Bioinformatics Tools for Pharmaceutical Drug Product Development

Authors

  • Johra Khan Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, 11952, Al Majmaah, Saudi Arabia. E-mail address: j.khan@gmail.com; Health and Basic Sciences Research Center, Majmaah University, Al Majmaah 11952, Saudi Arabia
  • Rajeev K. Singla Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China https://orcid.org/0000-0002-3353-7897

DOI:

https://doi.org/10.35652/IGJPS.2022.12037

Keywords:

Drug Discovery, Bioinformatics, Cheminformatics, Drug Development

Abstract

Drug discovery and production is a long and expensive process which starts with target identification followed by validation of targets to lead optimization, taking years to develop a drug which sometime false to reach marked resulting in loss of time, effort, and huge amount of money. Bioinformatics tools are becoming more and more important in drug product development. Repurposing large amount of data needs to be exploited and generated from genomics, epigenetics, cistromic, proteomics, transcriptomics, ribosomal profiling, and genomic based studies of drug targets. Bioinformatics analysis and data mining are effective tools to explore big series of biological and biomedical data, however the advance tools are often found difficult to understand making their use limited to difficult to access by the researchers working in drug discovery. In this review we focused on systematically presenting the different tools used for drug target identification and product development. The tools are broadly classified according to disease based computational tools, gene based tools, and web based tools and ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) study for drug repurposing. The focus was on the basic principle of these tools functioning, uses and limitations in drug target identification, validation, data analysis, comparison with other similar tools in target analysis. © 2022 Caproslaxy Media. All rights reserved.

Metrics

Metrics Loading ...

References

Power, A., A.C. Berger, and G.S. Ginsburg, Genomics-enabled drug repositioning and repurposing: insights from an IOM Roundtable activity. Jama, 2014. 311(20): p. 2063-2064.

Scannell, J.W., et al., Diagnosing the decline in pharmaceutical R&D efficiency. Nature reviews Drug discovery, 2012. 11(3): p. 191-200.

Padhy, B. and Y. Gupta, Drug repositioning: re-investigating existing drugs for new therapeutic indications. Journal of postgraduate medicine, 2011. 57(2): p. 153.

Buchan, N.S., et al., The role of translational bioinformatics in drug discovery. Drug discovery today, 2011. 16(9-10): p. 426-434.

Van Driel, M.A. and H.G. Brunner, Bioinformatics methods for identifying candidate disease genes. Human genomics, 2006. 2(6): p. 1-4.

Josset, L., et al., Gene expression signature-based screening identifies new broadly effective influenza a antivirals. PloS one, 2010. 5(10): p. e13169.

Shameer, K., et al., Systematic analyses of drugs and disease indications in RepurposeDB reveal pharmacological, biological and epidemiological factors influencing drug repositioning. Briefings in bioinformatics, 2018. 19(4): p. 656-678.

Li, J., et al., A survey of current trends in computational drug repositioning. Briefings in bioinformatics, 2016. 17(1): p. 2-12.

Dai, Y.-F. and X.-M. Zhao, A survey on the computational approaches to identify drug targets in the postgenomic era. BioMed research international, 2015. 2015.

March-Vila, E., et al., On the integration of in silico drug design methods for drug repurposing. Frontiers in pharmacology, 2017. 8: p. 298.

Corsello, S.M., et al., The Drug Repurposing Hub: a next-generation drug library and information resource. Nature medicine, 2017. 23(4): p. 405-408.

Wang, X., et al., PharmMapper 2017 update: a web server for potential drug target identification with a comprehensive target pharmacophore database. Nucleic acids research, 2017. 45(W1): p. W356-W360.

Li, C., et al., Applications of three-dimensional printing in surgery. Surgical innovation, 2017. 24(1): p. 82-88.

Jacobo, O.M., et al., Three-dimensional printing modeling: application in maxillofacial and hand fractures and resident training. European Journal of Plastic Surgery, 2018. 41(2): p. 137-146.

Papanikolaw, J., Bioinformatics emerges as key technology for developing new drugs. Chemical Market Reporter; May 24, 1999; 255, 21; ABI/INFORM Global pg, 1999. 22.

Wittenberger, T., H.C. Schaller, and S. Hellebrand, An expressed sequence tag (EST) data mining strategy succeeding in the discovery of new G-protein coupled receptors. Journal of molecular biology, 2001. 307(3): p. 799-813.

Marvanova, M., et al., Synexpression analysis of ESTs in the rat brain reveals distinct patterns and potential drug targets. Molecular brain research, 2002. 104(2): p. 176-183.

Nayal, M. and B. Honig, On the nature of cavities on protein surfaces: application to the identification of drug‐binding sites. Proteins: Structure, Function, and Bioinformatics, 2006. 63(4): p. 892-906.

Mehlin, C., et al., Heterologous expression of proteins from Plasmodium falciparum: results from 1000 genes. Molecular and biochemical parasitology, 2006. 148(2): p. 144-160.

Dong, Y., et al., Anopheles gambiae immune responses to human and rodent Plasmodium parasite species. PLoS pathogens, 2006. 2(6): p. e52.

Dudley, J.T., T. Deshpande, and A.J. Butte, Exploiting drug–disease relationships for computational drug repositioning. Briefings in bioinformatics, 2011. 12(4): p. 303-311.

Veber, D.F., et al., Molecular properties that influence the oral bioavailability of drug candidates. Journal of medicinal chemistry, 2002. 45(12): p. 2615-2623.

Spangenberg, T., et al., The open access malaria box: a drug discovery catalyst for neglected diseases. PloS one, 2013. 8(6): p. e62906.

Garofalo, A.W., Patents targeting γ-secretase inhibition and modulation for the treatment of Alzheimer's disease: 2004–2008. Expert Opinion on Therapeutic Patents, 2008. 18(7): p. 693-703.

Ou-Yang, S.-s., et al., Computational drug discovery. Acta Pharmacologica Sinica, 2012. 33(9): p. 1131-1140.

Sliwoski, G., et al., Computational methods in drug discovery. Pharmacological reviews, 2014. 66(1): p. 334-395.

Schaduangrat, N., et al., Towards reproducible computational drug discovery. Journal of cheminformatics, 2020. 12(1): p. 1-30.

Gong, J., et al., ChemMapper: a versatile web server for exploring pharmacology and chemical structure association based on molecular 3D similarity method. Bioinformatics, 2013. 29(14): p. 1827-1829.

Liu, X., H. Jiang, and H. Li, SHAFTS: a hybrid approach for 3D molecular similarity calculation. 1. Method and assessment of virtual screening. Journal of chemical information and modeling, 2011. 51(9): p. 2372-2385.

Lu, W., et al., SHAFTS: a hybrid approach for 3D molecular similarity calculation. 2. Prospective case study in the discovery of diverse p90 ribosomal S6 protein kinase 2 inhibitors to suppress cell migration. Journal of medicinal chemistry, 2011. 54(10): p. 3564-3574.

Liu, X., et al., PharmMapper server: a web server for potential drug target identification using pharmacophore mapping approach. Nucleic acids research, 2010. 38(suppl_2): p. W609-W614.

Taboureau, O., et al., ChemProt: a disease chemical biology database. Nucleic acids research, 2010. 39(suppl_1): p. D367-D372.

Kim Kjærulff, S., et al., ChemProt-2.0: visual navigation in a disease chemical biology database. Nucleic acids research, 2012. 41(D1): p. D464-D469.

Kringelum, J., et al., ChemProt-3.0: a global chemical biology diseases mapping. Database, 2016. 2016.

Rose, P.W., et al., The RCSB protein data bank: integrative view of protein, gene and 3D structural information. Nucleic acids research, 2016: p. gkw1000.

Rose, P.W., et al., The RCSB Protein Data Bank: views of structural biology for basic and applied research and education. Nucleic acids research, 2015. 43(D1): p. D345-D356.

Pagadala, N.S., K. Syed, and J. Tuszynski, Software for molecular docking: a review. Biophysical reviews, 2017. 9(2): p. 91-102.

Hernández-Santoyo, A., et al., Protein-protein and protein-ligand docking. Protein engineering-technology and application, 2013: p. 63-81.

Zheng, H., et al., X-ray crystallography over the past decade for novel drug discovery–where are we heading next? Expert opinion on drug discovery, 2015. 10(9): p. 975-989.

Eweas, A.F., I.A. Maghrabi, and A.I. Namarneh, Advances in molecular modeling and docking as a tool for modern drug discovery. Der Pharma Chemica, 2014. 6(6): p. 211-228.

del Carmen Fernández-Alonso, M., et al., Protein-carbohydrate interactions studied by NMR: from molecular recognition to drug design. Current Protein and Peptide Science, 2012. 13(8): p. 816-830.

Lipfert, J. and S. Doniach, Small-angle X-ray scattering from RNA, proteins, and protein complexes. Annu. Rev. Biophys. Biomol. Struct., 2007. 36: p. 307-327.

Schiffelers, R.M., et al., Effects of treatment with small interfering RNA on joint inflammation in mice with collagen‐induced arthritis. Arthritis & Rheumatism, 2005. 52(4): p. 1314-1318.

Xu, J., Strategic Research Institute--first international siRNA conference. Prospect for new therapeutics and commercial opportunities for pharma and biotech. 24-25 March 2003, LaJolla, CA, USA. IDrugs: the investigational drugs journal, 2003. 6(5): p. 449-450.

Xie, F.Y., M.C. Woodle, and P.Y. Lu, Harnessing in vivo siRNA delivery for drug discovery and therapeutic development. Drug discovery today, 2006. 11(1-2): p. 67-73.

Iorns, E., et al., A new mouse model for the study of human breast cancer metastasis. PloS one, 2012. 7(10): p. e47995.

Gondi, C.S. and J.S. Rao, Concepts in in vivo siRNA delivery for cancer therapy. Journal of cellular physiology, 2009. 220(2): p. 285-291.

Gamo, F.-J., et al., Thousands of chemical starting points for antimalarial lead identification. Nature, 2010. 465(7296): p. 305-310.

Singh, J., et al., Application of genetic algorithms to combinatorial synthesis: A computational approach to lead identification and lead optimization. Journal of the American Chemical Society, 1996. 118(7): p. 1669-1676.

Nicola, G., T. Liu, and M.K. Gilson, Public domain databases for medicinal chemistry. Journal of medicinal chemistry, 2012. 55(16): p. 6987-7002.

Wishart, D.S., et al., DrugBank: a knowledgebase for drugs, drug actions and drug targets. Nucleic acids research, 2008. 36(suppl_1): p. D901-D906.

Thorn, C.F., T.E. Klein, and R.B. Altman, PharmGKB: the pharmacogenomics knowledge base, in Pharmacogenomics. 2013, Springer. p. 311-320.

Zhao, Y., et al., Toxicity of ionic liquids: database and prediction via quantitative structure–activity relationship method. Journal of hazardous materials, 2014. 278: p. 320-329.

Cobanoglu, M.C., et al., BalestraWeb: efficient online evaluation of drug–target interactions. Bioinformatics, 2015. 31(1): p. 131-133.

Sam, E. and P. Athri, Web-based drug repurposing tools: a survey. Briefings in bioinformatics, 2019. 20(1): p. 299-316.

Malik, S.I., et al. Mathematical Modeling and Docking of Medicinal Plants and Synthetic drugs to determine their effects on Abnormal Expression of Cholinesterase and Acetyl Cholinesterase Proteins in Alzheimer. in International Work-Conference on Bioinformatics and Biomedical Engineering. 2019. Springer.

Munir, A., et al., In silico repositioning of alendronate and cytarabine drugs to cure mutations of FPPS, HAP, PTPRS, PTPRE, PTN4, GGPPS gene and mutant DNA, DPOLB, TOP2a, DPOLA, DNMT, RNA, TYSY, RIR genes. International Journal Bioautomation, 2016. 20(3): p. 317.

Lo, Y.-C., et al., 3D chemical similarity networks for structure-based target prediction and scaffold hopping. ACS chemical biology, 2016. 11(8): p. 2244-2253.

Cherkasov, A., et al., Mapping the protein interaction network in methicillin-resistant Staphylococcus aureus. Journal of proteome research, 2011. 10(3): p. 1139-1150.

Lo, Y.-C., et al., Machine learning in chemoinformatics and drug discovery. Drug discovery today, 2018. 23(8): p. 1538-1546.

Rzuczek, S.G., M.R. Southern, and M.D. Disney, Studying a drug-like, RNA-focused small molecule library identifies compounds that inhibit RNA toxicity in myotonic dystrophy. ACS chemical biology, 2015. 10(12): p. 2706-2715.

Ba-Alawi, W., et al., DASPfind: new efficient method to predict drug–target interactions. Journal of cheminformatics, 2016. 8(1): p. 1-9.

Zaki, N., et al., Protein-protein interaction based on pairwise similarity. BMC bioinformatics, 2009. 10(1): p. 1-12.

Xu, Q., E.W. Xiang, and Q. Yang. Protein-protein interaction prediction via collective matrix factorization. in 2010 IEEE International Conference on Bioinformatics and Biomedicine (BIBM). 2010. IEEE.

Skrabanek, L., et al., Computational prediction of protein–protein interactions. Molecular biotechnology, 2008. 38(1): p. 1-17.

Alaimo, S., et al., DT-Web: a web-based application for drug-target interaction and drug combination prediction through domain-tuned network-based inference. BMC systems biology, 2015. 9(3): p. 1-11.

Alaimo, S., et al., Drug–target interaction prediction through domain-tuned network-based inference. Bioinformatics, 2013. 29(16): p. 2004-2008.

Alaimo, S., R. Giugno, and A. Pulvirenti, Recommendation techniques for drug–target interaction prediction and drug repositioning, in Data Mining Techniques for the Life Sciences. 2016, Springer. p. 441-462.

Martínez-Jiménez, F. and M.A. Marti-Renom, Ligand-target prediction by structural network biology using nAnnoLyze. PLOS computational biology, 2015. 11(3): p. e1004157.

Azar, A.T., et al., A random forest classifier for lymph diseases. Computer methods and programs in biomedicine, 2014. 113(2): p. 465-473.

Chaudhary, A., S. Kolhe, and R. Kamal, An improved random forest classifier for multi-class classification. Information Processing in Agriculture, 2016. 3(4): p. 215-222.

Burkhardt, H.A., et al. Predicting adverse drug-drug interactions with neural embedding of semantic predications. in AMIA Annual Symposium Proceedings. 2019. American Medical Informatics Association.

Von Eichborn, J., et al., PROMISCUOUS: a database for network-based drug-repositioning. Nucleic acids research, 2010. 39(suppl_1): p. D1060-D1066.

Fu, G., et al., Predicting drug target interactions using meta-path-based semantic network analysis. BMC bioinformatics, 2016. 17(1): p. 1-10.

Chen, B., Y. Ding, and D.J. Wild, Assessing drug target association using semantic linked data. PLoS computational biology, 2012. 8(7): p. e1002574.

Cheng, T., et al., Large-scale prediction of drug-target interaction: a data-centric review. The AAPS journal, 2017. 19(5): p. 1264-1275.

Kuhn, M., et al., STITCH: interaction networks of chemicals and proteins. Nucleic acids research, 2007. 36(suppl_1): p. D684-D688.

Kuhn, M., et al., STITCH 4: integration of protein–chemical interactions with user data. Nucleic acids research, 2014. 42(D1): p. D401-D407.

Gao, Y.-F., et al., Predicting metabolic pathways of small molecules and enzymes based on interaction information of chemicals and proteins. 2012.

Szklarczyk, D., et al., STITCH 5: augmenting protein–chemical interaction networks with tissue and affinity data. Nucleic acids research, 2016. 44(D1): p. D380-D384.

Konc, J. and D. Janežič, ProBiS algorithm for detection of structurally similar protein binding sites by local structural alignment. Bioinformatics, 2010. 26(9): p. 1160-1168.

Konc, J., et al., ProBiS-CHARMMing: web interface for prediction and optimization of ligands in protein binding sites. 2015, ACS Publications.

Jukič, M., et al., ProBiS H2O MD approach for identification of conserved water sites in protein structures for drug design. ACS medicinal chemistry letters, 2020. 11(5): p. 877-882.

Ito, J.-I., et al., PoSSuM: a database of similar protein–ligand binding and putative pockets. Nucleic acids research, 2012. 40(D1): p. D541-D548.

Ito, J.-i., et al., PoSSuM v. 2.0: data update and a new function for investigating ligand analogs and target proteins of small-molecule drugs. Nucleic acids research, 2015. 43(D1): p. D392-D398.

Brown, A.S. and C.J. Patel, MeSHDD: literature-based drug-drug similarity for drug repositioning. Journal of the American Medical Informatics Association, 2017. 24(3): p. 614-618.

Zeng, X., et al., Measure clinical drug–drug similarity using electronic medical records. International journal of medical informatics, 2019. 124: p. 97-103.

Moosavinasab, S., et al., ‘RE: fine drugs’: an interactive dashboard to access drug repurposing opportunities. Database, 2016. 2016.

Shukla, R., et al., Signature-based approaches for informed drug repurposing: Targeting CNS disorders. Neuropsychopharmacology, 2021. 46(1): p. 116-130.

Musa, A., et al., A review of connectivity map and computational approaches in pharmacogenomics. Briefings in bioinformatics, 2018. 19(3): p. 506-523.

Cheng, J., et al., Systematic evaluation of connectivity map for disease indications. Genome medicine, 2014. 6(12): p. 1-8.

Qu, X.A. and D.K. Rajpal, Applications of Connectivity Map in drug discovery and development. Drug discovery today, 2012. 17(23-24): p. 1289-1298.

Lee, B.K.B., et al., DeSigN: connecting gene expression with therapeutics for drug repurposing and development. BMC genomics, 2017. 18(1): p. 1-11.

Jones, J., et al., Gene signatures of progression and metastasis in renal cell cancer. Clinical cancer research, 2005. 11(16): p. 5730-5739.

Dabra, R., T.R. Singh, and R.M. Yennamalli, In Silico Screening of Putative Drug Mplecules to Target MSI Pathway for Colorectal Cancer and HNPCC. 2017.

Louhimo, R., Biomedical Data Integration in Cancer Genomics. 2015.

Duan, Q., et al., L1000CDS 2: LINCS L1000 characteristic direction signatures search engine. NPJ systems biology and applications, 2016. 2(1): p. 1-12.

Clark, N.R., et al., The characteristic direction: a geometrical approach to identify differentially expressed genes. BMC bioinformatics, 2014. 15(1): p. 1-16.

Carrella, D., et al., Mantra 2.0: an online collaborative resource for drug mode of action and repurposing by network analysis. Bioinformatics, 2014. 30(12): p. 1787-1788.

Iorio, F., et al., Discovery of drug mode of action and drug repositioning from transcriptional responses. Proceedings of the National Academy of Sciences, 2010. 107(33): p. 14621-14626.

Setoain, J., et al., NFFinder: an online bioinformatics tool for searching similar transcriptomics experiments in the context of drug repositioning. Nucleic acids research, 2015. 43(W1): p. W193-W199.

Vazquez, M., et al., MARQ: an online tool to mine GEO for experiments with similar or opposite gene expression signatures. Nucleic acids research, 2010. 38(suppl_2): p. W228-W232.

Yu, H., et al. Prediction of drugs having opposite effects on disease genes in a directed network. in BMC systems biology. 2016. Springer.

Fang, M., et al., Drug perturbation gene set enrichment analysis (dpGSEA): a new transcriptomic drug screening approach. BMC bioinformatics, 2021. 22(1): p. 1-14.

Davis, A.M. and R.J. Riley, Predictive ADMET studies, the challenges and the opportunities. Current opinion in chemical biology, 2004. 8(4): p. 378-386.

Rudrapal, M., et al., Repurposing of phytomedicine-derived bioactive compounds with promising anti-SARS-CoV-2 potential: Molecular docking, MD simulation and drug-likeness/ADMET studies. Saudi journal of biological sciences, 2021.

Tareq Hassan Khan, M., Predictions of the ADMET properties of candidate drug molecules utilizing different QSAR/QSPR modelling approaches. Current drug metabolism, 2010. 11(4): p. 285-295.

Khan, M.T. and I. Sylte, Predictive QSAR modeling for the successful predictions of the ADMET properties of candidate drug molecules. Current drug discovery technologies, 2007. 4(3): p. 141-149.

Yang, H., et al., admetSAR 2.0: web-service for prediction and optimization of chemical ADMET properties. Bioinformatics, 2019. 35(6): p. 1067-1069.

Daina, A., O. Michielin, and V. Zoete, SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific reports, 2017. 7(1): p. 1-13.

Mahanthesh, M., et al., Swiss ADME prediction of phytochemicals present in Butea monosperma (Lam.) Taub. J. Pharmacogn. Phytochem, 2020. 9: p. 1799-1809.

Mishra, S. and R. Dahima, In vitro ADME studies of TUG-891, a GPR-120 inhibitor using SWISS ADME predictor. Journal of Drug Delivery and Therapeutics, 2019. 9(2-s): p. 366-369.

Kumar, M.S., et al. Credit card fraud detection using random forest algorithm. in 2019 3rd International Conference on Computing and Communications Technologies (ICCCT). 2019. IEEE.

Chawla, N.V., et al., SMOTE: synthetic minority over-sampling technique. Journal of artificial intelligence research, 2002. 16: p. 321-357.

Chicco, D., M.J. Warrens, and G. Jurman, The coefficient of determination R-squared is more informative than SMAPE, MAE, MAPE, MSE and RMSE in regression analysis evaluation. PeerJ Computer Science, 2021. 7: p. e623.

Chavda, Vivek P., et al. Advanced computational methodologies used in the discovery of new natural anticancer compounds. Frontiers in Pharmacology, 2021. 12.

Downloads

Published

2022-12-24

How to Cite

Khan, J., & Singla, R. K. (2022). Bioinformatics Tools for Pharmaceutical Drug Product Development. Indo Global Journal of Pharmaceutical Sciences, 12, 281–294. https://doi.org/10.35652/IGJPS.2022.12037

Issue

Section

Review Article