11 research outputs found

    Field-based Proteochemometric Models Derived from 3D Protein Structures : A Novel Approach to Visualize Affinity and Selectivity Features

    Get PDF
    Designing drugs that are selective is crucial in pharmaceutical research to avoid unwanted side effects. To decipher selectivity of drug targets, computational approaches that utilize the sequence and structural information of the protein binding pockets are frequently exploited. In addition to methods that rely only on protein information, quantitative approaches such as proteochemometrics (PCM) use the combination of protein and ligand descriptions to derive quantitative relationships with binding affinity. PCM aims to explain cross-interactions between the different proteins and ligands, hence facilitating our understanding of selectivity. The main goal of this dissertation is to develop and apply field-based PCM to improve the understanding of relevant molecular interactions through visual illustrations. Field-based description that depends on the 3D structural information of proteins enhances visual interpretability of PCM models relative to the frequently used sequence-based descriptors for proteins. In these field-based PCM studies, knowledge-based fields that explain polarity and lipophilicity of the binding pockets and WaterMap-derived fields that elucidate the positions and energetics of water molecules are used together with the various 2D / 3D ligand descriptors to investigate the selectivity profiles of kinases and serine proteases. Field-based PCM is first applied to protein kinases, for which designing selective inhibitors has always been a challenge, owing to their highly similar ATP binding pockets. Our studies show that the method could be successfully applied to pinpoint the regions influencing the binding affinity and selectivity of kinases. As an extension of the initial studies conducted on a set of 50 kinases and 80 inhibitors, field-based PCM was used to build classification models on a large dataset (95 kinases and 1572 inhibitors) to distinguish active from inactive ligands. The prediction of the bioactivities of external test set compounds or kinases with accuracies over 80% (Matthews correlation coefficient, MCC: ~0.50) and area under the ROC curve (AUC) above 0.8 together with the visual inspection of the regions promoting activity demonstrates the ability of field-based PCM to generate both predictive and visually interpretable models. Further, the application of this method to serine proteases provides an overview of the sub-pocket specificities, which is crucial for inhibitor design. Additionally, alignment-independent Zernike descriptors derived from fields were used in PCM models to study the influence of protein superimpositions on field comparisons and subsequent PCM modelling.Lääketutkimuksessa selektiivisten lääkeaineiden suunnittelu on ratkaisevan tärkeää haittavaikutusten välttämiseksi. Kohdeselektiivisyyden selvittämiseen käytetään usein tietokoneavusteisia menetelmiä, jotka hyödyntävät proteiinien sitoutumiskohtien sekvenssi- ja rakennetietoja. Proteiinilähtöisten menetelmien lisäksi kvantitatiiviset menetelmät kuten proteokemometria (proteochemometrics, PCM) yhdistävät sekä proteiinin että ligandin tietoja muodostaessaan kvantitatiivisen suhteen sitoutumisaffiniteettiin. PCM pyrkii selittämään eri proteiinien ja ligandien vuorovaikutuksia ja näin auttaa ymmärtämään selektiivisyyttä. Väitöstutkimuksen tavoitteena oli kehittää ja hyödyntää kenttäpohjaista proteokemometriaa, joka auttaa ymmärtämään relevantteja molekyylitasoisia vuorovaikutuksia visuaalisen esitystavan kautta. Proteiinin kolmiulotteisesta rakenteesta riippuva kenttäpohjainen kuvaus helpottaa PCM-mallien tulkintaa, etenkin usein käytettyihin sekvenssipohjaisiin kuvauksiin verrattuna. Näissä kenttäpohjaisissa PCM-mallinnuksissa käytettiin tietoperustaisia sitoutumistaskun polaarisuutta ja lipofiilisyyttä kuvaavia kenttiä ja WaterMap-ohjelman tuottamia vesimolekyylien sijaintia ja energiaa havainnollistavia kenttiä yhdessä lukuisten ligandia kuvaavien 2D- ja 3D-deskriptorien kanssa. Malleja sovellettiin kinaasien ja seriiniproteaasien selektiivisyysprofiilien tutkimukseen. Tutkimuksen ensimmäisessä osassa kenttäpohjaista PCM-mallinnusta sovellettiin proteiinikinaaseihin, joille selektiivisten inhibiittorien suunnittelu on haastavaa samankaltaisten ATP sitoutumistaskujen takia. Tutkimuksemme osoitti menetelmän soveltuvan kinaasien sitoutumisaffiniteettia ja selektiivisyyttä ohjaavien alueiden osoittamiseen. Jatkona 50 kinaasia ja 80 inhibiittoria käsittäneelle alkuperäiselle tutkimukselle rakensimme kenttäpohjaisia PCM-luokittelumalleja suuremmalle joukolle kinaaseja (95) ja inhibiittoreita (1572) erotellaksemme aktiiviset ja inaktiiviset ligandit toisistaan. Ulkoisen testiyhdiste- tai testikinaasijoukon bioaktiivisuuksien ennustaminen yli 80 % tarkkuudella (Matthews korrelaatiokerroin, MCC noin 0,50) ja ROC-käyrän alle jäävä ala (AUC) yli 0,8 yhdessä aktiivisuutta tukevien alueiden visuaalisen tarkastelun kanssa osoittivat kenttäpohjaisen PCM:n pystyvän tuottamaan sekä ennustavia että visuaalisesti ymmärrettäviä malleja. Tutkimuksen toisessa osassa metodin soveltaminen seriiniproteaaseihin tuotti yleisnäkemyksen sitoutumistaskun eri osien spesifisyyksistä, mikä on ensiarvoisen tärkeää inhibiittorien suunnittelulle. Lisäksi kentistä johdettuja, proteiinien päällekkäinasettelusta riippumattomia Zernike-deskriptoreita hyödynnettiin PCM-malleissa arvioidaksemme proteiinien päällekkäinasettelun vaikutusta kenttien vertailuun ja sen jälkeiseen PCM-mallinnukseen

    Fragment Hotspot Mapping to Identify Selectivity-Determining Regions between Related Proteins.

    Get PDF
    Funder: ExscientiaFunder: Diamond Light SourceFunder: Kungliga Tekniska HoegskolanFunder: Chinese Center for Disease Control and PreventionFunder: European Federation of Pharmaceutical Industries and AssociationsFunder: European CommissionFunder: Kennedy Trust for Rheumatology ResearchFunder: Ontario Institute for Cancer ResearchFunder: Royal Institution for the Advancement of Learning McGill UniversityFunder: UCBSelectivity is a crucial property in small molecule development. Binding site comparisons within a protein family are a key piece of information when aiming to modulate the selectivity profile of a compound. Binding site differences can be exploited to confer selectivity for a specific target, while shared areas can provide insights into polypharmacology. As the quantity of structural data grows, automated methods are needed to process, summarize, and present these data to users. We present a computational method that provides quantitative and data-driven summaries of the available binding site information from an ensemble of structures of the same protein. The resulting ensemble maps identify the key interactions important for ligand binding in the ensemble. The comparison of ensemble maps of related proteins enables the identification of selectivity-determining regions within a protein family. We applied the method to three examples from the well-researched human bromodomain and kinase families, demonstrating that the method is able to identify selectivity-determining regions that have been used to introduce selectivity in past drug discovery campaigns. We then illustrate how the resulting maps can be used to automate comparisons across a target protein family

    In silico Methods for Design of Kinase Inhibitors as Anticancer Drugs

    Get PDF
    Rational drug design implies usage of molecular modeling techniques such as pharmacophore modeling, molecular dynamics, virtual screening, and molecular docking to explain the activity of biomolecules, define molecular determinants for interaction with the drug target, and design more efficient drug candidates. Kinases play an essential role in cell function and therefore are extensively studied targets in drug design and discovery. Kinase inhibitors are clinically very important and widely used antineoplastic drugs. In this review, computational methods used in rational drug design of kinase inhibitors are discussed and compared, considering some representative case studies

    11th German Conference on Chemoinformatics (GCC 2015) : Fulda, Germany. 8-10 November 2015.

    Get PDF

    Développement de nouvelles approches protéo-chimiométriques appliquées à l'étude des interactions et de la sélectivité des inhibiteurs de kinases

    Get PDF
    The human kinome contains 518 proteins. They share a common mechanism of protein phosphorylation known to play an important role in cellular signaling pathways. Impaired kinase function is recognized to be involved in severe diseases like cancer. Due to high structural similarity between protein kinases, development of potent and selective kinase inhibitors is a challenging task. The selectivity of kinase inhibitors may lead to side effects potentially harmful. In this thesis, we first developed new selectivity metrics to determine inhibitor selectivity directly from biological inhibition data. Compared to existing metrics, the new selectivity scores can be applied on diverse inhibition data types. Second, we developed a proteometric approach in order to understand why some protein kinases are never inhibited by Type II inhibitors. The statistical model built for this purpose allowed us to identify several discriminant residues of which few of them correspond to experimentally described residues of interest. Third, using a new 3D protein kinase descriptor, we developed and validated novel proteo-chemometrics approaches to study and discover new kinase inhibitors.Le kinome humain comprend 518 protéines. Elles participent au processus de phosphorylation des protéines qui joue un rôle important dans les voies de signalisation cellulaire. Leur dérégulation est connue comme étant une cause de nombreuses maladies graves telle que les cancers. Du fait de leur grande similarité structurale des protéines kinases, il est difficile de développer des inhibiteurs qui soient à la fois efficaces et sélectifs. L’absence de sélectivité conduit le plus souvent à des effets secondaires particulièrement néfastes pour l’organisme. Au cours de cette thèse, nous avons d’abord développé de nouvelles métriques dont le but est de déterminer la sélectivité d’inhibiteurs à partir de données d’inhibition. Elles présentent l’avantage, comparées à d’autres métriques, d’être applicables sur n’importe quel type de données. Dans un deuxième temps, nous avons développé une approche protéométrique dans le but de comprendre pourquoi certaines protéines kinases ne sont jamais inhibées par des inhibiteurs de Type II. Le modèle statistique mis en place nous a permis d’identifier plusieurs résidus discriminants dont certains déjà décrits expérimentalement dans la littérature. Dans un troisième temps, nous avons développé un nouveau descripteur 3D de protéines kinases avec lequel nous avons mis en place et validé des modèles protéo-chimiométriques visant à étudier et découvrir de nouveaux inhibiteurs

    Structural Cheminformatics for Kinase-Centric Drug Design

    Get PDF
    Drug development is a long, expensive, and iterative process with a high failure rate, while patients wait impatiently for treatment. Kinases are one of the main drug targets studied for the last decades to combat cancer, the second leading cause of death worldwide. These efforts resulted in a plethora of structural, chemical, and pharmacological kinase data, which are collected in the KLIFS database. In this thesis, we apply ideas from structural cheminformatics to the rich KLIFS dataset, aiming to provide computational tools that speed up the complex drug discovery process. We focus on methods for target prediction and fragment-based drug design that study characteristics of kinase binding sites (also called pockets). First, we introduce the concept of computational target prediction, which is vital in the early stages of drug discovery. This approach identifies biological entities such as proteins that may (i) modulate a disease of interest (targets or on-targets) or (ii) cause unwanted side effects due to their similarity to on-targets (off-targets). We focus on the research field of binding site comparison, which lacked a freely available and efficient tool to determine similarities between the highly conserved kinase pockets. We fill this gap with the novel method KiSSim, which encodes and compares spatial and physicochemical pocket properties for all kinases (kinome) that are structurally resolved. We study kinase similarities in the form of kinome-wide phylogenetic trees and detect expected and unexpected off-targets. To allow multiple perspectives on kinase similarity, we propose an automated and production-ready pipeline; user-defined kinases can be inspected complementarily based on their pocket sequence and structure (KiSSim), pocket-ligand interactions, and ligand profiles. Second, we introduce the concept of fragment-based drug design, which is useful to identify and optimize active and promising molecules (hits and leads). This approach identifies low-molecular-weight molecules (fragments) that bind weakly to a target and are then grown into larger high-affinity drug-like molecules. With the novel method KinFragLib, we provide a fragment dataset for kinases (fragment library) by viewing kinase inhibitors as combinations of fragments. Kinases have a highly conserved pocket with well-defined regions (subpockets); based on the subpockets that they occupy, we fragment kinase inhibitors in experimentally resolved protein-ligand complexes. The resulting dataset is used to generate novel kinase-focused molecules that are recombinations of the previously fragmented kinase inhibitors while considering their subpockets. The KinFragLib and KiSSim methods are published as freely available Python tools. Third, we advocate for open and reproducible research that applies FAIR principles ---data and software shall be findable, accessible, interoperable, and reusable--- and software best practices. In this context, we present the TeachOpenCADD platform that contains pipelines for computer-aided drug design. We use open source software and data to demonstrate ligand-based applications from cheminformatics and structure-based applications from structural bioinformatics. To emphasize the importance of FAIR data, we dedicate several topics to accessing life science databases such as ChEMBL, PubChem, PDB, and KLIFS. These pipelines are not only useful to novices in the field to gain domain-specific skills but can also serve as a starting point to study research questions. Furthermore, we show an example of how to build a stand-alone tool that formalizes reoccurring project-overarching tasks: OpenCADD-KLIFS offers a clean and user-friendly Python API to interact with the KLIFS database and fetch different kinase data types. This tool has been used in this thesis and beyond to support kinase-focused projects. We believe that the FAIR-based methods, tools, and pipelines presented in this thesis (i) are valuable additions to the toolbox for kinase research, (ii) provide relevant material for scientists who seek to learn, teach, or answer questions in the realm of computer-aided drug design, and (iii) contribute to making drug discovery more efficient, reproducible, and reusable

    Network-driven strategies to integrate and exploit biomedical data

    Get PDF
    [eng] In the quest for understanding complex biological systems, the scientific community has been delving into protein, chemical and disease biology, populating biomedical databases with a wealth of data and knowledge. Currently, the field of biomedicine has entered a Big Data era, in which computational-driven research can largely benefit from existing knowledge to better understand and characterize biological and chemical entities. And yet, the heterogeneity and complexity of biomedical data trigger the need for a proper integration and representation of this knowledge, so that it can be effectively and efficiently exploited. In this thesis, we aim at developing new strategies to leverage the current biomedical knowledge, so that meaningful information can be extracted and fused into downstream applications. To this goal, we have capitalized on network analysis algorithms to integrate and exploit biomedical data in a wide variety of scenarios, providing a better understanding of pharmacoomics experiments while helping accelerate the drug discovery process. More specifically, we have (i) devised an approach to identify functional gene sets associated with drug response mechanisms of action, (ii) created a resource of biomedical descriptors able to anticipate cellular drug response and identify new drug repurposing opportunities, (iii) designed a tool to annotate biomedical support for a given set of experimental observations, and (iv) reviewed different chemical and biological descriptors relevant for drug discovery, illustrating how they can be used to provide solutions to current challenges in biomedicine.[cat] En la cerca d’una millor comprensió dels sistemes biològics complexos, la comunitat científica ha estat aprofundint en la biologia de les proteïnes, fàrmacs i malalties, poblant les bases de dades biomèdiques amb un gran volum de dades i coneixement. En l’actualitat, el camp de la biomedicina es troba en una era de “dades massives” (Big Data), on la investigació duta a terme per ordinadors se’n pot beneficiar per entendre i caracteritzar millor les entitats químiques i biològiques. No obstant, la heterogeneïtat i complexitat de les dades biomèdiques requereix que aquestes s’integrin i es representin d’una manera idònia, permetent així explotar aquesta informació d’una manera efectiva i eficient. L’objectiu d’aquesta tesis doctoral és desenvolupar noves estratègies que permetin explotar el coneixement biomèdic actual i així extreure informació rellevant per aplicacions biomèdiques futures. Per aquesta finalitat, em fet servir algoritmes de xarxes per tal d’integrar i explotar el coneixement biomèdic en diferents tasques, proporcionant un millor enteniment dels experiments farmacoòmics per tal d’ajudar accelerar el procés de descobriment de nous fàrmacs. Com a resultat, en aquesta tesi hem (i) dissenyat una estratègia per identificar grups funcionals de gens associats a la resposta de línies cel·lulars als fàrmacs, (ii) creat una col·lecció de descriptors biomèdics capaços, entre altres coses, d’anticipar com les cèl·lules responen als fàrmacs o trobar nous usos per fàrmacs existents, (iii) desenvolupat una eina per descobrir quins contextos biològics corresponen a una associació biològica observada experimentalment i, finalment, (iv) hem explorat diferents descriptors químics i biològics rellevants pel procés de descobriment de nous fàrmacs, mostrant com aquests poden ser utilitzats per trobar solucions a reptes actuals dins el camp de la biomedicina

    IN SILICO METHODS FOR DRUG DESIGN AND DISCOVERY

    Get PDF
    Computer-aided drug design (CADD) methodologies are playing an ever-increasing role in drug discovery that are critical in the cost-effective identification of promising drug candidates. These computational methods are relevant in limiting the use of animal models in pharmacological research, for aiding the rational design of novel and safe drug candidates, and for repositioning marketed drugs, supporting medicinal chemists and pharmacologists during the drug discovery trajectory.Within this field of research, we launched a Research Topic in Frontiers in Chemistry in March 2019 entitled “In silico Methods for Drug Design and Discovery,” which involved two sections of the journal: Medicinal and Pharmaceutical Chemistry and Theoretical and Computational Chemistry. For the reasons mentioned, this Research Topic attracted the attention of scientists and received a large number of submitted manuscripts. Among them 27 Original Research articles, five Review articles, and two Perspective articles have been published within the Research Topic. The Original Research articles cover most of the topics in CADD, reporting advanced in silico methods in drug discovery, while the Review articles offer a point of view of some computer-driven techniques applied to drug research. Finally, the Perspective articles provide a vision of specific computational approaches with an outlook in the modern era of CADD

    Visually Interpretable Models of Kinase Selectivity Related Features Derived from Field-Based Proteochemometrics

    No full text
    Achieving selectivity for small organic molecules toward biological targets is a main focus of drug discovery but has been proven difficult, for example, for kinases because of the high similarity of their ATP binding pockets. To support the design of more selective inhibitors with fewer side effects or with altered target profiles for improved efficacy, we developed a method combining ligand- and receptor-based information. Conventional QSAR models enable one to study the interactions of multiple ligands toward a single protein target, but in order to understand the interactions between multiple ligands and multiple proteins, we have used proteochemometrics, a multivariate statistics method that aims to combine and correlate both ligand and protein descriptions with affinity to receptors. The superimposed binding sites of 50 unique kinases were described by molecular interaction fields derived from knowledge-based potentials and Schrödinger’s WaterMap software. Eighty ligands were described by Mold<sup>2</sup>, Open Babel, and Volsurf descriptors. Partial least-squares regression including cross-terms, which describe the selectivity, was used for model building. This combination of methods allows interpretation and easy visualization of the models within the context of ligand binding pockets, which can be translated readily into the design of novel inhibitors
    corecore