31 research outputs found

    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

    Drug Design for CNS Diseases: Polypharmacological Profiling of Compounds Using Cheminformatic, 3D-QSAR and Virtual Screening Methodologies.

    Get PDF
    HIGHLIGHTS Many CNS targets are being explored for multi-target drug designNew databases and cheminformatic methods enable prediction of primary pharmaceutical target and off-targets of compoundsQSAR, virtual screening and docking methods increase the potential of rational drug design The diverse cerebral mechanisms implicated in Central Nervous System (CNS) diseases together with the heterogeneous and overlapping nature of phenotypes indicated that multitarget strategies may be appropriate for the improved treatment of complex brain diseases. Understanding how the neurotransmitter systems interact is also important in optimizing therapeutic strategies. Pharmacological intervention on one target will often influence another one, such as the well-established serotonin-dopamine interaction or the dopamine-glutamate interaction. It is now accepted that drug action can involve plural targets and that polypharmacological interaction with multiple targets, to address disease in more subtle and effective ways, is a key concept for development of novel drug candidates against complex CNS diseases. A multi-target therapeutic strategy for Alzheimer's disease resulted in the development of very effective Multi-Target Designed Ligands (MTDL) that act on both the cholinergic and monoaminergic systems, and also retard the progression of neurodegeneration by inhibiting amyloid aggregation. Many compounds already in databases have been investigated as ligands for multiple targets in drug-discovery programs. A probabilistic method, the Parzen-Rosenblatt Window approach, was used to build a "predictor" model using data collected from the ChEMBL database. The model can be used to predict both the primary pharmaceutical target and off-targets of a compound based on its structure. Several multi-target ligands were selected for further study, as compounds with possible additional beneficial pharmacological activities. Based on all these findings, it is concluded that multipotent ligands targeting AChE/MAO-A/MAO-B and also D1-R/D2-R/5-HT2A -R/H3-R are promising novel drug candidates with improved efficacy and beneficial neuroleptic and procognitive activities in treatment of Alzheimer's and related neurodegenerative diseases. Structural information for drug targets permits docking and virtual screening and exploration of the molecular determinants of binding, hence facilitating the design of multi-targeted drugs. The crystal structures and models of enzymes of the monoaminergic and cholinergic systems have been used to investigate the structural origins of target selectivity and to identify molecular determinants, in order to design MTDLs

    Studying sirtuin inhibitors with in silico and in vitro approaches

    Get PDF

    IN SILICO APPROACHES IN DRUG DESIGN AND DEVELOPMENT: APPLICATIONS TO RATIONAL LIGAND DESIGN AND METABOLISM PREDICTION

    Get PDF
    In the last decades, the applications of computational methods in medicinal chemistry have experienced significant changes which have incredibly expanded their approaches, and more importantly their objectives. The overall aim of the present research project is to explore the different fields of the modelling studies by using well-known computational methods as well as different and innovative techniques. Indeed, computational methods traditionally consisted in ligand-based and the structure-based approaches substantially aimed at optimizing the ligand structure in terms of affinity, potency and selectivity. The studies concerning the muscarinic receptors in the present thesis applied these approaches for the rational design of novel improved bioactive molecules, interacting both in the orthosteric (e.g., 1,4-dioxane agonist) and in the allosteric sites. The research includes also the application of a novel method for target optimization, which consists in the generation of the so-called conformational chimeras to explore the flexibility of the modelled GPCR structures. In parallel, computational methods are finding successful applications in the research phase which precedes the ligand design and which is focused on a detailed validation and characterization of the biological target. A proper example of this kind of studies is given by the study regarding the purinergic receptors, which is aimed at the identification and characterization of potential allosteric binding pockets for the already reported inhibitors, exploiting also innovative approaches for binding site predictions (e.g., PELE, SPILLO-PBSS). Over time, computational applications felt a rich extension of their objectives and one of the clearest examples is represented by the ever increasing attempts to optimize the ADME/Tox profile of the novel compounds, so reducing the marked attrition in drug discovery caused by unsuitable pharmacokinetic profiles. Coherently, the first and main project of the present thesis regards the field of metabolism prediction and is founded on the meta-analysis and the corresponding database called MetaSar, manually collected from the recent specialized literature. This ongoing extended project includes different studies which are overall aimed at developing a comprehensive method for metabolism prediction. In detail, this Thesis reports an interesting application of the database which exploits an innovative predictive technique, the Proteochemometric modelling (PCM). This approach is indeed at the forefront of the latest modelling techniques, as it perfectly fits the growing request of new solutions to deal with the incredibly huge amount of data recently produced by the \u201comics\u201d disciplines. In this context, MetaSar represents an alternative and still appropriate source of data for PCM studies, which also enables the extension of its fields of application to a new avenue, such as the prediction of metabolism biotransformation. In the present thesis, we present the first example of these applications, which involves the building of a classification model for the prediction of the glucuronidation reaction. The field of glucuronidation reactions is exhaustively explored also through an homology modelling study aimed at defining the complete three-dimensional structure of the enzyme UGT2B7, the main isoform of glucuronidation enzymes in humans, in complex with the cofactor UDPGA and a typical substrate, such as Naproxen. The paths of the substrate entering to the binding site and the egress of the product have been investigated by performing Steered Molecular Dynamics (SMD) simulations, which were also useful to gain deeper insights regarding the full mechanism of action and the movements of the cofactor

    Computational prediction of metabolism: sites, products, SAR, P450 enzyme dynamics, and mechanisms.

    Get PDF
    Metabolism of xenobiotics remains a central challenge for the discovery and development of drugs, cosmetics, nutritional supplements, and agrochemicals. Metabolic transformations are frequently related to the incidence of toxic effects that may result from the emergence of reactive species, the systemic accumulation of metabolites, or by induction of metabolic pathways. Experimental investigation of the metabolism of small organic molecules is particularly resource demanding; hence, computational methods are of considerable interest to complement experimental approaches. This review provides a broad overview of structure- and ligand-based computational methods for the prediction of xenobiotic metabolism. Current computational approaches to address xenobiotic metabolism are discussed from three major perspectives: (i) prediction of sites of metabolism (SOMs), (ii) elucidation of potential metabolites and their chemical structures, and (iii) prediction of direct and indirect effects of xenobiotics on metabolizing enzymes, where the focus is on the cytochrome P450 (CYP) superfamily of enzymes, the cardinal xenobiotics metabolizing enzymes. For each of these domains, a variety of approaches and their applications are systematically reviewed, including expert systems, data mining approaches, quantitative structure-activity relationships (QSARs), and machine learning-based methods, pharmacophore-based algorithms, shape-focused techniques, molecular interaction fields (MIFs), reactivity-focused techniques, protein-ligand docking, molecular dynamics (MD) simulations, and combinations of methods. Predictive metabolism is a developing area, and there is still enormous potential for improvement. However, it is clear that the combination of rapidly increasing amounts of available ligand- and structure-related experimental data (in particular, quantitative data) with novel and diverse simulation and modeling approaches is accelerating the development of effective tools for prediction of in vivo metabolism, which is reflected by the diverse and comprehensive data sources and methods for metabolism prediction reviewed here. This review attempts to survey the range and scope of computational methods applied to metabolism prediction and also to compare and contrast their applicability and performance.JK, MJW, JT, PJB, AB and RCG thank Unilever for funding

    Computational Approaches to Drug Profiling and Drug-Protein Interactions

    Get PDF
    Despite substantial increases in R&D spending within the pharmaceutical industry, denovo drug design has become a time-consuming endeavour. High attrition rates led to a long period of stagnation in drug approvals. Due to the extreme costs associated with introducing a drug to the market, locating and understanding the reasons for clinical failure is key to future productivity. As part of this PhD, three main contributions were made in this respect. First, the web platform, LigNFam enables users to interactively explore similarity relationships between ‘drug like’ molecules and the proteins they bind. Secondly, two deep-learning-based binding site comparison tools were developed, competing with the state-of-the-art over benchmark datasets. The models have the ability to predict offtarget interactions and potential candidates for target-based drug repurposing. Finally, the open-source ScaffoldGraph software was presented for the analysis of hierarchical scaffold relationships and has already been used in multiple projects, including integration into a virtual screening pipeline to increase the tractability of ultra-large screening experiments. Together, and with existing tools, the contributions made will aid in the understanding of drug-protein relationships, particularly in the fields of off-target prediction and drug repurposing, helping to design better drugs faster

    A review on machine learning approaches and trends in drug discovery

    Get PDF
    Abstract: Drug discovery aims at finding new compounds with specific chemical properties for the treatment of diseases. In the last years, the approach used in this search presents an important component in computer science with the skyrocketing of machine learning techniques due to its democratization. With the objectives set by the Precision Medicine initiative and the new challenges generated, it is necessary to establish robust, standard and reproducible computational methodologies to achieve the objectives set. Currently, predictive models based on Machine Learning have gained great importance in the step prior to preclinical studies. This stage manages to drastically reduce costs and research times in the discovery of new drugs. This review article focuses on how these new methodologies are being used in recent years of research. Analyzing the state of the art in this field will give us an idea of where cheminformatics will be developed in the short term, the limitations it presents and the positive results it has achieved. This review will focus mainly on the methods used to model the molecular data, as well as the biological problems addressed and the Machine Learning algorithms used for drug discovery in recent years.Instituto de Salud Carlos III; PI17/01826Instituto de Salud Carlos III; PI17/01561Xunta de Galicia; Ref. ED431D 2017/16Xunta de Galicia; Ref. ED431D 2017/23Xunta de Galicia; Ref. ED431C 2018/4

    Machine Learning Methodologies for Interpretable Compound Activity Predictions

    Get PDF
    Machine learning (ML) models have gained attention for mining the pharmaceutical data that are currently generated at unprecedented rates and potentially accelerate the discovery of new drugs. The advent of deep learning (DL) has also raised expectations in pharmaceutical research. A central task in drug discovery is the initial search of compounds with desired biological activity. ML algorithms are able to find patterns in compound structures that are related to bioactivity, the so-called structure-activity relationships (SARs). ML-based predictions can complement biological testing to prioritize further experiments. Moreover, insights into model decisions are highly desired for further validation and identification of activity-relevant substructures. However, the interpretation of complex ML models remains essentially prohibitive. This thesis focuses on ML-based predictions of compound activity against multiple biological targets. Single-target and multi-target models are generated for relevant tasks including the prediction of profiling matrices from screening data and the discrimination between weak and strong inhibitors for more than a hundred kinases. Moreover, the relative performance of distinct modeling strategies is systematically analyzed under varying training conditions, and practical guidelines are reported. Since explainable model decisions are a clear requirement for the utility of ML bioactivity models in pharmaceutical research, methods for the interpretation and intuitive visualization of activity predictions from any ML or DL model are introduced. Taken together, this dissertation presents contributions that advance in the application and rationalization of ML models for biological activity and SAR predictions

    Inhibitor selectivity: profiling and prediction

    Get PDF
    Less than 1 in 10 drug candidates that enter phase 1 clinical trials actually gets approved for human use. The high failure rate is in part due to unforeseen side effects or toxicity. A better understanding of the role of selectivity and a better insight in the off-target activities of drug candidates could greatly aid in preventing candidates to fail for these reasons. This thesis has tried to address some aspects in this challenging part of drug discovery. The use of activity-based protein profiling as presented in Chapters 2 and 3 in drug discovery and hit-to-lead optimization, and in Chapter 5 and 6 for the interaction profiling of a drug candidate, highlights the versatility and importance of this chemical biology technique. Combined with knowledge derived from biochemical assays, such as that developed in Chapter 4, ABPP can greatly aid the medicinal chemist. The recent surge in popularity of machine learning algorithms, backed by exponential growth of the amount of biological data available, holds great promise for drug discovery. Chapters 7 and 8 showed the applicability of one such algorithm, which was able to quite reliably predict interaction profiles. The challenges in finding, determining and predicting selectivity are far from solved, but, by incrementally expanding our understanding of the binding of small molecules to their (off-)targets, truly selective inhibitors might at some point become a reality or their necessity might be mitigated.Medical Biochemistr
    corecore