2,429 research outputs found

    Computational structure‐based drug design: Predicting target flexibility

    Get PDF
    The role of molecular modeling in drug design has experienced a significant revamp in the last decade. The increase in computational resources and molecular models, along with software developments, is finally introducing a competitive advantage in early phases of drug discovery. Medium and small companies with strong focus on computational chemistry are being created, some of them having introduced important leads in drug design pipelines. An important source for this success is the extraordinary development of faster and more efficient techniques for describing flexibility in three‐dimensional structural molecular modeling. At different levels, from docking techniques to atomistic molecular dynamics, conformational sampling between receptor and drug results in improved predictions, such as screening enrichment, discovery of transient cavities, etc. In this review article we perform an extensive analysis of these modeling techniques, dividing them into high and low throughput, and emphasizing in their application to drug design studies. We finalize the review with a section describing our Monte Carlo method, PELE, recently highlighted as an outstanding advance in an international blind competition and industrial benchmarks.We acknowledge the BSC-CRG-IRB Joint Research Program in Computational Biology. This work was supported by a grant from the Spanish Government CTQ2016-79138-R.J.I. acknowledges support from SVP-2014-068797, awarded by the Spanish Government.Peer ReviewedPostprint (author's final draft

    Molecular dynamics and virtual screening approaches in drug discovery

    Get PDF
    Computer-aided drug discovery (CADD) methods are now routinely used in the preclinical phase of drug development. Powerful high-performance computing facilities and the extremely fast CADD methods constantly scale up the coverage of drug-like chemical space achievable in rational drug development. In this thesis, CADD approaches were applied to address several early-phase drug discovery problems. Namely, small molecule binding site detection on a novel target protein, virtual screening (VS) of molecular databases, and characterization of small molecule interactions with metabolic enzymes were studied. Various CADD methods, including molecular dynamics (MD) simulations in mixed solvents, molecular docking, and binding free energy calculations, were employed. Co-solvent MD simulations detected biologically relevant binding sites and provided guidance for screening potential protein-protein interaction inhibitors for a crucial protein of the SARS-CoV-2. VS with fragment- and negative image-based (F-NIB) models identified three active and structurally novel inhibitors of the putative drug target phosphodiesterase 10A. MD simulations and docking provided detailed insights on the effects of active site structural flexibility and variation on the binding and resultant metabolism of small molecules with the cytochrome P450 enzymes. The results presented in this thesis contribute to the increasing evidence that supports employment and further development of CADD approaches in drug discovery. Ultimately, rational drug development coupled with CADD may enable higher quality drug candidates to the human studies in the future, reducing the risk of financially and temporally costly clinical failure. KEYWORDS: Structure-based drug development, Computer-aided drug discovery (CADD), Molecular dynamics (MD) simulation, Virtual screening (VS), Fragmentand negative image-based (F-NIB) model, Structure-activity relationship (QSAR), Cytochrome P450 ligand binding predictionMolekyylidynamiikka- ja virtuaaliseulontamenetelmät lääkeaine-etsinnässä Tietokoneavusteista lääkeaine-etsintää käytetään nykyisin yleisesti prekliinisessä lääketutkimuksessa. Suurteholaskenta ja äärimmäisen nopeat tietokoneavusteiset lääkeaine-etsintämenetelmät mahdollistavat jatkuvasti kattavamman lääkkeenkaltaisten molekyylien kemiallisen avaruuden seulonnan. Tässä väitöskirjassa tietokonepohjaisia menetelmiä hyödynnettiin lääketutkimuksen prekliiniseen vaiheeseen liittyvissä tyypillisissä tutkimusongelmissa. Työhön kuului pienmolekyylien sitoutumisalueiden tunnistus uuden kohdeproteiinin rakenteesta, molekyylitietokantojen virtuaaliseulonta sekä pienmolekyylien ja metabolian entsyymien välisten vuorovaikutusten tietokonemallinnus. Työssä käytettiin useita tietokoneavusteisen lääkeaine-etsinnän menetelmiä, sisältäen molekyylidynamiikkasimulaatiot (MD-simulaatiot) vaihtuvissa liuottimissa, molekulaarisen telakoinnin ja sitoutumisenergian laskennan. Orgaanisen liuottimen ja veden sekoituksessa tehdyt MD-simulaatiot tunnistivat biologisesti merkittäviä sitoutumisalueita SARS-CoV-2:n tärkeästä proteiinista ja ohjasivat infektioon liittyvän proteiini-proteiinivuorovaikutuksen potentiaalisten estäjien etsintää. Virtuaaliseulonnalla tunnistettiin kolme rakenteellisesti uudenlaista tunnetun lääkekehityskohteen, fosfodiesteraasi 10A:n, estäjää hyödyntäen fragmentti- ja negatiivikuvamalleja. MD-simulaatiot ja telakointi tuottivat yksityiskohtaista tietoa sytokromi P450 entsyymien aktiivisen kohdan rakenteen jouston ja muutosten vaikutuksesta pienmolekyylien sitoutumiseen ja metaboliaan. Tämän väitöskirjan tulokset tukevat kasvavaa todistusaineistoa tietokoneavusteisen lääkeaine-etsinnän käytön ja kehityksen hyödyllisyydestä prekliinisessä lääketutkimuksessa. Tietokoneavusteinen lääkeaine-etsintä voi lopulta mahdollistaa korkeampilaatuisten lääkekandidaattien päätymisen ihmiskokeisiin, pienentäen taloudellisesti ja ajallisesti kalliin kliinisen tutkimuksen epäonnistumisen riskiä. AVAINSANAT: Rakennepohjainen lääkeainekehitys, Tietokoneavusteinen lääkeaine-etsintä, Molekyylidynamiikkasimulaatio (MD-simulaatio), Virtuaaliseulonta, Fragmentti- ja negatiivikuvamalli, Rakenne-aktiivisuussuhde, Sytokromi P450 ligandien sitoutumisen ennustu

    Distinguishing Binders from False Positives by Free Energy Calculations: Fragment Screening Against the Flap Site of HIV Protease

    Full text link
    Molecular docking is a powerful tool used in drug discovery and structural biology for predicting the structures of ligand–receptor complexes. However, the accuracy of docking calculations can be limited by factors such as the neglect of protein reorganization in the scoring function; as a result, ligand screening can produce a high rate of false positive hits. Although absolute binding free energy methods still have difficulty in accurately rank-ordering binders, we believe that they can be fruitfully employed to distinguish binders from nonbinders and reduce the false positive rate. Here we study a set of ligands that dock favorably to a newly discovered, potentially allosteric site on the flap of HIV-1 protease. Fragment binding to this site stabilizes a closed form of protease, which could be exploited for the design of allosteric inhibitors. Twenty-three top-ranked protein–ligand complexes from AutoDock were subject to the free energy screening using two methods, the recently developed binding energy analysis method (BEDAM) and the standard double decoupling method (DDM). Free energy calculations correctly identified most of the false positives (≥83%) and recovered all the confirmed binders. The results show a gap averaging ≥3.7 kcal/mol, separating the binders and the false positives. We present a formula that decomposes the binding free energy into contributions from the receptor conformational macrostates, which provides insights into the roles of different binding modes. Our binding free energy component analysis further suggests that improving the treatment for the desolvation penalty associated with the unfulfilled polar groups could reduce the rate of false positive hits in docking. The current study demonstrates that the combination of docking with free energy methods can be very useful for more accurate ligand screening against valuable drug targets

    Protein-Ligand Complex Generator & Drug Screening via Tiered Tensor Transform

    Full text link
    The generation of small molecule candidate (ligand) binding poses in its target protein pocket is important for computer-aided drug discovery. Typical rigid-body docking methods ignore the pocket flexibility of protein, while the more accurate pose generation using molecular dynamics is hindered by slow protein dynamics. We develop a tiered tensor transform (3T) algorithm to rapidly generate diverse protein-ligand complex conformations for both pose and affinity estimation in drug screening, requiring neither machine learning training nor lengthy dynamics computation, while maintaining both coarse-grain-like coordinated protein dynamics and atomistic-level details of the complex pocket. The 3T conformation structures we generate achieve significantly higher accuracy in active ligand classification than traditional ensemble docking using hundreds of experimental protein conformations. Furthermore, we demonstrate that 3T can be used to explore distant protein-ligand binding poses within the protein pocket. 3T structure transformation is decoupled from the system physics, making future usage in other computational scientific domains possible

    HOMOLOGY MODELING AND DEVELOPMENT OF DIHYDRODIPICONILATE REDUCTASE INHIBITORS OF KLEBSIELLA PNEUMONIA: A COMPUTATIONAL APPROACH

    Get PDF
    Objective: In order to development of novel, potent and selective inhibitors of Dihydropiconilate reductase (KpDHDPR) of multidrug resistant Klebsiella pneumonia. Methods: Protein sequence of KpDHDPR was retrieved from the UNIPROT and the primary and secondary structure was analyzed using Prot Param, SOPMA, GOR4 and Chou and Fasman. Afterword's, 3D structure of KpDHDPR was built by using MODELLEER9.14. The Molecular dynamics simulation was carried out using NAMD2.9 with CHARMM27 force field for 10 picoseconds and production run with for 400 picoseconds time period covered with water box. Molecular docking and virtual screening was carried out using Auto Dock Vina4.0 with PyRx interface. Bond angles, bond lengths, bond distances and binding interactions were analyzed using PyMol. Toxicity assessment and Lipinski rule of five of ligand were assessed using MOLINSPIRATION and OSIRIS Server. Results: 3D structure of KpDHDPR was resolved on the basis of EcDHDPR that revealed N-terminal nucleotide domain and C-terminal substrate binding domain which are connected by a short hinge region. Nucleotide binding domain is formed with seven α-helices and the Substrate binding domain is composed with three α-helices and Rossman fold is observed with four α-helices and seven β-strands. Molecular docking analysis revealed that NADPH has exhibited more binding affinity to KpDHDPR than NADH. As results of virtual screening and docking, six compounds viz. ZINC04280533, ZINC04280532, ZINC04280468, ZINC33378709, ZINC05280538 and ZINC25694354 were identified. Bioavailability of these inhibitors are comply with the Lipinski rule of five, good pharmacokinetic and drug likeness properties. Conclusion: In conclusion, in silico studies revealed that these lead scaffolds could helpful in the development of KpDHDP R inhibitors. Hence, these drug candidates might be promoted as promising antibacterial agents for the treatment of drug resistant gram negative bacterial infections

    Technological developments in Virtual Screening for the discovery of small molecules with novel mechanisms of action

    Get PDF
    Programa de Doctorat en Recerca, Desenvolupament i Control de Medicaments[eng] Advances in structural and molecular biology have favoured the rational development of novel drugs thru structure-based drug design (SBDD). Particularly, computational tools have proven to be rapid and efficient tools for hit discovery and optimization. The main motivation of this thesis is to improve and develop new methods in the area of computer-based drug discovery in order to study challenging targets. Specifically, this thesis is focused on docking and Virtual Screening (VS) methodologies to be able to exploit non-standard sites, like protein-protein interfaces or allosteric sites, and discover bioactive molecules with novel mechanisms of action. First, I developed an automatic pipeline for binding mode prediction that applies knowledge- based restraints and validated the approach by participating in the CELPP Challenge, a blind pose prediction challenge. The aim of the first VS in this thesis is to find small molecules able to not only disrupt the RANK-RANKL interaction but also inhibit the constitutive activation of the receptor. With a combination of computational, biophysical, and cell-based assays we were able to identify the first small molecule binders for RANK that could be used as a treatment for Triple Negative Breast Cancer. When working with challenging targets, or with non-standard mechanisms of action, the relationship between binding and the biological response is unpredictable, because the biological response (if any) will depend on the biological function of the particular allosteric site, which is generally unknown. For this reason, we then tested the applicability of the combination of ultrahigh-throughput VS with low-throughput high content assay. This allowed us to characterize a novel allosteric pocket in PTEN and also describe the first allosteric modulators for this protein. Finally, as the accessible Chemical Space grows at a rapid pace, we developed an algorithm to efficiently explore ultra-large Chemical Collections using a Bottom-up approach. We prospectively validated the approach in BRD4 and identified novel BRD4 inhibitors with an affinity comparable to advanced drug candidates for this target.[spa] Els avenços en biologia estructural i molecular han afavorit el desenvolupament racional de nous fàrmacs a través del disseny de fàrmacs basat en l'estructura (SBDD). En particular, les eines computacionals han demostrat ser ràpides i eficients per al descobriment i l'optimització de fàrmacs. La principal motivació d'aquesta tesi és millorar i desenvolupar nous mètodes en l'àrea del descobriment de fàrmacs per ordinador per tal d'estudiar proteïnes complexes. Concretament, aquesta tesi se centra en les metodologies d'acoblament i de cribratge virtual (CV) per poder explotar llocs no estàndard, com interfícies proteïna-proteïna o llocs al·lostèrics, i descobrir molècules bioactives amb nous mecanismes d'acció. En primer lloc, vaig desenvolupar un protocol automàtic per a la predicció del mode d’unió aplicant restriccions basades en el coneixement i vaig validar l'enfocament participant en el repte CELPP, un repte de predicció del mode d’unió a cegues. L'objectiu del primer CV d'aquesta tesi és trobar petites molècules capaces no només d'interrompre la interacció RANK-RANKL sinó també d'inhibir l'activació constitutiva del receptor. Amb una combinació d'assajos computacionals, biofísics i basats en cèl·lules, vam poder identificar les primeres molècules petites per a RANK que es podrien utilitzar com a tractament per al càncer de mama triple negatiu. Quan es treballa amb proteïnes complexes, o amb mecanismes d'acció no estàndard, la relació entre la unió i la resposta biològica és impredictible, perquè la resposta biològica (si n'hi ha) dependrà de la funció biològica del lloc al·lostèric particular, que generalment és desconeguda. Per aquest motiu, després vam provar l'aplicabilitat de la combinació de CV d'alt rendiment amb assaig de contingut alt de baix rendiment. Això ens va permetre caracteritzar un nou lloc d’unió al·lostèric en PTEN i també descriure els primers moduladors al·lostèrics d'aquesta proteïna. Finalment, a mesura que l'espai químic accessible creix a un ritme ràpid, hem desenvolupat un algorisme per explorar de manera eficient col·leccions de productes químics molt grans mitjançant un enfocament de baix a dalt. Vam validar aquest enfocament amb BRD4 i vam identificar nous inhibidors de BRD4 amb una afinitat comparable als candidats a fàrmacs més avançats per a aquesta proteïna
    corecore