Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Nucl Med ; 60(7): 992-997, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30530832

RESUMEN

ß-secretase 1 (BACE1) is a key enzyme in the generation of ß-amyloid, which is accumulated in the brain of Alzheimer disease patients. PF-06684511 was identified as a candidate PET ligand for imaging BACE1 in the brain and showed high specific binding in an initial assessment in a nonhuman primate (NHP) PET study using 18F-PF-06684511. In this effort, we aimed to quantitatively evaluate the regional brain distribution of 18F-PF-06684511 in NHPs under baseline and blocking conditions and to assess the target occupancy of BACE1 inhibitors. In addition, NHP whole-body PET measurements were performed to estimate the effective radiation dose. Methods: Initial brain PET measurements were performed at baseline and after oral administration of 5 mg/kg of LY2886721, a BACE1 inhibitor, in 2 cynomolgus monkeys. Kinetic analysis was performed with the radiometabolite-corrected plasma input function. In addition, a wide dose range of another BACE1 inhibitor, PF-06663195, was examined to investigate the relationship between the brain target occupancy and plasma concentration of the drug. Finally, the effective radiation dose of 18F-PF-06684511 was estimated on the basis of the whole-body PET measurements in NHPs. Results: Radiolabeling was accomplished successfully with an incorporation radiochemical yield of 4%-12% (decay-corrected) from 18F ion. The radiochemical purity was greater than 99%. The whole-brain uptake of 18F-PF-06684511 peaked (∼220% SUV) at approximately 20 min and decreased thereafter (∼100% SUV at 180 min). A 2-tissue-compartment model described the time-activity curves well. Pretreatment with LY2886721 reduced the total distribution volume of 18F-PF-06684511 by 48%-80% depending on the brain region, confirming its in vivo specificity. BACE1 occupancy of PF-06663195, estimated using the Lassen occupancy plot, showed a dose-dependent increase. The effective dose of 18F-PF-06684511 was 0.043 mSv/MBq for humans. Conclusion: 18F-PF-06684511 is the first successful PET radioligand for BACE1 brain imaging that demonstrates favorable in vivo binding and brain kinetics in NHPs.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Radioisótopos de Flúor/metabolismo , Tomografía de Emisión de Positrones , Pirazinas/metabolismo , Tiazinas/metabolismo , Animales , Femenino , Cinética , Ligandos , Macaca fascicularis , Masculino , Modelos Biológicos , Radioquímica , Imagen de Cuerpo Entero
8.
J Neurosci ; 27(12): 3090-7, 2007 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-17376970

RESUMEN

Postoperative cognitive dysfunction, confusion, and delirium are common after general anesthesia in the elderly, with symptoms persisting for months or years in some patients. Even middle-aged patients are likely to have postoperative cognitive dysfunction for months after surgery, and Alzheimer's disease (AD) patients appear to be particularly at risk of deterioration after anesthesia. Several investigators have thus examined whether general anesthesia is associated with AD, with some studies suggesting that exposure to anesthetics may increase the risk of AD. However, little is known on the biochemical consequences of anesthesia on pathogenic pathways in vivo. Here, we investigated the effect of anesthesia on tau phosphorylation and amyloid precursor protein (APP) metabolism in mouse brain. We found that, regardless of the anesthetic used, anesthesia induced rapid and massive hyperphosphorylation of tau, rapid and prolonged hypothermia, inhibition of Ser/Thr PP2A (protein phosphatase 2A), but no changes in APP metabolism or Abeta (beta-amyloid peptide) accumulation. Reestablishing normothermia during anesthesia completely rescued tau phosphorylation to normal levels. Our results indicate that changes in tau phosphorylation were not a result of anesthesia per se, but a consequence of anesthesia-induced hypothermia, which led to inhibition of phosphatase activity and subsequent hyperphosphorylation of tau. These findings call for careful monitoring of core temperature during anesthesia in laboratory animals to avoid artifactual elevation of protein phosphorylation. Furthermore, a thorough examination of the effect of anesthesia-induced hypothermia on the risk and progression of AD is warranted.


Asunto(s)
Anestesia/efectos adversos , Hipotermia/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Proteínas tau/metabolismo , Anestésicos/administración & dosificación , Anestésicos/efectos adversos , Animales , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Hipocampo/metabolismo , Hipotermia/enzimología , Masculino , Ratones , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2
9.
J Med Chem ; 61(8): 3296-3308, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29356535

RESUMEN

Alzheimer's disease (AD) is characterized by accumulation of ß-amyloid (Aß) plaques and neurofibrillary tau tangles in the brain. ß-Site amyloid precursor protein cleaving enzyme 1 (BACE1) plays a key role in the generation of Aß fragments via extracellular cleavage of the amyloid precursor protein (APP). We became interested in developing a BACE1 PET ligand to facilitate clinical assessment of BACE1 inhibitors and explore its potential in the profiling and selection of patients for AD trials. Using a set of PET ligand design parameters, compound 3 (PF-06684511) was rapidly identified as a lead with favorable in vitro attributes and structural handles for PET radiolabeling. Further evaluation in an LC-MS/MS "cold tracer" study in rodents revealed high specific binding to BACE1 in brain. Upon radiolabeling, [18F]3 demonstrated favorable brain uptake and high in vivo specificity in nonhuman primate (NHP), suggesting its potential for imaging BACE1 in humans.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/metabolismo , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Radiofármacos/farmacología , Tiazinas/farmacología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Radioisótopos de Flúor , Ligandos , Masculino , Ratones , Tomografía de Emisión de Positrones , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacocinética , Pirazinas/síntesis química , Pirazinas/química , Pirazinas/farmacocinética , Radiofármacos/síntesis química , Radiofármacos/química , Radiofármacos/farmacocinética , Tiazinas/síntesis química , Tiazinas/química , Tiazinas/farmacocinética
10.
J Med Chem ; 61(10): 4476-4504, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29613789

RESUMEN

A major challenge in the development of ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors for the treatment of Alzheimer's disease is the alignment of potency, drug-like properties, and selectivity over related aspartyl proteases such as Cathepsin D (CatD) and BACE2. The potential liabilities of inhibiting BACE2 chronically have only recently begun to emerge as BACE2 impacts the processing of the premelanosome protein (PMEL17) and disrupts melanosome morphology resulting in a depigmentation phenotype. Herein, we describe the identification of clinical candidate PF-06751979 (64), which displays excellent brain penetration, potent in vivo efficacy, and broad selectivity over related aspartyl proteases including BACE2. Chronic dosing of 64 for up to 9 months in dog did not reveal any observation of hair coat color (pigmentation) changes and suggests a key differentiator over current BACE1 inhibitors that are nonselective against BACE2 in later stage clinical development.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Encéfalo/metabolismo , Diseño de Fármacos , Hipopigmentación , Inhibidores de Proteasas , Piranos , Pigmentación de la Piel/efectos de los fármacos , Tiazinas , Tiazoles , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Células Cultivadas , Perros , Humanos , Hipopigmentación/inducido químicamente , Masculino , Melanocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/efectos adversos , Inhibidores de Proteasas/química , Conformación Proteica , Piranos/administración & dosificación , Piranos/efectos adversos , Piranos/química , Tiazinas/administración & dosificación , Tiazinas/efectos adversos , Tiazinas/química , Tiazoles/administración & dosificación , Tiazoles/efectos adversos , Tiazoles/química
11.
J Med Chem ; 60(1): 386-402, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-27997172

RESUMEN

A growing subset of ß-secretase (BACE1) inhibitors for the treatment of Alzheimer's disease (AD) utilizes an anilide chemotype that engages a key residue (Gly230) in the BACE1 binding site. Although the anilide moiety affords excellent potency, it simultaneously introduces a third hydrogen bond donor that limits brain availability and provides a potential metabolic site leading to the formation of an aniline, a structural motif of prospective safety concern. We report herein an alternative aminomethyl linker that delivers similar potency and improved brain penetration relative to the amide moiety. Optimization of this series identified analogues with an excellent balance of ADME properties and potency; however, potential drug-drug interactions (DDI) were predicted based on CYP 2D6 affinities. Generation and analysis of key BACE1 and CYP 2D6 crystal structures identified strategies to obviate the DDI liability, leading to compound 16, which exhibits robust in vivo efficacy as a BACE1 inhibitor.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Anilidas/química , Inhibidores Enzimáticos/farmacología , Glicina/química , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/química , Animales , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión , Cristalización , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Masculino , Ratones , Técnicas de Placa-Clamp , Relación Estructura-Actividad , Espectrometría de Masas en Tándem
12.
Nat Commun ; 7: 13042, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27727204

RESUMEN

Inhibition of ß-secretase BACE1 is considered one of the most promising approaches for treating Alzheimer's disease. Several structurally distinct BACE1 inhibitors have been withdrawn from development after inducing ocular toxicity in animal models, but the target mediating this toxicity has not been identified. Here we use a clickable photoaffinity probe to identify cathepsin D (CatD) as a principal off-target of BACE1 inhibitors in human cells. We find that several BACE1 inhibitors blocked CatD activity in cells with much greater potency than that displayed in cell-free assays with purified protein. Through a series of exploratory toxicology studies, we show that quantifying CatD target engagement in cells with the probe is predictive of ocular toxicity in vivo. Taken together, our findings designate off-target inhibition of CatD as a principal driver of ocular toxicity for BACE1 inhibitors and more generally underscore the power of chemical proteomics for discerning mechanisms of drug action.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Catepsina D/metabolismo , Inhibidores Enzimáticos/toxicidad , Ojo/patología , Proteómica/métodos , Pruebas de Toxicidad , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ojo/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Espectrometría de Masas , Ratones Noqueados , Sondas Moleculares/síntesis química , Sondas Moleculares/química , Péptidos/metabolismo , Unión Proteica , Ratas Wistar , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Coloración y Etiquetado
13.
J Med Chem ; 58(6): 2678-702, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25695670

RESUMEN

The identification of centrally efficacious ß-secretase (BACE1) inhibitors for the treatment of Alzheimer's disease (AD) has historically been thwarted by an inability to maintain alignment of potency, brain availability, and desired absorption, distribution, metabolism, and excretion (ADME) properties. In this paper, we describe a series of truncated, fused thioamidines that are efficiently selective in garnering BACE1 activity without simultaneously inhibiting the closely related cathepsin D or negatively impacting brain penetration and ADME alignment, as exemplified by 36. Upon oral administration, these inhibitors exhibit robust brain availability and are efficacious in lowering central Amyloid ß (Aß) levels in mouse and dog. In addition, chronic treatment in aged PS1/APP mice effects a decrease in the number and size of Aß-derived plaques. Most importantly, evaluation of 36 in a 2-week exploratory toxicology study revealed no accumulation of autofluorescent material in retinal pigment epithelium or histology findings in the eye, issues observed with earlier BACE1 inhibitors.


Asunto(s)
Amidinas/química , Amidinas/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Placa Amiloide/tratamiento farmacológico , Enfermedad de Alzheimer/tratamiento farmacológico , Amidinas/farmacocinética , Amidinas/farmacología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Perros , Diseño de Fármacos , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Ratones , Modelos Moleculares , Placa Amiloide/metabolismo , Placa Amiloide/patología , Ratas , Ratas Wistar , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacocinética , Compuestos de Sulfhidrilo/farmacología , Compuestos de Sulfhidrilo/uso terapéutico
14.
J Med Chem ; 58(7): 3223-52, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25781223

RESUMEN

In recent years, the first generation of ß-secretase (BACE1) inhibitors advanced into clinical development for the treatment of Alzheimer's disease (AD). However, the alignment of drug-like properties and selectivity remains a major challenge. Herein, we describe the discovery of a novel class of potent, low clearance, CNS penetrant BACE1 inhibitors represented by thioamidine 5. Further profiling suggested that a high fraction of the metabolism (>95%) was due to CYP2D6, increasing the potential risk for victim-based drug-drug interactions (DDI) and variable exposure in the clinic due to the polymorphic nature of this enzyme. To guide future design, we solved crystal structures of CYP2D6 complexes with substrate 5 and its corresponding metabolic product pyrazole 6, which provided insight into the binding mode and movements between substrate/inhibitor complexes. Guided by the BACE1 and CYP2D6 crystal structures, we designed and synthesized analogues with reduced risk for DDI, central efficacy, and improved hERG therapeutic margins.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/química , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/química , Citocromo P-450 CYP2D6/química , Interacciones Farmacológicas , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Secuencia de Aminoácidos , Proteínas Amiloidogénicas/metabolismo , Animales , Cristalografía por Rayos X , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Diseño de Fármacos , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Concentración 50 Inhibidora , Masculino , Ratones Endogámicos , Modelos Moleculares , Datos de Secuencia Molecular , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacocinética , Pirazoles/química , Relación Estructura-Actividad
15.
Front Pharmacol ; 4: 72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785331

RESUMEN

Neurological and psychiatric disorders are frequently associated with disruption of various cognitive functions, but development of effective drug treatments for these conditions has proven challenging. One of the main obstacles is the poor predictive validity of our preclinical animal models. In the present study the effects of the γ-secretase inhibitor semagacestat was evaluated in preclinical in vivo electrophysiological models. Recently disclosed Phase III findings on semagacestat indicated that Alzheimer's disease (AD) patients on this drug showed significantly worsened cognitive function compared to those treated with placebo. Since previous studies have shown that drugs impairing cognitive function (including scopolamine, NMDA (N-methyl-D-aspartate) receptor antagonists, and nociceptin receptor agonists) disrupt or decrease power of elicited theta oscillation in the hippocampus, we tested the effects of acute and sub-chronic administration of semagacestat in this assay. Field potentials were recorded across the hippocampal formation with NeuroNexus multi-site silicon probes in urethane anesthetized male C57BL/6 mice; hippocampal CA1 theta oscillation was elicited by electrical stimulation of the brainstem nucleus pontis oralis. Sub-chronic administration of semagacestat twice daily over 12 days at a dose known to reduce beta-amyloid peptide (Aß) level [100 mg/kg, p.o. (per oral)] diminished power of elicited hippocampal theta oscillation. Acute, subcutaneous administration of semagacestat (100 mg/kg) produced a similar effect on hippocampal activity. We propose that the disruptive effect of semagacestat on hippocampal function could be one of the contributing mechanisms to its worsening of cognition in patients with AD. As it has been expected, both acute and sub-chronic administrations of semagacestat significantly decreased Aß40 and Aß42 levels but the current findings do not reveal the mode of action of semagacestat in disrupting hippocampal oscillation.

16.
J Med Chem ; 55(21): 9224-39, 2012 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22984865

RESUMEN

ß-Secretase 1 (BACE-1) is an attractive therapeutic target for the treatment and prevention of Alzheimer's disease (AD). Herein, we describe the discovery of a novel class of BACE-1 inhibitors represented by sulfamide 14g, using a medicinal chemistry strategy to optimize central nervous system (CNS) penetration by minimizing hydrogen bond donors (HBDs) and reducing P-glycoprotein (P-gp) mediated efflux. We have also taken advantage of the combination of structure based drug design (SBDD) to guide the optimization of the sulfamide analogues and the in silico tool WaterMap to explain the observed SAR. Compound 14g is a potent inhibitor of BACE-1 with excellent permeability and a moderate P-gp liability. Administration of 14g to mice produced a significant, dose-dependent reduction in central Aß(X-40) levels at a free drug exposure equivalent to the whole cell IC(50) (100 nM). Furthermore, studies of the P-gp knockout mouse provided evidence that efflux transporters affected the amount of Aß lowering versus that observed in wild-type (WT) mouse at an equivalent dose.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Compuestos Aza/síntesis química , Encéfalo/metabolismo , Compuestos de Espiro/síntesis química , Sulfonamidas/síntesis química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Secretasas de la Proteína Precursora del Amiloide/química , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas/química , Compuestos Aza/farmacocinética , Compuestos Aza/farmacología , Cristalografía por Rayos X , Perros , Diseño de Fármacos , Femenino , Humanos , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Noqueados , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Estructura Molecular , Permeabilidad , Compuestos de Espiro/farmacocinética , Compuestos de Espiro/farmacología , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Transfección
17.
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
18.
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
19.
J Pharmacol Exp Ther ; 319(2): 924-33, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16920992

RESUMEN

LY-450139 is a gamma-secretase inhibitor shown to have efficacy in multiple cellular and animal models. Paradoxically, robust elevations of plasma amyloid-beta (Abeta) have been reported in dogs and humans after administration of subefficacious doses. The present study sought to further evaluate Abeta responses to LY-450139 in the guinea pig, a nontransgenic model that has an Abeta sequence identical to that of human. Male guinea pigs were treated with LY-450139 (0.2-60 mg/kg), and brain, cerebrospinal fluid, and plasma Abeta levels were characterized at 1, 3, 6, 9, and 14 h postdose. Low doses significantly elevated plasma Abeta levels at early time points, with return to baseline within hours. Higher doses inhibited Abeta levels in all compartments at early time points, but elevated plasma Abeta levels at later time points. To determine whether this phenomenon occurs under steady-state drug exposure, guinea pigs were implanted with subcutaneous minipumps delivering LY-450139 (0.3-30 mg/kg/day) for 5 days. Plasma Abeta was significantly inhibited at 10-30 mg/kg/day, but significantly elevated at 1 mg/kg/day. To further understand the mechanism of Abeta elevation by LY-450139, H4 cells overexpressing the Swedish mutant of amyloid-precursor protein and a mouse embryonic stem cell-derived neuronal cell line were studied. In both cellular models, elevated levels of secreted Abeta were observed at subefficacious concentrations, whereas dose-responsive inhibition was observed at higher concentrations. These results suggest that LY-450139 modulates the gamma-secretase complex, eliciting Abeta lowering at high concentrations but Abeta elevation at low concentrations.


Asunto(s)
Alanina/análogos & derivados , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/sangre , Azepinas/farmacología , Inhibidores Enzimáticos/farmacología , Alanina/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Cobayas , Masculino , Ratones , Factores de Tiempo
20.
Bioorg Med Chem Lett ; 14(17): 4511-4, 2004 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-15357982

RESUMEN

The synthesis and nNOS and eNOS activity of 6-(4-(dimethylaminoalkyl)-/6-(4-(dimethylaminoalkoxy)-5-ethyl-2-methoxyphenyl)-pyridin-2-ylamines and 6-(4-(dimethylaminoalkyl)-/6-(4-(dimethylaminoalkoxy)-2,5-dimethoxyphenyl)-pyridin-2-ylamines 1-8 are described. These compounds are potent inhibitors of the human nNOS isoform.


Asunto(s)
Aminas/química , Inhibidores Enzimáticos/química , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Óxido Nítrico Sintasa/antagonistas & inhibidores , Piridinas/química , Aminas/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Inhibición Neural/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I , Piridinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA