Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
ChemMedChem ; 15(10): 900-905, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32181986

RESUMEN

Blood coagulation factor XIII (FXIII, F13) is considered to be a promising target for anticoagulants with reduced bleeding risk because of its unique position in the coagulation cascade downstream of thrombin. However, until now, no potent drug addressing FXIII has been available, indeed no compound has even entered clinical trials yet. In 2013, we published the co-crystal structure of FXIII in the active state (FXIIIa°), thereby providing a detailed map of the active site for the rational design of potent FXIIIa blockers. Here we report, for the first time, a structure-based approach to improving the affinity of FXIIIa inhibitors. FXIII was crystallized in complex with a methyl thiazole moiety to address a novel transient hydrophobic pocket close to the catalytic center. By subsequent structure-based design to rationalize the introduction of an ethyl ester, the potency of the inhibitor was improved significantly compared to that of the parent lead compound. The occupancy of the hydrophobic pocket described here might turn out to be a key step in the development of a potent reversible and orally available FXIIIa blocker.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Factor XIII/antagonistas & inhibidores , Sitios de Unión/efectos de los fármacos , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Factor XIII/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformación Molecular
3.
J Med Chem ; 59(16): 7561-75, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27463859

RESUMEN

Successful optimization of a given lead scaffold requires thorough binding-site mapping of the target protein particular in regions remote from the catalytic center where high conservation across protein families is given. We screened a 361-entry fragment library for binding to the aspartic protease endothiapepsin by crystallography. This enzyme is frequently used as a surrogate for the design of renin and ß-secretase inhibitors. A hit rate of 20% was achieved, providing 71 crystal structures. Here, we discuss 45 binding poses of fragments accommodated in pockets remote from the catalytic dyad. Three major hot spots are discovered in remote binding areas: Asp81, Asp119, and Phe291. Compared to the dyad binders, bulkier fragments occupy these regions. Many of the discovered fragments suggest an optimization concept on how to grow them into larger ligands occupying adjacent binding pockets that will possibly endow them with the desired selectivity for one given member of a protein family.


Asunto(s)
Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Ácido Aspártico Endopeptidasas/metabolismo , Biocatálisis , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Inhibidores de Proteasas/química , Relación Estructura-Actividad
4.
ACS Chem Biol ; 11(6): 1693-701, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27028906

RESUMEN

Fragment-based lead discovery (FBLD) has become a pillar in drug development. Typical applications of this method comprise at least two biophysical screens as prefilter and a follow-up crystallographic experiment on a subset of fragments. Clearly, structural information is pivotal in FBLD, but a key question is whether such a screening cascade strategy will retrieve the majority of fragment-bound structures. We therefore set out to screen 361 fragments for binding to endothiapepsin, a representative of the challenging group of aspartic proteases, employing six screening techniques and crystallography in parallel. Crystallography resulted in the very high number of 71 structures. Yet alarmingly, 44% of these hits were not detected by any biophysical screening approach. Moreover, any screening cascade, building on the results from two or more screening methods, would have failed to predict at least 73% of these hits. We thus conclude that, at least in the present case, the frequently applied biophysical prescreening filters deteriorate the number of possible X-ray hits while only the immediate use of crystallography enables exhaustive retrieval of a maximum of fragment structures, which represent a rich source guiding hit-to-lead-to-drug evolution.


Asunto(s)
Descubrimiento de Drogas/métodos , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/química , Biofisica , Calorimetría , Cristalografía por Rayos X , Bases de Datos de Compuestos Químicos , Modelos Moleculares , Inhibidores de Proteasas/química , Unión Proteica , Relación Estructura-Actividad
5.
J Med Chem ; 59(21): 9743-9759, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27726357

RESUMEN

Crystallography is frequently used as follow-up method to validate hits identified by biophysical screening cascades. The capacity of crystallography to directly screen fragment libraries is often underestimated, due to its supposed low-throughput and need for high-quality crystals. We applied crystallographic fragment screening to map the protein-binding site of the aspartic protease endothiapepsin by individual soaking experiments. Here, we report on 41 fragments binding to the catalytic dyad and adjacent specificity pockets. The analysis identifies already known warheads but also reveals hydrazide, pyrazole, or carboxylic acid fragments as novel functional groups binding to the dyad. A remarkable swapping of the S1 and S1' pocket between structurally related fragments is explained by either steric demand, required displacement of a well-bound water molecule, or changes of trigonal-planar to tetrahedral geometry of an oxygen functional group in a side chain. Some warheads simultaneously occupying both S1 and S1' are promising starting points for fragment-growing strategies.


Asunto(s)
Proteasas de Ácido Aspártico/química , Proteasas de Ácido Aspártico/metabolismo , Biocatálisis , Ácidos Carboxílicos/metabolismo , Hidrazinas/metabolismo , Pirazoles/metabolismo , Calorimetría , Ácidos Carboxílicos/química , Dominio Catalítico , Cristalografía por Rayos X , Hidrazinas/química , Modelos Moleculares , Pirazoles/química
6.
Structure ; 24(8): 1398-1409, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27452405

RESUMEN

Today the identification of lead structures for drug development often starts from small fragment-like molecules raising the chances to find compounds that successfully pass clinical trials. At the heart of the screening for fragments binding to a specific target, crystallography delivers structural information essential for subsequent drug design. While it is common to search for bound ligands in electron densities calculated directly after an initial refinement cycle, we raise the important question whether this strategy is viable for fragments characterized by low affinities. Here, we describe and provide a collection of high-quality diffraction data obtained from 364 protein crystals treated with diverse fragments. Subsequent data analysis showed that ∼25% of all hits would have been missed without further refining the resulting structures. To enable fast and reliable hit identification, we have designed an automated refinement pipeline that will inspire the development of optimized tools facilitating the successful application of fragment-based methods.


Asunto(s)
Cristalografía por Rayos X/estadística & datos numéricos , Ensayos Analíticos de Alto Rendimiento , Bibliotecas de Moléculas Pequeñas/química , Agua/química , Cristalografía por Rayos X/métodos , Conjuntos de Datos como Asunto , Diseño de Fármacos , Humanos , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA