442 research outputs found

    Pharmacy Handbook 2009

    Get PDF
    Item does not contain fulltext14 p

    Identification of Ligands with Tailored Selectivity: Strategies & Application

    Get PDF
    In the field of computer-aided drug design, docking is a computational tool, often used to evaluate the sterical and chemical complementarity between two molecules. This technique can be used to estimate the binding or non-binding of a small molecule to a protein binding site. The classical application of docking is to find those molecules within a large set of molecules that bind a certain target protein and modulate its biological activity. This setup can be considered as established for a single target protein. In contrast to this, the docking to multiple target structures offers new possible applications. It can be used, for example, to assess the binding profile of a ligand against a number of proteins. In this work, the applicability of docking is assessed in such a scenario where multiple target structures are used. The corresponding proteins mostly belong to the family of G protein-coupled receptors. This protein family is very large and numerous GPCRs have been identified as potential drug targets, explaining the their relevance in pharmaceutical research. The protein structures used herein have different relationships and thus represent different application scenarios. The first case study uses two structures belonging to different proteins. These proteins are CXCR3 and CXCR4, a pair of chemokine GPCRs. In this chapter, new ligands are identified that bind to these proteins and modulate their biological activity. More importantly, for each of these newly identified ligands it could be predicted using docking, whether this ligand binds only to one of the two target proteins or to both. This study proves the applicability of docking to identify ligands with tailored selectivity. In addition, these ligands show excellent binding affinities to their respective target or targets. In the following two studies, the docking to different structures of the same target protein is investigated. The first application aims at identifying ligands selective for either one of two isoforms of the zebrafish CXC receptor 4. Subsequently, multiple conformations of the chemokine receptor CCR5 are used to show that different starting structures can identify different ligands. Next to the plain identification of chemically new ligands, experimental hurdles to prove the biological activity of these molecules in a functional assay is discussed. These difficulties are based on the fact that docking evaluates the structural complementarity between molecules and protein structures rather than predicting the effect of these molecules on the proteins. In addition, GPCRs form a challenging set of target proteins, since their ligands can induce a variety of different effects. Finally, the general applicability of multi-target docking to a very large number of structures is investigated. For this evaluation, kinases are used as protein family since many more structures have been experimentally determined for these proteins compared to GPCRs as membrane proteins. First, using published experimental data, a dataset is created consisting of several hundred kinase structures and a set of small-molecule kinase inhibitors. This dataset is characterised by the availability of experimental binding data for each single kinase-inhibitor combination. These experimental data were subsequently compared to the docking results of each ligand into each single kinase structure. The results indicate that a reliable selectivity prediction for a ligand is highly demanding in such a large-scale setup and beyond current possibilities. However, it can be shown that the prediction accuracy of docking can be improved by normalising the docking scores over multiple ligands and proteins. Based on these findings, the idea of "protein decoys" is developed, which might in the future allow more accurate predictions of selectivity profiles using docking

    Pharmacy handbook

    Get PDF
    2005 handbook for the faculty of Pharmac

    Exploring Molecular Diversity: There is Plenty of Room at Markush's

    Get PDF
    L'estratègia de les etapes inicials del descobriment de fàrmacs està normalment basada en un procés anomenat hit-to-lead que implica un extens estudi entorn de la síntesi de derivats d'una molècula original que prèviament hagi mostrat certa activitat biològica davant d'una diana concreta. Per tant, aquest procés comporta la síntesi de molts anàlegs que descriurien una subquimioteca, que generalment evidencia que aquests estudis estan molt focalitzats al voltant de l'espai químic del compost original. Així i tot, quan aquesta molècula és finalment patentada, es descriu un espai químic molt més vast per mitjà d'estructures Markush donant per suposat que alguns dels seus derivats puguin presentar també activitat biològica. Tot i això, la presència d'aquestes estructures no implica la síntesi comprovada de tota la biblioteca molecular sinó només una petita mostra de la mateixa. La nostra hipòtesi és que hi ha una gran part de l’espai químic d’aquestes biblioteques que està sense explorar i pot amagar possibles candidats que poden fins i tot superar l’activitat del hit original. A través d'aquest projecte, es proposa una alternativa que sosté que una selecció racional de poques molècules – basat en l'agrupament segons semblança molecular – pot representar de manera més significativa l'espai químic establert, oferint la possibilitat d'explorar regions desconegudes que podrien amagar més potencial biològic. Després de revisar els darrers fàrmacs aprovats per la FDA en el període del 2008 al 2020 i la base de dades de molècules bioactives de ChEMBL, s'ha dut a terme una exploració de l'ampli espai químic resultant de molècules petites amb propietats similars a les dels medicaments per definir nous espais accessibles que podrien ocultar activitat. Els resultats obtinguts de set casos d'estudis reals han demostrat que tant la selecció racional com l’aleatòria representen més significativament les biblioteques combinatòries declarades a les patents, que les molècules descrites fins ara. S'han realitzat dos estudis pràctics que implementen aquesta metodologia suggerida per descriure millor l'espai químic del fàrmac antipalúdic Tafenoquina i del Dacomitinib, un inhibidor de tirosina cinases de segona generació per al tractament del càncer de pulmó de cèl·lules no petites. L’exploració de l’espai químic d’aquestes dues famílies ha portat a la síntesi racional de set anàlegs antipalúdics i vuit inhibidors de cinases que han mostrat interessants activitats inhibidores. Aquests resultats demostren que l'aplicació de la quimioinformàtica per a la selecció de biblioteques pot millorar la capacitat d'inspeccionar millor els conjunts de dades químiques per identificar nous compostos precandidats i representar grans biblioteques per a posteriors campanyes de reposicionament.La estrategia de las etapas iniciales del descubrimiento de fármacos está normalmente basada en un proceso denominado hit-to-lead que implica un extenso estudio entorno a la síntesis de derivados de una molécula original que previamente haya expresado cierta actividad biológica frente a una diana concreta. Por ende, este proceso conlleva la síntesis de muchos análogos que describirían una sublibrería química, la cual generalmente evidencia que estos estudios están muy focalizados alrededor del espacio químico del compuesto original. Aún y así, cuando esta molécula es finalmente patentada, se describe un espacio químico mucho más vasto por medio de estructuras Markush teorizando que algunos de sus derivados puedan presentar también actividad biológica. Sin embargo, la presencia de estas estructuras no implica la síntesis comprobada de toda la biblioteca molecular sino solo una pequeña muestra de la misma. Nuestra hipótesis es que hay una gran parte del espacio químico de estas bibliotecas que está sin explorar y puede ocultar posibles candidatos que pueden hasta superar la actividad del hit original. A través de este proyecto, se propone una alternativa que sostiene que una selección racional de pocas moléculas – fundada en el agrupamiento según su similitud química – puede representar de manera más significativa el espacio químico establecido, ofreciendo la posibilidad de explorar regiones desconocidas que podrían ocultar más potencial biológico. Después de revisar los últimos fármacos aprobados por la FDA en el período de 2008 a 2020 y la base de datos de moléculas bioactivas de ChEMBL, se ha llevado a cabo una exploración del amplio espacio químico resultante de moléculas pequeñas con propiedades similares a las de los medicamentos para definir nuevos espacios accesible que podrían ocultar actividad. Los resultados obtenidos de siete casos de estudios reales han demostrado que tanto la selección racional como la aleatoria representan más significativamente las bibliotecas combinatorias declaradas en las patentes que las moléculas descritas hasta la fecha. Se han desarrollado dos estudios prácticos que implementan esta metodología sugerida para describir mejor el espacio químico del fármaco antipalúdico Tafenoquina y Dacomitinib, un inhibidor de la tirosina quinasa de segunda generación para el tratamiento del cáncer de pulmón de células no pequeñas. La exploración del espacio químico de estas dos familias ha llevado a la síntesis racional de siete análogos antipalúdicos y ocho inhibidores de quinasas que han mostrado interesantes actividades inhibidoras. Estos resultados demuestran que la aplicación de la quimioinformática para la selección de bibliotecas puede mejorar la capacidad de inspeccionar mejor los conjuntos de datos químicos para identificar nuevos potenciales hits y representar grandes bibliotecas para fines de reposicionamiento.The early Drug Discovery strategy is commonly based on a hit-to-lead process which involves large research on the synthesis of derivatives of an original molecule that had previously shown biological activity against a specific biological target. Therefore, this process implies the synthesis of many analogs leading to the description of a chemical sub-library which generally leads to a highly focused study on the chemical space nearby the hit compound. However, when this drug is finally patented, a wider chemical space derived from a Markush structure is described, theorizing that some analogs within may present biological activity. Nevertheless, this claim involving the Markush structure does not imply the proven synthesis of all the chemical library but just a small population of it. We hypothesize that there is a great part of the chemical space of these libraries that is unexplored and can hide potential lead candidates which may even surpass the activity of the original hit. Through this project, an alternative is proposed claiming that a rational selection of a short sample of small molecules – founded on similarity-based clustering – can represent more significatively the stated chemical space offering the possibility to explore the unknown space that could hide more potential biological activity. After a review on the latest approved drugs by the FDA in the period from 2008 to 2020 and the ChEMBL database of bioactive molecules, an exploration of the resulting wide chemical space of small molecules with drug-like properties has been assessed in order to define accessible spots that might hide biological activity. The obtained results from seven real cases of study have proven that random and rationally selected molecules represent more significantly the combinatorial libraries stated in the patents rather than the reported molecules until date. Furthermore, two practical studies implementing our suggested methodology have been developed to better describe the chemical space of the antimalarial drug Tafenoquine and Dacomitinib, a second-generation tyrosine kinase inhibitor for non-small-cell lung cancer treatment. The assessment driven by a better chemical space exploration of these two families have led to the rational synthesis of seven antimalarial analogs and eight kinase inhibitors which have shown interesting inhibitory activities. Our results evince that the application of cheminformatics for library selection may improve the ability to better inspect chemical datasets in order to identify new potential hits and represent large libraries for further reprofiling purposes

    Prots: A fragment based protein thermo‐stability potential

    Get PDF
    Designing proteins with enhanced thermo‐stability has been a main focus of protein engineering because of its theoretical and practical significance. Despite extensive studies in the past years, a general strategy for stabilizing proteins still remains elusive. Thus effective and robust computational algorithms for designing thermo‐stable proteins are in critical demand. Here we report PROTS, a sequential and structural four‐residue fragment based protein thermo‐stability potential. PROTS is derived from a nonredundant representative collection of thousands of thermophilic and mesophilic protein structures and a large set of point mutations with experimentally determined changes of melting temperatures. To the best of our knowledge, PROTS is the first protein stability predictor based on integrated analysis and mining of these two types of data. Besides conventional cross validation and blind testing, we introduce hypothetical reverse mutations as a means of testing the robustness of protein thermo‐stability predictors. In all tests, PROTS demonstrates the ability to reliably predict mutation induced thermo‐stability changes as well as classify thermophilic and mesophilic proteins. In addition, this white‐box predictor allows easy interpretation of the factors that influence mutation induced protein stability changes at the residue level. Proteins 2012; © 2011 Wiley Periodicals, Inc.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/89526/1/23163_ftp.pd
    corecore