Review Article

Application of Machine Learning Approaches for the Design and Study of Anticancer Drugs

Author(s): Yan Hu, Yi Lu, Shuo Wang, Mengying Zhang, Xiaosheng Qu* and Bing Niu*

Volume 20, Issue 5, 2019

Page: [488 - 500] Pages: 13

DOI: 10.2174/1389450119666180809122244

Price: $65

Abstract

Background: Globally the number of cancer patients and deaths are continuing to increase yearly, and cancer has, therefore, become one of the world's highest causes of morbidity and mortality. In recent years, the study of anticancer drugs has become one of the most popular medical topics.

Objective: In this review, in order to study the application of machine learning in predicting anticancer drugs activity, some machine learning approaches such as Linear Discriminant Analysis (LDA), Principal components analysis (PCA), Support Vector Machine (SVM), Random forest (RF), k-Nearest Neighbor (kNN), and Naïve Bayes (NB) were selected, and the examples of their applications in anticancer drugs design are listed.

Results: Machine learning contributes a lot to anticancer drugs design and helps researchers by saving time and is cost effective. However, it can only be an assisting tool for drug design.

Conclusion: This paper introduces the application of machine learning approaches in anticancer drug design. Many examples of success in identification and prediction in the area of anticancer drugs activity prediction are discussed, and the anticancer drugs research is still in active progress. Moreover, the merits of some web servers related to anticancer drugs are mentioned.

Keywords: Machine learning (ML), anticancer drugs, linear discriminant analysis (LDA), principal components analysis (PCA), support vector machine (SVM), random forest (RF), k-nearest neighbor (kNN), naïve bayes (NB), deep learning, web servers.

Graphical Abstract
[1]
Some Studies in Machine Learning Using the Game of Checkers, IBM J Res Develop 1959; 210-29.
[2]
Koza JR. Automated design of both the topology and sizing of analog electrical circuits using genetic programming. Artificial Intelligence in Design’96 1996; 151-70.
[3]
Ron K, Foster P. Glossary of Terms. Mach Learn 1998; 271-4.
[4]
Dong Z, Zhang N, Li C, et al. Anticancer drug sensitivity prediction in cell lines from baseline gene expression through recursive feature selection. BMC Cancer 2015; 15: 489.
[5]
Chou KC, Jones D, Heinrikson RL. Prediction of the tertiary structure and substrate binding site of caspase-8. FEBS Lett 1997; 419: 49-54.
[6]
Chou KC, Wei DQ, Zhong WZ. Binding mechanism of coronavirus main proteinase with ligands and its implication to drug design against SARS. Biochem Biophys Res Commun 2003; 308: 148-51.
[7]
Chou KC, Tomasselli AG, Heinrikson RL. Prediction of the tertiary structure of a caspase-9/inhibitor complex. FEBS Lett 2000; 470: 249-56.
[8]
Li XB, Wang SQ, Xu WR, et al. Novel inhibitor design for hemagglutinin against H1N1 influenza virus by core hopping method. PLoS One 2011; 6(11): e28111.
[9]
Liao QH, Gao QZ, Wei J, et al. Docking and molecular dynamics study on the inhibitory activity of novel inhibitors on epidermal growth factor receptor (EGFR). Med Chem 2011; 7: 24-31.
[10]
Ma Y, Wang SQ, Xu WR, et al. Design novel dual agonists for treating type-2 diabetes by targeting peroxisome proliferator-activated receptors with core hopping approach. PLoS One 2012; 7(6): e38546.
[11]
Wang JF, Chou KC. Insights from modeling the 3d structure of new delhi metallo-beta-lactamse and its binding interactions with antibiotic drugs. PLoS One 2011; 6(4): e18414.
[12]
Wang JF, Chou KC. Insights into the Mutation-Induced HHH syndrome from modeling human mitochondrial ornithine transporter-1. PLoS One 2012; 7(1): e31048.
[13]
Chou KC. Insights from modeling three-dimensional structures of the human potassium and sodium channels. J Proteome Res 2004; 3: 856-61.
[14]
Chou KC. Insights from modelling the 3D structure of the extracellular domain of alpha 7 nicotinic acetylcholine receptor. Biochem Biophys Res Commun 2004; 319: 433-8.
[15]
Chou KC. Insights from modeling the tertiary structure of human BACE2. J Proteome Res 2004; 3: 1069-72.
[16]
Chou KC. Coupling interaction between thromboxane A2 receptor and alpha-13 subunit of guanine nucleotide-binding protein. J Proteome Res 2005; 4: 1681-6.
[17]
Chou KC. Insights from modeling the 3D structure of DNA-CBF3b complex. J Proteome Res 2005; 4: 1657-60.
[18]
Wang JF, Chou KC. Insights from studying the mutation-induced allostery in the M2 proton channel by molecular dynamics. Protein Eng Des Sel 2010; 23: 663-6.
[19]
Wang JF, Wei DQ, Lin Y, et al. Insights from modeling the 3D structure of NAD(P)H-dependent D-Xylose reductase of Pichia stipitis and its binding interactions with NAD and NADP. Biochem Biophys Res Commun 2007; 359: 323-9.
[20]
Wang SQ, Du QS, Huang RB, et al. Insights from investigating the interaction of oseltamivir (Tamiflu) with neuraminidase of the 2009 H1N1 swine flu virus. Biochem Biophys Res Commun 2009; 386: 432-6.
[21]
Chou KC. Structural bioinformatics and its impact to biomedical science. Curr Med Chem 2004; 11: 2105-34.
[22]
Fan YN, Xiao X, Min JL, et al. iNR-Drug: predicting the interaction of drugs with nuclear receptors in cellular networking.In J Mol Sci In: 2014; 15: pp. 4915-37.
[23]
Min JL, Xiao X, Chou KC. iEzy-Drug: A web server for identifying the interaction between enzymes and drugs in cellular networking. BioMed Res Int 2013; 2013: 701317.
[24]
Xiao X, Min JL, Lin WZ, et al. iDrug-Target: predicting the interactions between drug compounds and target proteins in cellular networking via benchmark dataset optimization approach. J Biomol Struct Dyn 2015; 33: 2221-33.
[25]
Xiao X, Min JL, Wang P, et al. iGPCR-Drug: A web server for predicting interaction between GPCRs and drugs in cellular networking. PLoS One 2013; 8.
[26]
Xiao X, Min JL, Wang P, et al. iCDI-PseFpt: Identify the channel-drug interaction in cellular networking with PseAAC and molecular fingerprints. J Theor Biol 2013; 337: 71-9.
[27]
Chen W, Feng PM, Ding H, et al. iRNA-Methyl: Identifying N-6-methyladenosine sites using pseudo nucleotide composition. Anal Biochem 2015; 490: 26-33.
[28]
Chou KC. Impacts of bioinformatics to medicinal chemistry. Med Chem 2015; 11: 218-34.
[29]
Jia CZ, Lin X, Wang ZP. Prediction of protein s-nitrosylation sites based on adapted normal distribution bi-profile bayes and chou’s pseudo amino acid composition. Int J Mol Sci 2014; 15: 10410-23.
[30]
Qiu WR, Xiao X, Lin WZ, et al. iMethyl-PseAAC: Identification of protein methylation sites via a pseudo amino acid composition approach. BioMed Res Int 2014; 2014: 947416.
[31]
Qiu WR, Xiao X, Lin WZ, et al. iUbiq-Lys: prediction of lysine ubiquitination sites in proteins by extracting sequence evolution information via a gray system model. J Biomol Struct Dyn 2015; 33: 1731-42.
[32]
Xie HL, Fu L, Nie XD. Using ensemble SVM to identify human GPCRs N-linked glycosylation sites based on the general form of Chous PseAAC. Protein Eng Des Sel 2013; 26: 735-42.
[33]
Xu Y, Ding J, Wu LY, et al. iSNO-PseAAC: Predict cysteine s-nitrosylation sites in proteins by incorporating position specific amino acid propensity into pseudo amino acid composition. PLoS One 2013; 8.
[34]
Xu Y, Shao XJ, Wu LY, et al. iSNO-AAPair: incorporating amino acid pairwise coupling into PseAAC for predicting cysteine S-nitrosylation sites in proteins. PeerJ 2013; 1.
[35]
Xu Y, Wen X, Shao XJ, et al. iHyd-PseAAC: Predicting hydroxyproline and hydroxylysine in proteins by incorporating dipeptide position-specific propensity into pseudo amino acid composition. Int J Mol Sci 2014; 15: 7594-610.
[36]
Xu Y, Wen X, Wen LS, et al. iNitro-Tyr: Prediction of nitrotyrosine sites in proteins with general pseudo amino acid composition. PLoS One 2014; 9.
[37]
Zhang J, Zhao XW, Sun PP, et al. PSNO: Predicting cysteine s-nitrosylation sites by incorporating various sequence-derived features into the general form of chou’s PseAAC. Int J Mol Sci 2014; 15: 11204-19.
[38]
Chen W, Feng P, Yang H, et al. iRNA-3typeA: identifying 3-types of modification at RNA’s adenosine sites. Mol Ther Nucleic Acids 2018; 11: 468-74.
[39]
Chou KC. An unprecedented revolution in medicinal chemistry driven by the progress of biological science. Curr Top Med Chem 2017; 17: 2337-58.
[40]
Feng P, Yang H, Ding H, et al. iDNA6mA-PseKNC: Identifying DNA N 6 -methyladenosine sites by incorporating nucleotide physicochemical properties into PseKNC. Genomics 2018.
[41]
Feng PM, Ding H, Yang H, et al. iRNA-PseColl: Identifying the Occurrence Sites of Different RNA Modifications by Incorporating Collective Effects of Nucleotides into PseKNC. Mol Ther Nucleic Acids 2017; 7: 155-63.
[42]
Jia JH, Liu Z, Xiao X, et al. iSuc-PseOpt: Identifying lysine succinylation sites in proteins by incorporating sequence-coupling effects into pseudo components and optimizing imbalanced training dataset. Anal Biochem 2016; 497: 48-56.
[43]
Jia JH, Liu Z, Xiao X, et al. pSuc-Lys: Predict lysine succinylation sites in proteins with PseAAC and ensemble random forest approach. J Theor Biol 2016; 394: 223-30.
[44]
Jia JH, Liu Z, Xiao X, et al. iCar-PseCp: identify carbonylation sites in proteins by Monto Carlo sampling and incorporating sequence coupled effects into general PseAAC. Oncotarget 2016; 7: 34558-70.
[45]
Jia JH, Zhang LX, Liu Z, et al. pSumo-CD: predicting sumoylation sites in proteins with covariance discriminant algorithm by incorporating sequence-coupled effects into general PseAAC. Bioinformatics 2016; 32: 3133-41.
[46]
Lee K, Jung SY, Hwang H, et al. A novel concept for integrating and delivering health information using a comprehensive digital dashboard: An analysis of healthcare professionals’ intention to adopt a new system and the trend of its real usage. Int J Med Inform 2017; 97: 98-108.
[47]
Ju Z, Wang SY. Prediction of citrullination sites by incorporating k-spaced amino acid pairs into Chou’s general pseudo amino acid composition. Gene 2018.
[48]
Khan YD, Rasool N, Hussain W, et al. iPhosT-PseAAC: Identify phosphothreonine sites by incorporating sequence statistical moments into PseAAC. Anal Biochem 2018; 550: 109-16.
[49]
Liu LM, Xu Y, Chou KC. iPGK-PseAAC: Identify lysine phosphoglycerylation sites in proteins by incorporating four different tiers of amino acid pairwise coupling information into the general PseAAC. Med Chem 2017; 13: 552-9.
[50]
Liu Z, Xiao X, Yu DJ, et al. pRNAm-PC: Predicting N-6-methyladenosine sites in RNA sequences via physical-chemical properties. Anal Biochem 2016; 497: 60-7.
[51]
Qiu WR, Jiang SY, Sun BQ, et al. iRNA-2methyl: Identify RNA 2′-O-methylation Sites by Incorporating Sequence-Coupled Effects into General PseKNC and Ensemble Classifier. Med Chem 2017; 13: 734-43.
[52]
Qiu WR, Jiang SY, Xu ZC, et al. iRNAm5C-PseDNC: identifying RNA 5-methylcytosine sites by incorporating physical-chemical properties into pseudo dinucleotide composition. Oncotarget 2017; 8: 41178-88.
[53]
Qiu WR, Sun BQ, Xiao X, et al. iPhos-PseEvo: Identifying Human Phosphorylated Proteins by Incorporating Evolutionary Information into General PseAAC via Grey System Theory. Mol Inform 2017; 36.
[54]
Qiu WR, Sun BQ, Xiao X, et al. iHyd-PseCp: Identify hydroxyproline and hydroxylysine in proteins by incorporating sequence-coupled effects into general PseAAC. Oncotarget 2016; 7: 44310-21.
[55]
Qiu WR, Sun BQ, Xiao X, et al. iPTM-mLys: identifying multiple lysine PTM sites and their different types. Bioinformatics 2016; 32: 3116-23.
[56]
Qiu WR, Sun BQ, Xuan X, et al. iKcr-PseEns: Identify lysine crotonylation sites in histone proteins with pseudo components and ensemble classifier. Genomics 2017.
[57]
Qiu WR, Xiao X, Xu ZC, et al. iPhos-PseEn: Identifying phosphorylation sites in proteins by fusing different pseudo components into an ensemble classifier. Oncotarget 2016; 7: 51270-83.
[58]
Sabooh MF, Iqbal N, Khan M, et al. Identifying 5-methylcytosine sites in RNA sequence using composite encoding feature into Chou’s PseKNC. J Theor Biol 2018; 7(452): 1-9.
[59]
Xu Y, Chou KC. Recent progress in predicting posttranslational modification sites in proteins. Curr Top Med Chem 2016; 16: 591-603.
[60]
Xu Y, Wang Z, Li CH, et al. iPreny-PseAAC: Identify C-terminal Cysteine Prenylation Sites in Proteins by Incorporating Two Tiers of Sequence Couplings into PseAAC. Med Chem 2017; 13: 544-51.
[61]
Chou KC. Some remarks on protein attribute prediction and pseudo amino acid composition. J Theor Biol 2011; 273: 236-47.
[62]
Chen W, Lin H, Chou KC. Pseudo nucleotide composition or PseKNC: an effective formulation for analyzing genomic sequences. Mol Biosyst 2015; 11: 2620-34.
[63]
Cheng X, Xiao X, Chou KC. pLoc-mPlant: predict subcellular localization of multi-location plant proteins by incorporating the optimal GO information into general PseAAC. Mol Biosyst 2017; 13: 1722-7.
[64]
Cheng X, Xiao X, Chou KC. pLoc-mVirus: Predict subcellular localization of multi-location virus proteins via incorporating the optimal GO information into general PseAAC. Gene 2017; 628: 315-21.
[65]
Cheng X, Zhao SG, Lin WZ, et al. pLoc-mAnimal: predict subcellular localization of animal proteins with both single and multiple sites. Bioinformatics 2017; 33: 3524-31.
[66]
Cheng X, Zhao SG, Xiao X, et al. iATC-mISF: a multi-label classifier for predicting the classes of anatomical therapeutic chemicals. Bioinformatics 2017; 33: 341-6.
[67]
Cheng X, Xiao X, Chou KC. pLoc-mGneg: Predict subcellular localization of Gram-negative bacterial proteins by deep gene ontology learning via general PseAAC. Genomics 2017.
[68]
Cheng X, Xiao X, Chou KC. pLoc-mEuk: Predict subcellular localization of multi-label eukaryotic proteins by extracting the key GO information into general PseAAC. Genomics 2018; 110: 50-8.
[69]
Cheng X, Xiao X, Chou KC. pLoc-mHum: predict subcellular localization of multi-location human proteins via general PseAAC to winnow out the crucial GO information. Bioinformatics 2018; 34: 1448-56.
[70]
Xiao X, Cheng X, Chen G, et al. pLoc-mGpos: Predict subcellular localization of Gram-positive bacterial proteins by quasi-balancing training dataset and PseAAC. Genomics 2018.
[71]
Xiao X, Cheng X, Su S, et al. pLoc-mGpos: Incorporate Key Gene Ontology Information into General PseAAC for Predicting Subcellular Localization of Gram-Positive Bacterial Proteins. Nat Sci 2017; 09: 330-49.
[72]
Chou KC. Some remarks on predicting multi-label attributes in molecular biosystems. Mol Biosyst 2013; 9: 1092-100.
[73]
Chen W, Feng PM, Lin H, et al. iRSpot-PseDNC: identify recombination spots with pseudo dinucleotide composition. Nucleic Acids Res 2013; 41.
[74]
Chen W, Feng PM, Yang H, et al. iRNA-AI: identifying the adenosine to inosine editing sites in RNA sequences. Oncotarget 2017; 8: 4208-17.
[75]
Cheng X, Zhao SG, Xiao X, et al. iATC-mHyb: a hybrid multi-label classifier for predicting the classification of anatomical therapeutic chemicals. Oncotarget 2017; 8: 58494-503.
[76]
Du QS, Wang SQ, Xie NZ, et al. 2L-PCA: a two-level principal component analyzer for quantitative drug design and its applications. Oncotarget 2017; 8: 70564-78.
[77]
Lin H, Deng EZ, Ding H, et al. iPro54-PseKNC: a sequence-based predictor for identifying sigma-54 promoters in prokaryote with pseudo k-tuple nucleotide composition. Nucleic Acids Res 2014; 42: 12961-72.
[78]
Liu B, Liu FL, Wang XL, et al. Pse-in-One: a web server for generating various modes of pseudo components of DNA, RNA, and protein sequences. Nucleic Acids Res 2015; 43: W65-71.
[79]
Liu B, Wang SY, Long R, et al. iRSpot-EL: identify recombination spots with an ensemble learning approach. Bioinformatics 2017; 33: 35-41.
[80]
Liu B, Wu H, Zhang DY, et al. Pse-Analysis: a python package for DNA/RNA and protein/peptide sequence analysis based on pseudo components and kernel methods. Oncotarget 2017; 8: 13338-43.
[81]
Liu B, Yang F, Chou KC. 2L-piRNA: A Two-Layer Ensemble Classifier for Identifying Piwi-Interacting RNAs and Their Function. Mol Ther Nucleic Acids 2017; 7: 267-77.
[82]
Niu B, Zhang MY, Du P, et al. Small molecular floribundiquinone B derived from medicinal plants inhibits acetylcholinesterase activity. Oncotarget 2017; 8: 57149-62.
[83]
Qiu WR, Xiao X, Chou KC. iRSpot-TNCPseAAC: Identify recombination spots with trinucleotide composition and pseudo amino acid components. Int J Mol Sci 2014; 15: 1746-66.
[84]
Su Q, Lu WC, Du DS, et al. Prediction of the aquatic toxicity of aromatic compounds to tetrahymena pyriformis through support vector regression. Oncotarget 2017; 8: 49359-69.
[85]
Wang JW, Yang BJ, Revote J, et al. POSSUM: a bioinformatics toolkit for generating numerical sequence feature descriptors based on PSSM profiles. Bioinformatics 2017; 33: 2756-8.
[86]
Xu ZC, Qiu WR, Xiao X. iRSpotH-TNCPseAAC: Identifying recombination spots in human by using pseudo trinucleotide composition with an ensemble of support vector machine classifiers. Lett Org Chem 2017; 14: 703-13.
[87]
Zhang ZD, Liang K, Li K, et al. Chlorella vulgaris Induces Apoptosis of Human Non-Small Cell Lung Carcinoma (NSCLC) Cells. Med Chem 2017; 13: 560-8.
[88]
Kotsiantis SB. Supervised machine learning: a review of classification techniques Informatica. (Ljubl) 2007; pp. 249-68.
[89]
Rennie J, Shih L, Teevan J, et al. Tackling the poor assumptions of naive bayes text Classifiers 2003.
[90]
Duda RO, Hart PE, Stork DG. Pattern Classification, ch. 10: Unsupervised learning and clustering 2001.
[91]
Kotsiantis S, Pintelas P. Recent advances in clustering: A brief survey. Wseas Transactions Inform Sci 2004; 1(1): 73-81.
[92]
Laskaris R. Artificial intelligence: A modern approach, 3rd edition. In: Lib J. 2015; 140: pp. 45-5.
[93]
Mehryar Mohri AR, Talwalkar A. Machine learning; Computer algorithms. 2012.
[94]
Gould KA. The elements of statistical learning (2nd edition): Data mining, inference, and prediction. In: Dimensions Critical Care Nursing. 2016; 35: pp. 52-2.
[95]
Bousquet O, Luxburg U, Rätsch G. Advanced lectures on machine learning. Springer 2003.
[96]
Lewis R. Chapter 4: The development of molecular modelling programs: the use and limitations of physical models.
[97]
rajamani R. Good A. Ranking poses in structure-based lead discovery and optimization: current trends in scoring function development. Curr Opin Drug Dis Develop 2007; 308-15.
[98]
Hughes JP, Rees S, Kalindjian SB, et al. Principles of early drug discovery. British J Pharmacol 2011; 162: 1239-49.
[99]
F.R.S. Rafsd. The use of multiple measurements in taxonomic problems. Ann Human Genetics 1936; 7(2): 179-88.
[100]
Gohulkumar M, Kumar P, Murali Krishna C, et al. Evaluation of Raman spectroscopy for prediction of antitumor response to silibinin and its nanoparticulates in DMBA-induced oral carcinogenesis. J Raman Spectroscopy 2016; 47: 375-83.
[101]
Adhikaria1 N, Haldera1 AK, Sahab A, et al. Structural findings of phenylindoles as cytotoxic antimitotic agents in human breast cancer cell lines through multiple validated QSAR studies. Toxicol In Vitro 2015: 1392-404. 2015; 1392-404.
[102]
Maldonado-Rojas W, Olivero-Verbel J, Marrero-Ponce Y. Computational fishing of new DNA methyltransferase inhibitors from natural products. J Mol Graph Model 2015; 43-54.
[103]
Goel PN, Singh SP, Murali Krishna C, et al. Investigating the effects of Pentoxifylline on human breast cancer cells using Raman spectroscopy. J Innov Opt Health Sci 2015; 08: 1550004.
[104]
Covell DG. Integrating constitutive gene expression and chemoactivity: Mining the NCI60 Anticancer Screen. PLoS One 2012; 7.
[105]
F.R.S KP LIII. On lines and planes of closest fit to systems of points in space. Philos Mag 1901; 2(11): 559-72.
[106]
Paguigan ND, Al-Huniti MH, Raja HA, et al. Chemoselective fluorination and chemoinformatic analysis of griseofulvin: Natural vs fluorinated fungal metabolites. Bioorg Med Chem 2017; 25: 5238-46.
[107]
Taguchi YH, Wang HY. Genetic association between amyotrophic lateral sclerosis and cancer. Genes (Basel) 2017; 8.
[108]
Wang Z, Liu JQ, Xu JD, et al. UPLC/ESI-QTOF-MS-based metabolomics survey on the toxicity of triptolide and detoxication of licorice. Chin J Nat Med 2017; 15: 474-80.
[109]
Su J, Liu X, Zhang S, et al. A computational insight into binding modes of inhibitors XD29, XD35, and XD28 to bromodomain-containing protein 4 based on molecular dynamics simulations. J Biomol Struct Dyn 2018; 36(5): 1212-24.
[110]
Chen JZ. Clarifying binding difference of ATP and ADP to extracellular signal-regulated kinase 2 by using molecular dynamics simulations. Chem Biol Drug Des 2017; 89: 548-58.
[111]
Demir O, Ieong PU, Amaro RE. Full-length p53 tetramer bound to DNA and its quaternary dynamics. Oncogene 2017; 36: 1451-60.
[112]
Shafique S, Rashid S. Antiviral drug acyclovir exhibits antitumor activity via targeting beta TrCP1: Molecular docking and dynamics simulation study. J Mol Graph Model 2017; 72: 96-105.
[113]
Yao YR, Zhang P, Wang J, et al. Dissecting target toxic tissue and tissue specific responses of irinotecan in rats using metabolomics approach. Front Pharmacol 2017; 8.
[114]
Wali VB, Langdon CG, Held MA, et al. Systematic drug screening identifies tractable targeted combination therapies in triple-negative breast cancer. Cancer Res 2017; 77: 566-78.
[115]
Altman NS. An introduction to Kernel and nearest-neighbor nonparametric regression. The American Statistician 1992; 175.
[116]
Amin SA, Adhikari N, Agrawal RK, et al. Possible binding mode analysis of pyrazolo-triazole hybrids as potential anticancer agents through validated molecular docking and 3d-qsar modeling approaches. Lett Drug Des Dis 2017; pp. 515-27.
[117]
Bhandari DSV. 2D, 3D, G-QSAR and Docking Studies of Thiazolyl- Pyrazoline Analogues as Potent (Epidermal Growth Factor Receptor-Tyrosine Kinase) EGFRTK Inhibitors. Lett Drug Des Dis 2017; 14.
[118]
Aboalhaija NH, Zihlif MA, Taha MO. Discovery of new selective cytotoxic agents against Bcl-2 expressing cancer cells using ligand-based modeling. Chem Biol Interact 2016; 250: 12-6.
[119]
AlQudah DA, Zihlif MA, Taha MO. Ligand-based modeling of diverse aryalkylamines yields new potent P-glycoprotein inhibitors. Eur J Med Chem 2016; 110: 204-23.
[120]
Vapnik VN. An overview of statistical learning theory. IEEE Trans Neural Netw 1999; 988-9.
[121]
Cortes C, Vapnik V. Support-vector networks. Mach Learn 1995; 273-97.
[122]
A P, P S, B Ż, et al. Novel 2-(2-alkylthiobenzenesulfonyl)-3-(phenylprop-2-ynylideneamino)guanidine derivatives as potent anticancer agents - Synthesis, molecular structure, QSAR studies and metabolic stability, Eur J Med Chem 2017; 357-70.
[123]
Li FM, Wang XQ. Identifying anticancer peptides by using improved hybrid compositions. Sci Rep 2016; 6.
[124]
Singh H, Kumar R, Singh S, et al. Prediction of anticancer molecules using hybrid model developed on molecules screened against NCI-60 cancer cell lines. BMC Cancer 2016; 16.
[125]
Dong ZL, Zhang NQ, Li C, et al. Anticancer drug sensitivity prediction in cell lines from baseline gene expression through recursive feature selection. BMC Cancer 2015; 15.
[126]
Zhang M, Su Q, Lu Y, et al. Application of machine learning approaches for protein-protein interactions prediction. Med Chem 2017; 13: 506-14.
[127]
Zhang P, Chen JQ, Huang WQ, et al. Renal medulla is more sensitive to cisplatin than cortex revealed by untargeted mass spectrometry-based metabolomics in rats. Sci Rep 2017; 7.
[128]
Dhiman K, Agarwal SM. NPred: QSAR classification model for identifying plant based naturally occurring anti-cancerous inhibitors. RSC Advances 2016; 6: 49395-400.
[129]
Wangabd L, Liabd Y, Xuc M, et al. Chemical fragment-based CDK4/6 inhibitors prediction and web server. RSC Adv 2016; pp. 16972-81.
[130]
Hand DJ, Yu KM. Idiot’s Bayes-Not so stupid after all?. Int Stat Rev 2001; 69: 385-98.
[131]
Rish I. An empirical study of the naive Bayes classifier 2001.
[132]
Rennie JDM, Shih L, Teevan J, et al. Tackling the Poor Assumptions of Naive Bayes Text Classifiers 2003.
[133]
Krishna S, Shukla S, Lakra AD, et al. Identification of potent inhibitors of DNA methyltransferase 1 (DNMT1) through a pharmacophore-based virtual screening approach. J Mol Graph Model 2017; 75: 174-88.
[134]
Tran WT, Gangeh MJ, Sannachi L, et al. Predicting breast cancer response to neoadjuvant chemotherapy using pretreatment diffuse optical spectroscopic texture analysis. Br J Cancer 2017; 116: 1329-39.
[135]
Liu Z, He W, Gao J, et al. Computational prediction and experimental validation of a novel synthesized pan-PIM inhibitor PI003 and its apoptosis-inducing mechanisms in cervical cancer. Oncotarget 2015; 6: 8019-35.
[136]
Yin J-Y, Li X, Li X-P, et al. Prediction models for platinum-based chemotherapy response and toxicity in advanced NSCLC patients. Cancer Lett 2016; 377: 65-73.
[137]
Zhang H, Cao Z-X, Li M, et al. Novel naive Bayes classification models for predicting the carcinogenicity of chemicals. Food And Chem Toxicol 2016; 97: 141-9.
[138]
Ali S, Majid A. Can-Evo-Ens: Classifier stacking based evolutionary ensemble system for prediction of human breast cancer using amino acid sequences. J Biomed Informatics 2015; 54: 256-69.
[139]
Amirkhah R, Farazmand A, Gupta SK, et al. Naive Bayes classifier predicts functional microRNA target interactions in colorectal cancer. Mol Biosyst 2015; 11: 2126-34.
[140]
Yang R, Zhang C, Gao R, et al. A machine learning approach to identify dna replication proteins from sequence-derived features. 2015 Ieee 28th Canadian Conf Electrical Comput Engin 2015; 13-8.
[141]
Begum S, Chakraborty D, Sarkar R, et al. Identifying cancer biomarkers from leukemia data using feature selection and supervised learning 2016.
[142]
Bengio Y, Courville A, Vincent P. Representation Learning: A review and new perspectives. IEEE Trans Pattern Anal Mach Intell 2013; 35: 1798-828.
[143]
LeCun Y, Bengio Y, Hinton G. Deep learning. Nature 2015; 521: 436-44.
[144]
Schmidhuber J. Deep learning in neural networks: An overview. Neural Network 2015; 61: 85-17.
[145]
Ghasemi F, Mehridehnavi AR, Fassihi A, et al. Deep neural network in biological activity prediction using deep belief network. Appl Soft Comput 2017; 62.
[146]
Schmidhuber J. Multi-column deep neural networks for image classification. In: Computer Vision and Pattern Recognition. 2012; pp. 3642-9.
[147]
Krizhevsky A, Sutskever I, Hinton GE. ImageNet classification with deep convolutional neural networks. In: International Conference on Neural Information Processing Systems 2012; 1097-105.
[148]
Cao RZ, Bhattacharya D, Hou J, et al. Deep QA: improving the estimation of single protein model quality with deep belief networks. BMC Bioinformatics 2016; 17.
[149]
Cao RZ, Freitas C, Chan L, et al. ProLanGO: Protein function prediction using neural machine translation based on a recurrent Neural Network. Mol 2017; 22.
[150]
van Gerven M, Bohte S. Editorial: artificial neural networks as models of neural information processing. Front Comput Neurosci 2017; 11.
[151]
Abadi RSK, Alizadehdakhel A, Moosapour F. Linear and non-linear QSAR models on platinum (II) anticancer drugs with N-donor ligands. Ind J Chem Section B-Org Chem Including Med Chem 2017; 56: 677-86.
[152]
Abadi RSK, Alizadehdakhel A, Shiraz SD. Ab initio and QSAR study of several etoposides as anticancer drugs: Solvent effect. Russian J Phys Chem 2017; 11: 307-17.
[153]
Amin SA, Adhikari N, Gayen S, et al. First report on the structural exploration and prediction of new BPTES analogs as glutaminase inhibitors. J Mol Struct 2017; 1143: 49-64.
[154]
Ramaiah MJ, Naushad SM, Lavanya A, et al. Scriptaid cause histone deacetylase inhibition and cell cycle arrest in HeLa cancer cells: A study on structural and functional aspects. Gene 2017; 627: 379-86.
[155]
Chou K, Shen H. REVIEW: Recent advances in developing web-servers for predicting protein attributes. Nat Sci 2009; 1: 63-92.
[156]
Jia J, Liu Z, Xiao X, et al. iPPI-Esml: An ensemble classifier for identifying the interactions of proteins by incorporating their physicochemical properties and wavelet transforms into PseAAC. J Theor Biol 2015; 377: 47-56.
[157]
Liu B, Fang L, Liu F, et al. Identification of real microrna precursors with a pseudo structure status composition approach. PLoS One 2015; 10.
[158]
Liu B, Fang L, Long R, et al. iEnhancer-2L: a two-layer predictor for identifying enhancers and their strength by pseudo k-tuple nucleotide composition. Bioinformatics 2016; 32: 362-9.
[159]
Chen W, Ding H, Feng P, et al. IACP: a sequence-based tool for identifying anticancer peptides. Oncotarget 2016; 7: 16895-909.
[160]
Sharma A, Singla D, Rashid M, et al. Designing of peptides with desired half-life in intestine-like environment. BMC Bioinformatics 2014; 15.
[161]
Wang L, Li Y, Xu M, et al. Chemical fragment-based CDK4/6 inhibitors prediction and web server. RSC Adv 2016; 6: 16972-81.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy