Добавил:
Опубликованный материал нарушает ваши авторские права? Сообщите нам.
Вуз: Предмет: Файл:
Becker O.M., MacKerell A.D., Roux B., Watanabe M. (eds.) Computational biochemistry and biophysic.pdf
Скачиваний:
68
Добавлен:
15.08.2013
Размер:
5.59 Mб
Скачать

364

Tropsha and Zheng

libraries that are then tested against a variety of biological targets. This design strategy is suited for lead identification programs. Thus, two categories of computational tools should be developed and validated to meet the needs of the two different types of projects.

In a targeted screening project, computational library design involves the selection of a subset of chemical building blocks from an available pool of chemical structures. This subset of selected building blocks affords a limited virtual library with a high content of compounds similar to a lead molecule. Molecular similarity is quantified by using a chosen set of molecular descriptors and similarity metrics [64,65]. Building blocks can also be chosen such that the resulting virtual library could have a high percentage of compounds that are predicted to be active from a preconstructed QSAR model [66]. When the structure of the biological target is known, one can select building blocks such that the resulting library compounds are stereochemically complementary to the binding site structure of the underlying target [67,68]. Other approaches to targeted library design using different criteria have also been considered [69]. Similar approaches have long been used in targeted database mining that were based on the principle of either molecular similarity [70,71] or structure-based drug design [72]. In a broad screening project, computational library design or database mining involves the selection of a subset of compounds that are optimally diverse and representative of available classes of compounds, leading to a nonredundant chemical library or a set of nonredundant compounds for biological testing. Reported methods can be generally classified into several categories.

1.Cluster sampling methods, which first identify a set of compound clusters, followed by the selection of several compounds from each cluster [73].

2.Grid-based sampling, which places all the compounds into a low-dimensional descriptor space divided into many cells and then chooses a few compounds from each cell [74].

3.Direct sampling methods, which try to obtain a subset of optimally diverse compounds from an available pool by directly analyzing the diversity of the selected molecules [75,76].

Many reports have been published that address various aspects of diversity analysis in the context of chemical library design and database mining [77–84].

IV. SUMMARY AND CONCLUSIONS

We have reviewed several methods of computer-aided drug design that are formally divided into ligand-based and receptor-based methods. Ligand-based methods of analysis are used widely because they are not very computationally intensive and afford rapid generation of QSAR models from which the biological activity of newly designed compounds can be predicted. Most of the existing methods require the generation of a 3D pharmacophore hypothesis (i.e., unique 3D arrangements of important functional groups common to all or the majority of the receptor ligands). In many cases, when the receptor ligands are not very structurally diverse and include conformationally rigid compounds, a pharmacophore can be generated in a reasonably unbiased and unique way, using either automated (e.g., DISCO [85]) or semiautomated [3] pharmacophore prediction methods. The pharmacophore hypothesis can help medicinal chemists gain an insight into the key interactions between ligand and receptor when a 3D ligand–receptor complex structure has not been determined. It can be used as the search query for 3D database mining, and

Computer Aided Drug Design

365

this approach has been demonstrated as very productive for lead compound discovery [86]. It can also be used for 3D QSAR analysis, grouping together the compounds that follow the same binding mode and indicating the possible 3D alignment rules [87]. However, this task of unique pharmacophore generation becomes less feasible for more structurally diverse and/or conformationally flexible compounds. In general, in the absence of detailed structural information about the receptor binding site, any pharmacophore inferred from only the ligand structure remains hypothetical.

Structure-based design methods present an appealing alternative to more traditional approaches to CADD. Molecular docking algorithms can generate fairly accurate orientations of known or designed receptor ligands in the active site (e.g., GRID [88], DOCK [16]). However, their application in practice is limited by the availability of macromolecular target structure. The protein and nucleic acid structural databases have been growing exponentially over the past decade, with the size of the Protein Data Bank (PDB) [89] currently exceeding 10,000 entries. Nevertheless, many of these entries are highly homologous proteins, and the structure of many pharmacologically important molecules such as transmembrane receptor proteins cannot be resolved experimentally at the present time. Furthermore, predictions of the binding affinity of receptor ligands are fast but rather inaccurate because the scoring functions used in docking calculations are not robust enough. Alternatively, these predictions can be fairly accurate but very computationally intensive when free energy simulation methods are used [21,90]. Therefore, because of these practical limitations, ligand-based design methods will probably remain the main arena of CADD efforts, supplemented, when possible, by structure-based approaches.

We have shown that successful molecular modeling and the design of new drugs can be achieved by integration of different computational chemistry and molecular modeling techniques. Owing to the rapid increase in computational power due to advances in both hardware and software, molecular modeling has become an important integral part of multidisciplinary efforts in the design and synthesis of new potent pharmaceuticals. Practical knowledge of these techniques and their limitations is a necessary component of modern research in drug discovery.

REFERENCES

1.C Hansch, A Leo. Exploring QSAR: Fundamentals and Applications in Chemistry and Biology. Washington, DC: American Chemical Society, 1995.

2.DB Boyd. In: KB Lipkowitz, DB Boyd, eds. Reviews in Computational Chemistry. New York: VCH, 1991.

3.GR Marshall, CD Barry, HE Bosshard, RA Dammkoehler, DA Dunn. The conformational parameter in drug design: The active analog approach. ACS Symp Ser 112:205–226, 1979.

4.YC Martin. Overview of concepts and methods in computer-assisted rational drug design. Methods Enzymol 203:587–613, 1991.

5.RD Cramer III, DE Patterson, JD Bunce. Comparative molecular field analysis (CoMFA). 1. Effect of shape on binding of steroids to carrier proteins. J Am Chem Soc 110:5959–5967, 1988.

6.DB Boyd. Successes of computer-assisted molecular design. In: KB Lipkowitz, DB Boyd, eds. Reviews in Computational Chemistry I. New York: VCH, 1990, pp 355–372.

7.CE Bugg, WM Carson, JA Montgomery. Drugs by design. Sci Am 269:92–98, 1993.

8.SG Deeks, M Smith, M Holdniy, JO Kahn. HIV-1 protease inhibitors: A review for clinicians. J Am Med Assoc 277:145–153, 1997.

366

Tropsha and Zheng

9.GR Marshall, CD Barry, HE Bosshard, RA Dammkoehler, DA Dunn. The conformational parameter in drug design: The active analog approach. ACS Symp Ser 112:205–226, 1979.

10.JL Fauchere, ed. QSAR: Quantitative Structure–Activity Relationships in Drug Design. New York: Alan R Liss, 1989, pp 177–181.

11.H Kubinyi, G Folkers, YC Martin, eds. 3D QSAR in Drug Design: Recent Advances, Vols 2 and 3. Dordrecht, The Netherlands: Kluwer, 1998.

12.KB Lipkowitz, DB Boyd, eds. Reviews in Computational Chemistry, Vols I–XII. New York, VCH, 1991– (ongoing series).

13.DM Mottola, S Laiter, VJ Watts, A Tropsha, SW Wyrick, DE Nichols, P Mailman. Conformational analysis of d1 dopamine receptor agonists: Pharmacophore assessment and receptor mapping. J Med Chem 39:285–296, 1996.

14.RB Mailman, DE Nichols, A Tropsha. Molecular drug design and dopamine receptors. In: K Neve, R Neve, eds. Dopamine Receptors. Totowa, NJ: Humana Press, 1996, pp 105–133.

15.BT Hoffman, SJ Cho, W Zheng, S Wyrick, DE Nichols, RB Mailman, A Tropsha. QSAR modeling of dopamine d1 agonists using comparative molecular field analysis, genetic algo- rithms-partial least squares, and K nearest neighbor methods. J Med Chem 42:3217–3226, 1999.

16.EC Meng, BK Shoichet, ID Kuntz. Automated docking with grid-based energy evaluation. J Comput Chem 13:505–524, 1992.

17.DS Goodsell, AJ Olson. Automated docking of substrates to proteins by simulated annealing. Proteins Struct Funct Genet 8:195–202, 1990.

18.M Karplus, GA Petsko. Molecular dynamics simulations in biology. Nature 347:631–637, 1990.

19.TL Tembe, JA McCammon. Ligand–receptor interactions. Comput Chem 8:281–283, 1984.

20.S Hirono, PA Kollman. Calculation of the relative binding free energy of 2-GMP and 2- AMP to ribonuclease T1 using molecular dynamics/free energy perturbation approaches. J Mol Biol 212:197–209, 1990.

21.A Tropsha, J Hermans. Application of free energy simulations to the binding of a transition- state-analogue inhibitor to HIV protease. Protein Eng 51:29–34, 1992.

22.DP Marriott, IG Dougall, P Meghani, Y-J Liu, DR Flower. Lead generation using pharmacophore mapping and three-dimensional database searching: Application to muscarinic M3 receptor antagonists. J Med Chem 42:3210–3216, 1999.

23.C Hansch, RM Muir, T Fujita, PP Maloney, E Geiger, M Streich. The correlation of biological activity of plant growth regulators and chloromycetin derivatives with Hammett constants and partition coefficients. J Am Chem Soc 85:2817–2824, 1963.

24.T Fujita, J Iwasa, C Hansch. A new substituent constant, π, derived from partition coefficients. J Am Chem Soc 86:5175–5180, 1964.

25.LP Hammett. Some relations between reaction rates and equilibrium constants. Chem Rev 17: 125–136, 1935.

26.C Hansch, A Leo. Exploring QSAR: Fundamentals and Applications in Chemistry and Biology (Chapters 1–4). Washington, DC: Am Chem Soc, 1995.

27.C Hansch, A Leo, D Hoekman. Exploring QSAR: Hydrophobic, Electronic, and Steric Constants. Washington, DC: Am Chem Soc, 1995.

28.A Verloop, W Hoogenstraaten, J Tipker. In: EJ Ariens, ed. Drug Design, Vol VII. New York: Academic Press, 1976, p 165.

29.M Randic. On characterization of molecular branching. J Am Chem Soc 97:6609–6615, 1975.

30.LB Kier, LH Hall. Molecular Connectivity in Structure–Activity Analysis. Chichester, England: Research Studies Press, 1986.

31.LH Hall, LB Kier. The molecular connectivity chi indexes and kappa shape indexes in struc- ture–property modeling. In: KB Lipkowitz, DB Boyd, eds. Reviews in Computational Chemistry, Vol. 2. New York: VCH, 1991, pp 367–422.

Computer Aided Drug Design

367

32.S Wold, A Ruhe, H Wold, WJ Dunn III. The collinearity problem in linear regression. The partial least squares (PLS) approach to generalized inverses. SIAM J Sci Stat Comput 5:735– 743, 1984.

33.S Hellberg, M Sjostrom, B Skagerberg, S Wold. Peptide quantitative structure–activity relationships, a multivariate approach. J Med Chem 30:1126–1135, 1987.

34.AJ Hopfinger. A QSAR investigation of dihydrofolate reductase inhibition by Baker triazines based upon molecular shape analysis. J Am Chem Soc 102:7196–7206, 1980.

35.GM Crippen. Distance geometry approach to rationalizing binding data. J Med Chem 22(8): 988–997, 1979.

36.GM Crippen. Quantitative structure–activity relationships by distance geometry: Systematic analysis of dihydrofolate reductase inhibitors. J Med Chem 23:599–606, 1980.

37.LG Boulu, GM Crippen. Voronoi binding site models: Calculation of binding models and influence of drug binding data accuracy. J Comput Chem 10(5):673–682, 1989.

38.H Kubinyi, G Folkers, YC Martin, eds. 3D QSAR in Drug Design: Recent Advances, Vols 2 and 3. Dordrecht, The Netherlands: Kluwer Academic, 1998.

39.JM Sutter, SL Dixon, PC Jurs. Automated descriptor selection for quantitative structure–activ- ity relationships using generalized simulated annealing. J Chem Inf Comput Sci 35(1):77–84, 1995.

40.D Rogers, AJ Hopfinger. Application of genetic function approximation to quantitative struc- ture–activity relationships and quantitative structure–property relationships. J Chem Inf Comput Sci 34(4):854–866, 1994.

41.H Kubinyi. Variable selection in QSAR studies. I. An evolutionary algorithm. Quant StructAct Relat 13:285–294, 1994.

42.H Kubinyi. Variable selection in QSAR studies. II. A highly efficient combination of systematic search and evolution. Quant Struct-Act Relat 13:393–401, 1994.

43.BT Luke. Evolutionary programming applied to the development of quantitative structure– activity relationships and quantitative structure–property relationships. J Chem Inf Comput Sci 34(6):1279–1287, 1994.

44.SS So, M Karplus. Evolutionary optimization in quantitative structure–activity relationship: An application of genetic neural networks. J Med Chem 39:1521–1530, 1996.

45.M Baroni, G Costantino, G Cruciani, D Riganelli, R Valigi, S Clementi. Generating optimal linear PLS estimations (GOLPE): An advanced chemometric tool for handling 3D-QSAR problems. Quant Struct-Act Relat 12:9–20, 1993.

46.SJ Cho, A Tropsha. Cross-validated R2-guided region selection for comparative molecular field analysis: A simple method to achieve consistent results. J Med Chem 38:1060–1066, 1995.

47.T Kimura, K Hasegawa, K Funatsu. GA strategy for variable selection in QSAR studies: GAbased region selection for CoMFA modeling. J Chem Inf Comput Sci 38:276–282, 1998.

48.SJ Cho, ML Garsia, J Bier, A Tropsha. Structure-based alignment and comparative molecular field analysis of acetylcholinesterase inhibitors. J Med Chem 39(26):5064–5071, 1996.

49.TA Andrea, H Kalayeh. Applications of neural networks in quantitative structure–activity relationships of dihydrofolate reductase inhibitors. J Med Chem 34:2824–2836, 1991.

50.G Bolis, L Pace, F Fabrocini. A machine learning approach to computer-aided molecular design. J Comput Aided Mol Des 5(6):617–628, 1991.

51.PJ Whittle, TL Blundell. Protein structure-based drug design. Annu Rev Biophys Biomol Struct 23:349–375, 1994.

52.DM Lorber, BK Shoichet. Flexible ligand docking using conformational ensembles. Protein Sci 7:938–950, 1998.

53.TJA Ewing, ID Kuntz. Critical evaluation of search algorithms for automated molecular docking and database screening. J Comput Chem 18:1175–1189, 1997.

54.J Aqvist, C Medina, J-E Samuelsson. A new method for predicting binding affinity in com- puter-aided drug design. Protein Eng 7:385–391, 1994.

368

Tropsha and Zheng

55.HA Carlson, WL Jorgensen. An extended linear response method for determining free energies of hydration. J Phys Chem 99:10667, 1995.

56.X Chen, A Tropsha. A generalized linear response method: Application to the hydration free energy calculations. J Comput Chem 20:749–759, 1999.

57.J Apostolakis, A Caflisch. Computational ligand design. Comb Chem High Throughput Screen 2:91–104, 1999.

58.JW Pitera, NR Munagala, CC Wang, PA Kollman. Understanding substrate specificity in human and parasite phosphoribosyltransferases through calculation and experiment. Biochemistry 38:10298–10306, 1999.

59.J Marelius, M Graffner-Nordberg, T Hansson, A Hallberg, J Aqvist. Computation of affinity and selectivity: Binding of 2,4-diaminopteridine and 2,4-diaminoquinazoline inhibitors to dihydrofolate reductases. J Comput-Aided Mol Des 12:119–131, 1998.

60.RH Smith Jr, WL Jorgensen, J Tirado-Rives, ML Lamb, PAJ Janssen, CJ Michejda, MBK Smith. Prediction of binding affinities for TIBO inhibitors of HIV-1 reverse transcriptase using Monte Carlo simulations in a linear response method. J Med Chem 41:5272–5286, 1998.

61.EM Gordon, RW Barret, WJ Dower, SPA Fodor, MA Gallop. Applications of combinatorial technologies to drug discovery. 2. Combinatorial organic synthesis, library screening strategies, and future directions. J Med Chem 37:1385–1401, 1994.

62.MA Gallop, RW Barret, WJ Dower, SPA Fodor, EM Gordon. Applications of combinatorial technologies to drug discovery. 1. Background and peptide combinatorial libraries. J Med Chem 37:1233–1251, 1994.

63.WA Warr. Combinatorial chemistry and molecular diversity. An overview. J Chem Inf Comput Sci 37:134–140, 1997.

64.RP Sheridan, SK Kearsley. Using a genetic algorithm to suggest combinatorial libraries. J Chem Inf Comput Sci 35:310–320, 1995.

65.W Zheng, SJ Cho, A Tropsha. Rational combinatorial library design 1. Focus-2D: A new approach to the design of targeted combinatorial chemical libraries. J Chem Inf Comput Sci 38:251–258, 1998.

66.SJ Cho, W Zheng, A Tropsha. Rational combinatorial library design 2. Rational design of targeted combinatorial peptide libraries using chemical similarity probe and the inverse QSAR approaches. J Chem Inf Comput Sci 38:259–268, 1998.

67.Q Zheng, DJ Kyle. Computational screening of combinatorial libraries. Bioorg Med Chem 4(5):631–638, 1996.

68.EK Kick, DC Roe, AG Skillman, G Liu, TJ Ewing, Y Sun, ID Kuntz, JA Ellman. Structurebased design and combinatorial chemistry yield low nanomolar inhibitors of cathepsin D. Chem Biol 4(4):297–307, 1997.

69.RD Brown, YC Martin. Designing combinatorial library mixtures using a genetic algorithm. J Med Chem 40(15):2304–2313, 1997.

70.P Willett. Algorithms for the calculation of similarity in chemical structure databases. In: MA Johnson, GM Maggiora, eds. Concepts and Applications of Molecular Similarity. New York: Wiley, 1990, pp 43–63.

71.NC Perry, VJ van Geerestein. Database searching on the basis of three-dimensional molecular similarity using the SPERM program. J Chem Inf Comput Sci 32(6):607, 1992.

72.DA Gschwend, AC Good, ID Kuntz. Molecular docking towards drug discovery. J Mol Recogn 9(2):175–186, 1996.

73.P Willett, V Winterman, D Bawden. Implementation of nonhierarchic cluster analysis methods in chemical information systems: Selection of compounds for biological testing and clustering of substructures search output. J Chem Inf Comput Sci 26:109–118, 1986.

74.RS Pearlman. Novel software tools for addressing chemical diversity. Network Sci 1996. http:/ /www.awod.com/netsci/Science/Combichem/feature08.html

75.EJ Martin, JM Blaney, MA Siani, DC Spellmeyer, AK Wong, WH Moos. Measuring diversity:

Computer Aided Drug Design

369

Experimental design of combinatorial libraries for drug discovery. J Med Chem 38(9):1431– 1436, 1995.

76.W Zheng, SJ Cho, CL Waller, A Tropsha. Rational combinatorial library design. 3. Simulated annealing guided evaluation (SAGE) of molecular diversity: A novel computational tool for diverse library design and database mining. J Chem Inf Comput Sci 39:738–746, 1999.

77.H Bauknecht, A Zell, H Bayer, P Levi, M Wagener, J Sadowski, J Gasteiger. Locating biologically active compounds in medium-sized heterogeneous datasets by topological autocorrelation vectors: Dopamine and benzodiazepine agonists. J Chem Inf Comput Sci 36(6):1205–1213, 1996.

78.A Polinski, RD Feinstein, S Shi, A Kuki. LiBrain: Software for automated design of exploratory and targeted combinatorial libraries. In: IM Chaiken, KD Janda, eds. Molecular Diversity and Combinatorial Chemistry: Libraries and Drug Discovery. ACS Conf Proc Ser. Washington, DC: Am Chem Soc, 1996, pp 219–232.

79.DB Turner, SM Tyrell, P Willett. Rapid quantification of molecular diversity for selective database acquisition. J Chem Inf Comput Sci 37:18–22, 1997.

80.DJ Cummins, CW Andrews, JA Bentley, M Cory. Molecular diversity in chemical databases: Comparison of medicinal chemistry knowledge bases and databases of commercially available compounds. J Chem Inf Comput Sci 36:750–763, 1996.

81.S Pickett, JS Mason, IM McLay. Diversity profiling and design using 3D pharmacophores: Pharmacophore-derived queries (PDQ). J Chem Inf Comput Sci 36:1214–1223, 1996.

82.PL Myers, JW Greene, J Saunders, SL Teig. Rapid, reliable drug discovery. Today’s Chemist at Work July/August 1997, pp 46–53.

83.AC Good, R Lewis. New methodology for profiling combinatorial libraries and screening sets: Cleaning up the design process with HARPick. J Med Chem 40:3926–3936, 1997.

84.VJ Gillet, P Willett, J Bradshaw. The effectiveness of reactant pools for generating structurallydiverse combinatorial libraries. J Chem Inf Comput Sci 37:731–740, 1997.

85.YC Martin, MG Bures, EA Danaher, J DeLazzer, I Lico, PA Pavlik. A fast new approach to pharmacophore mapping and its application to dopaminergic and benzodiazepine agonists. J Comput Aided Mol Des 7:83–102, 1993.

86.S Wang, DW Zaharevitz, R Sharma, VE Marquez, NE Lewin, L Du, PM Blumberg, GWA Milne. The discovery of novel, structurally diverse protein kinase C agonists through computer 3D-database pharmacophore search. J Med Chem 37:4479–4489, 1994.

87.SA DePriest, D Mayer, CB Naylor, GR Marshall. 3D-QSAR of angiotensin-converting enzyme and thermolysin inhibitors: A comparison of CoMFA models based on deduced and experimentally determined active site geometries. J Am Chem Soc 115:5372–5384, 1993.

88.PJ Goodford. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules. J Med Chem 28:849–957, 1985.

89.FC Bernstein, TF Koetzle, GJB Williams, EF Meyer, MD Brice, JR Rodgers, O Kennard, T Shimanouchi, M Tasumi. Protein Data Bank: Computer based archival file for macromolecular structure. J Mol Biol 112:535–542, 1977.

90.DM Ferguson, RJ Radmer, PA Kollman. Determination of the relative binding free energies of peptide inhibitors to the HIV-1 protease. J Med Chem 34:2654–2659, 1991.