• Tidak ada hasil yang ditemukan

FROM HIT TO LEAD: SUMMARY OF COMPOUND OPTIMIZATION IN DRUG DISCOVERY

Optimization Studies

5.4 FROM HIT TO LEAD: SUMMARY OF COMPOUND OPTIMIZATION IN DRUG DISCOVERY

at various points in the product lifecycle. However, there are still concerns associated with the management of enormous data generated through anal- ysis of multiple large databases simultaneously, placing a special demand for software, hardware, and behavior developments. Before the inception of bioinformatics and cheminformatics, handling of large individual databases remained a daunting task, which currently is being addressed through the informatics technologies, although the challenges of efficient correlation of such diversified information still exist [67].

The United States National Center for Biotechnology Information has been responsible for keeping databases for genome projects. GenBank, an international collaboration of three databases between the United States, the European Molecular Biology Institute, and the DNA Data Bank of Japan, constantly engages in interchange of database information. This has resulted in problems requiring continual update of records in the old data- base regarding the use of terms and tags that are not consistent with the new format. Private companies are building web interfaces for their database offerings. Celera and Incyte offer web subscriptions to their proprietary databases and customized analysis tools. It appears that the bioinformatics child will continue to speak the web language.

Bank It is a web submission program that includes the top 100 most- accessed information. Sequin is GenBank’s submission program, running on several platforms, with complicated entries and the ability to locate errors such as missing organism information, incorrect coding region lengths, mis- matched amino acids, or internal stop codons in coding regions, and more.

Information exchange in drug discovery would have been more con- strained if it were not for the increasing application of informatics [68].

5.4 FROM HIT TO LEAD: SUMMARY OF COMPOUND

Social Aspects of Drug Discovery, Development and Commercialization 124

Once a drug hit shows a possibility with its target, series of further chemical modifications take place to ensure in vivo compliance and ad- equate functionality. Multiple chemical series are generated based on the structural framework of the hit molecules, leading to an enormous number of compounds with wide chemical and structural diversity. The classical medicinal chemistry approach is used to build a SAR by modifying sub- stituents on a structural scaffold using mostly biochemical knowledge to obtain the desired affinity for the target. Most chemical libraries focus on the chemical series that contain many variations on the same molecular scaffold or molecular backbone. Medicinal chemists utilize many strategies like combinatorial chemistry to introduce structural changes in order to improve a compound’s pharmacodynamics properties.

Combinatorial chemistry involves preparation of diverse compounds and structures by synthesis methods. It could be simply defined as a tech- nology involved in the creation of large diversity libraries, speeding up the production of a wide combination of reactive chemical entities. It has aided the construction the chemicals in a way that improve the biological proper- ties that are determined through bioassays (Figure 5.2). Together with HTS it is expected to provide more biological data in the same proportion with the efficiency of the technique used.

As the chemical series are being evaluated, the pharmacological profile further narrows the series and helps to prioritize the hit compounds. Paral- lel models in drug compound optimization seek to synchronize biological

Figure 5.2 Iterative Process of Compound Design, Synthesis, and Testing.

with chemical studies that increasingly transform the drug compounds to become more drug-like for future clinical application. Successive screenings of the drug molecules lead to a more extended HTS of arrayed targets and ligands (Figure 5.3).

The newly found pharmacologically active moieties might not pos- sess the drug-like properties. Thus, as the bioassays are being conducted, the medicinal chemists use relevant robust data as a foundation for deci- sion making on the chemical modification (Figure 5.3), which gives rise to drug-like qualities. Molecular modification often involves adapting the molecular structure into one that has improved drug qualities, which would exhibit properties like potency, minimized side effects, bioactivity, and im- proved PK. Such data would be used to identify the undesirable activities and to determine SARs at the molecular targets to aid drug design while excluding the undesirable attributes from the chemical series. The modified versions of the lead compound could be synthesized in order to reduce side effects, enhance affinity for one receptor over another, and/or improve activity.

The iterative screening and optimization schemes have taken varied for- mats, where the building knowledge enables the development process, result- ing in a lead compound optimized for potency, selectivity, and PK qualities.

When the selectivity profile has been established, and pharmacology attri- butes and druggability have been duly confirmed for a particular drug, the drug development cycle progresses to the preclinical phase.

Figure 5.3 The Pathway Leading to Compound Optimization in Drug Discovery.

Social Aspects of Drug Discovery, Development and Commercialization 126

REFERENCES

[1] Horrobin DF. Realism in drug discovery – could Cassandra be right? Nat Biotechnol 2001;19:1099.

[2] Drew KL, Baiman H, Khwaounjoo P, Yu B, Reynisson J. Size estimation of chemical space: how big is it? J Pharm Pharmacol 2012;64:490–5.

[3] Fox S, Farr-Jones S, Sopchak L, Boggs A, Wang Nicely H, Khoury R, Biroshigh M. High throughput screening: update on practices and success. J Biomol Screen 2006;11:864–9.

[4] Lahana R. How many leads from HTS? Drug Discov Today 1999;4:447–8.

[5] Coma I, Clark L, Diez E, Harper G, Herranz J, Hofmann G, Lennon M, Richmond N, Valmaseda M, Macarron R. Process validation and screen reproducibility in high- throughput screening. J Biomol Screen 2009;14:66–76.

[6] Nichols A. High content screening as a screening tool in drug discovery. Methods Mol Biol 2007;356:379–87.

[7] Bleicher KH, Böhm HJ, Müller K, Alanine AI. Hit and lead generation: beyond high- throughput screening. Nature Rev Drug Discov 2003;2:369–78.

[8] Houston JG, Banks MN, Binnie A, Brenner S, O’Connell JO, Petrillo EW. Case study:

impact of technology investment on lead discovery at Bristol-Myers Squibb, 1998–

2006. Drug Discov Today 2008;13:44–51.

[9] Zhang JH, Chung TD, Oldenburg KR. A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J Biomol Screen 1999;4:67–73.

[10] Makarenkov V, Zentilli P, Kevorkov D, Gagarin A, Malo N, Nadon R. An efficient method for the detection and elimination of systematic error in high-throughput screening. Bioinformatics 2007;23:1648–57.

[11] Kaiser J. Industrial-style screening meets academic biology. Science 2008;321:764–6.

[12] Silber BM. Driving drug discovery: the fundamental role of academic labs. Sci Transl Med 2010;2:30cm16.

[13] Posner BA. High-throughput screening-driven lead discovery: meeting the challenges of finding new therapeutics. Curr Opin Drug Discov Devel 2005;8(4):487–94.

[14] Zhang Z, Guan N, Li T, Mais DE, Wang M. Quality control of cell-based high- throughput drug screening. Acta Pharmaceut Sin B 2012;2(5):429–38.

[15] Hann MM, Leach AR, Harper G. Molecular complexity and its impact on the probability of finding leads for drug discovery. J Chem Inform Comput Sci 2001;41:856–64.

[16] Irwin JJ. How good is your screening library? Curr Opin Chem Biol 2006;10:352–6.

[17] Lahana R. Who wants to be irrational? Drug Discov Today 2003;8:655–6.

[18] Michelini E, Cevenini L, Mezzanotte L, Coppa A, Roda A. Cell based assays: fuelling drug discovery. Anal Biochem 2010;397:1–10.

[19] Dunne A, Jowett M, Rees S. Use of primary cells in high throughput screens. Meth Mol Biol 2009;565:239–57.

[20] Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early drug discovery. J Phar- macol Mar 2011;162(6):1239–49.

[21] Moore K, Rees S. Cell-based versus isolated target screening: how lucky do you feel?

J Biomol Scr 2001;6:69–74.

[22] Hunter PJ, Borg TK. Integration from proteins to organs: the Physiome Project. Nat Rev Mol Cell Biol 2003;4:237–43.

[23] Noble D. Modelling the heart – from genes to cells to the whole organ. Science 2002;295:1678–82.

[24] Bassingthwaighte JB, Vinnakota KC. The computational integrated myocyte: a view into the virtual heart. Ann NY Acad Sci 2004;1015:391–404.

[25] Wang F, Wong PK. Intelligent systems and technology for integrative and predictive medicine: an ACP approach. ACM Trans Intell Syst Technol 2013;4(2):1–6, article 32.

[26] Li S, Zhang B, Zhang N. Network target for screening synergistic drug combinations with application to traditional Chinese medicine. BMC Syst Biol 2011;5(Suppl. 1):S10.

[27] Vedani A, Smiesko M. In silico toxicology in drug discovery – concepts based on three- dimensional models. Altern Lab Anim 2009;37:477–96.

[28] Kuhn M, Campillos M, Letunic I, Jensen LJ, Bork P. A side effect resource to capture phenotypic effects of drugs. Mol Syst Biol 2010;6:343.

[29] Ekins S, Williams AJ, Krasowski MD, Freundlich JS. In silico repositioning of approved drugs for rare and neglected diseases. Drug Discov Today 2011;16:298–310.

[30] Mendrick DL. Transcriptional profiling to identify biomarkers of disease and drug response. Pharmacogenomics 2011;12:235–49.

[31] Mah JT, Low ES, Lee E. In silico SNP analysis and bioinformatics tools: a review of the state of the art to aid drug discovery. Drug Discov Today 2011;16:800–9.

[32] Tanrikulu Y, Kruger B, Proschak E. The holistic integration of virtual screening in drug discovery. Drug Discov Today 2013;18(7/8):358–64.

[33] Stumpfe D, Ripphausen P, Bajorath J. Virtual compound screening in drug discovery.

Fut Med Chem 2012;4:593–602.

[34] Kitchen DB, Decornez H, Furr JR, Bajorath J. Docking and scoring in virtual screen- ing for drug discovery: methods and applications.. Nat Rev Drug Discov 2004;3:

935–48.

[35] McInnes C. Virtual screening strategies in drug discovery. Curr Opin Chem Biol 2007;11(5):494–502.

[36] Hajduk PJ, Huth JR, Tse C. Predicting protein druggability. Drug Discov Today 2005;10:1675–82.

[37] Laurie ATR, Jackson RM. Methods for the prediction of protein-ligand binding sites for structure-based drug design and virtual ligand screening. Curr Protein Pept Sci 2006;7:395.

[38] Prinz F, Schlange T, Asadullah K. Believe it or not: how much can we rely on published data on potential drug targets? Nat Rev Drug Discov 2011;10:712.

[39] Faller B, Ottaviani G, Ertl P, Berellini G, Collis A. Evolution of the physicochemical properties of marketed drugs: can history foretell the future? Drug Discov Today 2011;16:976–84.

[40] Park BK, Boobis A, Clarke S, Goldring CE, Jones D, Kenna JG, Lambert C, Laverty HG, Naisbitt DJ, Nelson S, Deborah A, Nicoll-Griffith R, Scott O, Philip R, Smith DA, Tweedie DJ, Vermeulen N, Williams DP, Wilson ID, Thomas AB. Managing the challenge of chemically reactive metabolites in drug development. Nat Rev Drug Discov 2011;10:292–306.

[41] Bauer RA, Wurst JM, Tan DS. Expanding the range of “druggable” targets with natu- ral product-based libraries: an academic perspective. Curr Opin Chem Biol 2010;14:

308–14.

[42] Leeson PD, St-Gallay SA. The influence of the “organizational factor” on compound quality in drug discovery. Nat Rev Drug Discov 2011;10:749–65.

[43] Van Drie JH. Computer-aided drug design: the next 20 years. J Comput Aided Mol Des 2007;21:591–601.

[44] Bocker A, Derksen S, Schmidt E, Teckentrup A, Schneider G. A hierarchical clustering approach for large compound libraries. J Chem Inform Model 2005;45:807–15.

[45] Kapetanovic IM. Computer-aided drug discovery and development (CADDD):

in silico-chemico-biological approach. ChemBiol Interact 2008;171(2):165–76.

[46] Kalyaanamoorthy S, Chen YP. Structure-based drug design to augment hit discovery.

Drug Discov Today 2011;16:831.

[47] Jorgensen WL. Drug discovery: pulled from a protein’s embrace. Nature 2010;466:6–8.

[48] Plowright AT, Johnstone C, Kihlberg J, Pettersson J, Robb G, Thompson RA. Hypothesis driven drug design: improving quality and effectiveness of the design-make-test-analyse cycle. Drug Discov Today 2012;17:56–62.

[49] Andersson S, Armstrong A, Björe A, Bowker S, Chapman S, Davies R, Donald C, Egner B, Elebring T, Holmqvist S, Inghardt T, Johannesson P, Johansson M, Johnstone C,

Social Aspects of Drug Discovery, Development and Commercialization 128

Kemmitt P, Kihlberg J, Korsgren P, Lemurell M, Moore J, Jonas AP, Pointon H, Pontén F, Schofield P, Selmi N, Whittamore P. Making medicinal chemistry more effective – application of lean sigma to improve processes, speed and quality. Drug Discov Today 2009;14:598–604.

[50] Johnstone C, Pairaudeau G, Pettersson JA. Creativity, innovation and lean sigma: a controversial combination? Drug Discov Today 2011;16:50–7.

[51] Erickson JA, Mader MM, Watson IA, Webster YW, Higgs RE, Bell MA, Vieth M.

Structure-guided expansion of kinase fragment libraries driven by support vector machine models. Biochim Biophys Acta 2010;1804:642–52.

[52] Vinter A, Gardner S, Rose S. Overcoming the limitations of chemical structure. Mol Model 2009;59.

[53] Gleeson MP, Hersey AS, Hannongbua S. In-silico ADME models: a general assessment of their utility in drug discovery applications. Curr Top Med Chem 2011;10:11358–81.

[54] Li Q, Jørgensen FS, Oprea T, Brunak S, Taboureau O. hERG classification model based on a combination of support vector machine method and GRIND descriptors. Mol Pharm 2008;5(1):117–27.

[55] Gavaghan CL, Arnby CH, Blomberg N, Strandlund G, Boyer S. Development, interpretation and temporal evaluation of a global QSAR of hERG electrophysiology screening data. J Comput Aided Mol Des 2007;21:189–206.

[56] Acharya C, Coop A, Polli JE, Mackerell AD Jr. Recent advances in ligand based drug design: relevance and utility of the conformationally sampled pharmacophore approach.

Curr Comput Aided Drug Des 2011;7:10–22.

[57] Yang SY. Pharmacophore modelling and applications in drug discovery: challenges and recent advances. Drug Discov Today 2010;15:444.

[58] Kurogi Y, Güner OF. Pharmacophore modelling and three-dimensional database searching for drug design using catalyst. Curr Med Chem 2001;8:1035–55.

[59] Jones G, Willett P, Glen RC. A genetic algorithm for flexible molecular overlay and pharmacophore elucidation. J Comput Aided Mol Des 1995;9:532.

[60] Dixon SL, Smondyrev AM, Knoll EH, Rao SN, Shaw DE, Friesner RA. PHASE: a new engine for pharmacophore perception, 3D QSAR model development, and 3D database screening: 1. Methodology and preliminary results. J Comput Aided Mol Des 2006;20:647.

[61] Michielan L, Moro S. Pharmaceutical perspectives of nonlinear QSAR strategies.

J Chem Inf Model 2010;50:961–78.

[62] Lagorce D, Maupetit J, Baell J, Sperandio O, Tufféry P, Miteva MA, Galons H, Villoutreix BO. The FAF-Drugs2 server: a multistep engine to prepare electronic chemical compound collections. Bioinformatics 2011;27:2018–20.

[63] Bhogal N, Grindon C, Combes R, Balls M. Toxicity testing: creating a revolution based on new technologies. Trends Biotechnol 2005;23:299–307.

[64] Modi S, Hughes M, Garrow A, White A. The value of in silico chemistry in the safety assessment of chemicals in the consumer goods and pharmaceutical industries. Drug Discov Today 2012;17(3-4):135–42.

[65] Yang Y, Adelstein SJ, Kassis AI. Target discovery from data mining approaches. Drug Discov Today 2009;14:147–54.

[66] Bertram L, Tanzi RE. Thirty years of Alzheimer’s disease genetics: the implications of systematic meta-analyses. Nat Rev Neurosci 2008;9:768–78.

[67] Fayyad U. Diving into databases. Database Program Design 1998;11(3):24–31.

[68] Baell JB. Observations on screening-based research and some concerning trends in the literature. Future Med Chem 2010;2:1529–46.

Social Aspects of Drug Discovery, Development and Commercialization Copyright © 2016 Elsevier Inc.

http://dx.doi.org/10.1016/B978-0-12-802220-7.00006-5 All rights reserved. 129