Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Docking and scoring in virtual screening for drug discovery: methods and applications

Key Points

  • Computational methodologies have become a crucial component of many drug discovery programmes, from hit identification to lead optimization and beyond.

  • One key such methodology — docking of small molecules to protein binding sites — was pioneered during the early 1980s, and remains a highly active area of research.

  • The docking process involves the prediction of ligand conformation and orientation (or posing) within a targeted binding site. In general, there are two aims of docking studies: accurate structural modelling and correct prediction of activity.

  • Docking is generally devised as a multi-step process in which each step introduces one or more additional degrees of complexity. The process begins with the application of docking algorithms that pose small molecules in the active site. These algorithms are complemented by scoring functions that are designed to predict the biological activity through the evaluation of interactions between compounds and potential targets.

  • This article reviews basic concepts and specific features of small-molecule–protein docking methods and several selected applications, with particular emphasis on hit identification and lead optimization.

  • We attempt to distinguish between the problems of docking compounds into target sites and of scoring docked conformations, because the available data indicate that numerous robust and accurate docking algorithms are available, whereas imperfections of scoring functions continue to be a major limiting factor.

Abstract

Computational approaches that 'dock' small molecules into the structures of macromolecular targets and 'score' their potential complementarity to binding sites are widely used in hit identification and lead optimization. Indeed, there are now a number of drugs whose development was heavily influenced by or based on structure-based design and screening strategies, such as HIV protease inhibitors. Nevertheless, there remain significant challenges in the application of these approaches, in particular in relation to current scoring schemes. Here, we review key concepts and specific features of small-molecule–protein docking methods, highlight selected applications and discuss recent advances that aim to address the acknowledged limitations of established approaches.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Grid representations.
Figure 2: Electrostatic potential of a bound inhibitor.
Figure 3: Modelling molecular recognition.
Figure 4: Complexity of protein–ligand interactions.
Figure 5: Design of specific inhibitors.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Berman, H. M. et al. The protein data bank and the challenge of structural genomics. Nature Struct. Biol. 7, 957–959 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Westbrook, J., Feng, Z., Chen, L., Yang, H. & Berman, H. M. The protein data bank and structural genomics. Nucleic Acid Res. 31, 489–491 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Blundell, T. L., Jhoti, H. & Abell, C. High-throughput crystallography for lead discovery in drug design. Nature Rev. Drug Discov. 1, 45–54 (2002).

    Article  CAS  Google Scholar 

  4. Bajorath, J. Integration of virtual and high-throughput screening. Nature Rev. Drug Discov. 1, 882–894 (2002).

    Article  CAS  Google Scholar 

  5. Walters, W. P., Stahl, M. T. & Murcko, M. A. Virtual screening — an overview. Drug Discov. Today 3, 160–178 (1998).

    Article  CAS  Google Scholar 

  6. Langer, T. & Hoffmann, R. D. Virtual screening: an effective tool for lead structure discovery. Curr. Pharm. Design 7, 509–527 (2001).

    Article  CAS  Google Scholar 

  7. Gohlke, H. & Klebe, G. Approaches to the description and prediction of the binding affinity of small-molecule ligands to macromolecular receptors. Angew. Chem. Int. Ed. 41, 2644–2676 (2002). A very extensive and informative review with emphasis on quantitative analysis of protein–ligand interactions.

    Article  CAS  Google Scholar 

  8. Kuntz, I. D., Blaney, J. M., Oatley, S. J., Langridge, R. & Ferrin, T. E. A geometric approach to macromolecule–ligand interactions. J. Mol. Biol. 161, 269–288 (1982).

    Article  CAS  PubMed  Google Scholar 

  9. Venhorst, J. et al. Homology modeling of rat and human cytochrome P450 2D (CYP2D) isoforms and computational rationalization of experimental ligand-binding specificities. J. Med. Chem. 46, 74–86 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Williams, P. A. et al. Crystal structure of human cytochrome P450 2C9 with bound warfarin. Nature 424, 464–468 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Brooijmans, N. & Kuntz, I. D. Molecular recognition and docking algorithms. Annu. Rev. Biophys. Biolmol. Struct. 32, 335–373 (2003). Excellent review of research in the docking arena that contains an instructive section on the conceptually different processes involved in ligand–protein docking.

    Article  CAS  Google Scholar 

  12. Halperin, I., Ma, B., Wolfson, H. & Nussinov, R. Principles of docking: an overview of search algorithms and a guide to scoring functions. Proteins 47, 409–443 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Burnett, R. M. & Taylor, J. S. DARWIN: a program for docking flexible molecules. Proteins 41, 173–191 (2000).

    Article  PubMed  Google Scholar 

  14. Norel, R., Lin, S. L., Wolfson, H. & Nussinov, R. Shape complementarity at protein–protein interfaces. Biopolymers 34, 933–940 (1994).

    Article  CAS  PubMed  Google Scholar 

  15. Norel, R., Petrey, D., Wolfson, H. & Nussinov, R. Examination of shape complementarity in docking of unbound proteins. Proteins 35, 403–419 (1999).

    Google Scholar 

  16. Connolly, M. L. Analytical molecular surface calculation. J. Appl. Cryst. 16, 548–558 (1983).

    Article  CAS  Google Scholar 

  17. Connolly, M. Solvent-accessible surface of proteins and nucleic acids. Science 221, 709–713 (1983). References 16 and 17 outline the theoretical foundation of molecular surface calculations that have also become a crucial component of many shape-based docking algorithms.

    Article  CAS  PubMed  Google Scholar 

  18. Norel, R., Wolfson, H. & Nussinov, R. Small molecular recognition: solid angles surface representation and shape complementarity. Comb. Chem. High Throughput Screen 2, 177–191 (1999).

    Article  Google Scholar 

  19. Goodford, P. J. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules. J. Med. Chem. 28, 849–857 (1985). This seminal paper introduced the idea of potential energy grids and its application to understanding protein–ligand interactions. This concept has been applied and extended in many contemporary docking programs.

    Article  CAS  PubMed  Google Scholar 

  20. Leach, A. R. Molecular Modelling: Principles and Applications (Addison Wesley Longman Limited, Harlow, 1996).

    Google Scholar 

  21. DesJarlais, R. L. Docking flexible ligands to macromolecular receptors by shape. J. Med Chem. 29, 2149–2153 (1986).

    Article  CAS  PubMed  Google Scholar 

  22. Klebe, G. & Rarey, M. A fast flexible docking method using an incremental construction algorithm. J. Mol. Biol. 261, 470–489 (1996).

    Article  PubMed  Google Scholar 

  23. Kuntz, I. D. & Leach, A. R. Conformational analysis of flexible ligands in macromolecular receptor sites. J. Comput. Chem. 13, 730–748 (1992).

    Article  Google Scholar 

  24. Ewing, T. J. A., Makino, S., Skillman, A. G. & Kuntz, I. D. DOCK 4.0: search strategies for automated molecular docking of flexible molecule databases. J. Comput. Aided Mol. Des. 15, 411–428 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Conformation search [online], <http://dock.compbio.ucsf.edu/dock4/html/Manual.f.html> (1998).

  26. Kramer, B., Rarey, M., Lengauer, T. Evaluation of the FlexX incremental construction algorithm for protein–ligand docking. Proteins 37, 228–241 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Linnainmaa, S., Harwood, D. & Davis, L. S. Pose determination of a three-dimensional object using triangle pairs. IEEE Trans. Comput. Anal. Machine Intelligence 10, 634–646 (1988). An in-depth study of a computer vision technique (pose clustering) that is utilized, for example, in FlexX.

    Article  Google Scholar 

  28. Welch, W., Ruppert, J. & Jain, A. N. Hammerhead: fast, fully automated docking of flexible ligands to protein binding sites. Chem. Biol. 3, 449–462 (1996).

    Article  CAS  PubMed  Google Scholar 

  29. Kearsly, S. K., Underwood, D. J., Sheridan, R. P. & Miller, M. D. Flexibase: a way to enhance the use of molecular docking methods. J. Comput. Aided Mol. Des. 8, 565–582 (1994).

    Article  Google Scholar 

  30. Olson, A. J. & Goodsell, D. S. Automated docking in crystallography: analysis of the substrates of aconitase. Proteins 17, 1–10 (1993).

    Article  PubMed  Google Scholar 

  31. Read, R. J. & Hart, T. N. A multiple-start Monte Carlo docking method. Proteins 13, 206–222 (1992).

    Article  PubMed  Google Scholar 

  32. Dixon, J. S. & Oshiro, C. M. Flexible ligand docking using a genetic algorithm. J. Comput. Aided Mol. Des. 9, 113–130 (1995).

    Article  PubMed  Google Scholar 

  33. Morris, G. M., Goodsell, D. S., Halliday, R. S., Huey, R. & Hart, W. E. Automated docking using a Lamarckian genetic algorithm and an empirical free energy function. J. Comput. Chem. 19, 1639–1662 (1998).

    Article  CAS  Google Scholar 

  34. Jones, G., Willet, P., Glen, R. C., Leach, A. R. & Taylor, R. Development and validation of a genetic algorithm for flexible docking. J. Mol. Biol. 267, 727–748 (1997).

    Article  CAS  PubMed  Google Scholar 

  35. Westhead, D. R., Clark, D. E. & Murray, C. W. A comparison of heuristic search algorithms for molecular docking. J. Comput. Aided Mol. Des. 11, 209–228 (1997).

    Article  CAS  PubMed  Google Scholar 

  36. Baxter, C. A., Murray, C. W., Clark, D. E., Westhead, D. R. & Eldridge, M. D. Flexible docking using tabu search and an empirical estimate of binding affinity. Proteins 33, 367–382 (1998).

    Article  CAS  PubMed  Google Scholar 

  37. Di Nola, A., Berendsen, H. J. C. & Roccatano, D. Molecular dynamics simulation of the docking of substrates to proteins. Proteins 19, 174–182 (1994).

    Article  CAS  PubMed  Google Scholar 

  38. Trosset, J. -Y. & Scheraga, H. A. Reaching the global minimum in docking simulations: a Monte Carlo energy minimization approach using Bezier Splines. Proc. Natl Acad. Sci. USA 95, 8011–8015 (1995).

    Article  Google Scholar 

  39. Carlson, H. A. & McGammon, J. A. Accommodating protein flexibility in computational drug design. Mol. Pharmacol. 57, 213–218 (2000). Informative review of approaches to treat protein flexibility in the computational study of protein–ligand interactions.

    Article  CAS  PubMed  Google Scholar 

  40. Leach, A. R. Ligand docking to proteins with discrete side-chain flexibility. J. Mol. Biol. 235, 345–356 (1994).

    Article  CAS  PubMed  Google Scholar 

  41. Desmet, J., Maeyer, M. D., Hazes, B. & Lasters, I. The dead end elimination theorem and its use in protein side-chain positioning. Nature 356, 539–542 (1992).

    Article  CAS  PubMed  Google Scholar 

  42. Knegtel, R. M. A., Kuntz, I. D. & Oshiro, C. M. Molecular docking to ensembles of protein structures. J. Mol. Biol. 266, 242–440 (1997).

    Article  Google Scholar 

  43. Kollman, P. A. Free energy calculations: applications to chemical and biochemical phenomena. Chem. Rev. 93, 2395–2417 (1993). Review of the theory of free-energy calculations and their areas of application, including ligand binding.

    Article  CAS  Google Scholar 

  44. Simonson, T., Archontis, G. & Karplus, M. Free energy simulations come of age: protein–ligand recognition. Acc. Chem. Res. 35, 430–437 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Morris, G. M. et al. Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. J. Comput. Chem. 19, 1639–1662 (1998).

    Article  CAS  Google Scholar 

  46. Weiner, S. J., Kollman, P. A., Nguyen, D. T. & Case, D. A. An all-atom force field for simulations of proteins and nucleic acids. J. Comput. Chem. 7, 252 (1986).

    Article  Google Scholar 

  47. Verdonk, M. L., Cole, J. C., Hartshorn, M. J., Murray, C. W. & Taylor, R. D. Improved protein–ligand docking using GOLD. Proteins 52, 609–623 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Böhm, H. -J. LUDI: rule-based automatic design of new substituents for enzyme inhibitor leads. J. Comput. Aided Mol. Des. 6, 593–606 (1992).

    Article  PubMed  Google Scholar 

  49. Eldridge, M. D., Murray, C. W., Auton, T. R., Paolini, G. V. & Mee, R. P. Empirical scoring functions: I. The development of a fast empirical scoring function to estimate the binding affinity of ligands in receptor complexes. J. Comput. Aided Mol. Des. 11, 425–445 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Rarey, M., Kramer, B., Lengauer, T. & Klebe, G. A fast flexible docking method using an incremental construction algorithm. J. Mol. Biol. 261, 470–489 (1996).

    Article  CAS  PubMed  Google Scholar 

  51. Rognan, D., Lauemoller, S. L., Holm, A., Buus, S. & Tschinke, V. Predicting binding affinities of protein ligands from three-dimensional models: application to peptide binding to class I major histocompatibility proteins. J. Med. Chem. 42, 4650–4658 (1999).

    Article  CAS  PubMed  Google Scholar 

  52. Sitkoff, D. F., Sharp, K. A. & Honig, B. Accurate calculation of hydration free energies using macroscopic continuum models. J. Phys. Chem. 98, 1978–1983 (1998).

    Article  Google Scholar 

  53. Huo, S., Wang, J., Cieplak, P., Kollman, P. A. & Kuntz, I. D. Molecular dynamics and free energy analyses of cathepsin D–inhibitor interactions: insight into structure-based ligand design. J. Med. Chem. 45, 1412–1419 (2002).

    Article  CAS  PubMed  Google Scholar 

  54. Muegge, I. A knowledge-based scoring function for protein–ligand interactions: probing the reference state. Perspect. Drug Discov. Des. 20, 99–114 (2000).

    Article  CAS  Google Scholar 

  55. Muegge, I. Effect of ligand volume correction on PMF scoring. J. Comput. Chem. 22, 418–425 (2001).

    Article  CAS  Google Scholar 

  56. Muegge, I. & Martin, Y. C. A general and fast scoring function for protein-ligand interactions: a simplified potential approach. J. Med. Chem. 42, 791–804 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Gohlke, H., Hendlich, M. & Klebe, G. Knowledge-based scoring function to predict protein-ligand interactions. J. Mol. Biol. 295, 337–356 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. DeWitte, R. S. & Shakhnovich, E. I. SMoG: de novo design method based on simple, fast, and accurate free energy estimates. 1. Methodology and supporting evidence. J. Am. Chem. Soc. 118, 11733–11744 (1996).

    Article  CAS  Google Scholar 

  59. Charifson, P. S., Corkery, J. J., Murcko, M. A. & Walters, W. P. Consensus scoring: a method for obtaining improved hit rates from docking databases of three-dimensional structures into proteins. J. Med. Chem. 42, 5100–5109 (1999). This study introduced the concept of consensus scoring as an approach to balance imperfections of single scoring functions and improve prediction accuracy.

    Article  CAS  PubMed  Google Scholar 

  60. Wang, R., Lai, L. & Wang, S. Further development and validation of empirical scoring functions for structure-based binding affinity prediction. J. Comput. Aided Mol. Des. 16, 11–26 (2002).

    Article  CAS  PubMed  Google Scholar 

  61. Perez, C. & Ortiz, A. R. Evaluation of docking functions for protein–ligand docking. J. Med. Chem. 44, 3768–3785 (2001).

    Article  CAS  PubMed  Google Scholar 

  62. Good, A. C. et al. Analysis and optimization of structure-based virtual screening protocols 2. Examination of docked ligand orientations sampling methodology: mapping a pharmacophore for success. J. Mol. Graph. Model. 22, 31–40 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Baxter, C. A. et al. New approach to molecular docking and its application to virtual screening of chemical databases. J. Chem. Inf. Comput. Sci. 40, 254–262 (2000).

    Article  CAS  PubMed  Google Scholar 

  64. GOLD Version 1.2. [online], <http://www.ccdc.cam.ac.uk/products/life_sciences/gold/> (2003).

  65. Sotriffer, C. A., Gohlke, H. & Klebe, G. Docking into knowledge-based potential fields: a comparative evaluation of DrugScore. J. Med. Chem. 45, 1967–1970 (2002).

    Article  CAS  PubMed  Google Scholar 

  66. Wang, R., Lu, Y. & Wang, S. Comparative evaluation of 11 scoring functions for molecular docking. J. Med. Chem. 46, 2287–2303 (2003).

    Article  CAS  PubMed  Google Scholar 

  67. McGann, M. R., Almond, H. R., Nicholls, A., Grant, J. A. & Brown, F. K. Gaussian docking functions. Biopolymers 68, 76–90 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Schulz-Gasch, T. & Stahl, M. Binding site characteristics in structure-based virtual screening: evaluation of current docking tools. J. Mol. Model 9, 47–57 (2003).

    Article  CAS  PubMed  Google Scholar 

  69. Erickson, J. A., Jalaie, M., Robertson, D. H., Lewis, R. A. & Vieth, M. Lessons in molecular recognition: the effects of ligand and protein flexibility on molecular docking accuracy. J. Med. Chem. 47, 45–55 (2004).

    Article  CAS  PubMed  Google Scholar 

  70. Kontoyianni, M., McClellan, L. M. & Sokol, G. S. Evaluation of docking performance: comparative data on docking algorithms. J. Med. Chem. 47, 558–565 (2004).

    Article  CAS  PubMed  Google Scholar 

  71. Smith, R., Hubbard, R. E., Gschwend, D. A., Leach, A. R. & Good, A. C. Analysis and optimization of structure-based virtual screening protocols 3. New Methods and old problems in scoring function design. J. Mol. Graph. Model. 22, 41–53 (2003).

    Article  CAS  PubMed  Google Scholar 

  72. Still, W. C., Tempczyk, A., Hawley, R. C. & Hendrickson, T. Semianalytical treatment of solvation for molecular mechanics and dynamics. J. Am. Chem. Soc. 112, 6127–6129 (1990).

    Article  CAS  Google Scholar 

  73. Ghosh, A., Rapp, C. S. & Friesner, R. A. A generalized Born model based on a surface integral formulation. J. Phys. Chem. B 102, 10983–10990 (1998).

    Article  CAS  Google Scholar 

  74. Nissink, J. W. M. et al. A new test set for validating predictions of protein–ligand interaction. Proteins 49, 457–471 (2002).

    Article  CAS  PubMed  Google Scholar 

  75. Grzybowski, B. A., Ishchenko, A. V., Shimada, J. & Shakhnovich, E. I. From knowledge-based potentials to combinatorial lead design in silico. Acc. Chem. Res. 35, 261–269 (2002).

    Article  CAS  PubMed  Google Scholar 

  76. Diller, D. J. & Li, Y. Kinases, homology models, and high throughput docking. J. Med. Chem. 46, 4638–4647 (2003).

    Article  CAS  PubMed  Google Scholar 

  77. DesJarlais, R. L. et al. Using shape complementarity as an initial screen in designing ligands for a receptor binding site of known three-dimensional structure. J. Med. Chem. 31, 722–729 (1988).

    Article  CAS  PubMed  Google Scholar 

  78. Dean, P. M. & Poornima, C. S. Hydration in drug design. 1. Multiple hydrogen-bonding features of water molecules in mediating protein–ligand interactions. J. Comput. Aided Mol. Des. 9, 500–512 (1995).

    Article  PubMed  Google Scholar 

  79. McGovern, S. L., Caselli, E., Grigorieff, N. & Shoichet, B. K. A common mechanism underlying promiscous inhibitors from virtual and high-throughput screening. J. Med. Chem. 45, 1712–1722 (2002).

    Article  CAS  PubMed  Google Scholar 

  80. Roche, O. et al. Development of a virtual screening method for identification of 'frequent hitters' in compound libraries. J. Med. Chem. 45, 137–142 (2002).

    Article  CAS  PubMed  Google Scholar 

  81. Doman, T. N. et al. Molecular docking and high-throughput screening for novel inhibitors of protein tyrosine phosphatase-1B. J. Med. Chem. 45, 2213–2221 (2002). An impressive example of the performance of structure-based virtual screening.

    Article  CAS  PubMed  Google Scholar 

  82. McGovern, S. L. & Shoichet, B. K. Information decay in molecular docking screens against holo, apo and modeled conformations of enzymes. J. Med Chem. 46, 2895–2907 (2003). Informative analysis of the influence of chosen protein-structure templates on the quality of docking and scoring.

    Article  CAS  PubMed  Google Scholar 

  83. Lipinski, C. A. & Christopher, A. L. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 23, 3–25 (1997).

    Article  CAS  Google Scholar 

  84. Nilakantan, R., Bauman, N. & Venkataraghavan, R. New method for rapid characterization of molecular shapes: applications in drug design. J. Chem. Inf. Comput. Sci. 33, 79–85 (1993).

    Article  CAS  PubMed  Google Scholar 

  85. Good, A. C., Ewing, T. J. A., Gschwend, D. A. & Kuntz, I. D. New molecular shape descriptors: application in database screening. J. Comput. Aided Mol. Des. 9, 1–12 (1995).

    Article  CAS  PubMed  Google Scholar 

  86. Zauhar, R. J., Moyna, G., Tian, L., Li, Z. & Welsh, W. J. Shape signatures: a new approach to computer-aided ligand-and receptor-based drug design. J. Med. Chem. 46, 5674–5690 (2003).

    Article  CAS  PubMed  Google Scholar 

  87. Rastelli, G. et al. Docking and database screening reveal new classes of Plasmodium falciparum dihydrofolate reductase inhibitors. J. Med. Chem. 46, 2834–2845 (2003).

    Article  CAS  PubMed  Google Scholar 

  88. Choong, I. C. et al. Identification of potent and selective small-molecule inhibitors of caspase-3 through the use of extended tethering and structure-based drug design. J. Med. Chem. 45, 5005–5022 (2002).

    Article  CAS  PubMed  Google Scholar 

  89. Kick, E. K. et al. Structure-based design and combinatorial chemistry yield low nanomolar inhibitors of cathepsin D. Chem. Biol. 4, 297–307 (1997). An instructive study highlighting the potential of interfacing docking analysis and targeted library design.

    Article  CAS  PubMed  Google Scholar 

  90. Karplus, M. & Miranker, A. Functionality maps of binding sites: a multiple copy simultaneous search method. Proteins 11, 29–34 (1991).

    Article  PubMed  Google Scholar 

  91. Caflisch, A. Computational combinatorial ligand design: application to human α-thrombin. J. Comput. Aided Mol. Des. 10, 372–396 (1996).

    Article  CAS  PubMed  Google Scholar 

  92. Bohm, H. J. The development of a simple empirical scoring function to estimate the binding constant for a protein–ligand complex of known three-dimensional structure. J. Comput. Aided Mol. Des. 8, 243–256 (1994). Pioneering development of an empirical scoring function using multiple linear regression to calculate coefficients for the most important terms contributing to ligand binding.

    Article  CAS  PubMed  Google Scholar 

  93. Böhm, H. J. Prediction of binding constants of protein ligands: a fast method for the polarization of hits obtained from de novo design on 3D database search programs. J. Comput. Aided Mol. Des. 12, 309–323 (1998).

    Article  PubMed  Google Scholar 

  94. Murcko, M. A. & Rotstein, S. H. GroupBuild: a fragment-based method for de novo drug design. J. Med. Chem. 36, 1700–1710 (1993).

    Article  PubMed  Google Scholar 

  95. Murcko, M. A. & Rotstein, S. H. GenStar: a method for de novo drug design. J. Comput. Aided Mol. Des. 7, 23–43 (1993).

    Article  PubMed  Google Scholar 

  96. Howe, W. J. & Moon, J. B. 3D database searching and de novo construction methods in molecular design. Comput. Meth. 3, 697–711 (1990).

    Google Scholar 

  97. Bohacek, R. S. & McMartin, C. Multiple highly diverse structures complementary to enzyme binding sites: results of extensive application of a de novo design method incorporating combinatorial growth. J. Am. Chem. Soc. 116, 5560–5571 (1994).

    Article  CAS  Google Scholar 

  98. Vinkers, H. M. et al. SYNOPSIS: SYNthesize and OPtimize system in silico. J. Med. Chem. 46, 2765–2773 (2003).

    Article  CAS  PubMed  Google Scholar 

  99. Guimaraes, C. R. W. & de Alencastro, R. B. Thrombin inhibition by novel benzamidine derivatives: a free-energy perturbation study. J. Med. Chem. 45, 4995–5004 (2003).

    Article  CAS  Google Scholar 

  100. Pearlman, D. A. & Charifson, P. S. Improved scoring of ligand–protein interactions using OWFEG free energy grids. J. Med. Chem. 44, 502–511 (2001).

    Article  CAS  PubMed  Google Scholar 

  101. Aqvist, J., Medina, C. & Samuelsson, J. E. A new method for predicting binding affinity in computer-aided drug design. Protein Eng. 7, 385–391 (1994). This article presents an early formulation and use of linear response and linear interaction approximations in estimating binding affinity of protein ligands.

    Article  CAS  PubMed  Google Scholar 

  102. Tounge, B. A. & Reynolds, C. H. Calculation of the binding affinity of β-secretase inhibitors using the linear interaction energy method. J. Med. Chem. 46, 2074–2082 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Rizzo, R. C., Wang, D. -P., Tirado-Rives, J. & Jorgensen, W. L. Validation of a model for the complex of HIV-1 reverse transcriptase with sustiva through computation of resistance profiles. J. Am. Chem. Soc. 122, 12898–12900 (2003).

    Article  CAS  Google Scholar 

  104. Rizzo, R. C., Tirado-Rives, J. & Jorgensen, W. L. Estimation of binding affinities for HEPT and nevirapine analogues with HIV-1 reverse transcriptase via Monte Carlo simulations. J. Med. Chem. 44, 145–154 (2003).

    Article  CAS  Google Scholar 

  105. Kroeger-Smith, M. B. et al. Molecular modeling calculations of HIV-1 reverse transcriptase nonnucleoside inhibitors: correlation of binding energy with biological activity for novel 2-aryl-substituted benzimidazole analogues. J. Med. Chem. 46, 1940–1947 (2003).

    Article  CAS  PubMed  Google Scholar 

  106. Udier-Blagovic, M., Tirado-Rives, J. & Jorgensen, W. L. Validation of a model for the complex of HIV-1 reverse transcriptase with nonnucleoside inhibitor TMC125. J. Am. Chem. Soc. 125, 6016–6017 (2003).

    Article  CAS  PubMed  Google Scholar 

  107. Rizzo, R. C. et al. Prediction of activity for nonnucleoside inhibitors with HIV-1 reverse transcriptase based on Monte Carlo simulations. J. Med. Chem. 45, 2970–2987 (2002).

    Article  CAS  PubMed  Google Scholar 

  108. Ostrovsky, D., Udier-Blagovic, M. & Jorgensen, W. L. Analyses of activity for Factor Xa inhibitors based on Monte Carlo simulations. J. Med. Chem. 46, 5691–5699 (2003).

    Article  CAS  PubMed  Google Scholar 

  109. van Lipzig, M. M. et al. Prediction of ligand binding affinity and orientation of xenoestrogens to the estrogen receptor by molecular dynamics simulations and the linear interaction energy method. J. Med. Chem. 47, 1018–1030 (2004). This work provides a good example of linear interaction methods applied to binding energies ranging over many orders of magnitude.

    Article  CAS  PubMed  Google Scholar 

  110. Kollman, P. A. et al. Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models. Acc. Chem. Res. 33, 889–897 (2000).

    Article  CAS  PubMed  Google Scholar 

  111. Masukawa, K. M., Kollman, P. A. & Kuntz, I. D. Investigation of neuraminidase-substrate recognition using molecular dynamics and free energy calculations. J. Med. Chem. 46, 5628–5637 (2003).

    Article  CAS  PubMed  Google Scholar 

  112. Sheridan, R., Holloway, M. K., McGaughey, G. B., Mosley, R. T. & Singh, S. B. A simple method for visualizing the differences between related receptor sites. J. Mol. Graph. Model. 21, 71–79 (2002).

    Article  CAS  PubMed  Google Scholar 

  113. Deng, Z., Chuaqui, C. & Singh, J. Structural interaction fingerprint (SIFt): a novel method for analyzing three-dimensional protein–ligand binding interactions. J. Med. Chem. 47, 337–344 (2004).

    Article  CAS  PubMed  Google Scholar 

  114. Horvath, D. A virtual screening approach applied to the search for trypanothione reductase inhibitors. J. Med. Chem. 40, 2412–2423 (1997). The study details many possible scoring terms for protein–ligand complexes and is a good example of the value of refitting parameters for a particular protein class and series of ligands.

    Article  CAS  PubMed  Google Scholar 

  115. Matter, H. et al. Design and quantitative structure–activity relationship of 3-amidinobenzyl-1H-indole-2-carboxamides as potent, nonchiral and selective inhibitors of blood coagulation factor Xa. J. Med. Chem. 45, 2749–2769 (2002).

    Article  CAS  PubMed  Google Scholar 

  116. Murcia, M. & Ortiz, A. R. Virtual screening with flexible docking and COMBINE-based models. Application to a series of factor Xa inhibitors. J. Med. Chem. 47, 805–820 (2004).

    Article  CAS  PubMed  Google Scholar 

  117. van de Waterbeemd, H. & Gifford, E. ADMET in silico modelling: towards prediction paradise? Nature Rev. Drug Discov. 2, 192–204 (2003).

    Article  CAS  Google Scholar 

  118. Omiecinski, C. J. Concise review of the cytochrome P450s and their roles in toxicology. Toxicol. Sci. 48, 151–156 (1999).

    Article  CAS  PubMed  Google Scholar 

  119. de Groot, M. J., Ackland, M. J., Horne, V. A., Alex, A. A. & Jones, B. C. Novel approach to predicting P450-mediated drug metabolism: development of a combined protein and pharmacophore model for CYP2D6. J. Med. Chem. 42, 1515–1524 (1999).

    Article  CAS  PubMed  Google Scholar 

  120. de Groot, M. J., Ackland, M. J., Horne, V. A., Alex, A. A. & Jones, B. C. A novel approach to predicting P450 mediated drug metabolism. CYP2D6 catalyzed n-dealkylation reactions and qualitative metabolite predictions using a combined protein and pharmacophore model for CYP2D6. J. Med. Chem. 42, 4062–4070 (1999).

    Article  CAS  PubMed  Google Scholar 

  121. de Groot, M. J. Development of a combined protein and pharmacophore model for cytochrome P450 2C9. J. Med. Chem. 45, 1983–1993 (2002).

    Article  CAS  PubMed  Google Scholar 

  122. Park, J. -Y. & Harris, D. Construction and assessment of models of CYP2E1: Predictions of metabolism from docking, molecular dynamics, and density functional theoretical calculations. J. Med. Chem. 46, 1645–1660 (2003).

    Article  CAS  PubMed  Google Scholar 

  123. Godden, J. W., Stahura, F. L. & Bajorath, J. Statistical analysis of computational docking of large compound databases to distinct protein binding sites. J. Comput. Chem. 20, 1634–1643 (1999).

    Article  CAS  Google Scholar 

  124. Briem, H. & Kuntz, I. D. Molecular similarity based on DOCK-generated fingerprints. J. Med. Chem. 39, 3401–3408 (1996).

    Article  CAS  PubMed  Google Scholar 

  125. Su, A. I. et al. Docking molecules by families to increase the diversity of hits in database screens: computational strategy and experimental evaluation. Proteins 42, 279–293 (2001).

    Article  CAS  PubMed  Google Scholar 

  126. Rognan, D., Lauemoller, S. L., Holm, A., Buus, S., Tschinke V. Predicting binding affinities of protein ligands from three-dimensional models: application to peptide binding to class I major histocompatibility proteins. J. Med. Chem. 42, 4650–4658 (1999).

    Article  CAS  PubMed  Google Scholar 

  127. Wei, B. Q., Baase, W. A., Weaver, L. H., Matthews, B. W. & Shoichet, B. K. A model binding site for testing scoring functions in molecular docking. J. Mol. Biol. 322, 339–355 (2002).

    Article  CAS  PubMed  Google Scholar 

  128. Fradera, X., Knegtel, M. A., Mestres, J. Similarity-driven flexible ligand docking. Proteins 40, 623–626 (2000).

    Article  CAS  PubMed  Google Scholar 

  129. Lamb, M. L. et al. Design, docking, and evaluation of multiple libraries against multiple targets. Proteins 42, 296–318 (2001).

    Article  CAS  PubMed  Google Scholar 

  130. Aronov, A. M., Munagala, N. R., Kuntz, I. D. & Wang, C. C. Virtual screening of combinatorial libraries across a gene family in search of inhibitors of Giardia lamblia guanine phosphoribosyltransferase. Antimicrob. Agents Chemother. 45, 2571–2576 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wang, R., Liu, L., Lai, L. & Tang, Y. SCORE: a new empirical method for estimating the binding affinity of a protein-ligand complex. J. Mol. Model 4, 379–394 (1998).

    Article  CAS  Google Scholar 

  132. Tao, P. & Lai, L. Protein ligand docking based on empirical method for binding affinity estimation. J. Comput. Aided Mol. Des. 15, 429–446 (2001).

    Article  CAS  PubMed  Google Scholar 

  133. Chemical Computing Group. MOE. 2003. Montreal, Quebec, Canada.

  134. Friesner, R. A. et al. Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 47, 1739–1749 (2004).

    Article  CAS  PubMed  Google Scholar 

  135. Kearsley, S. K., Underwood, D. J., Sheridan, R. P. & Miller, M. D. Flexibases: a way to enhance the use of molecular docking methods. J. Comput. Aided Mol. Des. 8, 565–582 (1994).

    Article  CAS  PubMed  Google Scholar 

  136. Peng, H. et al. Identification of novel inhibitors of BCR-ABL tyrosine kinase via virtual screening. Bioorg. Med. Chem. Lett. 13, 3693–3699 (2003).

    Article  CAS  PubMed  Google Scholar 

  137. McNally, V. A. et al. Identification of a novel class of inhibitor of human and Escherichia coli thymidine phosphorylase by in silico screening. Bio. Med. Chem. Lett. 13, 3705–3709 (2003).

    Article  CAS  Google Scholar 

  138. Brenk, R. et al. Virtual screening for submicromolar leads of tRNA-guanine transglycosylase based on a new unexpected binding mode detected by crystal structure analysis. J. Med. Chem. 46, 1133–1143 (2003).

    Article  CAS  PubMed  Google Scholar 

  139. Kamionka, M. et al. In silico and NMR identification of inhibitors of the IGF-I and IGF-Binding protein-5 interaction. J. Med. Chem. 45, 5655–5660 (2002).

    Article  CAS  PubMed  Google Scholar 

  140. Vangrevelinghe, E. et al. Discovery of a potent and selective protein kinase CK2 inhibitor by high-througput docking. J. Med. Chem. 46, 2656–2662 (2003).

    Article  CAS  PubMed  Google Scholar 

  141. Enyedy, I. J. et al. Discovery of small-molecule inhibitors of Bcl-2 through structure-based computer screening. J. Med. Chem. 44, 4313–4324 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

H.D. and J.R.F. contributed equally to this paper. This manuscript is dedicated to Wolfram Saenger, Free University Berlin, on the occasion of his sixty-fifth birthday.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jürgen Bajorath.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Related links

Related links

FURTHER INFORMATION

Protein Structure Prediction Center

Research Collaboratory for Structural Biology Protein Data Bank

Biomolecular Interaction Network Database

Drug Design Resources

Glossary

POSING

The process of determining whether a given conformation and orientation of a ligand fits the active site. This is usually a fuzzy procedure that returns many alternative results.

SCORING

Both posing and ranking involve scoring. The pose score is often a rough measure of the fit of a ligand into the active site. The rank score is generally more complex and might attempt to estimate binding energies.

RANKING

A more advanced process than pose scoring that typically takes several results from an initial scoring phase and re-evaluates them. This process usually attempts to estimate the free energy of binding as accurately as possible. Although the posing phase might use simple energy calculations (electrostatic and van der Waals), ranking procedures typically involve more elaborate calculations (perhaps including properties such as entropy or explicit solvation).

POSE SPACE

All degrees of freedom involved in the process of placing one molecule relative to another. For example, for two rigid molecules the pose space simply consists of relative orientations. When one of the molecules, the ligand, is allowed to be flexible, the pose space comprises both the conformational space of the ligand and orientational space of ligand and receptor.

FORCE-FIELD

A function expressing the energy of a system as a sum of diverse molecular mechanics (or other) terms.

TORSIONAL ENTROPY

Entropy associated with a rotatable bond in a molecule. Immobilization of a rotatable bond on binding leads to loss of its torsional (or rotational) entropy.

REGRESSION ANALYSIS

Determination of parameter values for a chosen (linear or nonlinear) function to best fit a set of observations.

POTENTIAL OF MEAN FORCE

(PMF). In the context of docking and scoring, PMFs are derived from statistical analysis of experimentally observed distributions and frequencies of specific atom-pair interactions in a large collection of protein–ligand structures. Interaction potentials between each atom pair in two molecules (for example, ligand and protein) approximate the free energy of each pair-wise interaction as a function of inter-atomic distance.

LINEAR DISCRIMINANT ANALYSIS

Mathematical analysis based on two classes of data and two independent variables (a, b) that attempts to find a line that best separates the data. This line is orthogonal to the discriminant function that is a linear combination of the original variables, in this case: F = caa + cbb (ca, cb; coefficients).

PHARMACOPHORE

The spatial arrangement of atoms or groups in a molecule known or predicted to be responsible for specific biological activity.

HOLO-, APO-ENZYME

Holo-: ligand-bound form of an enzyme; apo-: uncomplexed form. The original definitions referred to enzymes and cofactors, rather than ligands, but ligands and cofactors are often synonymously used.

NORMAL MODE

An oscillation in which all particles of a system move with the same frequency and phase.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kitchen, D., Decornez, H., Furr, J. et al. Docking and scoring in virtual screening for drug discovery: methods and applications. Nat Rev Drug Discov 3, 935–949 (2004). https://doi.org/10.1038/nrd1549

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd1549

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing