262 research outputs found

    Application of Computer-Aided Drug Discovery Methodologies Towards the Rational Design of Drugs Against Infectious Diseases

    Get PDF
    Computer-aided drug discovery involves the application of computer science and programming to solve chemical and biological problems. Specifically, the QSAR (Quantitative Structure Activity Relationships) methodology is used in drug development to provide a rational basis of drug synthesis, rather than a trial and error approach. Molecular dynamics (MD) studies focus on investigating the details of drug-target interactions to elucidate various biophysical characteristics of interest. Infectious diseases like Trypanosoma brucei rhodesiense (TBR) and P. falciparum (malaria) are responsible for millions of deaths annually around the globe. This necessitates an immediate need to design and develop new drugs that efficiently battle these diseases. As a part of the initiatives to improve drug efficacy QSAR studies accomplished the formulation of chemical hypothesis to assist development of drugs against TBR. Results show that CoMSIA 3D QSAR models, with a Pearson’s correlation coefficient of 0.95, predict a compound with meta nitrogens on the phenyl groups, in the combinatorial space based on a biphenyl-furan diamidine design template, to have higher activity against TBR relative to the existing compound set within the same space. Molecular dynamics study, conducted on a linear benzimidazole-biphenyl diamidine that has non-classical structural similarity to earlier known paradigms of minor groove binders, gave insights into the unique water mediated interactions between the DNA minor groove and this ligand. Earlier experiments suggested the interfacial water molecules near the terminal ends of the ligand to be responsible for the exceptianlly high binding constant of the ligand. Results from MD studies show two other modes of binding. The first conformation has a single water molecule with a residency time of 6ns (average) that is closer to the central part of the ligand, which stabilizes the structure in addition to the terminal water. The second conformation that was detected had the ligand completely away from the floor of the minor groove, and hydrogen bonded to the sugar oxygens

    Structure Based 3D-QSAR Studies on Cholinesterase Inhibitors

    Get PDF

    Fundamentals of drug design from a biophysical viewpoint

    Get PDF
    Drug design means many things to many people. Commercially the aim is the development of compounds that can be patented and meet a variety of regulatory standards. In drug design, for medical purposes, toxicity and bio-availability are major consideration

    Lead optimization for new antimalarials and Successful lead identification for metalloproteinases: A Fragment-based approach Using Virtual Screening

    Get PDF
    Lead optimization for new antimalarials and Successful lead identification for metalloproteinases: A Fragment-based approach Using Virtual Screening Computer-aided drug design is an essential part of the modern medicinal chemistry, and has led to the acceleration of many projects. The herein described thesis presents examples for its application in the field of lead optimization and lead identification for three metalloproteins. DOXP-reductoisomerase (DXR) is a key enzyme of the mevalonate independent isoprenoid biosynthesis. Structure-activity relationships for 43 DXR inhibitors are established, derived from protein-based docking, ligand-based 3D QSAR and a combination of both approaches as realized by AFMoC. As part of an effort to optimize the properties of the established inhibitor Fosmidomycin, analogues have been synthesized and tested to gain further insights into the primary determinants of structural affinity. Unfortunately, these structures still leave the active Fosmidomycin conformation and detailed reaction mechanism undetermined. This fact, together with the small inhibitor data set provides a major challenge for presently available docking programs and 3D QSAR tools. Using the recently developed protein tailored scoring protocol AFMoC precise prediction of binding affinities for related ligands as well as the capability to estimate the affinities of structurally distinct inhibitors has been achieved. Farnesyltransferase is a zinc-metallo enzyme that catalyzes the posttranslational modification of numerous proteins involved in intracellular signal transduction. The development of farnesyltransferase inhibitors is directed towards the so-called non-thiol inhibitors because of adverse drug effects connected to free thiols. A first step on the way to non-thiol farnesyltransferase inhibitors was the development of an CAAX-benzophenone peptidomimetic based on a pharmacophore model. On its basis bisubstrate analogues were developed as one class of non-thiol farnesyltransferase inhibitors. In further studies two aryl binding and two distinct specificity sites were postulated. Flexible docking of model compounds was applied to investigate the sub-pockets and design highly active non-thiol farnesyltransferase inhibitor. In addition to affinity, special attention was paid towards in vivo activity and species specificity. The second part of this thesis describes a possible strategy for computer-aided lead discovery. Assembling a complex ligand from simple fragments has recently been introduced as an alternative to traditional HTS. While frequently applied experimentally, only a few examples are known for computational fragment-based approaches. Mostly, computational tools are applied to compile the libraries and to finally assess the assembled ligands. Using the metalloproteinase thermolysin (TLN) as a model target, a computational fragment-based screening protocol has been established. Starting with a data set of commercially available chemical compounds, a fragment library has been compiled considering (1) fragment likeness and (2) similarity to known drugs. The library is screened for target specificity, resulting in 112 fragments to target the zinc binding area and 75 fragments targeting the hydrophobic specificity pocket of the enzyme. After analyzing the performance of multiple docking programs and scoring functions forand the most 14 candidates are selected for further analysis. Soaking experiments were performed for reference fragment to derive a general applicable crystallization protocol for TLN and subsequently for new protein-fragment complex structures. 3-Methylsaspirin could be determined to bind to TLN. Additional studies addressed a retrospective performance analysis of the applied scoring functions and modification on the screening hit. Curios about the differences of aspirin and 3-methylaspirin, 3-chloroaspirin has been synthesized and affinities could be determined to be 2.42 mM; 1.73 mM und 522 μM respectively. The results of the thesis show, that computer aided drug design approaches could successfully support projects in lead optimization and lead identification. fragments in general, the fragments derived from the screening are docke

    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

    Histamine H1-receptor agonists and antagonists molecular modeling and drug design

    Get PDF

    A computational study of the substrate conversion and selective inhibition of aldosterone synthase

    Get PDF
    When a functional or structural impairment of cardiac output has occurred, the cardiovascular system will attempt to compensate for the reduced blood flow. Unfortunately, many of the resulting processes, such as the renin angiotensin aldosterone system, will progressively weaken the heart, resulting in the condition called heart failure. The renin angiotensin aldosterone regulatory system is currently targeted with medicine for heart failure. Many successes for the prolongation of patient age have been achieved by inhibition of angiotensin II synthesis and action. It has become apparent that this approach is suboptimal. Antagonists of aldosterone have provided better treatment options, however, side-effects are still observed. In the search for an alternative therapeutic application, we have studied a novel treatment involving the selective inhibition of aldosterone biosynthesis. The scope of this study has involved the in silico design and prediction of novel inhibitors, the synthesis of these inhibitors and analogues, and finally the in vitro measurement of their potency. The biosynthesis of aldosterone is performed by two cytochrome p450 enzymes, 11B1 and 11B2, denoted as CYP11B1 and CYP11B2, respectively. From these two family members, only CYP11B2 can perform the final synthesis step that converts 18-hydroxycorticosterone into aldosterone. CYP11B1 performs the synthesis of glucocorticoids that are responsible for metabolic, immunologic and homeostatic functions. Because these glucocorticoid actions should not be inhibited, the newly designed medicine must be CYP11B2 selective. Since CYP11B1 is highly homologous to CYP11B2, we have performed an in silico study that allows us to model the interactions of substrates and inhibitors in both the active sites of CYP11B1 and CYP11B2. Using comparative modelling, we have constructed models for the three dimensional architecture of both proteins. These models have been validated by investigating the torsional properties of the protein backbone and residue side chains, the overall protein packing and the dynamic behaviour of the protein models. Subsequently, the models have been used to evaluate the binding mechanisms and conversion mechanisms for the natural steroidal ligands of CYP11B1 and CYP11B2. A hypothetical binding mode has been proposed for 18-hydroxycorticosterone in CYP11B2, featuring the presence of stabilising hydrogen bonding interactions required for its conversion. Quantum mechanical analyses on the conversion of the steroids involved have shown a favourable conversion for this conformation, thereby supporting our hypothesis. In addition, the quantum mechanical analyses have provided insights on steroid conformations in the active sites during conversion. The suitability of the protein models for inhibitor design has been tested by subjecting the models to a case study with four known inhibitors of CYP11B1 and CYP11B2. Using molecular dynamics and molecular docking, the inhibitor potencies for CYP11B1 and CYP11B2 have been predicted, and their interactions with the proteins have been evaluated. The trends in inhibitor potency found by these computational methods have been confirmed by in vitro inhibition measurements. As a next step, the molecular docking study has been expanded to improve the confidence in the predictive power of the models. Using the protein states evaluated by the molecular dynamics study, the molecular docking results of inhibitor analogues have been investigated and the predictive power of the models has been qualitatively improved. In a final approach, we have performed a ligand-based investigation of the inhibitor analogues to determine which ligand characteristics are important for the potency for CYP11B1 and CYP11B2. To this end, we have conducted decision tree analyses on the physico-chemical properties of inhibitor substituents, resulting in a collection of descriptors that can be used for the prediction and design of novel inhibitors. We have shown that a combination of synthesis, molecular modelling and experimental measurements form a promising approach towards the design of potentially new inhibitors
    corecore