Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 61(10): 4635-4640, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29718668

RESUMEN

We disclose the discovery and X-ray cocrystal data of potent, selective quinazoline inhibitors of PDE1. Inhibitor ( S)-3 readily attains free plasma concentrations above PDE1 IC50 values and has restricted brain access. The racemic compound 3 inhibits >75% of PDE hydrolytic activity in soluble samples of human myocardium, consistent with heightened PDE1 activity in this tissue. These compounds represent promising new tools to probe the value of PDE1 inhibition in the treatment of cardiovascular disease.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Descubrimiento de Drogas , Miocardio/enzimología , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Quinazolinas/química , AMP Cíclico/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Conformación Proteica
2.
Neurodegener Dis ; 12(1): 36-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22922480

RESUMEN

BACKGROUND: Reducing brain ß-amyloid (Aß) via inhibition of ß-secretase, or inhibition/modulation of γ-secretase, has been widely pursued as a potential disease-modifying treatment for Alzheimer's disease. Compounds that act through these mechanisms have been screened and characterized with Aß lowering in the brain and/or cerebrospinal fluid (CSF) as the primary pharmacological end point. Interpretation and translation of the pharmacokinetic (PK)/pharmacodynamic (PD) relationship for these compounds is complicated by the relatively slow Aß turnover process in these compartments. OBJECTIVE: To understand Aß turnover kinetics in preclinical species and humans. METHODS: We collected CSF Aß dynamic data after ß- or γ-secretase inhibitor treatment from in-house experiments and the public domain, and analyzed the data using PK/PD modeling to obtain CSF Aß turnover rates (kout) in the mouse, dog, monkey and human. RESULTS: The kout for CSF Aß40 follows allometry (kout = 0.395 × body weight(-0.351)). The kout for CSF Aß40 is approximately 2-fold higher than the turnover of CSF in rodents, but in higher species, the two are comparable. CONCLUSION: The turnover of CSF Aß40 was systematically examined, for the first time, in multiple species through quantitative modeling of multiple data sets. Our result suggests that the clearance mechanisms for CSF Aß in rodents may be different from those in the higher species. The understanding of Aß turnover has considerable implications for the discovery and development of Aß-lowering therapeutics, as illustrated from the perspectives of preclinical PK/PD characterization and preclinical-to-clinical translation.


Asunto(s)
Péptidos beta-Amiloides/líquido cefalorraquídeo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/sangre , Animales , Perros , Humanos , Macaca fascicularis , Ratones , Oligopéptidos/farmacología , Oxadiazoles/farmacología , Pirimidinas/farmacología , Sulfonamidas/farmacología , Tiazinas/farmacología
3.
J Med Chem ; 55(21): 9055-68, 2012 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-23025719

RESUMEN

Phosphodiesterase 9A inhibitors have shown activity in preclinical models of cognition with potential application as novel therapies for treating Alzheimer's disease. Our clinical candidate, PF-04447943 (2), demonstrated acceptable CNS permeability in rats with modest asymmetry between central and peripheral compartments (free brain/free plasma = 0.32; CSF/free plasma = 0.19) yet had physicochemical properties outside the range associated with traditional CNS drugs. To address the potential risk of restricted CNS penetration with 2 in human clinical trials, we sought to identify a preclinical candidate with no asymmetry in rat brain penetration and that could advance into development. Merging the medicinal chemistry strategies of structure-based design with parallel chemistry, a novel series of PDE9A inhibitors was identified that showed improved selectivity over PDE1C. Optimization afforded preclinical candidate 19 that demonstrated free brain/free plasma ≥ 1 in rat and reduced microsomal clearance along with the ability to increase cyclic guanosine monophosphosphate levels in rat CSF.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Azetidinas/química , Barrera Hematoencefálica/metabolismo , Pirazoles/química , Pirazoles/síntesis química , Pirimidinas/química , Pirimidinas/síntesis química , Pirimidinonas/química , 3',5'-AMP Cíclico Fosfodiesterasas/química , Administración Oral , Animales , Azetidinas/síntesis química , Azetidinas/farmacocinética , Cristalografía por Rayos X , GMP Cíclico/líquido cefalorraquídeo , Ciclopentanos/síntesis química , Ciclopentanos/química , Ciclopentanos/farmacocinética , Bases de Datos Factuales , Perros , Diseño de Fármacos , Humanos , Modelos Moleculares , Estructura Molecular , Pirazoles/farmacocinética , Pirimidinas/farmacocinética , Pirimidinonas/síntesis química , Pirimidinonas/farmacocinética , Ratas , Estereoisomerismo , Relación Estructura-Actividad
4.
J Pharmacol Exp Ther ; 342(2): 366-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22562771

RESUMEN

Reducing the generation of amyloid-ß (Aß) in the brain via inhibition of ß-secretase or inhibition/modulation of γ-secretase has been pursued as a potential disease-modifying treatment for Alzheimer's disease. For the discovery and development of ß-secretase inhibitors (BACEi), γ-secretase inhibitors (GSI), and γ-secretase modulators (GSM), Aß in cerebrospinal fluid (CSF) has been presumed to be an effect biomarker for Aß lowering in the brain. However, this presumption is challenged by the lack of quantitative understanding of the relationship between brain and CSF Aß lowering. In this study, we strived to elucidate how the intrinsic pharmacokinetic (PK)/pharmacodynamic (PD) relationship for CSF Aß lowering is related to that for brain Aß through quantitative modeling of preclinical data for numerous BACEi, GSI, and GSM across multiple species. Our results indicate that the intrinsic PK/PD relationship in CSF is predictive of that in brain, at least in the postulated pharmacologically relevant range, with excellent consistency across mechanisms and species. As such, the validity of CSF Aß as an effect biomarker for brain Aß lowering is confirmed preclinically. Meanwhile, we have been able to reproduce the dose-dependent separation between brain and CSF effect profiles using simulations. We further discuss the implications of our findings to drug discovery and development with regard to preclinical PK/PD characterization and clinical prediction of Aß lowering in the brain.


Asunto(s)
Péptidos beta-Amiloides/líquido cefalorraquídeo , Encéfalo/metabolismo , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/líquido cefalorraquídeo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/metabolismo , Líquido Cefalorraquídeo/química , Cobayas , Masculino , Ratones , Ratones de la Cepa 129 , Inhibidores de Proteasas/farmacología , Ratas , Ratas Sprague-Dawley
5.
J Med Chem ; 55(7): 3414-24, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22420884

RESUMEN

Replacement of the central, para-substituted fluorophenyl ring in the γ-secretase inhibitor 1 (BMS-708,163) with the bicyclo[1.1.1]pentane motif led to the discovery of compound 3, an equipotent enzyme inhibitor with significant improvements in passive permeability and aqueous solubility. The modified biopharmaceutical properties of 3 translated into excellent oral absorption characteristics (~4-fold ↑ C(max) and AUC values relative to 1) in a mouse model of γ-secretase inhibition. In addition, SAR studies into other fluorophenyl replacements indicate the intrinsic advantages of the bicyclo[1.1.1]pentane moiety over conventional phenyl ring replacements with respect to achieving an optimal balance of properties (e.g., γ-secretase inhibition, aqueous solubility/permeability, in vitro metabolic stability). Overall, this work enhances the scope of the [1.1.1]-bicycle beyond that of a mere "spacer" unit and presents a compelling case for its broader application as a phenyl group replacement in scenarios where the aromatic ring count impacts physicochemical parameters and overall drug-likeness.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Compuestos Bicíclicos con Puentes/síntesis química , Oxadiazoles/síntesis química , Pentanos/síntesis química , Sulfonamidas/síntesis química , Administración Oral , Animales , Disponibilidad Biológica , Encéfalo/metabolismo , Compuestos Bicíclicos con Puentes/farmacocinética , Compuestos Bicíclicos con Puentes/farmacología , Línea Celular , Perros , Femenino , Humanos , Ratones , Microsomas Hepáticos/metabolismo , Oxadiazoles/farmacocinética , Oxadiazoles/farmacología , Pentanos/farmacocinética , Pentanos/farmacología , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Distribución Tisular
6.
J Med Chem ; 54(22): 7772-83, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21995460

RESUMEN

A metabolism-based approach toward the optimization of a series of N-arylsulfonamide-based γ-secretase inhibitors is reported. The lead cyclohexyl analogue 6 suffered from extensive oxidation on the cycloalkyl motif by cytochrome P450 3A4, translating into poor human liver microsomal stability. Knowledge of the metabolic pathways of 6 triggered a structure-activity relationship study aimed at lowering lipophilicity through the introduction of polarity. This effort led to several tetrahydropyran and tetrahydrofuran analogues, wherein the 3- and 4-substituted variants exhibited greater microsomal stability relative to their 2-substituted counterparts. Further reduction in lipophilicity led to the potent γ-secretase inhibitor and 3-substituted oxetane 1 with a reduced propensity toward oxidative metabolism, relative to its 2-substituted isomer. The slower rates of metabolism with 3-substituted cyclic ethers most likely originate from reductions in lipophilicity and/or unfavorable CYP active site interactions with the heteroatom. Preliminary animal pharmacology studies with a representative oxetane indicate that the series is generally capable of lowering Aß in vivo. As such, the study also illustrates the improvement in druglikeness of molecules through the use of the oxetane motif.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Éteres Cíclicos/síntesis química , Sulfonamidas/síntesis química , Péptidos beta-Amiloides/líquido cefalorraquídeo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular , Cristalografía por Rayos X , Perros , Diseño de Fármacos , Éteres Cíclicos/metabolismo , Éteres Cíclicos/farmacología , Humanos , Técnicas In Vitro , Ratones , Microsomas Hepáticos/metabolismo , Oxidación-Reducción , Receptores Notch/metabolismo , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Distribución Tisular
7.
J Pharmacol Exp Ther ; 339(3): 922-34, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21930801

RESUMEN

Alzheimer's disease (AD) poses a serious public health threat to the United States. Disease-modifying drugs slowing AD progression are in urgent need, but they are still unavailable. According to the amyloid cascade hypothesis, inhibition of ß- or γ-secretase, key enzymes for the production of amyloid ß (Aß), may be viable mechanisms for the treatment of AD. For the discovery of γ-secretase inhibitors (GSIs), the APP-overexpressing Tg2576 mouse has been the preclinical model of choice, in part because of the ease of detection of Aß species in its brain, plasma, and cerebrospinal fluid (CSF). Some biological observations and practical considerations, however, argue against the use of the Tg2576 mouse. We reasoned that an animal model would be suitable for GSI discovery if the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a compound for Aß lowering in this model is predictive of that in human. In this study, we assessed whether the background 129/SVE strain is a suitable preclinical pharmacology model for identifying new GSIs by evaluating the translatability of the intrinsic PK/PD relationships for brain and CSF Aß across the Tg2576 and 129/SVE mouse and human. Using semimechanistically based PK/PD modeling, our analyses indicated that the intrinsic PK/PD relationship for brain Aßx-42 and CSF Aßx-40 in the 129/SVE mouse is indicative of that for human CSF Aß. This result, in conjunction with practical considerations, strongly suggests that the 129/SVE mouse is a suitable model for GSI discovery. Concurrently, the necessity and utilities of PK/PD modeling for rational interpretation of Aß data are established.


Asunto(s)
Alanina/análogos & derivados , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Azepinas/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Oxadiazoles/farmacología , Sulfonamidas/farmacología , Alanina/sangre , Alanina/farmacocinética , Alanina/farmacología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/sangre , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Azepinas/sangre , Azepinas/farmacocinética , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacocinética , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Modelos Animales , Oxadiazoles/sangre , Oxadiazoles/farmacocinética , Bibliotecas de Moléculas Pequeñas , Sulfonamidas/sangre , Sulfonamidas/farmacocinética
8.
Neuropharmacology ; 61(5-6): 1001-15, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21763704

RESUMEN

Observations that N-Methyl-D-Aspartate (NMDA) antagonists produce symptoms in humans that are similar to those seen in schizophrenia have led to the current hypothesis that schizophrenia might result from NMDA receptor hypofunction. Inhibition of D-amino acid oxidase (DAAO), the enzyme responsible for degradation of D-serine, should lead to increased levels of this co-agonist at the NMDA receptor, and thereby provide a therapeutic approach to schizophrenia. We have profiled some of the preclinical biochemical, electrophysiological, and behavioral consequences of administering potent and selective inhibitors of DAAO to rodents to begin to test this hypothesis. Inhibition of DAAO activity resulted in a significant dose and time dependent increase in D-serine only in the cerebellum, although a time delay was observed between peak plasma or brain drug concentration and cerebellum D-serine response. Pharmacokinetic/pharmacodynamic (PK/PD) modeling employing a mechanism-based indirect response model was used to characterize the correlation between free brain drug concentration and D-serine accumulation. DAAO inhibitors had little or no activity in rodent models considered predictive for antipsychotic activity. The inhibitors did, however, affect cortical activity in the Mescaline-Induced Scratching model, produced a modest but significant increase in NMDA receptor-mediated synaptic currents in primary neuronal cultures from rat hippocampus, and resulted in a significant increase in evoked hippocampal theta rhythm, an in vivo electrophysiological model of hippocampal activity. These findings demonstrate that although DAAO inhibition did not cause a measurable increase in D-serine in forebrain, it did affect hippocampal and cortical activity, possibly through augmentation of NMDA receptor-mediated currents.


Asunto(s)
Encéfalo/metabolismo , D-Aminoácido Oxidasa/antagonistas & inhibidores , Memoria a Corto Plazo/fisiología , Agitación Psicomotora/tratamiento farmacológico , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Encéfalo/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología , GMP Cíclico/análisis , GMP Cíclico/biosíntesis , D-Aminoácido Oxidasa/metabolismo , D-Aminoácido Oxidasa/fisiología , Evaluación Preclínica de Medicamentos , Electroencefalografía , Habituación Psicofisiológica/efectos de los fármacos , Habituación Psicofisiológica/fisiología , Harmalina/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/efectos de los fármacos , Mescalina/farmacología , Ratones , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Modelos Biológicos , Modelos Químicos , Terapia Molecular Dirigida , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Prurito/inducido químicamente , Prurito/prevención & control , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/fisiología , Serina/sangre , Agonistas de Receptores de Serotonina/farmacología
9.
Bioorg Med Chem Lett ; 21(9): 2631-6, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21269825

RESUMEN

The synthesis and structure-activity relationship (SAR) of a novel series of di-substituted imidazoles, derived from modification of DAPT, are described. Subsequent optimization led to identification of a highly potent series of inhibitors that contain a ß-amine in the imidazole side-chain resulting in a robust in vivo reduction of plasma and brain Aß in guinea pigs. The therapeutic index between Aß reductions and changes in B-cell populations were studied for compound 10 h.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Imidazoles/síntesis química , Imidazoles/farmacología , Aminación/efectos de los fármacos , Péptidos beta-Amiloides/sangre , Péptidos beta-Amiloides/metabolismo , Animales , Bioensayo , Diamida/síntesis química , Diamida/química , Diamida/farmacología , Inhibidores Enzimáticos/química , Cobayas , Células HeLa , Humanos , Imidazoles/química , Concentración 50 Inhibidora , Estructura Molecular , Relación Estructura-Actividad
10.
Bioorg Med Chem Lett ; 21(9): 2637-40, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21269827

RESUMEN

A novel series of tetralin containing amino imidazoles, derived from modification of the corresponding phenyl acetic acid derivatives is described. Replacement of the amide led to identification of a potent series of tetralin-amino imidazoles with robust central efficacy. The reduction of brain Aß in guinea pigs in the absence of changes in B-cells suggested a potential therapeutic index with respect to APP processing compared with biomarkers of notch related toxicity. Optimization of the FTOC to plasma concentrations at the brain Aß EC(50) lead to the identification of compound 14f (PF-3084014) which was selected for clinical development.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Tetrahidronaftalenos/síntesis química , Tetrahidronaftalenos/farmacología , Valina/análogos & derivados , Animales , Bioensayo , Diseño de Fármacos , Inhibidores Enzimáticos/química , Cobayas , Imidazoles/síntesis química , Imidazoles/química , Imidazoles/farmacología , Concentración 50 Inhibidora , Estructura Molecular , Relación Estructura-Actividad , Tetrahidronaftalenos/química , Valina/síntesis química , Valina/química , Valina/farmacología
11.
J Pharmacol Exp Ther ; 334(1): 269-77, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20363853

RESUMEN

PF-3084014 [(S)-2-((S)-5,7-difluoro-1,2,3,4-tetrahydronaphthalen-3-ylamino)-N-(1-(2-methyl-1-(neopentylamino)propan-2-yl)-1H-imidazol-4-yl)pentanamide] is a novel gamma-secretase inhibitor that reduces amyloid-beta (Abeta) production with an in vitro IC(50) of 1.2 nM (whole-cell assay) to 6.2 nM (cell-free assay). This compound inhibits Notch-related T- and B-cell maturation in an in vitro thymocyte assay with an EC(50) of 2.1 microM. A single acute dose showed dose-dependent reduction in brain, cerebrospinal fluid (CSF), and plasma Abeta in Tg2576 mice as measured by enzyme-linked immunosorbent assay and immunoprecipitation (IP)/mass spectrometry (MS). Guinea pigs were dosed with PF-3084014 for 5 days via osmotic minipump at 0.03 to 3 mg/kg/day and exhibited dose-dependent reduction in brain, CSF, and plasma Abeta. To further characterize Abeta dynamics in brain, CSF, and plasma in relation to drug exposure and Notch-related toxicities, guinea pigs were dosed with 0.03 to 10 mg/kg PF-3084014, and tissues were collected at regular intervals from 0.75 to 30 h after dose. Brain, CSF, and plasma all exhibited dose-dependent reductions in Abeta, and the magnitude and duration of Abeta lowering exceeded those of the reductions in B-cell endpoints. Other gamma-secretase inhibitors have shown high potency at elevating Abeta in the conditioned media of whole cells and the plasma of multiple animal models and humans. Such potentiation was not observed with PF-3084014. IP/MS analysis, however, revealed dose-dependent increases in Abeta11-40 and Abeta1-43 at doses that potently inhibited Abeta1-40 and Abeta1-42. PF-3084014, like previously described gamma-secretase inhibitors, preferentially reduced Abeta1-40 relative to Abeta1-42. Potency at Abeta relative to Notch-related endpoints in vitro and in vivo suggests that a therapeutic index can be achieved with this compound.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/farmacocinética , Tetrahidronaftalenos/farmacología , Tetrahidronaftalenos/farmacocinética , Valina/análogos & derivados , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Línea Celular , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/química , Escherichia coli/genética , Femenino , Cobayas , Humanos , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos , Estructura Molecular , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Bazo/citología , Bazo/efectos de los fármacos , Tetrahidronaftalenos/efectos adversos , Tetrahidronaftalenos/química , Distribución Tisular , Transfección , Valina/efectos adversos , Valina/química , Valina/farmacocinética , Valina/farmacología
12.
J Med Chem ; 52(11): 3576-85, 2009 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-19438227

RESUMEN

3-Hydroxyquinolin-2(1H)-one (2) was discovered by high throughput screening in a functional assay to be a potent inhibitor of human DAAO, and its binding affinity was confirmed in a Biacore assay. Cocrystallization of 2 with the human DAAO enzyme defined the binding site and guided the design of new analogues. The SAR, pharmacokinetics, brain exposure, and effects on cerebellum D-serine are described. Subsequent evaluation against the rat DAAO enzyme revealed a divergent SAR versus the human enzyme and may explain the high exposures of drug necessary to achieve significant changes in rat or mouse cerebellum D-serine.


Asunto(s)
D-Aminoácido Oxidasa/antagonistas & inhibidores , Hidroxiquinolinas/farmacología , Hidroxiquinolinas/farmacocinética , Animales , Cerebelo/metabolismo , Cristalografía por Rayos X , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Humanos , Hidroxiquinolinas/síntesis química , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , Relación Estructura-Actividad
13.
Drug Metab Dispos ; 34(9): 1443-7, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16760229

RESUMEN

This study was designed to evaluate the use of cerebrospinal fluid (CSF) drug concentration and plasma unbound concentration (C(u,plasma)) to predict brain unbound concentration (C(u,brain)). The concentration-time profiles in CSF, plasma, and brain of seven model compounds were determined after subcutaneous administration in rats. The C(u,brain) was estimated from the product of total brain concentrations and unbound fractions, which were determined using brain tissue slice and brain homogenate methods. For theobromine, theophylline, caffeine, fluoxetine, and propranolol, which represent rapid brain penetration compounds with a simple diffusion mechanism, the ratios of the area under the curve of C(u,brain)/C(CSF) and C(u,brain)/C(u,plasma) were 0.27 to 1.5 and 0.29 to 2.1, respectively, using the brain slice method, and were 0.27 to 2.9 and 0.36 to 3.9, respectively, using the brain homogenate method. A P-glycoprotein substrate, CP-141938 (methoxy-3-[(2-phenyl-piperadinyl-3-amino)-methyl]-phenyl-N-methyl-methane-sulfonamide), had C(u,brain)/C(CSF) and C(u,brain)/C(u,plasma) ratios of 0.57 and 0.066, using the brain slice method, and 1.1 and 0.13, using the brain homogenate method, respectively. The slow brain-penetrating compound, N[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl-]sarcosine, had C(u,brain)/C(CSF) and C(u,brain)/C(u,plasma) ratios of 0.94 and 0.12 using the brain slice method and 0.15 and 0.018 using the brain homogenate method, respectively. Therefore, for quick brain penetration with simple diffusion mechanism compounds, C(CSF) and C(u,plasma) represent C(u,brain) equally well; for efflux substrates or slow brain penetration compounds, C(CSF) appears to be equivalent to or more accurate than C(u,plasma) to represent C(u,brain). Thus, we hypothesize that C(CSF) is equivalent to or better than C(u,plasma) to predict C(u,brain). This hypothesis is supported by the literature data.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Fluoxetina/líquido cefalorraquídeo , Teobromina/líquido cefalorraquídeo , Teofilina/líquido cefalorraquídeo , Animales , Evaluación Preclínica de Medicamentos/métodos , Fluoxetina/sangre , Fluoxetina/farmacocinética , Masculino , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Teobromina/sangre , Teobromina/farmacocinética , Teofilina/sangre , Teofilina/farmacocinética
14.
J Pharmacol Exp Ther ; 313(3): 1254-62, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15743928

RESUMEN

This study was designed 1) to examine the effects of blood-brain barrier (BBB) permeability [quantified as permeability-surface area product (PS)], unbound fraction in plasma (f(u,plasma)), and brain tissue (f(u,brain)) on the time to reach equilibrium between brain and plasma and 2) to investigate the drug discovery strategies to design and select compounds that can rapidly penetrate the BBB and distribute to the site of action. The pharmacokinetics of seven model compounds: caffeine, CP-141938 [methoxy-3-[(2-phenyl-piperadinyl-3-amino)-methyl]-phenyl-N-methyl-methane-sulfonamide], fluoxetine, NFPS [N[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine], propranolol, theobromine, and theophylline in rat brain and plasma after subcutaneous administration were studied. The in vivo log PS and log f(u,brain) calculated using a physiologically based pharmacokinetic model correlates with in situ log PS (R(2) = 0.83) and in vitro log f(u,brain) (R(2) = 0.69), where the in situ PS and in vitro f(u,brain) was determined using in situ brain perfusion and equilibrium dialysis using brain homogenate, respectively. The time to achieve brain equilibrium can be quantitated with a proposed parameter, intrinsic brain equilibrium half-life [t(1/2eq,in) = V(b)ln2/(PS . f(u,brain))], where V(b) is the physiological volume of brain. The in vivo log t(1/2eq,in) does not correlate with in situ log PS (R(2) < 0.01) but correlates inversely with log(PS . f(u,brain)) (R(2) = 0.85). The present study demonstrates that rapid brain equilibration requires a combination of high BBB permeability and low brain tissue binding. A high BBB permeability alone cannot guarantee a rapid equilibration. The strategy to select compounds with rapid brain equilibration in drug discovery should identify compounds with high BBB permeability and low nonspecific binding in brain tissue.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Barrera Hematoencefálica , Encéfalo/metabolismo , Farmacocinética , Animales , Semivida , Masculino , Modelos Biológicos , Permeabilidad , Unión Proteica , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...