Docking (molecular)

Last updated
Docking glossary
Receptor or host or lock
The "receiving" molecule, most commonly a protein or other biopolymer.
Ligand or guest or key
The complementary partner molecule which binds to the receptor. Ligands are most often small molecules but could also be another biopolymer.
Docking
Computational simulation of a candidate ligand binding to a receptor.
Binding mode
The orientation of the ligand relative to the receptor as well as the conformation of the ligand and receptor when bound to each other.
Pose
A candidate binding mode.
Scoring
The process of evaluating a particular pose by counting the number of favorable intermolecular interactions such as hydrogen bonds and hydrophobic contacts.
Ranking
The process of classifying which ligands are most likely to interact favorably to a particular receptor based on the predicted free-energy of binding.
Docking assessment (DA)
Procedure to quantify the predictive capability of a docking protocol.
edit

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. [1] 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.

Contents

Schematic illustration of docking a small molecule ligand (green) to a protein target (black) producing a stable complex. Docking representation 2.png
Schematic illustration of docking a small molecule ligand (green) to a protein target (black) producing a stable complex.
Docking of a small molecule (green) into the crystal structure of the beta-2 adrenergic G-protein coupled receptor ( PDB: 3SN6 )

The associations between biologically relevant molecules such as proteins, peptides, nucleic acids, carbohydrates, and lipids play a central role in signal transduction. Furthermore, the relative orientation of the two interacting partners may affect the type of signal produced (e.g., agonism vs antagonism). Therefore, docking is useful for predicting both the strength and type of signal produced.

Molecular docking is one of the most frequently used methods in structure-based drug design, due to its ability to predict the binding-conformation of small molecule ligands to the appropriate target binding site. Characterisation of the binding behaviour plays an important role in rational design of drugs as well as to elucidate fundamental biochemical processes. [2] [3]

Definition of problem

One can think of molecular docking as a problem of “lock-and-key”, in which one wants to find the correct relative orientation of the “key” which will open up the “lock” (where on the surface of the lock is the key hole, which direction to turn the key after it is inserted, etc.). Here, the protein can be thought of as the “lock” and the ligand can be thought of as a “key”. Molecular docking may be defined as an optimization problem, which would describe the “best-fit” orientation of a ligand that binds to a particular protein of interest. However, since both the ligand and the protein are flexible, a “hand-in-glove” analogy is more appropriate than “lock-and-key”. [4] During the course of the docking process, the ligand and the protein adjust their conformation to achieve an overall "best-fit" and this kind of conformational adjustment resulting in the overall binding is referred to as "induced-fit". [5]

Molecular docking research focuses on computationally simulating the molecular recognition process. It aims to achieve an optimized conformation for both the protein and ligand and relative orientation between protein and ligand such that the free energy of the overall system is minimized.

Docking approaches

Two approaches are particularly popular within the molecular docking community.

Both approaches have significant advantages as well as some limitations. These are outlined below.

Shape complementarity

Geometric matching/shape complementarity methods describe the protein and ligand as a set of features that make them dockable. [10] These features may include molecular surface/complementary surface descriptors. In this case, the receptor's molecular surface is described in terms of its solvent-accessible surface area and the ligand's molecular surface is described in terms of its matching surface description. The complementarity between the two surfaces amounts to the shape matching description that may help finding the complementary pose of docking the target and the ligand molecules. Another approach is to describe the hydrophobic features of the protein using turns in the main-chain atoms. Yet another approach is to use a Fourier shape descriptor technique. [11] [12] [13] Whereas the shape complementarity based approaches are typically fast and robust, they cannot usually model the movements or dynamic changes in the ligand/protein conformations accurately, although recent developments allow these methods to investigate ligand flexibility. Shape complementarity methods can quickly scan through several thousand ligands in a matter of seconds and actually figure out whether they can bind at the protein's active site, and are usually scalable to even protein-protein interactions. They are also much more amenable to pharmacophore based approaches, since they use geometric descriptions of the ligands to find optimal binding.

Simulation

Simulating the docking process is much more complicated. In this approach, the protein and the ligand are separated by some physical distance, and the ligand finds its position into the protein's active site after a certain number of “moves” in its conformational space. The moves incorporate rigid body transformations such as translations and rotations, as well as internal changes to the ligand's structure including torsion angle rotations. Each of these moves in the conformation space of the ligand induces a total energetic cost of the system. Hence, the system's total energy is calculated after every move.

The obvious advantage of docking simulation is that ligand flexibility is easily incorporated, whereas shape complementarity techniques must use ingenious methods to incorporate flexibility in ligands. Also, it more accurately models reality, whereas shape complementary techniques are more of an abstraction.

Clearly, simulation is computationally expensive, having to explore a large energy landscape. Grid-based techniques, optimization methods, and increased computer speed have made docking simulation more realistic.

Mechanics of docking

Docking flow-chart overview Overview docking.png
Docking flow-chart overview

To perform a docking screen, the first requirement is a structure of the protein of interest. Usually the structure has been determined using a biophysical technique such as

but can also derive from homology modeling construction. This protein structure and a database of potential ligands serve as inputs to a docking program. The success of a docking program depends on two components: the search algorithm and the scoring function.

Search algorithm

The search space in theory consists of all possible orientations and conformations of the protein paired with the ligand. However, in practice with current computational resources, it is impossible to exhaustively explore the search space — this would involve enumerating all possible distortions of each molecule (molecules are dynamic and exist in an ensemble of conformational states) and all possible rotational and translational orientations of the ligand relative to the protein at a given level of granularity. Most docking programs in use account for the whole conformational space of the ligand (flexible ligand), and several attempt to model a flexible protein receptor. Each "snapshot" of the pair is referred to as a pose. [14]

A variety of conformational search strategies have been applied to the ligand and to the receptor. These include:

Ligand flexibility

Conformations of the ligand may be generated in the absence of the receptor and subsequently docked [15] or conformations may be generated on-the-fly in the presence of the receptor binding cavity, [16] or with full rotational flexibility of every dihedral angle using fragment based docking. [17] Force field energy evaluation are most often used to select energetically reasonable conformations, [18] but knowledge-based methods have also been used. [19]

Peptides are both highly flexible and relatively large-sized molecules, which makes modeling their flexibility a challenging task. A number of methods were developed to allow for efficient modeling of flexibility of peptides during protein-peptide docking. [20]

Receptor flexibility

Computational capacity has increased dramatically over the last decade making possible the use of more sophisticated and computationally intensive methods in computer-assisted drug design. However, dealing with receptor flexibility in docking methodologies is still a thorny issue. [21] The main reason behind this difficulty is the large number of degrees of freedom that have to be considered in this kind of calculations. Neglecting it, however, in some of the cases may lead to poor docking results in terms of binding pose prediction. [22]

Multiple static structures experimentally determined for the same protein in different conformations are often used to emulate receptor flexibility. [23] Alternatively rotamer libraries of amino acid side chains that surround the binding cavity may be searched to generate alternate but energetically reasonable protein conformations. [24] [25]

Scoring function

Docking programs generate a large number of potential ligand poses, of which some can be immediately rejected due to clashes with the protein. The remainder are evaluated using some scoring function, which takes a pose as input and returns a number indicating the likelihood that the pose represents a favorable binding interaction and ranks one ligand relative to another.

Most scoring functions are physics-based molecular mechanics force fields that estimate the energy of the pose within the binding site. The various contributions to binding can be written as an additive equation:

The components consist of solvent effects, conformational changes in the protein and ligand, free energy due to protein-ligand interactions, internal rotations, association energy of ligand and receptor to form a single complex and free energy due to changes in vibrational modes. [26] A low (negative) energy indicates a stable system and thus a likely binding interaction.

Alternative approaches use modified scoring functions to include constraints based on known key protein-ligand interactions, [27] or knowledge-based potentials derived from interactions observed in large databases of protein-ligand structures (e.g. the Protein Data Bank). [28]

There are a large number of structures from X-ray crystallography for complexes between proteins and high affinity ligands, but comparatively fewer for low affinity ligands as the latter complexes tend to be less stable and therefore more difficult to crystallize. Scoring functions trained with this data can dock high affinity ligands correctly, but they will also give plausible docked conformations for ligands that do not bind. This gives a large number of false positive hits, i.e., ligands predicted to bind to the protein that actually don't when placed together in a test tube.

One way to reduce the number of false positives is to recalculate the energy of the top scoring poses using (potentially) more accurate but computationally more intensive techniques such as Generalized Born or Poisson-Boltzmann methods. [9]

Docking assessment

The interdependence between sampling and scoring function affects the docking capability in predicting plausible poses or binding affinities for novel compounds. Thus, an assessment of a docking protocol is generally required (when experimental data is available) to determine its predictive capability. Docking assessment can be performed using different strategies, such as:

Docking accuracy

Docking accuracy [30] [31] represents one measure to quantify the fitness of a docking program by rationalizing the ability to predict the right pose of a ligand with respect to that experimentally observed. [32]

Enrichment factor

Docking screens can also be evaluated by the enrichment of annotated ligands of known binders from among a large database of presumed non-binding, “decoy” molecules. [29] In this way, the success of a docking screen is evaluated by its capacity to enrich the small number of known active compounds in the top ranks of a screen from among a much greater number of decoy molecules in the database. The area under the receiver operating characteristic (ROC) curve is widely used to evaluate its performance.

Prospective

Resulting hits from docking screens are subjected to pharmacological validation (e.g. IC50 , affinity or potency measurements). Only prospective studies constitute conclusive proof of the suitability of a technique for a particular target. [33] In the case of G protein-coupled receptors (GPCRs), which are targets of more than 30% of marketed drugs, molecular docking led to the discovery of more than 500 GPCR ligands. [34]

Benchmarking

The potential of docking programs to reproduce binding modes as determined by X-ray crystallography can be assessed by a range of docking benchmark sets.

For small molecules, several benchmark data sets for docking and virtual screening exist e.g. Astex Diverse Set consisting of high quality protein−ligand X-ray crystal structures [35] or the Directory of Useful Decoys (DUD) for evaluation of virtual screening performance. [29]

An evaluation of docking programs for their potential to reproduce peptide binding modes can be assessed by Lessons for Efficiency Assessment of Docking and Scoring (LEADS-PEP). [36]

Applications

A binding interaction between a small molecule ligand and an enzyme protein may result in activation or inhibition of the enzyme. If the protein is a receptor, ligand binding may result in agonism or antagonism. Docking is most commonly used in the field of drug design — most drugs are small organic molecules, and docking may be applied to:

See also

Related Research Articles

<span class="mw-page-title-main">Allosteric regulation</span> Regulation of enzyme activity

In biochemistry, allosteric regulation is the regulation of an enzyme by binding an effector molecule at a site other than the enzyme's active site.

<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.

<span class="mw-page-title-main">Binding site</span> Molecule-specific coordinate bonding area in biological systems

In biochemistry and molecular biology, a binding site is a region on a macromolecule such as a protein that binds to another molecule with specificity. The binding partner of the macromolecule is often referred to as a ligand. Ligands may include other proteins, enzyme substrates, second messengers, hormones, or allosteric modulators. The binding event is often, but not always, accompanied by a conformational change that alters the protein's function. Binding to protein binding sites is most often reversible, but can also be covalent reversible or irreversible.

<span class="mw-page-title-main">Molecular mechanics</span> Use of classical mechanics to model molecular systems

Molecular mechanics uses classical mechanics to model molecular systems. The Born–Oppenheimer approximation is assumed valid and the potential energy of all systems is calculated as a function of the nuclear coordinates using force fields. Molecular mechanics can be used to study molecule systems ranging in size and complexity from small to large biological systems or material assemblies with many thousands to millions of atoms.

<span class="mw-page-title-main">Drug design</span> Invention of new medications based on 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.

Protein design is the rational design of new protein molecules to design novel activity, behavior, or purpose, and to advance basic understanding of protein function. Proteins can be designed from scratch or by making calculated variants of a known protein structure and its sequence. Rational protein design approaches make protein-sequence predictions that will fold to specific structures. These predicted sequences can then be validated experimentally through methods such as peptide synthesis, site-directed mutagenesis, or artificial gene synthesis.

<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.

Macromolecular docking is the computational modelling of the quaternary structure of complexes formed by two or more interacting biological macromolecules. Protein–protein complexes are the most commonly attempted targets of such modelling, followed by protein–nucleic acid complexes.

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.

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

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.

In molecular modelling, docking is a method which predicts the preferred orientation of one molecule to another when bound together in a stable complex. In the case of protein docking, the search space consists of all possible orientations of the protein with respect to the ligand. Flexible docking in addition considers all possible conformations of the protein paired with all possible conformations of the ligand.

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.

<span class="mw-page-title-main">Cell surface receptor</span> Class of ligand activated receptors localized in surface of plama cell membrane

Cell surface receptors are receptors that are embedded in the plasma membrane of cells. They act in cell signaling by receiving extracellular molecules. They are specialized integral membrane proteins that allow communication between the cell and the extracellular space. The extracellular molecules may be hormones, neurotransmitters, cytokines, growth factors, cell adhesion molecules, or nutrients; they react with the receptor to induce changes in the metabolism and activity of a cell. In the process of signal transduction, ligand binding affects a cascading chemical change through the cell membrane.

The program UCSF DOCK was created in the 1980s by Irwin "Tack" Kuntz's Group, and was the first docking program. DOCK uses geometric algorithms to predict the binding modes of small molecules. Brian K. Shoichet, David A. Case, and Robert C.Rizzo are codevelopers of DOCK.

Computational Resources for Drug Discovery (CRDD) is one of the important silico modules of Open Source for Drug Discovery (OSDD). The CRDD web portal provides computer resources related to drug discovery on a single platform. It caters to researchers of computer-aided drug design, providing computational resources, a discussion forum, and wiki resources related to drug discovery, predicting inhibitors, and predicting the ADME-Tox properties of molecules. One of the major objectives of CRDD is to promote open source software in the field of cheminformatics and pharmacoinformatics.

Lead Finder is a computational chemistry tool designed for modeling protein-ligand interactions. This application is useful for conducting molecular docking studies and quantitatively assessing ligand binding and biological activity. Lead Finder offers free access to individual users, especially those in non-commercial and academic settings.

SAMPL is a set of community-wide blind challenges aimed to advance computational techniques as standard predictive tools in rational drug design. A broad range of biologically relevant systems with different sizes and levels of complexities including proteins, host–guest complexes, and drug-like small molecules have been selected to test the latest modeling methods and force fields in SAMPL. New experimental data, such as binding affinity and hydration free energy, are withheld from participants until the prediction submission deadline, so that the true predictive power of methods can be revealed. The most recent SAMPL5 challenge contains two prediction categories: the binding affinity of host–guest systems, and the distribution coefficients of drug-like molecules between water and cyclohexane. Since 2008, the SAMPL challenge series has attracte interest from scientists engaged in the field of computer-aided drug design (CADD) The current SAMPL organizers include John Chodera, Michael K. Gilson, David Mobley, and Michael Shirts.

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.

LeDock is a molecular docking software designed for ligand-protein interactions, compatible with Linux, macOS, and Windows. It supports the Tripos Mol2 file format and employs a simulated annealing and genetic algorithm approach for docking. Utilizing a knowledge-based scoring scheme, it is categorized as a flexible docking method.

FlexAID is a molecular docking software that can use small molecules and peptides as ligands and proteins and nucleic acids as docking targets. As the name suggests, FlexAID supports full ligand flexibility as well side-chain flexibility of the target. It does using a soft scoring function based on the complementarity of the two surfaces.

References

  1. Lengauer T, Rarey M (Jun 1996). "Computational methods for biomolecular docking". Current Opinion in Structural Biology. 6 (3): 402–6. doi:10.1016/S0959-440X(96)80061-3. PMID   8804827.
  2. Kitchen DB, Decornez H, Furr JR, Bajorath J (Nov 2004). "Docking and scoring in virtual screening for drug discovery: methods and applications". Nature Reviews. Drug Discovery. 3 (11): 935–49. doi:10.1038/nrd1549. PMID   15520816. S2CID   1069493.
  3. Mostashari-Rad T, Arian R, Mehridehnavi A, Fassihi A, Ghasemi F (June 13, 2019). "Study of CXCR4 chemokine receptor inhibitors using QSPR andmolecular docking methodologies". Journal of Theoretical and Computational Chemistry. 178 (4). doi:10.1142/S0219633619500184. S2CID   164985789.
  4. Jorgensen WL (Nov 1991). "Rusting of the lock and key model for protein-ligand binding". Science. 254 (5034): 954–5. Bibcode:1991Sci...254..954J. doi:10.1126/science.1719636. PMID   1719636.
  5. Wei BQ, Weaver LH, Ferrari AM, Matthews BW, Shoichet BK (Apr 2004). "Testing a flexible-receptor docking algorithm in a model binding site". Journal of Molecular Biology. 337 (5): 1161–82. doi:10.1016/j.jmb.2004.02.015. PMID   15046985.
  6. Goldman BB, Wipke WT (2000). "QSD quadratic shape descriptors. 2. Molecular docking using quadratic shape descriptors (QSDock)". Proteins. 38 (1): 79–94. doi: 10.1002/(SICI)1097-0134(20000101)38:1<79::AID-PROT9>3.0.CO;2-U . PMID   10651041.
  7. Meng EC, Shoichet BK, Kuntz ID (1992). "Automated docking with grid-based energy evaluation". Journal of Computational Chemistry. 13 (4): 505–524. doi:10.1002/jcc.540130412. S2CID   97778840.
  8. Morris GM, Goodsell DS, Halliday RS, Huey R, Hart WE, Belew RK, Olson AJ (1998). "Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function". Journal of Computational Chemistry. 19 (14): 1639–1662. CiteSeerX   10.1.1.471.5900 . doi:10.1002/(SICI)1096-987X(19981115)19:14<1639::AID-JCC10>3.0.CO;2-B.
  9. 1 2 Feig M, Onufriev A, Lee MS, Im W, Case DA, Brooks CL (Jan 2004). "Performance comparison of generalized born and Poisson methods in the calculation of electrostatic solvation energies for protein structures". Journal of Computational Chemistry. 25 (2): 265–84. doi:10.1002/jcc.10378. PMID   14648625. S2CID   3191066.
  10. Shoichet BK, Kuntz ID, Bodian DL (2004). "Molecular docking using shape descriptors". Journal of Computational Chemistry. 13 (3): 380–397. doi:10.1002/jcc.540130311. S2CID   42749294.
  11. Cai W, Shao X, Maigret B (Jan 2002). "Protein-ligand recognition using spherical harmonic molecular surfaces: towards a fast and efficient filter for large virtual throughput screening". Journal of Molecular Graphics & Modelling. 20 (4): 313–28. doi:10.1016/S1093-3263(01)00134-6. PMID   11858640.
  12. Morris RJ, Najmanovich RJ, Kahraman A, Thornton JM (May 2005). "Real spherical harmonic expansion coefficients as 3D shape descriptors for protein binding pocket and ligand comparisons". Bioinformatics. 21 (10): 2347–55. doi: 10.1093/bioinformatics/bti337 . PMID   15728116.
  13. Kahraman A, Morris RJ, Laskowski RA, Thornton JM (Apr 2007). "Shape variation in protein binding pockets and their ligands". Journal of Molecular Biology. 368 (1): 283–301. doi:10.1016/j.jmb.2007.01.086. PMID   17337005.
  14. Torres PH, Sodero AC, Jofily P, Silva-Jr FP (September 2019). "Key Topics in Molecular Docking for Drug Design". International Journal of Molecular Sciences. 20 (18): 4574. doi: 10.3390/ijms20184574 . PMC   6769580 . PMID   31540192.
  15. Kearsley SK, Underwood DJ, Sheridan RP, Miller MD (Oct 1994). "Flexibases: a way to enhance the use of molecular docking methods". Journal of Computer-Aided Molecular Design. 8 (5): 565–82. Bibcode:1994JCAMD...8..565K. doi:10.1007/BF00123666. PMID   7876901. S2CID   8834526.
  16. Friesner RA, Banks JL, Murphy RB, Halgren TA, Klicic JJ, Mainz DT, Repasky MP, Knoll EH, Shelley M, Perry JK, Shaw DE, Francis P, Shenkin PS (Mar 2004). "Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy". Journal of Medicinal Chemistry. 47 (7): 1739–49. doi:10.1021/jm0306430. PMID   15027865.
  17. Zsoldos Z, Reid D, Simon A, Sadjad SB, Johnson AP (Jul 2007). "eHiTS: a new fast, exhaustive flexible ligand docking system". Journal of Molecular Graphics & Modelling. 26 (1): 198–212. doi:10.1016/j.jmgm.2006.06.002. PMID   16860582.
  18. Wang Q, Pang YP (September 2007). Romesberg F (ed.). "Preference of small molecules for local minimum conformations when binding to proteins". PLOS ONE. 2 (9): e820. Bibcode:2007PLoSO...2..820W. doi: 10.1371/journal.pone.0000820 . PMC   1959118 . PMID   17786192.
  19. Klebe G, Mietzner T (October 1994). "A fast and efficient method to generate biologically relevant conformations". Journal of Computer-Aided Molecular Design. 8 (5): 583–606. Bibcode:1994JCAMD...8..583K. doi:10.1007/BF00123667. PMID   7876902. S2CID   206768542.
  20. Ciemny M, Kurcinski M, Kamel K, Kolinski A, Alam N, Schueler-Furman O, Kmiecik S (May 2018). "Protein-peptide docking: opportunities and challenges". Drug Discovery Today. 23 (8): 1530–1537. doi: 10.1016/j.drudis.2018.05.006 . PMID   29733895.
  21. Antunes DA, Devaurs D, Kavraki LE (December 2015). "Understanding the challenges of protein flexibility in drug design" (PDF). Expert Opinion on Drug Discovery. 10 (12): 1301–13. doi:10.1517/17460441.2015.1094458. hdl: 1911/88215 . PMID   26414598. S2CID   6589810.
  22. Cerqueira NM, Bras NF, Fernandes PA, Ramos MJ (January 2009). "MADAMM: a multistaged docking with an automated molecular modeling protocol". Proteins. 74 (1): 192–206. doi:10.1002/prot.22146. PMID   18618708. S2CID   36656063.
  23. Totrov M, Abagyan R (Apr 2008). "Flexible ligand docking to multiple receptor conformations: a practical alternative". Current Opinion in Structural Biology. 18 (2): 178–84. doi:10.1016/j.sbi.2008.01.004. PMC   2396190 . PMID   18302984.
  24. Hartmann C, Antes I, Lengauer T (Feb 2009). "Docking and scoring with alternative side-chain conformations". Proteins. 74 (3): 712–26. doi:10.1002/prot.22189. PMID   18704939. S2CID   36088213.
  25. Taylor RD, Jewsbury PJ, Essex JW (Oct 2003). "FDS: flexible ligand and receptor docking with a continuum solvent model and soft-core energy function". Journal of Computational Chemistry. 24 (13): 1637–56. CiteSeerX   10.1.1.147.1131 . doi:10.1002/jcc.10295. PMID   12926007. S2CID   15814316.
  26. Murcko MA (Dec 1995). "Computational Methods to Predict Binding Free Energy in Ligand-Receptor Complexes". Journal of Medicinal Chemistry. 38 (26): 4953–67. doi:10.1021/jm00026a001. PMID   8544170.
  27. Arcon JP, Turjanski AG, Martí MA, Forli S (2021). Ballante F (ed.). Biased Docking for Protein-Ligand Pose Prediction. Methods in Molecular Biology. Vol. 2266. New York, NY: Springer US. pp. 39–72. doi:10.1007/978-1-0716-1209-5_3. ISBN   978-1-0716-1209-5. PMID   33759120. S2CID   232340746.
  28. Gohlke H, Hendlich M, Klebe G (January 2000). "Knowledge-based scoring function to predict protein-ligand interactions". Journal of Molecular Biology. 295 (2): 337–356. doi:10.1006/jmbi.1999.3371. PMID   10623530.
  29. 1 2 3 Huang N, Shoichet BK, Irwin JJ (Nov 2006). "Benchmarking sets for molecular docking". Journal of Medicinal Chemistry. 49 (23): 6789–801. doi:10.1021/jm0608356. PMC   3383317 . PMID   17154509.
  30. Ballante F, Marshall GR (January 2016). "An Automated Strategy for Binding-Pose Selection and Docking Assessment in Structure-Based Drug Design". Journal of Chemical Information and Modeling. 56 (1): 54–72. doi:10.1021/acs.jcim.5b00603. PMID   26682916.
  31. Bursulaya BD, Totrov M, Abagyan R, Brooks CL (November 2003). "Comparative study of several algorithms for flexible ligand docking". Journal of Computer-Aided Molecular Design. 17 (11): 755–763. Bibcode:2003JCAMD..17..755B. doi:10.1023/B:JCAM.0000017496.76572.6f. PMID   15072435. S2CID   12569345.
  32. Ballante F (2018). "Protein-Ligand Docking in Drug Design: Performance Assessment and Binding-Pose Selection". Rational Drug Design. Methods in Molecular Biology. Vol. 1824. pp. 67–88. doi:10.1007/978-1-4939-8630-9_5. ISBN   978-1-4939-8629-3. PMID   30039402.
  33. Irwin JJ (2008-02-14). "Community benchmarks for virtual screening". Journal of Computer-Aided Molecular Design. 22 (3–4): 193–199. Bibcode:2008JCAMD..22..193I. doi:10.1007/s10822-008-9189-4. PMID   18273555. S2CID   26260725.
  34. Ballante F, Kooistra AJ, Kampen S, de Graaf C, Carlsson J (October 2021). "Structure-Based Virtual Screening for Ligands of G Protein-Coupled Receptors: What Can Molecular Docking Do for You?". Pharmacological Reviews. 73 (4): 527–565. doi: 10.1124/pharmrev.120.000246 . PMID   34907092. S2CID   245163594.
  35. Hartshorn MJ, Verdonk ML, Chessari G, Brewerton SC, Mooij WT, Mortenson PN, Murray CW (Feb 2007). "Diverse, high-quality test set for the validation of protein-ligand docking performance". Journal of Medicinal Chemistry. 50 (4): 726–41. doi:10.1021/jm061277y. PMID   17300160.
  36. Hauser AS, Windshügel B (Dec 2015). "A Benchmark Data Set for Assessment of Peptide Docking Performance". Journal of Chemical Information and Modeling. 56 (1): 188–200. doi:10.1021/acs.jcim.5b00234. PMID   26651532.
  37. Suresh PS, Kumar A, Kumar R, Singh VP (Jan 2008). "An in silico [correction of insilico] approach to bioremediation: laccase as a case study". Journal of Molecular Graphics & Modelling. 26 (5): 845–9. doi:10.1016/j.jmgm.2007.05.005. PMID   17606396.
  38. Basharat Z, Yasmin A, Bibi M (2020). "Implications of Molecular Docking Assay for Bioremediation". Data Analytics in Medicine: Concepts, Methodologies, Tools, and Applications. Advances in Environmental Engineering and Green Technologies. IGI Global. pp. 1556–1577. doi:10.4018/978-1-5225-2325-3.ch002. ISBN   978-1799812043. S2CID   63136337.