responsible for clearance, and estimating potential drug-drug interactions. A combination of in vitro and in vivo results was successfully extrapolated and used to develop a robust physiologically based pharmacokinetic model of risdiplam. These results were verified through early clinical studies, further strengthening the understanding of the ADME properties of risdiplam in humans. These approaches can be applied to other compounds with similar ADME profiles, which may be difficult to investigate using standard methods. SIGNIFICANCE STATEMENT: Risdiplam is the first approved, small-molecule, survival of motor neuron 2 mRNA splicing modifier for the treatment of spinal muscular atrophy. The approach taken to characterize the absorption, distribution, metabolism, and excretion (ADME) properties of risdiplam during clinical development incorporated in vitro-in vivo-in silico techniques, which may be applicable to other small molecules with challenging ADME. These strategies may be useful in improving the speed at which future drug molecules can be developed.


Asunto(s)
Compuestos Azo/metabolismo , Compuestos Azo/farmacocinética , Preparaciones Farmacéuticas/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacocinética , Empalme del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Distribución Tisular , Animales , Humanos , Técnicas In Vitro , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
2.
Br J Clin Pharmacol ; 85(1): 181-193, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30302786

RESUMEN

AIMS: Risdiplam (RG7916, RO7034067) is an orally administered, centrally and peripherally distributed, survival of motor neuron 2 (SMN2) mRNA splicing modifier for the treatment of spinal muscular atrophy (SMA). The objectives of this entry-into-human study were to assess the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics of risdiplam, and the effect of the strong CYP3A inhibitor itraconazole on the PK of risdiplam in healthy male volunteers. METHODS: Part 1 had a randomized, double-blind, adaptive design with 25 subjects receiving single ascending oral doses of risdiplam (ranging from 0.6-18.0 mg, n = 18) or placebo (n = 7). A Bayesian framework was applied to estimate risdiplam's effect on SMN2 mRNA. The effect of multiple doses of itraconazole on the PK of risdiplam was also assessed using a two-period cross-over design (n = 8). RESULTS: Risdiplam in the fasted or fed state was well tolerated. Risdiplam exhibited linear PK over the dose range with a multi-phasic decline with a mean terminal half-life of 40-69 h. Food had no relevant effect, and itraconazole had only a minor effect on plasma PK indicating a low fraction of risdiplam metabolized by CYP3A. The highest tested dose of 18.0 mg risdiplam led to approximately 41% (95% confidence interval 27-55%) of the estimated maximum increase in SMN2 mRNA. CONCLUSIONS: Risdiplam was well tolerated and proof of mechanism was demonstrated by the intended shift in SMN2 splicing towards full-length SMN2 mRNA. Based on these data, Phase 2/3 studies of risdiplam in patients with SMA are now ongoing.


Asunto(s)
Compuestos Azo/administración & dosificación , Fármacos Neuromusculares/administración & dosificación , Pirimidinas/administración & dosificación , Empalme del ARN/efectos de los fármacos , Administración Oral , Adolescente , Adulto , Área Bajo la Curva , Compuestos Azo/efectos adversos , Compuestos Azo/farmacocinética , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Método Doble Ciego , Interacciones Farmacológicas , Voluntarios Sanos , Humanos , Itraconazol/farmacocinética , Masculino , Persona de Mediana Edad , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Fármacos Neuromusculares/efectos adversos , Fármacos Neuromusculares/farmacocinética , Pirimidinas/efectos adversos , Pirimidinas/farmacocinética , ARN Mensajero/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Adulto Joven
3.
Xenobiotica ; 45(3): 230-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25350082

RESUMEN

Abstract 1. The metabolism and drug-drug interaction (DDI) risk of tofogliflozin, a potent and highly specific sodium-glucose co-transporter 2 inhibitor, were evaluated by in vitro studies using human liver microsomes, human hepatocytes, and recombinant human CYPs. 2. The main metabolite of tofogliflozin was the carboxylated derivative (M1) in human hepatocytes, which was the same as in vivo. The metabolic pathway of tofogliflozin to M1 was considered to be as follows: first, tofogliflozin was catalyzed to the primary hydroxylated derivative (M4) by CYP2C18, CYP4A11 and CYP4F3B, then M4 was oxidized to M1. 3. Tofogliflozin had no induction potential on CYP1A2 and CYP3A4. Neither tofogliflozin nor M1 had inhibition potential on CYPs, with the exception of a weak CYP2C19 inhibition by M1. 4. Not only are multiple metabolic enzymes involved in the tofogliflozin metabolism, but the drug is also excreted into urine after oral administration, indicating that tofogliflozin is eliminated through multiple pathways. Thus, the exposure of tofogliflozin would not be significantly altered by DDI caused by any co-administered drugs. Also, tofogliflozin seems not to cause significant DDI of co-administered drugs because tofogliflozin has no CYP induction or inhibition potency, and the main metabolite M1 has no clinically relevant CYP inhibition potency.


Asunto(s)
Compuestos de Bencidrilo/metabolismo , Glucósidos/metabolismo , Hepatocitos/metabolismo , Metabolómica/métodos , Microsomas Hepáticos/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Compuestos de Bencidrilo/química , Radioisótopos de Carbono , Coenzimas/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/biosíntesis , Interacciones Farmacológicas , Inducción Enzimática/efectos de los fármacos , Glucósidos/química , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Concentración 50 Inhibidora , Redes y Vías Metabólicas/efectos de los fármacos , Metaboloma/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Factores de Tiempo
4.
Drug Metab Dispos ; 42(9): 1466-77, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24989889

RESUMEN

Although the multiplicity in transport proteins assessed during drug development is continuously increasing, the clinical relevance of the breast cancer resistance protein (BCRP) is still under debate. Here, our aim is to rationalize the need to consider BCRP substrate and inhibitor interactions and to define optimum selection and acceptance criteria between cell-based and vesicle-based assays in vitro. Information on the preclinical and clinical pharmacokinetics (PK), drug-drug interactions, and pharmacogenomics data was collated for 13 marketed drugs whose PK is reportedly associated with BCRP interaction. Clinical examples where BCRP impacts drug PK and efficacy appear to be rare and confounded by interactions with other transporters. Thirty-seven compounds were selected to be tested as BCRP substrates in a cell-based assay using MDCKII cells (Madin-Darby canine kidney cells) and 18 in membrane vesicles. Depending on the physicochemical compound properties, we observed both in vitro systems to give false-negative readouts. In addition, the inhibition potential of 19 compounds against BCRP was assessed in vesicles and in MDCKII cells, where we observed significant system and substrate-dependent IC50 values. Therefore, neither of the two test systems is superior to the other. Instead, one system may offer advantages under certain situations (e.g., low permeability) and thus should be selected based on the physicochemical compound properties. Finally, given the clinical relevance of BCRP, we propose that its evaluation should remain issue-driven: for low permeable, low bioavailable drugs, in particular when other more common processes do not allow a mechanistic understanding of any unexpected absorption or brain disposition, and for drugs with a low therapeutic window.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Preparaciones Farmacéuticas/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Animales , Disponibilidad Biológica , Línea Celular , Perros , Descubrimiento de Drogas/métodos , Interacciones Farmacológicas/fisiología , Humanos , Células LLC-PK1 , Células de Riñón Canino Madin Darby , Proteínas de Transporte de Membrana/metabolismo , Porcinos
5.
Drug Metab Dispos ; 42(9): 1411-22, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24939652

RESUMEN

The multidrug resistance protein 1 (MDR1) is known to limit brain penetration of drugs and play a key role in drug-drug interactions (DDIs). Theoretical cut-offs from regulatory guidelines are used to extrapolate MDR1 interactions from in vitro to in vivo. However, these cut-offs do not account for interlaboratory variability. Our aim was to calibrate our experimental system to allow better in vivo predictions. We selected 166 central nervous system (CNS) and non-CNS drugs to calibrate the MDR1 transport screening assay using Lewis lung cancer porcine kidney 1 epithelial cells overexpressing MDR1 (L-MDR1). A threshold efflux ratio (ER) of 2 was established as one parameter to assess brain penetration in lead optimization. The inhibitory potential of 57 molecules was evaluated using IC50 values based on the digoxin ER-IC50(ER)-or apparent permeability-IC50(Papp)-in L-MDR1 cells. Published clinical data for 68 DDIs involving digoxin as the victim drug were collected. DDI risk assessments were based on intestinal concentrations ([I2]) as well as unbound [I1u] and total plasma [I1T] concentrations. A receiver operating characteristic analysis identified an [I2]/IC50(ER) of 6.5 as the best predictor of a potential interaction with digoxin in patients. The model was further evaluated with a test set of 11 digoxin DDIs and 16 nondigoxin DDIs, resulting in only one false negative for each test set, no false positives among the digoxin DDIs, and two among the nondigoxin DDIs. Future refinements might include using cerebrospinal fluid to unbound plasma concentration ratios rather than therapeutic class, better estimation of [I2], and dynamic modeling of MDR1-mediated DDIs.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Interacciones Farmacológicas/fisiología , Preparaciones Farmacéuticas/metabolismo , Animales , Bioensayo/métodos , Transporte Biológico/fisiología , Calibración , Línea Celular Tumoral , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Digoxina/metabolismo , Humanos , Técnicas In Vitro/métodos , Permeabilidad , Porcinos
6.
Drug Metab Dispos ; 40(8): 1556-65, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22584254

RESUMEN

It was reported that oseltamivir (Tamiflu) absorption was mediated by human peptide transporter (hPEPT) 1. Understanding the exact mechanism(s) of absorption is important in the context of drug-drug and diet-drug interactions. Hence, we investigated the mechanism governing the intestinal absorption of oseltamivir and its active metabolite (oseltamivir carboxylate) in wild-type [Chinese hamster ovary (CHO)-K1] and hPEPT1-transfected cells (CHO-PEPT1), in pharmacokinetic studies in juvenile and adult rats, and in healthy volunteers. In vitro cell culture studies showed that the intracellular accumulation of oseltamivir and its carboxylate into CHO-PEPT1 and CHO-K1 was always similar under a variety of experimental conditions, demonstrating that these compounds are not substrates of hPEPT1. Furthermore, neither oseltamivir nor its active metabolite was capable of inhibiting Gly-Sar uptake in CHO-PEPT1 cells. In vivo pharmacokinetic studies in juvenile and adult rats showed that the disposition of oseltamivir and oseltamivir carboxylate, after oral administration of oseltamivir, was sensitive to the feed status but insensitive to the presence of milk and Gly-Sar. Moreover, oseltamivir and oseltamivir carboxylate exhibited significantly higher exposure in rats under fasted conditions than under fed conditions. In humans, oral dosing after a high-fat meal resulted in a statistically significant but moderate lower exposure than after an overnight fasting. This change has no clinical implications. Taken together, the results do not implicate either rat Pept1 or hPEPT1 in the oral absorption of oseltamivir.


Asunto(s)
Antivirales/farmacocinética , Mucosa Intestinal/metabolismo , Oseltamivir/farmacocinética , Simportadores/fisiología , Animales , Células CHO , Cricetinae , Cricetulus , Técnicas In Vitro , Masculino , Transportador de Péptidos 1 , Ratas , Ratas Sprague-Dawley
8.
Mol Pharm ; 6(6): 1716-33, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19739673

RESUMEN

A mechanistic model was applied to quantitatively derive the kinetic parameters from in vitro hepatic uptake transport data. These parameters were used as input to simulate in vivo elimination using a fully mechanistic physiologically based pharmacokinetic (PBPK) model. Fexofenadine and napsagatran, both BDDCS class 3 drugs, were chosen as model compounds. In rat, both compounds are hardly metabolized and are eliminated unchanged mostly through biliary excretion. Uptake was estimated in this study based on plated rat hepatocytes, and a mechanistic model was used to derive the active and passive transport parameters, namely Michaelis-Menten uptake parameters (V(maxI) and K(mI,u)) together with passive diffusion (P(dif)) and nonspecific binding. Maximum transport velocity and passive diffusion were scaled to in vivo parameters (J(maxI) and PS(TC)) using hepatocellularity. Biliary excretion, through passive and active transport, was assessed from in vivo studies. These transport parameters were then used as input in a whole body physiologically based model in which the liver compartment was parametrized for the different passive and active transport processes. Each of the processes was linked to the free concentration in the relevant compartment. For napsagatran hepatic uptake, no passive diffusion and no binding were detected in vitro besides the active transport (K(mI,u) = 88.4 +/- 8.1 microM, V(maxI) = 384 +/- 19 pmol/mg/min). Fexofenadine was rapidly taken up into rat hepatocytes (K(mI,u) = 271 +/- 35 microM, V(maxI) = 3162 +/- 274 pmol/mg/min), and some contribution of passive diffusion to the uptake (P(dif) = 2.08 +/- 0.67 microL/mg/min) was observed. For fexofenadine, the biliary export rate was found to be slower than the uptake, leading to drug accumulation in liver. No accumulation was observed for napsagatran where excretion was faster than hepatic uptake. Observed plasma, liver and bile concentration time profiles were compared to PBPK simulations based on scaled in vitro transport kinetic parameters. An uncertainty analysis indicated that for both compounds the scaled in vitro uptake clearance had to be adjusted with an additional empirical scaling factor of 10 to match the plasma and liver concentrations and biliary excretion profiles. Applying this model, plasma clearance (CL(P)) and half-life (t(1/2)), maximum liver concentration (C(maxL)) and fraction excreted in bile (f(bile)) were predicted within 2-fold. In vitro uptake data had most impact on the simulated plasma and biliary excretion profiles, while accurate simulations of liver concentrations required also quantitative estimates of biliary excretion transport. This study indicated that the mechanistic model allowed for accurate evaluation of in vitro experiments; and the scaled kinetic parameters of hepatic uptake transport enabled the prediction of in vivo PK profiles and plasma clearances, using PBPK modeling.


Asunto(s)
Hepatocitos/metabolismo , Modelos Teóricos , Naftalenos/metabolismo , Piperidinas/metabolismo , Terfenadina/análogos & derivados , Animales , Células Cultivadas , Cinética , Masculino , Modelos Biológicos , Naftalenos/farmacocinética , Piperidinas/farmacocinética , Ratas , Ratas Wistar , Terfenadina/metabolismo , Terfenadina/farmacocinética
9.
J Pharmacokinet Pharmacodyn ; 36(6): 585-611, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19936896

RESUMEN

The aim of this study was to evaluate a strategy based on a physiologically based pharmacokinetic (PBPK) model for the prediction of PK profiles in human using in vitro data when elimination of compounds relies on active transport processes. The strategy was first applied to rat in vivo and in vitro data in order to refine the PBPK model. The model could then be applied to human in vitro uptake transport data using valsartan as a probe substrate. Plated rat and human hepatocytes, and cell lines overexpressing human OATP1B1 and OATP1B3 were used for in vitro uptake experiments. The uptake rate of valsartan was higher for rat hepatocytes (K (m,u) = 28.4 +/- 3.7 muM, V (max) = 1318 +/- 176 pmol/mg/min and P (dif) = 1.21 +/- 0.42 microl/mg/min) compared to human hepatocytes (K (m,u) = 44.4 +/- 14.6 microM, V (max) = 304 +/- 85 pmol/mg/min and P (dif) = 0.724 +/- 0.271 microl/mg/min). OATP1B1 and 1B3 parameters were correlated to human hepatocyte data using experimentally established relative activity factors (RAF). Resulting PBPK simulations using those in vitro data were compared for plasma (human and rat) and bile (rat) concentration-time profiles following i.v. bolus administration of valsartan. An uncertainty analysis indicated that the scaled in vitro uptake clearance had to be adjusted with an additional empirical scaling factor of 5 to match the plasma concentrations and biliary excretion profiles. Applying this model, plasma clearances (CL(P)) for rat and human were predicted within two-fold relative to predictions based on respective in vitro data. The corrected hepatic uptake transport kinetic parameters enabled the prediction of valsartan in vivo PK profiles and plasma clearances, using PBPK modeling. Moreover, the interspecies difference in elimination rate observed in vivo was correctly reflected in the transport parameters determined in vitro. More data are needed to support more general applications of the proposed approach including its use for metabolized compounds.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Hepatocitos/metabolismo , Modelos Biológicos , Transportadores de Anión Orgánico/metabolismo , Tetrazoles/farmacocinética , Valina/análogos & derivados , Administración Oral , Adulto , Anciano , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/sangre , Animales , Bilis/metabolismo , Transporte Biológico Activo , Células CHO , Cricetinae , Cricetulus , Estudios Cruzados , Interacciones Farmacológicas , Femenino , Humanos , Inyecciones Intravenosas , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Persona de Mediana Edad , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Reproducibilidad de los Resultados , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Especificidad de la Especie , Tetrazoles/administración & dosificación , Tetrazoles/sangre , Transfección , Valina/administración & dosificación , Valina/sangre , Valina/farmacocinética , Valsartán
10.
Chem Biodivers ; 6(11): 1975-87, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19937834

RESUMEN

The aim of this study was to evaluate a physiologically based pharmacokinetic (PBPK) model for predicting PK profiles in humans based on a model refined in rats and humans in vitro uptake-transport data using valsartan as a probe substrate. Valsartan is eliminated unchanged, mostly through biliary excretion, both in humans and rats. It was, therefore, chosen as model compound to predict in vivo elimination based on in vitro hepatic uptake-transport data using a fully mechanistic PBPK model. Plated rat and human hepatocytes, and cell lines overexpressing human OATP1B1 and OATP1B3 were used for in vitro uptake experiments. A mechanistic two-compartment model was used to derive the active and passive transport parameters, namely uptake Michaelis-Menten parameters (V(max) and K(m,u)) together with passive diffusion (P(dif)). These transport parameters were then used as input in a whole body physiologically based pharmacokinetic (PBPK) model. The uptake rate of valsartan was higher for rat hepatocytes (K(m,u)=28.4+/-3.7 microM, V(max)=1320+/-180 pmol/mg/min, and P(dif) =1.21+/-0.42 microl/mg/min) compared to human hepatocytes (K(m,u)=44.4+/-14.6 microM, V(max)=304+/-85 pmol/mg/min, and P(dif)=0.724+/-0.271 microl/mg/min). OATP1B1 and -1B3 parameters were correlated to human hepatocyte data, using experimentally established relative activity factors (RAF). Resulting PBPK simulations were compared for plasma- (humans and rats) and bile- (rats) concentration-time profiles following iv bolus administration of valsartan. Plasma clearances (CL(P)) for rats and humans were predicted within twofold relative to predictions based on respective in vitro data. The simulations were extended to simulate the impact of either OATP1B1 or -1B3 inhibition on plasma profile. The limited data set indicates that the mechanistic model allowed for accurate evaluation of in vitro transport data; and the resulting hepatic uptake transport kinetic parameters enabled the prediction of in vivo PK profiles and plasma clearances, using PBPK modelling. Moreover, the interspecies difference in elimination rate observed in vivo was correctly reflected in the transport parameters determined in vitro.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Tetrazoles/farmacocinética , Valina/análogos & derivados , Algoritmos , Animales , Bilis/metabolismo , Transporte Biológico Activo/fisiología , Línea Celular , Simulación por Computador , Interpretación Estadística de Datos , Predicción , Hepatocitos/metabolismo , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado , Modelos Biológicos , Tamaño de los Órganos/fisiología , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Ratas , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Distribución Tisular , Transfección , Valina/farmacocinética , Valsartán
11.
Drug Metab Dispos ; 36(12): 2434-44, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18809732

RESUMEN

The use of in vitro data for quantitative predictions of transporter-mediated elimination in vivo requires an accurate estimation of the transporter Michaelis-Menten parameters, V(max) and K(m), as a first step. Therefore, the experimental conditions of in vitro studies used to assess hepatic uptake transport were optimized regarding active transport processes, nonspecific binding, and passive diffusion (P(dif)). A mechanistic model was developed to analyze and accurately describe these active and passive processes. This two-compartmental model was parameterized to account for nonspecific binding, bidirectional passive diffusion, and active uptake processes based on the physiology of the cells. The model was used to estimate kinetic parameters of in vitro transport data from organic anion-transporting peptide model substrates (e.g., cholecystokinin octapeptide deltorphin II, fexofenadine, and pitavastatin). Data analysis by this mechanistic model significantly improved the accuracy and precision in all derived parameters [mean coefficient of variations (CVs) for V(max) and K(m) were 19 and 23%, respectively] compared with the conventional kinetic method of transport data analysis (mean CVs were 58 and 115%, respectively, using this method). Furthermore, permeability was found to be highly temperature-dependent in Chinese hamster ovary (CHO) control cells and artificial membranes (parallel artificial membrane permeability assay). Whereas for some compounds (taurocholate, estrone-3-sulfate, and propranolol) the effect was moderate (1.5-6-fold higher permeability at 37 degrees C compared with that at 4 degrees C), for fexofenadine a 16-fold higher passive permeability was seen at 37 degrees C. Therefore, P(dif) was better predicted if it was evaluated under the same experimental conditions as V(max) and K(m), i.e., in a single incubation of CHO overexpressed cells or rat hepatocytes at 37 degrees C, instead of a parallel control evaluation at 4 degrees C.


Asunto(s)
Simulación por Computador , Hepatocitos/metabolismo , Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Algoritmos , Animales , Transporte Biológico Activo , Células CHO , Cricetinae , Cricetulus , Difusión , Estrona/análogos & derivados , Estrona/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Fluvastatina , Indoles/metabolismo , Cinética , Masculino , Membranas Artificiales , Naftalenos/metabolismo , Oligopéptidos/metabolismo , Transportadores de Anión Orgánico/genética , Permeabilidad , Piperidinas/metabolismo , Quinolinas/metabolismo , Ratas , Ratas Wistar , Sincalida/metabolismo , Temperatura , Terfenadina/análogos & derivados , Terfenadina/metabolismo
12.
Pharmacol Res Perspect ; 6(6): e00447, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30519476

RESUMEN

Spinal muscular atrophy (SMA) is a rare, inherited neuromuscular disease caused by deletion and/or mutation of the Survival of Motor Neuron 1 (SMN1) gene. A second gene, SMN2, produces low levels of functional SMN protein that are insufficient to fully compensate for the lack of SMN1. Risdiplam (RG7916; RO7034067) is an orally administered, small-molecule SMN2 pre-mRNA splicing modifier that distributes into the central nervous system (CNS) and peripheral tissues. To further explore risdiplam distribution, we assessed in vitro characteristics and in vivo drug levels and effect of risdiplam on SMN protein expression in different tissues in animal models. Total drug levels were similar in plasma, muscle, and brain of mice (n = 90), rats (n = 148), and monkeys (n = 24). As expected mechanistically based on its high passive permeability and not being a human multidrug resistance protein 1 substrate, risdiplam CSF levels reflected free compound concentration in plasma in monkeys. Tissue distribution remained unchanged when monkeys received risdiplam once daily for 39 weeks. A parallel dose-dependent increase in SMN protein levels was seen in CNS and peripheral tissues in two SMA mouse models dosed with risdiplam. These in vitro and in vivo preclinical data strongly suggest that functional SMN protein increases seen in patients' blood following risdiplam treatment should reflect similar increases in functional SMN protein in the CNS, muscle, and other peripheral tissues.


Asunto(s)
Compuestos Azo/farmacocinética , Atrofia Muscular Espinal/tratamiento farmacológico , Fármacos Neuromusculares/farmacocinética , Pirimidinas/farmacocinética , Empalme del ARN/efectos de los fármacos , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Compuestos Azo/líquido cefalorraquídeo , Compuestos Azo/farmacología , Compuestos Azo/uso terapéutico , Encéfalo/metabolismo , Encéfalo/patología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Perros , Evaluación Preclínica de Medicamentos , Exones/efectos de los fármacos , Exones/genética , Femenino , Humanos , Macaca fascicularis , Células de Riñón Canino Madin Darby , Masculino , Ratones , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Fármacos Neuromusculares/líquido cefalorraquídeo , Fármacos Neuromusculares/farmacología , Fármacos Neuromusculares/uso terapéutico , Pirimidinas/líquido cefalorraquídeo , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Ratas , Ratas Wistar , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Porcinos , Distribución Tisular
13.
J Med Chem ; 61(15): 6501-6517, 2018 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-30044619

RESUMEN

SMA is an inherited disease that leads to loss of motor function and ambulation and a reduced life expectancy. We have been working to develop orally administrated, systemically distributed small molecules to increase levels of functional SMN protein. Compound 2 was the first SMN2 splicing modifier tested in clinical trials in healthy volunteers and SMA patients. It was safe and well tolerated and increased SMN protein levels up to 2-fold in patients. Nevertheless, its development was stopped as a precautionary measure because retinal toxicity was observed in cynomolgus monkeys after chronic daily oral dosing (39 weeks) at exposures in excess of those investigated in patients. Herein, we describe the discovery of 1 (risdiplam, RG7916, RO7034067) that focused on thorough pharmacology, DMPK and safety characterization and optimization. This compound is undergoing pivotal clinical trials and is a promising medicine for the treatment of patients in all ages and stages with SMA.


Asunto(s)
Compuestos Azo/farmacología , Descubrimiento de Drogas , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Pirimidinas/farmacología , Empalme del ARN/efectos de los fármacos , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Compuestos Azo/efectos adversos , Compuestos Azo/uso terapéutico , Humanos , Pirimidinas/efectos adversos , Pirimidinas/uso terapéutico , Seguridad
14.
Curr Opin Drug Discov Devel ; 10(1): 74-83, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17265745

RESUMEN

Pharmacokinetic drug-drug interactions (DDIs) are a major concern in drug development. Drug transport, along with drug metabolism via cytochrome P450s (CYPs), is increasingly being considered as an integral part of the overall pharnmacokinetics profile of a drug. Inhibition of transporters can lead to altered pharmacokinetics, potentially interfering with drug safety and efficacy. There is an increasing number of DDIs observed with statins, which are widely used in combination therapies, and this can be partly attributed to inhibition of individual hepatic transporters. Studies of these inhibitory interactions in vitro has indicated the importance of both hepatic solute carriers of the organic anion transporting polypeptide (OATP) superfamily and CYP inhibition. Mathematical models have been developed to gain more quantitative insights into the interplay between transport and metabolism of drugs. This article reviews new developments in the area of in vitro tools and modeling approaches that are used to study DDIs related to OATP transporters, with a focus on the clinical relevance of the transport-mediated DDIs involving statins.


Asunto(s)
Interacciones Farmacológicas , Transportadores de Anión Orgánico/metabolismo , Tecnología Farmacéutica/métodos , Animales , Ciclosporina/metabolismo , Ciclosporina/farmacocinética , Ciclosporina/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Modelos Teóricos , Rifampin/metabolismo , Rifampin/farmacocinética , Rifampin/farmacología
15.
Clin Pharmacokinet ; 55(2): 237-47, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26341813

RESUMEN

OBJECTIVE: To assess the effect of strong and moderate cytochrome P450 (CYP) 3A4 inhibition on exposure of bitopertin, a glycine reuptake inhibitor primarily metabolized by CYP3A4, and to compare the results with predictions based on physiologically based pharmacokinetic (PBPK) modelling. METHODS: The effects of ketoconazole and erythromycin were assessed in two male volunteer studies with open-label, two-period, fixed-sequence designs. Twelve subjects were enrolled in each of the studies. In period 1, a single dose of bitopertin was administered; in period 2, 400 mg ketoconazole was administered once daily for 17 days or 500 mg erythromycin was administered twice daily for 21 days. A single dose of bitopertin was coadministered on day 5. Pharmacokinetic parameters were derived by non-compartmental methods. Simulated bitopertin profiles using dynamic PBPK modelling for a typical healthy volunteer in GastroPlus(®) were used to predict changes in pharmacokinetic parameters. RESULTS: In healthy volunteers, coadministration of ketoconazole increased the bitopertin area under the plasma concentration-time curve (AUC) from 0 to 312 h (AUC0-312h) 4.2-fold (90 % confidence interval [CI] 3.5-5.0) and erythromycin increased the AUC from time zero to infinity (AUC0-inf) 2.1-fold (90 % CI 1.9-2.3). The peak concentration (C max) increased by <25 % in both studies. Simulated bitopertin profiles using PBPK modelling showed good agreement with the observed AUC ratios in both studies. The predicted AUC0-inf ratios for the interaction with ketoconazole and erythromycin were 7.7 and 1.9, respectively. CONCLUSION: Strong CYP3A4 inhibitors increase AUC0-inf of bitopertin 7- to 8-fold and hence should not be administered concomitantly with bitopertin. Moderate CYP3A4 inhibitors double AUC0-inf.


Asunto(s)
Inhibidores del Citocromo P-450 CYP3A/farmacología , Eritromicina/farmacología , Cetoconazol/farmacología , Modelos Biológicos , Piperazinas/farmacocinética , Sulfonas/farmacocinética , Adulto , Interacciones Farmacológicas , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Piperazinas/sangre , Sulfonas/sangre , Adulto Joven
16.
J Med Chem ; 59(13): 6086-100, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27299419

RESUMEN

Spinal muscular atrophy (SMA) is the leading genetic cause of infant and toddler mortality, and there is currently no approved therapy available. SMA is caused by mutation or deletion of the survival motor neuron 1 (SMN1) gene. These mutations or deletions result in low levels of functional SMN protein. SMN2, a paralogous gene to SMN1, undergoes alternative splicing and exclusion of exon 7, producing an unstable, truncated SMNΔ7 protein. Herein, we report the identification of a pyridopyrimidinone series of small molecules that modify the alternative splicing of SMN2, increasing the production of full-length SMN2 mRNA. Upon oral administration of our small molecules, the levels of full-length SMN protein were restored in two mouse models of SMA. In-depth lead optimization in the pyridopyrimidinone series culminated in the selection of compound 3 (RG7800), the first small molecule SMN2 splicing modifier to enter human clinical trials.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Atrofia Muscular Espinal/tratamiento farmacológico , Pirimidinonas/química , Pirimidinonas/farmacología , ARN Mensajero/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Exones/efectos de los fármacos , Humanos , Ratones , Atrofia Muscular Espinal/genética , Pirimidinonas/farmacocinética , Pirimidinonas/uso terapéutico
17.
Clin Pharmacokinet ; 54(5): 537-49, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25488594

RESUMEN

BACKGROUND AND OBJECTIVES: Understanding transmembrane transport provides a more complete understanding of the pharmacokinetics of a drug and mechanistic explanations for drug-drug interactions. Here, the transmembrane transport of danoprevir (hepatitis C virus protease inhibitor) and the effects of ritonavir and ciclosporin on transmembrane transport of danoprevir were evaluated and clinical pharmacokinetic studies of danoprevir co-administered with/without ritonavir and ciclosporin were conducted. METHODS: Transcellular transport of danoprevir was evaluated in Lewis lung cancer porcine kidney, Madin-Darby canine kidney, or Chinese hamster ovary cells transfected with human transport proteins, and in human hepatocytes. The pharmacokinetics of intravenous and oral danoprevir administered with/without ritonavir, and the impact of ciclosporin on danoprevir pharmacokinetics were evaluated in randomized, open-label, crossover studies in healthy subjects. RESULTS: Danoprevir transport in vitro involved organic anion transporting polypeptide (OATP) 1B1, OATP1B3, P-glycoprotein, and multidrug resistance protein-2, but not breast cancer resistance protein. Ritonavir and ciclosporin inhibited transport of danoprevir by human hepatocytes. The pharmacokinetics of intravenous danoprevir 6 mg were not altered by oral ritonavir 100 mg. In contrast, exposure to oral danoprevir 100 mg increased two- to threefold when co-administered with ritonavir. Absolute bioavailability of danoprevir 100 mg was low (1.15%), but increased more than threefold (3.86%) when co-administered with ritonavir. Oral ciclosporin 100 mg increased exposure to intravenous danoprevir 2 mg and oral ritonavir 100 mg. CONCLUSION: Collectively, these studies provide insight into the transmembrane transport and pharmacokinetics of danoprevir and the mechanisms that underlie a recently reported, three-way drug-drug interaction involving danoprevir, ritonavir, and ciclosporin.


Asunto(s)
Lactamas/farmacocinética , Proteínas de Transporte de Membrana/metabolismo , Inhibidores de Proteasas/farmacocinética , Sulfonamidas/farmacocinética , Adolescente , Adulto , Animales , Antivirales/farmacocinética , Antivirales/farmacología , Disponibilidad Biológica , Células CHO , Carcinoma Pulmonar de Lewis/metabolismo , Cricetinae , Cricetulus , Estudios Cruzados , Ciclopropanos , Ciclosporina/farmacología , Perros , Interacciones Farmacológicas , Femenino , Hepatocitos/metabolismo , Humanos , Isoindoles , Lactamas/farmacología , Lactamas Macrocíclicas , Células de Riñón Canino Madin Darby , Masculino , Persona de Mediana Edad , Prolina/análogos & derivados , Inhibidores de Proteasas/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Ritonavir/farmacología , Sulfonamidas/farmacología , Porcinos , Adulto Joven
18.
Biochem Pharmacol ; 85(11): 1684-99, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23454189

RESUMEN

The unbound drug concentration in brain parenchyma is considered to be the relevant driver for interaction with central nervous system (CNS) biological targets. Drug levels in cerebrospinal fluid (C_CSF) are frequently used surrogates for the unbound concentrations in brain. For drugs actively transported across the blood-brain barrier (BBB), C_CSF differs from unbound plasma concentration (Cu_p) to an extent that is commonly unknown. In this study, the relationship between CSF-to-unbound plasma drug partitioning in rats and the mouse Pgp (Mdr1a) efflux ratio (ER) obtained from in vitro transcellular studies has been investigated for a set of 61 CNS compounds exhibiting substantial diversity in chemical structure and physico-chemical properties. In order to understand the in vitro-in vivo extrapolation of Pgp efflux, a mechanistic model was derived relating in vivo CNS distribution kinetics to in vitro active transport. The model was applied to predict C_CSF from Cu_p and ER data for 19 proprietary Roche CNS drug candidates. The calculated CSF concentrations were correlated with CNS pharmacodynamic responses observed in rodent models. The correlation between in vitro and in vivo potency for different pharmacological endpoints indicated that the predicted C_CSF is a valuable surrogate of the concentration at the target site. Overall, C_CSF proved superior description of PK/PD data than unbound plasma or total brain concentration for Mdr1a substrates. Predicted C_CSF can be used as a default approach to understand the PK/PD relationships in CNS efficacy models and can support the extrapolation of efficacious brain exposure for new drug candidates from rodent to man.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Fármacos del Sistema Nervioso Central/farmacología , Fármacos del Sistema Nervioso Central/farmacocinética , Descubrimiento de Drogas , Animales , Proteínas Sanguíneas/metabolismo , Fármacos del Sistema Nervioso Central/líquido cefalorraquídeo , Análisis por Conglomerados , Células LLC-PK1 , Ratones , Modelos Teóricos , Ratas , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA