Skip to main content

Advertisement

Log in

Bigger data, collaborative tools and the future of predictive drug discovery

  • Published:
Journal of Computer-Aided Molecular Design Aims and scope Submit manuscript

Abstract

Over the past decade we have seen a growth in the provision of chemistry data and cheminformatics tools as either free websites or software as a service commercial offerings. These have transformed how we find molecule-related data and use such tools in our research. There have also been efforts to improve collaboration between researchers either openly or through secure transactions using commercial tools. A major challenge in the future will be how such databases and software approaches handle larger amounts of data as it accumulates from high throughput screening and enables the user to draw insights, enable predictions and move projects forward. We now discuss how information from some drug discovery datasets can be made more accessible and how privacy of data should not overwhelm the desire to share it at an appropriate time with collaborators. We also discuss additional software tools that could be made available and provide our thoughts on the future of predictive drug discovery in this age of big data. We use some examples from our own research on neglected diseases, collaborations, mobile apps and algorithm development to illustrate these ideas.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Villoutreix BO, Lagorce D, Labbe CM, Sperandio O, Miteva MA (2013) One hundred thousand mouse clicks down the road: selected online resources supporting drug discovery collected over a decade. Drug Discov Today 18:1081–1089

    Google Scholar 

  2. Pence HE, Williams AJ (2010) ChemSpider: an online chemical information resource. J Chem Educ 87:1123–1124

    CAS  Google Scholar 

  3. Ekins S, Williams AJ (2010) Precompetitive preclinical ADME/Tox data: set it free on the web to facilitate computational model building to assist drug development. Lab Chip 10:13–22

    CAS  Google Scholar 

  4. Ekins S, Andreyev S, Ryabov A, Kirillov E, Rakhmatulin EA, Bugrim A, Nikolskaya T (2005) Computational prediction of human drug metabolism. Expert Opin Drug Metab Toxicol 1:303–324

    CAS  Google Scholar 

  5. Ekins S, Andreyev S, Ryabov A, Kirillov E, Rakhmatulin EA, Sorokina S, Bugrim A, Nikolskaya T (2006) A combined approach to drug metabolism and toxicity assessment. Drug Metab Dispos 34:495–503

    CAS  Google Scholar 

  6. Ekins S, Bugrim A, Brovold L, Kirillov E, Nikolsky Y, Rakhmatulin EA, Sorokina S, Ryabov A, Serebryiskaya T, Melnikov A, Metz J, Nikolskaya T (2006) Algorithms for network analysis in systems-ADME/Tox using the MetaCore and MetaDrug platforms. Xenobiotica 36:877–901

    CAS  Google Scholar 

  7. Ekins S, Kirillov E, Rakhmatulin EA, Nikolskaya T (2005) A novel method for visualizing nuclear hormone receptor networks relevant to drug metabolism. Drug Metab Dispos 33:474–481

    CAS  Google Scholar 

  8. Ekins S, Nikolsky Y, Bugrim A, Kirillov E, Nikolskaya T (2007) Pathway mapping tools for analysis of high content data. Methods Mol Biol 356:319–350

    CAS  Google Scholar 

  9. Embrechts MJ, Ekins S (2007) Classification of metabolites with kernel-partial least squares (K-PLS). Drug Metab Dispos 35:325–327

    CAS  Google Scholar 

  10. Stranz DD, Miao S, Campbell S, Maydwell G, Ekins S (2008) Combined computational metabolite prediction and automated structure-based analysis of mass spectrometric data. Toxicol Mech Methods 18:243–250

    CAS  Google Scholar 

  11. Hohman M, Gregory K, Chibale K, Smith PJ, Ekins S, Bunin B (2009) Novel web-based tools combining chemistry informatics, biology and social networks for drug discovery. Drug Disc Today 14:261–270

    CAS  Google Scholar 

  12. Bost F, Jacobs RT, Kowalczyk P (2010) Informatics for neglected diseases collaborations. Curr Opin Drug Discov Dev 13:286–296

    CAS  Google Scholar 

  13. Bunin BA, Ekins S (2011) Alternative business models for drug discovery. Drug Disc Today 16:643–645

    Google Scholar 

  14. Sarker M, Talcott C, Madrid P, Chopra S, Bunin BA, Lamichhane G, Freundlich JS, Ekins S (2012) Combining cheminformatics methods and pathway analysis to identify molecules with whole-cell activity against Mycobacterium tuberculosis. Pharm Res 29:2115–2127

    CAS  Google Scholar 

  15. Ekins S, Hupcey MAZ, Williams AJ (2011) Collaborative computational technologies for biomedical research. Wiley, Hoboken, NJ

    Google Scholar 

  16. Ekins S, Hohman M, Bunin BA (2011). Pioneering use of the cloud for development of Collaborative Drug Discovery (CDD) Database. In: Ekins S, Hupcey MAZ, Williams AJ (eds) Collaborative computational technologies for biomedical research. Wiley, Hoboken

  17. Burrill GS (2010) 2010 4th annual CDD community meeting San Francisco. https://www.collaborativedrug.com/wp-content/uploads/2010/10/State-of-the-World_CDD-Community-Meeting_Oct-21-2010_EXCERPT.pdf

  18. Todd MH (2007) Open access and open source in chemistry. Chem Cent J 1:3

    Google Scholar 

  19. Ardal C, Rottingen JA (2012) Open source drug discovery in practice: a case study. PLoS Negl Trop Dis 6:e1827

    Google Scholar 

  20. Anon European Lead Factory. http://www.europeanleadfactory.eu/

  21. Anon Elixir. http://www.elixir-europe.org/

  22. Li Q, Cheng T, Wang Y, Bryant SH (2010) PubChem as a public resource for drug discovery. Drug Discov Today 15:1052–1057

    CAS  Google Scholar 

  23. Williams AJ, Ekins S (2011) A quality alert and call for improved curation of public chemistry databases. Drug Disc Today 16:747–750

    CAS  Google Scholar 

  24. Williams AJ, Ekins S, Tkachenko V (2012) Towards a gold standard: regarding quality in public domain chemistry databases and approaches to improving the situation. Drug Disc Today 17:685–701

    CAS  Google Scholar 

  25. Hotez PJ, Molyneux DH, Fenwick A, Kumaresan J, Sachs SE, Sachs JD, Savioli L (2007) Control of neglected tropical diseases. N Engl J Med 357:1018–1027

    CAS  Google Scholar 

  26. Guiguemde WA, Shelat AA, Bouck D, Duffy S, Crowther GJ, Davis PH, Smithson DC, Connelly M, Clark J, Zhu F, Jimenez-Diaz MB, Martinez MS, Wilson EB, Tripathi AK, Gut J, Sharlow ER, Bathurst I, El Mazouni F, Fowble JW, Forquer I, McGinley PL, Castro S, Angulo-Barturen I, Ferrer S, Rosenthal PJ, Derisi JL, Sullivan DJ, Lazo JS, Roos DS, Riscoe MK, Phillips MA, Rathod PK, Van Voorhis WC, Avery VM, Guy RK (2010) Chemical genetics of Plasmodium falciparum. Nature 465:311–315

    CAS  Google Scholar 

  27. Ribeiro I, Sevcsik AM, Alves F, Diap G, Don R, Harhay MO, Chang S, Pecoul B (2009) New, improved treatments for Chagas disease: from the R&D pipeline to the patients. PLoS Negl Trop Dis 3:e484

    Google Scholar 

  28. Bettiol E, Samanovic M, Murkin AS, Raper J, Buckner F, Rodriguez A (2009) Identification of three classes of heteroaromatic compounds with activity against intracellular Trypanosoma cruzi by chemical library screening. PLoS Negl Trop Dis 3:e384

    Google Scholar 

  29. Ponder EL, Freundlich JS, Sarker M, Ekins S (2014) Computational models for neglected diseases: gaps and opportunities. Pharm Res 31:271–277

    CAS  Google Scholar 

  30. Paul SM, Mytelka DS, Dunwiddie CT, Persinger CC, Munos BH, Lindborg SR, Schacht AL (2010) How to improve R&D productivity: the pharmaceutical industry’s grand challenge. Nat Rev Drug Discov 9:203–214

    CAS  Google Scholar 

  31. Ballell L, Bates RH, Young RJ, Alvarez-Gomez D, Alvarez-Ruiz E, Barroso V, Blanco D, Crespo B, Escribano J, Gonzalez R, Lozano S, Huss S, Santos-Villarejo A, Martin-Plaza JJ, Mendoza A, Rebollo-Lopez MJ, Remuinan-Blanco M, Lavandera JL, Perez-Herran E, Gamo-Benito FJ, Garcia-Bustos JF, Barros D, Castro JP, Cammack N (2013) Fueling open-source drug discovery: 177 small-molecule leads against tuberculosis. ChemMedChem 8:313–321

    CAS  Google Scholar 

  32. Gamo F-J, Sanz LM, Vidal J, de Cozar C, Alvarez E, Lavandera J-L, Vanderwall DE, Green DVS, Kumar V, Hasan S, Brown JR, Peishoff CE, Cardon LR, Garcia-Bustos JF (2010) Thousands of chemical starting points for antimalarial lead identification. Nature 465:305–310

    CAS  Google Scholar 

  33. Anon TB Alliance Preclinical Pipeline. http://www.tballiance.org/downloads/Pipeline/TBA%20Pipeline%20Q1%202014%282%29%20%28DA%29.pdf

  34. Anon TB Alliance Clinical Pipeline. http://www.tballiance.org/portfolio/

  35. Ekins S, Pottorf R, Reynolds RC, Williams AJ, Clark AM, Freundlich JS (2014) Looking back to the future: predicting in vivo efficacy of small molecules versus Mycobacterium tuberculosis. J Chem Inf Model 54:1070–1082

    CAS  Google Scholar 

  36. Ballell L, Field RA, Duncan K, Young RJ (2005) New small-molecule synthetic antimycobacterials. Antimicrob Agents Chemother 49:2153–2163

    CAS  Google Scholar 

  37. Reynolds RC, Ananthan S, Faaleolea E, Hobrath JV, Kwong CD, Maddox C, Rasmussen L, Sosa MI, Thammasuvimol E, White EL, Zhang W, Secrist JA, 3rd (2011) High throughput screening of a library based on kinase inhibitor scaffolds against Mycobacterium tuberculosis H37Rv. Tuberculosis (Edinb) 92:72–83

  38. Maddry JA, Ananthan S, Goldman RC, Hobrath JV, Kwong CD, Maddox C, Rasmussen L, Reynolds RC, Secrist JA 3rd, Sosa MI, White EL, Zhang W (2009) Antituberculosis activity of the molecular libraries screening center network library. Tuberculosis (Edinb) 89:354–363

    CAS  Google Scholar 

  39. Ananthan S, Faaleolea ER, Goldman RC, Hobrath JV, Kwong CD, Laughon BE, Maddry JA, Mehta A, Rasmussen L, Reynolds RC, Secrist JA 3rd, Shindo N, Showe DN, Sosa MI, Suling WJ, White EL (2009) High-throughput screening for inhibitors of Mycobacterium tuberculosis H37Rv. Tuberculosis (Edinb) 89:334–353

    CAS  Google Scholar 

  40. Ekins S, Freundlich JS, Hobrath JV, White EL, Reynolds RC (2014) Combining computational methods for hit to lead optimization in Mycobacterium tuberculosis drug discovery. Pharm Res 31:414–435

    CAS  Google Scholar 

  41. Reynolds RC, Ananthan S, Faaleolea E, Hobrath JV, Kwong CD, Maddox C, Rasmussen L, Sosa MI, Thammasuvimol E, White EL, Zhang W, Secrist JA 3rd (2012) High throughput screening of a library based on kinase inhibitor scaffolds against Mycobacterium tuberculosis H37Rv. Tuberculosis (Edinb) 92:72–83

    CAS  Google Scholar 

  42. Prakash O, Ghosh I (2006) Developing an antituberculosis compounds database and data mining in the search of a motif responsible for the activity of a diverse class of antituberculosis agents. J Chem Inf Model 46:17–23

    CAS  Google Scholar 

  43. Garcia-Garcia A, Galvez J, de Julian-Ortiz JV, Garcia-Domenech R, Munoz C, Guna R, Borras R (2005) Search of chemical scaffolds for novel antituberculosis agents. J Biomol Screen 10:206–214

    CAS  Google Scholar 

  44. Planche AS, Scotti MT, Lopez AG, de Paulo Emerenciano V, Perez EM, Uriarte E (2009) Design of novel antituberculosis compounds using graph-theoretical and substructural approaches. Mol Divers 13:445–458

    CAS  Google Scholar 

  45. Sundaramurthi JC, Brindha S, Reddy TB, Hanna LE (2012) Informatics resources for tuberculosis: towards drug discovery. Tuberculosis (Edinb) 92:133–138

    Google Scholar 

  46. Ekins S, Freundlich JS, Choi I, Sarker M, Talcott C (2011) Computational databases, pathway and cheminformatics tools for tuberculosis drug discovery. Trends Microbiol 19:65–74

    CAS  Google Scholar 

  47. Ekins S, Freundlich JS (2013) Computational models for tuberculosis drug discovery. Methods Mol Biol 993:245–262

    CAS  Google Scholar 

  48. Ekins S, Bradford J, Dole K, Spektor A, Gregory K, Blondeau D, Hohman M, Bunin B (2010) A collaborative database and computational models for tuberculosis drug discovery. Mol BioSyst 6:840–851

    CAS  Google Scholar 

  49. Ekins S, Freundlich JS (2011) Validating new tuberculosis computational models with public whole cell screening aerobic activity datasets. Pharm Res 28:1859–1869

    CAS  Google Scholar 

  50. Ekins S, Kaneko T, Lipinksi CA, Bradford J, Dole K, Spektor A, Gregory K, Blondeau D, Ernst S, Yang J, Goncharoff N, Hohman M, Bunin B (2010) Analysis and hit filtering of a very large library of compounds screened against Mycobacterium tuberculosis. Mol BioSyst 6:2316–2324

    CAS  Google Scholar 

  51. Ekins S, Reynolds RC, Franzblau SG, Wan B, Freundlich JS, Bunin BA (2013) Enhancing hit identification in Mycobacterium tuberculosis drug discovery using validated dual-event Bayesian models. PLOS One 8:e63240

    CAS  Google Scholar 

  52. Ekins S, Reynolds R, Kim H, Koo M-S, Ekonomidis M, Talaue M, Paget SD, Woolhiser LK, Lenaerts AJ, Bunin BA, Connell N, Freundlich JS (2013) Bayesian models leveraging bioactivity and cytotoxicity information for drug discovery. Chem Biol 20:370–378

    CAS  Google Scholar 

  53. Ekins S, Freundlich JS, Reynolds RC (2014) Are bigger datasets better for machine learning? Fusing single-point and dual-event dose response data for Mycobacterium tuberculosis (submitted)

  54. Anderson JW, Sarantakis D, Terpinski J, Kumar TR, Tsai HC, Kuo M, Ager AL, Jacobs WR Jr, Schiehser GA, Ekins S, Sacchettini JC, Jacobus DP, Fidock DA, Freundlich JS (2012) Novel diaryl ureas with efficacy in a mouse model of malaria. Bioorg Med Chem Lett 23:1022–1025

    Google Scholar 

  55. Alvarez G, Martinez J, Aguirre-Lopez B, Cabrera N, Perez-Diaz L, Gomez-Puyou MT, Gomez-Puyou A, Perez-Montfort R, Garat B, Merlino A, Gonzalez M, Cerecetto H (2012) New chemotypes as Trypanosoma cruzi triosephosphate isomerase inhibitors: a deeper insight into the mechanism of inhibition. J Enzyme Inhib Med Chem 29:198–204

  56. Pires DE, de Melo-Minardi RC, da Silveira CH, Campos FF, Meira W Jr (2013) aCSM: noise-free graph-based signatures to large-scale receptor-based ligand prediction. Bioinformatics 29:855–861

    CAS  Google Scholar 

  57. Gunatilleke SS, Calvet CM, Johnston JB, Chen CK, Erenburg G, Gut J, Engel JC, Ang KK, Mulvaney J, Chen S, Arkin MR, McKerrow JH, Podust LM (2012) Diverse inhibitor chemotypes targeting Trypanosoma cruzi CYP51. PLoS Negl Trop Dis 6:e1736

    CAS  Google Scholar 

  58. Schamberger J, Grimm M, Steinmeyer A, Hillisch A (2011) Rendezvous in chemical space? Comparing the small molecule compound libraries of Bayer and Schering. Drug Discov Today 16:636–641

    CAS  Google Scholar 

  59. Kogej T, Blomberg N, Greasley PJ, Mundt S, Vainio MJ, Schamberger J, Schmidt G, Huser J (2012) Big pharma screening collections: more of the same or unique libraries? The AstraZeneca-Bayer pharma AG case. Drug Discov Today 18:1014–1024

  60. Tu M, Rai BK, Mathiowetz AM, Didiuk M, Pfefferkorn JA, Guzman-Perez A, Benbow J, Guimaraes CR, Mente S, Hayward MM, Liras S (2012) Exploring aromatic chemical space with NEAT: novel and electronically equivalent aromatic template. J Chem Inf Model 52:1114–1123

    CAS  Google Scholar 

  61. Posner BA, Xi H, Mills JE (2009) Enhanced HTS hit selection via a local hit rate analysis. J Chem Inf Model 49:2202–2210

    CAS  Google Scholar 

  62. Gunter B, Brideau C, Pikounis B, Liaw A (2003) Statistical and graphical methods for quality control determination of high-throughput screening data. J Biomol Screen 8:624–633

    Google Scholar 

  63. Varin T, Gubler H, Parker CN, Zhang JH, Raman P, Ertl P, Schuffenhauer A (2010) Compound set enrichment: a novel approach to analysis of primary HTS data. J Chem Inf Model 50:2067–2078

    CAS  Google Scholar 

  64. Swamidass SJ, Calhoun BT, Bittker JA, Bodycombe NE, Clemons PA (2011) Enhancing the rate of scaffold discovery with diversity-oriented prioritization. Bioinformatics 27:2271–2278

    CAS  Google Scholar 

  65. Swamidass SJ, Calhoun BT, Bittker JA, Bodycombe NE, Clemons PA (2011) Utility-aware screening with clique-oriented prioritization. J Chem Inf Model 52:29–37

    Google Scholar 

  66. Swamidass SJ (2013) Using economic optimization to design high-throughput screens. Future Med Chem 5:9–11

    CAS  Google Scholar 

  67. Makarenkov V, Kevorkov D, Zentilli P, Gagarin A, Malo N, Nadon R (2006) HTS-Corrector: software for the statistical analysis and correction of experimental high-throughput screening data. Bioinformatics 22:1408–1409

    CAS  Google Scholar 

  68. Makarenkov V, Zentilli P, Kevorkov D, Gagarin A, Malo N, Nadon R (2007) An efficient method for the detection and elimination of systematic error in high-throughput screening. Bioinformatics 23:1648–1657

    CAS  Google Scholar 

  69. Seiler KP, George GA, Happ MP, Bodycombe NE, Carrinski HA, Norton S, Brudz S, Sullivan JP, Muhlich J, Serrano M, Ferraiolo P, Tolliday NJ, Schreiber SL, Clemons PA (2008) ChemBank: a small-molecule screening and cheminformatics resource database. Nucleic Acids Res 36:D351–D359

    CAS  Google Scholar 

  70. Calhoun BT, Browning MR, Chen BR, Bittker JA, Swamidass SJ (2012) Automatically detecting workflows in PubChem. J Biomol Screen 17:1071–1079

    Google Scholar 

  71. Browning MR, Calhoun BT, Swamidass SJ (2013) Managing missing measurements in small-molecule screens. J Comput Aided Mol Des 27:469–478

  72. Schuffenhauer A, Ertl P, Roggo S, Wetzel S, Koch MA, Waldmann H (2007) The scaffold tree-visualization of the scaffold universe by hierarchical scaffold classification. J Chem Inf Model 47:47–58

    CAS  Google Scholar 

  73. Dimova D, Wawer M, Wassermann AM, Bajorath J (2011) Design of multitarget activity landscapes that capture hierarchical activity cliff distributions. J Chem Inf Model 51:258–266

    CAS  Google Scholar 

  74. Wassermann AM, Bajorath J (2012) Directed R-group combination graph: a methodology to uncover structure-activity relationship patterns in a series of analogues. J Med Chem 55:1215–1226

    CAS  Google Scholar 

  75. Howells J, Gagliardi D, Malik K (2012) Sourcing knowledge: R&D outsourcing in UK pharmaceuticals. Int J Tech Man 59:139–161

    Google Scholar 

  76. Fox S, Farr-Jones S, Sopchak L, Boggs A, Nicely HW, Khoury R, Biros M (2006) High-throughput screening: update on practices and success. J Biomol Screen 11:864–869

    Google Scholar 

  77. McGee J (2012) Outsourcing and contract services. J Biomol Screen 17:1379–1381

    Google Scholar 

  78. Ekins S, Olechno J, Williams AJ (2013) Dispensing processes impact apparent biological activity as determined by computational and statistical analyses. PLoS One 8:e62325

    CAS  Google Scholar 

  79. Bradley D (2005) Share and share alike. Nat Rev Drug Discov 4:180

    CAS  Google Scholar 

  80. Masek BB, Shen L, Smith KM, Pearlman RS (2008) Sharing chemical information without sharing chemical structure. J Chem Inf Model 48:256–261

    CAS  Google Scholar 

  81. Balaban A (2005) Can topological indices transmit information on properties but not on structures? J Comput Aided Mol Des 19:651–660

    CAS  Google Scholar 

  82. Bologa C, Allu TK, Olah M, Kappler MA, Oprea TI (2005) Descriptor collision and confusion: toward the design of descriptors to mask chemical structures. J Comput Aided Mol Des 19:625–635

    CAS  Google Scholar 

  83. Clement OO, Guner OF (2005) Possibilities for transfer of relevant data without revealing structural information. J Comput Aided Mol Des 19:731–738

    CAS  Google Scholar 

  84. Filimonov D, Poroikov V (2005) Why relevant chemical information cannot be exchanged without disclosing structures. J Comput Aided Mol Des 19:705–713

    CAS  Google Scholar 

  85. Kaiser D, Zdrazil B, Ecker GF (2005) Similarity-based descriptors (SIBAR): a tool for safe exchange of chemical information? J Comput Aided Mol Des 19:687–692

    CAS  Google Scholar 

  86. Trepalin S, Osadchiy N (2005) The centroidal algorithm in molecular similarity and diversity calculations on confidential datasets. J Comput Aided Mol Des 19:715–729

    CAS  Google Scholar 

  87. Tetko IV, Abagyan R, Oprea TI (2005) Surrogate data: a secure way to share corporate data. J Comput Aided Mol Des 19:749–764

    CAS  Google Scholar 

  88. Karr AF, Feng J, Lin X, Sanil AP, Young SS, Reiter JP (2005) Secure analysis of distributed chemical databases without data integration. J Comput Aided Mol Des 19:739–747

    CAS  Google Scholar 

  89. Faulon JL, Brown WM, Martin S (2005) Reverse engineering chemical structures from molecular descriptors: how many solutions? J Comput Aided Mol Des 19:637–650

    CAS  Google Scholar 

  90. Matlock M, Swamidass SJ (2014) Sharing chemical relationships does not reveal structures. J Chem Inf Model 54:37–48

    CAS  Google Scholar 

  91. Swamidass SJ, Matlock M, Rozenblit L (2013) When should we share? Securely measuring he overlap between private datasets (submitted)

  92. Huang Y, Shen C, Evans D, Katz J, Shelat A (2011) Efficient secure computation with garbled circuits. In: Jajodia S, Mazumdar C (eds) Information systems security. Springer, Heidelberg, pp 28–48

  93. Kuzu M, Kantarcioglu M, Durham EA, Toth C, Malin B (2013) A practical approach to achieve private medical record linkage in light of public resources. J Am Med Inform Assoc 20:285–292

    Google Scholar 

  94. Johnson SB, Whitney G, McAuliffe M, Wang H, McCreedy E, Rozenblit L, Evans CC (2010) Using global unique identifiers to link autism collections. J Am Med Inform Assoc 17:689–695

    Google Scholar 

  95. Warner DJ, Griffen EJ, St-Gallay SA (2010) WizePairZ: a novel algorithm to identify, encode, and exploit matched molecular pairs with unspecified cores in medicinal chemistry. J Chem Inf Model 50:1350–1357

    CAS  Google Scholar 

  96. Anon (2013) Roche and AstraZeneca launch medicinal chemistry data-sharing consortium to further accelerate drug discovery. http://www.astrazeneca.com/Research/news/Article/260613-roche-and-astrazeneca-launch-medicinal-chemistry-datasha

  97. Zaretzki J, Matlock M, Swamidass SJ (2013) XenoSite: accurately predicting CYP-mediated sites of metabolism with neural networks. J Chem Inf Model 53:3373–3383

    CAS  Google Scholar 

  98. Ekins S (2014) Progress in computational toxicology. J Pharmacol Toxicol Methods 69:115–140

    CAS  Google Scholar 

  99. Aruoja V, Moosus M, Kahru A, Sihtmae M, Maran U (2014) Measurement of baseline toxicity and QSAR analysis of 50 non-polar and 58 polar narcotic chemicals for the alga Pseudokirchneriella subcapitata. Chemosphere 96:23–32

    CAS  Google Scholar 

  100. Sushko I, Novotarskyi S, Korner R, Pandey AK, Rupp M, Teetz W, Brandmaier S, Abdelaziz A, Prokopenko VV, Tanchuk VY, Todeschini R, Varnek A, Marcou G, Ertl P, Potemkin V, Grishina M, Gasteiger J, Schwab C, Baskin II, Palyulin VA, Radchenko EV, Welsh WJ, Kholodovych V, Chekmarev D, Cherkasov A, Aires-de-Sousa J, Zhang QY, Bender A, Nigsch F, Patiny L, Williams A, Tkachenko V, Tetko IV (2011) Online chemical modeling environment (OCHEM): web platform for data storage, model development and publishing of chemical information. J Comput Aided Mol Des 25:533–554

    CAS  Google Scholar 

  101. Walker T, Grulke CM, Pozefsky D, Tropsha A (2010) Chembench: a cheminformatics workbench. Bioinformatics 26:3000–3001

    CAS  Google Scholar 

  102. Gupta RR, Gifford EM, Liston T, Waller CL, Bunin B, Ekins S (2010) Using open source computational tools for predicting human metabolic stability and additional ADME/TOX properties. Drug Metab Dispos 38:2083–2090

    CAS  Google Scholar 

  103. Spjuth O, Willighagen EL, Guha R, Eklund M, Wikberg JE (2010) Towards interoperable and reproducible QSAR analyses: exchange of datasets. J Cheminform 2:5

    Google Scholar 

  104. Williams AJ, Ekins S, Spjuth O, Willighagen EL (2012) Accessing, using, and creating chemical property databases for computational toxicology modeling. Methods Mol Biol 929:221–241

    CAS  Google Scholar 

  105. Ekins S, Gupta RR, Gifford E, Bunin BA, Waller CL (2010) Chemical space: missing pieces in cheminformatics. Pharm Res 27:2035–2039

    CAS  Google Scholar 

  106. Guha R, Spjuth O, Willighagen EL (2011) Collaborative cheminformatics applications. In: Ekins S, Hupcey MAZ, Williams AJ (eds) Collaborative computational technologies for biomedical research. Wiley, Hoboken, pp 399–422

  107. Spjuth O, Carlsson L, Alvarsson J, Georgiev V, Willighagen E, Eklund M (2012) Open source drug discovery with bioclipse. Curr Top Med Chem 12:1980–1986

    CAS  Google Scholar 

  108. Williams AJ, Harland L, Groth P, Pettifer S, Chichester C, Willighagen EL, Evelo CT, Blomberg N, Ecker G, Goble C, Mons B (2012) Open PHACTS: semantic interoperability for drug discovery. Drug Disc Today 17:1188–1198

  109. Ekins S, Bugrim A, Nikolsky Y, Nikolskaya T (2005) Systems Biology: Applications in drug discovery. In: Gad (S) (ed) Drug discovery handbook. Wiley, New York, pp 123–183

  110. Anon ACS GCI Pharmaceutical Roundtable. http://portal.acs.org/portal/acs/corg/content?_nfpb=true&_pageLabel=PP_TRANSITIONMAIN&node_id=1422&use_sec=false&sec_url_var=region1&__uuid=46aca9b6-a985-42cd-a534-7d6cabf892a7

  111. Ekins S, Clark AM, Williams AJ (2013) Incorporating green chemistry concepts into mobile chemistry applications and their potential uses. ACS Sustain Chem Eng 1:8–13

    CAS  Google Scholar 

  112. Ekins S, Casey AC, Roberts D, Parish T, Bunin BA (2013) Bayesian models for screening and TB mobile for target inference with Mycobacterium tuberculosis. Tuberculosis (Edinb) 94:162–169

  113. Ekins S, Clark AM, Sarker M (2013) TB Mobile: a mobile app for anti-tuberculosis molecules with known targets. J Cheminform 5:13

    CAS  Google Scholar 

  114. Clark AM, Sarker M, Ekins S (2014) New target predictions and visualization tools incorporating open source molecular fingerprints for TB Mobile 2.0 (submitted)

  115. Ekins S, Clark AM, Williams AJ (2012) Open drug discovery teams: a chemistry mobile app for collaboration. Mol Inform 31:585–597

    CAS  Google Scholar 

  116. Clark AM, Ekins S, Williams AJ (2012) Redefining cheminformatics with intuitive collaborative mobile apps. Mol Inf 31:569–584

    CAS  Google Scholar 

  117. Clark AM, Williams AJ, Ekins S (2013) Cheminformatics workflows using mobile apps. Chem Bio Inf J 13:1–18

    Google Scholar 

  118. Ekins S, Waller CL, Bradley MP, Clark AM, Williams AJ (2013) Four disruptive strategies for removing drug discovery bottlenecks. Drug Disc Today 18:265–271

    Google Scholar 

  119. Anon Assay Depot. https://www.assaydepot.com/

  120. Anon Science Exchange. https://www.scienceexchange.com/

  121. Anon Euretos. http://euretos.com/brain

  122. van Haagen HH, t Hoen PA, Mons B, Schultes EA (2013) Generic information can retrieve known biological associations: implications for biomedical knowledge discovery. PLoS One 8:e78665

    Google Scholar 

  123. Ekins S, Polli JE, Swaan PW, Wright SH (2012) Computational modeling to accelerate the identification of substrates and inhibitors for transporters that affect drug disposition. Clin Pharmacol Ther 92:661–665

    CAS  Google Scholar 

  124. Beaulieu CL, Ekins S, Samuels M, Boycott KM, MacKenzie A (2012) Towards the development of a generalizable pre-clinical research pathway for orphan disease therapy. Orphanet J Rare Dis 7:39

    Google Scholar 

  125. Wood J, Sames L, Moore A, Ekins S (2013) Multifaceted roles of ultra-rare and rare disease patients/parents in drug discovery. Drug Discov Today 18:1043–1051

    Google Scholar 

  126. Williams AJ, Wilbanks J, Ekins S (2012) Why open drug discovery needs four simple rules for licensing data and models. PLoS Comput Biol 8:e1002706

    CAS  Google Scholar 

  127. Houlton S (2014) Science at your fingerprints. Chemistry World 27th Feb

  128. Ekins S, Clark AM, Williams AJ (2012) Open drug discovery teams: a chemistry mobile app for collaboration. Mol Inf 31:585–597

    CAS  Google Scholar 

Download references

Acknowledgments

S.E. gratefully acknowledges colleagues at CDD, Dr. Joel S. Freundlich (Rutgers), Dr. Malabika Sarker (SRI) and Dr. Katalin Nadassy (Accelrys) for valuable discussions and assistance in developing some of the projects discussed. S.E. acknowledges that the Bayesian models were developed with support from Award Number R43 LM011152-01 “Biocomputation across distributed private datasets to enhance drug discovery” from the National Library of Medicine. TB Mobile and the associated datasets was made possible with funding from Award Number 2R42AI088893-02 “Identification of novel therapeutics for tuberculosis combining cheminformatics, diverse databases and logic based pathway analysis” from the National Institutes of Allergy and Infectious Diseases.

Conflict of interest

N.L. is an employee and S.E. is a consultant for CDD Inc. S.E. is on the advisory board for Assay Depot. A.J.W. is an employee of the Royal Society of Chemistry. A.M.C. is the founder of Molecular Materials Informatics and a consultant for CDD.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sean Ekins.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ekins, S., Clark, A.M., Swamidass, S.J. et al. Bigger data, collaborative tools and the future of predictive drug discovery. J Comput Aided Mol Des 28, 997–1008 (2014). https://doi.org/10.1007/s10822-014-9762-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10822-014-9762-y

Keywords

Navigation