Virtual screening

Last updated
Figure 1. Flow Chart of Virtual Screening FlowChart.png
Figure 1. Flow Chart of Virtual Screening

Virtual screening (VS) is a computational technique used in drug discovery to search libraries of small molecules in order to identify those structures which are most likely to bind to a drug target, typically a protein receptor or enzyme. [2] [3]

Contents

Virtual screening has been defined as "automatically evaluating very large libraries of compounds" using computer programs. [4] As this definition suggests, VS has largely been a numbers game focusing on how the enormous chemical space of over 1060 conceivable compounds [5] can be filtered to a manageable number that can be synthesized, purchased, and tested. Although searching the entire chemical universe may be a theoretically interesting problem, more practical VS scenarios focus on designing and optimizing targeted combinatorial libraries and enriching libraries of available compounds from in-house compound repositories or vendor offerings. As the accuracy of the method has increased, virtual screening has become an integral part of the drug discovery process. [6] [1] Virtual Screening can be used to select in house database compounds for screening, choose compounds that can be purchased externally, and to choose which compound should be synthesized next.

Methods

There are two broad categories of screening techniques: ligand-based and structure-based. [7] The remainder of this page will reflect Figure 1 Flow Chart of Virtual Screening.

Ligand-based methods

Given a set of structurally diverse ligands that binds to a receptor, a model of the receptor can be built by exploiting the collective information contained in such set of ligands. Different computational techniques explore the structural, electronic, molecular shape, and physicochemical similarities of different ligands that could imply their mode of action against a specific molecular receptor or cell lines. [8] A candidate ligand can then be compared to the pharmacophore model to determine whether it is compatible with it and therefore likely to bind. [9] Different 2D chemical similarity analysis methods [10] have been used to scan a databases to find active ligands. Another popular approach used in ligand-based virtual screening consist on searching molecules with shape similar to that of known actives, as such molecules will fit the target's binding site and hence will be likely to bind the target. There are a number of prospective applications of this class of techniques in the literature. [11] [12] [13] Pharmacophoric extensions of these 3D methods are also freely-available as webservers. [14] [15] Also shape based virtual screening has gained significant popularity. [16]

Structure-based methods

Structure-based virtual screening approach includes different computational techniques that consider the structure of the receptor that is the molecular target of the investigated active ligands. Some of these techniques include molecular docking, structure-based pharmacophore prediction, and molecular dynamics simulations. [17] [18] [8] Molecular docking is the most used structure-based technique, and it applies a scoring function to estimate the fitness of each ligand against the binding site of the macromolecular receptor, helping to choose the ligands with the most high affinity. [19] [20] [21] Currently, there are some webservers oriented to prospective virtual screening. [22] [23]

Hybrid methods

Hybrid methods that rely on structural and ligand similarity were also developed to overcome the limitations of traditional VLS approaches. This methodologies utilizes evolution‐based ligand‐binding information to predict small-molecule binders [24] [25] and can employ both global structural similarity and pocket similarity. [24] A global structural similarity based approach employs both an experimental structure or a predicted protein model to find structural similarity with proteins in the PDB holo‐template library. Upon detecting significant structural similarity, 2D fingerprint based Tanimoto coefficient metric is applied to screen for small-molecules that are similar to ligands extracted from selected holo PDB templates. [26] [27] The predictions from this method have been experimentally assessed and shows good enrichment in identifying active small molecules.

The above specified method depends on global structural similarity and is not capable of a priori selecting a particular ligand‐binding site in the protein of interest. Further, since the methods rely on 2D similarity assessment for ligands, they are not capable of recognizing stereochemical similarity of small-molecules that are substantially different but demonstrate geometric shape similarity. To address these concerns, a new pocket centric approach, PoLi, capable of targeting specific binding pockets in holo‐protein templates, was developed and experimentally assessed.

Computing Infrastructure

The computation of pair-wise interactions between atoms, which is a prerequisite for the operation of many virtual screening programs, scales by , N is the number of atoms in the system. Due to the quadratic scaling, the computational costs increase quickly.

Ligand-based Approach

Ligand-based methods typically require a fraction of a second for a single structure comparison operation. Sometimes a single CPU is enough to perform a large screening within hours. However, several comparisons can be made in parallel in order to expedite the processing of a large database of compounds.

Structure-based Approach

The size of the task requires a parallel computing infrastructure, such as a cluster of Linux systems, running a batch queue processor to handle the work, such as Sun Grid Engine or Torque PBS.

A means of handling the input from large compound libraries is needed. This requires a form of compound database that can be queried by the parallel cluster, delivering compounds in parallel to the various compute nodes. Commercial database engines may be too ponderous, and a high speed indexing engine, such as Berkeley DB, may be a better choice. Furthermore, it may not be efficient to run one comparison per job, because the ramp up time of the cluster nodes could easily outstrip the amount of useful work. To work around this, it is necessary to process batches of compounds in each cluster job, aggregating the results into some kind of log file. A secondary process, to mine the log files and extract high scoring candidates, can then be run after the whole experiment has been run.

Accuracy

The aim of virtual screening is to identify molecules of novel chemical structure that bind to the macromolecular target of interest. Thus, success of a virtual screen is defined in terms of finding interesting new scaffolds rather than the total number of hits. Interpretations of virtual screening accuracy should, therefore, be considered with caution. Low hit rates of interesting scaffolds are clearly preferable over high hit rates of already known scaffolds.

Most tests of virtual screening studies in the literature are retrospective. In these studies, the performance of a VS technique is measured by its ability to retrieve a small set of previously known molecules with affinity to the target of interest (active molecules or just actives) from a library containing a much higher proportion of assumed inactives or decoys. There are several distinct ways to select decoys by matching the properties of the corresponding active molecule [28] and more recently decoys are also selected in a property-unmatched manner. [29] The actual impact of decoy selection, either for training or testing purposes, has also been discussed. [29] [30]

By contrast, in prospective applications of virtual screening, the resulting hits are subjected to experimental confirmation (e.g., IC50 measurements). There is consensus that retrospective benchmarks are not good predictors of prospective performance and consequently only prospective studies constitute conclusive proof of the suitability of a technique for a particular target. [31] [32] [33] [34] [35]

Application to drug discovery

Virtual screening is a very useful application when it comes to identifying hit molecules as a beginning for medicinal chemistry. As the virtual screening approach begins to become a more vital and substantial technique within the medicinal chemistry industry the approach has had an expeditious increase. [36]

Ligand-based methods

While not knowing the structure trying to predict how the ligands will bind to the receptor. With the use of pharmacophore features each ligand identified donor, and acceptors. Equating features are overlaid, however given it is unlikely there is a single correct solution. [1]

Pharmacophore models

This technique is used when merging the results of searches by using unlike reference compounds, same descriptors and coefficient, but different active compounds. This technique is beneficial because it is more efficient than just using a single reference structure along with the most accurate performance when it comes to diverse actives. [1]

Pharmacophore is an ensemble of steric and electronic features that are needed to have an optimal supramolecular interaction or interactions with a biological target structure in order to precipitate its biological response. Choose a representative as a set of actives, most methods will look for similar bindings. [37] It is preferred to have multiple rigid molecules and the ligands should be diversified, in other words ensure to have different features that don't occur during the binding phase. [1]

Shape-Based Virtual Screening

Shape-based molecular similarity approaches have been established as important and popular virtual screening techniques. At present, the highly optimized screening platform ROCS (Rapid Overlay of Chemical Structures) is considered the de facto industry standard for shape-based, ligand-centric virtual screening. [38] [39] [40] It uses a Gaussian function to define molecular volumes of small organic molecules. The selection of the query conformation is less important, rendering shape-based screening ideal for ligand-based modeling: As the availability of a bioactive conformation for the query is not the limiting factor for screening — it is more the selection of query compound(s) that is decisive for screening performance. [16]

Field-Based Virtual Screening

As an improvement to Shape-Based similarity methods, Field-Based methods try to take into account all the fields that influence a ligand-receptor interaction while being agnostic of the chemical structure used as a query. Examples of other fields that are used in these methods are Electrostatic or Hidrophobic fields.

Quantitative-Structure Activity Relationship

Quantitative-Structure Activity Relationship (QSAR) models consist of predictive models based on information extracted from a set of known active and known inactive compounds. [41] SAR's (Structure Activity Relationship) where data is treated qualitatively and can be used with structural classes and more than one binding mode. Models prioritize compounds for lead discovery. [1]

Machine learning algorithms

Machine learning algorithms have been widely used in virtual screening approaches. Supervised learning techniques use a training and test datasets composed of known active and known inactive compounds. Different ML algorithms have been applied with success in virtual screening strategies, such as recursive partitioning, support vector machines, k-nearest neighbors and neural networks. [42] [43] [44] These models find the probability that a compound is active and then ranking each compound based on its probability. [1]

Substructural analysis in Machine Learning

The first Machine Learning model used on large datasets is the Substructure Analysis that was created in 1973. Each fragment substructure make a continuous contribution an activity of specific type. [1] Substructure is a method that overcomes the difficulty of massive dimensionality when it comes to analyzing structures in drug design. An efficient substructure analysis is used for structures that have similarities to a multi-level building or tower. Geometry is used for numbering boundary joints for a given structure in the onset and towards the climax. When the method of special static condensation and substitutions routines are developed this method is proved to be more productive than the previous substructure analysis models. [45]

Recursive partitioning

Recursively partitioning is method that creates a decision tree using qualitative data. Understanding the way rules break classes up with a low error of misclassification while repeating each step until no sensible splits can be found. However, recursive partitioning can have poor prediction ability potentially creating fine models at the same rate. [1]

Structure-based methods known protein ligand docking

Ligand can bind into an active site within a protein by using a docking search algorithm, and scoring function in order to identify the most likely cause for an individual ligand while assigning a priority order. [1] [46]

See also

Related Research Articles

<span class="mw-page-title-main">Structural bioinformatics</span> Bioinformatics subfield

Structural bioinformatics is the branch of bioinformatics that is related to the analysis and prediction of the three-dimensional structure of biological macromolecules such as proteins, RNA, and DNA. It deals with generalizations about macromolecular 3D structures such as comparisons of overall folds and local motifs, principles of molecular folding, evolution, binding interactions, and structure/function relationships, working both from experimentally solved structures and from computational models. The term structural has the same meaning as in structural biology, and structural bioinformatics can be seen as a part of computational structural biology. The main objective of structural bioinformatics is the creation of new methods of analysing and manipulating biological macromolecular data in order to solve problems in biology and generate new knowledge.

A chemical database is a database specifically designed to store chemical information. This information is about chemical and crystal structures, spectra, reactions and syntheses, and thermophysical data.

<span class="mw-page-title-main">Drug discovery</span> Pharmaceutical procedure

In the fields of medicine, biotechnology and pharmacology, drug discovery is the process by which new candidate medications are discovered.

<span class="mw-page-title-main">Drug design</span> Inventive process of finding new medications based on the knowledge of a biological target

Drug design, often referred to as rational drug design or simply rational design, is the inventive process of finding new medications based on the knowledge of a biological target. The drug is most commonly an organic small molecule that activates or inhibits the function of a biomolecule such as a protein, which in turn results in a therapeutic benefit to the patient. In the most basic sense, drug design involves the design of molecules that are complementary in shape and charge to the biomolecular target with which they interact and therefore will bind to it. Drug design frequently but not necessarily relies on computer modeling techniques. This type of modeling is sometimes referred to as computer-aided drug design. Finally, drug design that relies on the knowledge of the three-dimensional structure of the biomolecular target is known as structure-based drug design. In addition to small molecules, biopharmaceuticals including peptides and especially therapeutic antibodies are an increasingly important class of drugs and computational methods for improving the affinity, selectivity, and stability of these protein-based therapeutics have also been developed.

Quantitative structure–activity relationship models are regression or classification models used in the chemical and biological sciences and engineering. Like other regression models, QSAR regression models relate a set of "predictor" variables (X) to the potency of the response variable (Y), while classification QSAR models relate the predictor variables to a categorical value of the response variable.

<span class="mw-page-title-main">Medicinal chemistry</span> Scientific branch of chemistry

Medicinal or pharmaceutical chemistry is a scientific discipline at the intersection of chemistry and pharmacy involved with designing and developing pharmaceutical drugs. Medicinal chemistry involves the identification, synthesis and development of new chemical entities suitable for therapeutic use. It also includes the study of existing drugs, their biological properties, and their quantitative structure-activity relationships (QSAR).

<span class="mw-page-title-main">Pharmacophore</span> Abstract description of molecular features

In medicinal chemistry and molecular biology, a pharmacophore is an abstract description of molecular features that are necessary for molecular recognition of a ligand by a biological macromolecule. IUPAC defines a pharmacophore to be "an ensemble of steric and electronic features that is necessary to ensure the optimal supramolecular interactions with a specific biological target and to trigger its biological response". A pharmacophore model explains how structurally diverse ligands can bind to a common receptor site. Furthermore, pharmacophore models can be used to identify through de novo design or virtual screening novel ligands that will bind to the same receptor.

<span class="mw-page-title-main">Ligand (biochemistry)</span> Substance that forms a complex with a biomolecule

In biochemistry and pharmacology, a ligand is a substance that forms a complex with a biomolecule to serve a biological purpose. The etymology stems from Latin ligare, which means 'to bind'. In protein-ligand binding, the ligand is usually a molecule which produces a signal by binding to a site on a target protein. The binding typically results in a change of conformational isomerism (conformation) of the target protein. In DNA-ligand binding studies, the ligand can be a small molecule, ion, or protein which binds to the DNA double helix. The relationship between ligand and binding partner is a function of charge, hydrophobicity, and molecular structure.

<span class="mw-page-title-main">Docking (molecular)</span> Prediction method in molecular modeling

In the field of molecular modeling, docking is a method which predicts the preferred orientation of one molecule to a second when a ligand and a target are bound to each other to form a stable complex. Knowledge of the preferred orientation in turn may be used to predict the strength of association or binding affinity between two molecules using, for example, scoring functions.

<span class="mw-page-title-main">Chemogenomics</span>

Chemogenomics, or chemical genomics, is the systematic screening of targeted chemical libraries of small molecules against individual drug target families with the ultimate goal of identification of novel drugs and drug targets. Typically some members of a target library have been well characterized where both the function has been determined and compounds that modulate the function of those targets have been identified. Other members of the target family may have unknown function with no known ligands and hence are classified as orphan receptors. By identifying screening hits that modulate the activity of the less well characterized members of the target family, the function of these novel targets can be elucidated. Furthermore, the hits for these targets can be used as a starting point for drug discovery. The completion of the human genome project has provided an abundance of potential targets for therapeutic intervention. Chemogenomics strives to study the intersection of all possible drugs on all of these potential targets.

Protein–ligand docking is a molecular modelling technique. The goal of protein–ligand docking is to predict the position and orientation of a ligand when it is bound to a protein receptor or enzyme. Pharmaceutical research employs docking techniques for a variety of purposes, most notably in the virtual screening of large databases of available chemicals in order to select likely drug candidates. There has been rapid development in computational ability to determine protein structure with programs such as AlphaFold, and the demand for the corresponding protein-ligand docking predictions is driving implementation of software that can find accurate models. Once the protein folding can be predicted accurately along with how the ligands of various structures will bind to the protein, the ability for drug development to progress at a much faster rate becomes possible.

In the fields of computational chemistry and molecular modelling, scoring functions are mathematical functions used to approximately predict the binding affinity between two molecules after they have been docked. Most commonly one of the molecules is a small organic compound such as a drug and the second is the drug's biological target such as a protein receptor. Scoring functions have also been developed to predict the strength of intermolecular interactions between two proteins or between protein and DNA.

SMILES arbitrary target specification (SMARTS) is a language for specifying substructural patterns in molecules. The SMARTS line notation is expressive and allows extremely precise and transparent substructural specification and atom typing.

<span class="mw-page-title-main">Chemical similarity</span> Chemical term

Chemical similarity refers to the similarity of chemical elements, molecules or chemical compounds with respect to either structural or functional qualities, i.e. the effect that the chemical compound has on reaction partners in inorganic or biological settings. Biological effects and thus also similarity of effects are usually quantified using the biological activity of a compound. In general terms, function can be related to the chemical activity of compounds.

Inte:Ligand was founded in Maria Enzersdorf, Lower Austria (Niederösterreich) in 2003. They established the company headquarters on Mariahilferstrasse in Vienna, Austria that same year.

Fragment-based lead discovery (FBLD) also known as fragment-based drug discovery (FBDD) is a method used for finding lead compounds as part of the drug discovery process. Fragments are small organic molecules which are small in size and low in molecular weight. It is based on identifying small chemical fragments, which may bind only weakly to the biological target, and then growing them or combining them to produce a lead with a higher affinity. FBLD can be compared with high-throughput screening (HTS). In HTS, libraries with up to millions of compounds, with molecular weights of around 500 Da, are screened, and nanomolar binding affinities are sought. In contrast, in the early phase of FBLD, libraries with a few thousand compounds with molecular weights of around 200 Da may be screened, and millimolar affinities can be considered useful. FBLD is a technique being used in research for discovering novel potent inhibitors. This methodology could help to design multitarget drugs for multiple diseases. The multitarget inhibitor approach is based on designing an inhibitor for the multiple targets. This type of drug design opens up new polypharmacological avenues for discovering innovative and effective therapies. Neurodegenerative diseases like Alzheimer’s (AD) and Parkinson’s, among others, also show rather complex etiopathologies. Multitarget inhibitors are more appropriate for addressing the complexity of AD and may provide new drugs for controlling the multifactorial nature of AD, stopping its progression.

Topological inhibitors are rigid three-dimensional molecules of inorganic, organic, and hybrid compounds that form multicentered supramolecular interactions in vacant cavities of protein macromolecules and their complexes . Extensive surface and very diverse geometry make cage compounds with an encapsulated metal ion (clathrochelates) suitable for targeting both the active and allosteric sites of enzymes as well as the interfaces of their macromolecular complexes. An efficient structure- and concentration-dependent transcription inhibition in a model in vitro systems based on RNA and DNA polymerases by the iron(II) mono- and bis-clathrochelates at their submicro- and nanomolar concentrations, respectively, is observed in. Molecular docking and preincubation experiments suggested that these cage compounds form supramolecular assemblies with protein residues as well as with DNA and RNA. Thus, they are prospective precursors for the design of antiviral and anticancer drug candidates.

Early twenty-first century pesticide research has focused on developing molecules that combine low use rates and that are more selective, safer, resistance-breaking and cost-effective. Obstacles include increasing pesticide resistance and an increasingly stringent regulatory environment.

Chemoproteomics entails a broad array of techniques used to identify and interrogate protein-small molecule interactions. Chemoproteomics complements phenotypic drug discovery, a paradigm that aims to discover lead compounds on the basis of alleviating a disease phenotype, as opposed to target-based drug discovery, in which lead compounds are designed to interact with predetermined disease-driving biological targets. As phenotypic drug discovery assays do not provide confirmation of a compound's mechanism of action, chemoproteomics provides valuable follow-up strategies to narrow down potential targets and eventually validate a molecule's mechanism of action. Chemoproteomics also attempts to address the inherent challenge of drug promiscuity in small molecule drug discovery by analyzing protein-small molecule interactions on a proteome-wide scale. A major goal of chemoproteomics is to characterize the interactome of drug candidates to gain insight into mechanisms of off-target toxicity and polypharmacology.

Molecular Operating Environment (MOE) is a drug discovery software platform that integrates visualization, modeling and simulations, as well as methodology development, in one package. MOE scientific applications are used by biologists, medicinal chemists and computational chemists in pharmaceutical, biotechnology and academic research. MOE runs on Windows, Linux, Unix, and macOS. Main application areas in MOE include structure-based design, fragment-based design, ligand-based design, pharmacophore discovery, medicinal chemistry applications, biologics applications, structural biology and bioinformatics, protein and antibody modeling, molecular modeling and simulations, virtual screening, cheminformatics & QSAR. The Scientific Vector Language (SVL) is the built-in command, scripting and application development language of MOE.

References

  1. 1 2 3 4 5 6 7 8 9 10 Gillet V (2013). "Ligand-Based and Structure-Based Virtual Screening" (PDF). The University of Sheffield.
  2. Rester U (July 2008). "From virtuality to reality - Virtual screening in lead discovery and lead optimization: a medicinal chemistry perspective". Current Opinion in Drug Discovery & Development. 11 (4): 559–68. PMID   18600572.
  3. Rollinger JM, Stuppner H, Langer T (2008). "Virtual screening for the discovery of bioactive natural products". Natural Compounds as Drugs Volume I. Progress in Drug Research. Vol. 65. pp. 211, 213–49. doi:10.1007/978-3-7643-8117-2_6. ISBN   978-3-7643-8098-4. PMC   7124045 . PMID   18084917.{{cite book}}: |journal= ignored (help)
  4. Walters WP, Stahl MT, Murcko MA (1998). "Virtual screening – an overview". Drug Discov. Today. 3 (4): 160–178. doi:10.1016/S1359-6446(97)01163-X.
  5. Bohacek RS, McMartin C, Guida WC (1996). "The art and practice of structure-based drug design: a molecular modeling perspective". Med. Res. Rev. 16 (1): 3–50. doi:10.1002/(SICI)1098-1128(199601)16:1<3::AID-MED1>3.0.CO;2-6. PMID   8788213.
  6. McGregor MJ, Luo Z, Jiang X (June 11, 2007). "Chapter 3: Virtual screening in drug discovery". In Huang Z (ed.). Drug Discovery Research. New Frontiers in the Post-Genomic Era. Wiley-VCH: Weinheim, Germany. pp. 63–88. ISBN   978-0-471-67200-5.
  7. McInnes C (October 2007). "Virtual screening strategies in drug discovery". Current Opinion in Chemical Biology. 11 (5): 494–502. doi:10.1016/j.cbpa.2007.08.033. PMID   17936059.
  8. 1 2 Santana K, do Nascimento LD, Lima e Lima A, Damasceno V, Nahum C, Braga RC, Lameira J (2021-04-29). "Applications of Virtual Screening in Bioprospecting: Facts, Shifts, and Perspectives to Explore the Chemo-Structural Diversity of Natural Products". Frontiers in Chemistry. 9: 662688. Bibcode:2021FrCh....9..155S. doi: 10.3389/fchem.2021.662688 . ISSN   2296-2646. PMC   8117418 . PMID   33996755.
  9. Sun H (2008). "Pharmacophore-based virtual screening". Current Medicinal Chemistry. 15 (10): 1018–24. doi:10.2174/092986708784049630. PMID   18393859.
  10. Willet P, Barnard JM, Downs GM (1998). "Chemical similarity searching". Journal of Chemical Information and Computer Sciences. 38 (6): 983–996. CiteSeerX   10.1.1.453.1788 . doi:10.1021/ci9800211.
  11. Rush TS, Grant JA, Mosyak L, Nicholls A (March 2005). "A shape-based 3-D scaffold hopping method and its application to a bacterial protein-protein interaction". Journal of Medicinal Chemistry. 48 (5): 1489–95. CiteSeerX   10.1.1.455.4728 . doi:10.1021/jm040163o. PMID   15743191.
  12. Ballester PJ, Westwood I, Laurieri N, Sim E, Richards WG (February 2010). "Prospective virtual screening with Ultrafast Shape Recognition: the identification of novel inhibitors of arylamine N-acetyltransferases". Journal of the Royal Society, Interface. 7 (43): 335–42. doi:10.1098/rsif.2009.0170. PMC   2842611 . PMID   19586957.
  13. Kumar A, Zhang KY (2018). "Advances in the Development of Shape Similarity Methods and Their Application in Drug Discovery". Frontiers in Chemistry. 6: 315. Bibcode:2018FrCh....6..315K. doi: 10.3389/fchem.2018.00315 . PMC   6068280 . PMID   30090808.
  14. Li H, Leung KS, Wong MH, Ballester PJ (July 2016). "USR-VS: a web server for large-scale prospective virtual screening using ultrafast shape recognition techniques". Nucleic Acids Research. 44 (W1): W436–41. doi:10.1093/nar/gkw320. PMC   4987897 . PMID   27106057.
  15. Sperandio O, Petitjean M, Tuffery P (July 2009). "wwLigCSRre: a 3D ligand-based server for hit identification and optimization". Nucleic Acids Research. 37 (Web Server issue): W504–9. doi:10.1093/nar/gkp324. PMC   2703967 . PMID   19429687.
  16. 1 2 Kirchmair J, Distinto S, Markt P, Schuster D, Spitzer GM, Liedl KR, Wolber G (2009). "How To Optimize Shape-Based Virtual Screening: Choosing the Right Query and Including Chemical Information". Journal of Chemical Information and Modeling. 49 (3): 678–692. doi:10.1021/ci8004226. PMID   19434901.
  17. Toledo Warshaviak D, Golan G, Borrelli KW, Zhu K, Kalid O (July 2014). "Structure-based virtual screening approach for discovery of covalently bound ligands". Journal of Chemical Information and Modeling. 54 (7): 1941–50. doi:10.1021/ci500175r. PMID   24932913.
  18. Maia EH, Assis LC, de Oliveira TA, da Silva AM, Taranto AG (2020-04-28). "Structure-Based Virtual Screening: From Classical to Artificial Intelligence". Frontiers in Chemistry. 8: 343. Bibcode:2020FrCh....8..343M. doi: 10.3389/fchem.2020.00343 . PMC   7200080 . PMID   32411671.
  19. Kroemer RT (August 2007). "Structure-based drug design: docking and scoring". Current Protein & Peptide Science. 8 (4): 312–28. CiteSeerX   10.1.1.225.959 . doi:10.2174/138920307781369382. PMID   17696866.
  20. Cavasotto CN, Orry AJ (2007). "Ligand docking and structure-based virtual screening in drug discovery". Current Topics in Medicinal Chemistry. 7 (10): 1006–14. doi:10.2174/156802607780906753. PMID   17508934.
  21. Kooistra AJ, Vischer HF, McNaught-Flores D, Leurs R, de Esch IJ, de Graaf C (June 2016). "Function-specific virtual screening for GPCR ligands using a combined scoring method". Scientific Reports. 6: 28288. Bibcode:2016NatSR...628288K. doi:10.1038/srep28288. PMC   4919634 . PMID   27339552.
  22. Irwin JJ, Shoichet BK, Mysinger MM, Huang N, Colizzi F, Wassam P, Cao Y (September 2009). "Automated docking screens: a feasibility study". Journal of Medicinal Chemistry. 52 (18): 5712–20. doi:10.1021/jm9006966. PMC   2745826 . PMID   19719084.
  23. Li H, Leung KS, Ballester PJ, Wong MH (2014-01-24). "istar: a web platform for large-scale protein-ligand docking". PLOS ONE. 9 (1): e85678. Bibcode:2014PLoSO...985678L. doi: 10.1371/journal.pone.0085678 . PMC   3901662 . PMID   24475049.
  24. 1 2 Zhou H, Skolnick J (January 2013). "FINDSITE(comb): a threading/structure-based, proteomic-scale virtual ligand screening approach". Journal of Chemical Information and Modeling. 53 (1): 230–40. doi:10.1021/ci300510n. PMC   3557555 . PMID   23240691.
  25. Roy A, Skolnick J (February 2015). "LIGSIFT: an open-source tool for ligand structural alignment and virtual screening". Bioinformatics. 31 (4): 539–44. doi:10.1093/bioinformatics/btu692. PMC   4325547 . PMID   25336501.
  26. Gaulton A, Bellis LJ, Bento AP, Chambers J, Davies M, Hersey A, Light Y, McGlinchey S, Michalovich D, Al-Lazikani B, Overington JP (January 2012). "ChEMBL: a large-scale bioactivity database for drug discovery". Nucleic Acids Research. 40 (Database issue): D1100–7. doi:10.1093/nar/gkr777. PMC   3245175 . PMID   21948594.
  27. Wishart DS, Knox C, Guo AC, Shrivastava S, Hassanali M, Stothard P, Chang Z, Woolsey J (January 2006). "DrugBank: a comprehensive resource for in silico drug discovery and exploration". Nucleic Acids Research. 34 (Database issue): D668–72. doi:10.1093/nar/gkj067. PMC   1347430 . PMID   16381955.
  28. Réau M, Langenfeld F, Zagury JF, Lagarde N, Montes M (2018). "Decoys Selection in Benchmarking Datasets: Overview and Perspectives". Frontiers in Pharmacology. 9: 11. doi: 10.3389/fphar.2018.00011 . PMC   5787549 . PMID   29416509.
  29. 1 2 Ballester PJ (December 2019). "Selecting machine-learning scoring functions for structure-based virtual screening". Drug Discovery Today: Technologies. 32–33: 81–87. doi: 10.1016/j.ddtec.2020.09.001 . PMID   33386098. S2CID   224968364.
  30. Li H, Sze KH, Lu G, Ballester PJ (2021). "Machine-learning scoring functions for structure-based virtual screening". WIREs Computational Molecular Science. 11 (1): e1478. doi:10.1002/wcms.1478. ISSN   1759-0884. S2CID   219089637.
  31. Wallach I, Heifets A (2018). "Most Ligand-based classification benchmarks reward memorization rather than generalization". Journal of Chemical Information and Modeling. 58 (5): 916–932. arXiv: 1706.06619 . doi:10.1021/acs.jcim.7b00403. PMID   29698607. S2CID   195345933.
  32. Irwin JJ (2008). "Community benchmarks for virtual screening". Journal of Computer-Aided Molecular Design. 22 (3–4): 193–9. Bibcode:2008JCAMD..22..193I. doi:10.1007/s10822-008-9189-4. PMID   18273555. S2CID   26260725.
  33. Good AC, Oprea TI (2008). "Optimization of CAMD techniques 3. Virtual screening enrichment studies: a help or hindrance in tool selection?". Journal of Computer-Aided Molecular Design. 22 (3–4): 169–78. Bibcode:2008JCAMD..22..169G. doi:10.1007/s10822-007-9167-2. PMID   18188508. S2CID   7738182.
  34. Schneider G (April 2010). "Virtual screening: an endless staircase?". Nature Reviews. Drug Discovery. 9 (4): 273–6. doi:10.1038/nrd3139. PMID   20357802. S2CID   205477076.
  35. Ballester PJ (January 2011). "Ultrafast shape recognition: method and applications". Future Medicinal Chemistry . 3 (1): 65–78. doi:10.4155/fmc.10.280. PMID   21428826.
  36. Lavecchia A, Di Giovanni C (2013). "Virtual screening strategies in drug discovery: a critical review". Current Medicinal Chemistry. 20 (23): 2839–60. doi:10.2174/09298673113209990001. PMID   23651302.
  37. Spitzer GM, Heiss M, Mangold M, Markt P, Kirchmair J, Wolber G, Liedl KR (2010). "One concept, three implementations of 3D pharmacophore-based virtual screening: distinct coverage of chemical search space". Journal of Chemical Information and Modeling. 50 (7): 1241–1247. doi:10.1021/ci100136b. PMID   20583761.
  38. Grant JA, Gallard MA, Pickup BT (1996). "A fast method of molecular shape comparison: a simple application of a Gaussian description of molecular shape". Journal of Computational Chemistry. 17 (14): 1653–1666. doi:10.1002/(SICI)1096-987X(19961115)17:14<1653::AID-JCC7>3.0.CO;2-K.
  39. Nicholls A, Grant JA (2005). "Molecular shape and electrostatics in the encoding of relevant chemical information". Journal of Computer-Aided Molecular Design. 19 (9–10): 661–686. doi:10.1007/s10822-005-9019-x. PMID   16328855.
  40. Rush TS, Grant JA, Mosyak L, Nicholls A (2005). "A shape-based 3-D scaffold hopping method and its application to a bacterial protein-protein interaction". Journal of Medicinal Chemistry. 48 (5): 1489–1495. doi:10.1021/jm040163o. PMID   15743191.
  41. Neves BJ, Braga RC, Melo-Filho CC, Moreira-Filho JT, Muratov EN, Andrade CH (2018-11-13). "QSAR-Based Virtual Screening: Advances and Applications in Drug Discovery". Frontiers in Pharmacology. 9: 1275. doi: 10.3389/fphar.2018.01275 . PMC   6262347 . PMID   30524275.
  42. Alsenan S, Al-Turaiki I, Hafez A (December 2020). "A Recurrent Neural Network model to predict blood-brain barrier permeability". Computational Biology and Chemistry. 89: 107377. doi: 10.1016/j.compbiolchem.2020.107377 . PMID   33010784.
  43. Dimitri GM, Lió P (June 2017). "DrugClust: A machine learning approach for drugs side effects prediction". Computational Biology and Chemistry. 68: 204–210. doi:10.1016/j.compbiolchem.2017.03.008. PMID   28391063.
  44. Shoombuatong W, Schaduangrat N, Pratiwi R, Nantasenamat C (June 2019). "THPep: A machine learning-based approach for predicting tumor homing peptides". Computational Biology and Chemistry. 80: 441–451. doi:10.1016/j.compbiolchem.2019.05.008. PMID   31151025.
  45. Gurujee CS, Deshpande VL (February 1978). "An improved method of substructure analysis". Computers & Structures. 8 (1): 147–152. doi:10.1016/0045-7949(78)90171-2.
  46. Pradeepkiran JA, Reddy PH (March 2019). "Structure Based Design and Molecular Docking Studies for Phosphorylated Tau Inhibitors in Alzheimer's Disease". Cells. 8 (3): 260. doi: 10.3390/cells8030260 . PMC   6468864 . PMID   30893872.

Further reading