Research article Special Issues

A GraphSAGE-based model with fingerprints only to predict drug-drug interactions


  • Received: 04 December 2023 Revised: 18 January 2024 Accepted: 21 January 2024 Published: 26 January 2024
  • Drugs are an effective way to treat various diseases. Some diseases are so complicated that the effect of a single drug for such diseases is limited, which has led to the emergence of combination drug therapy. The use multiple drugs to treat these diseases can improve the drug efficacy, but it can also bring adverse effects. Thus, it is essential to determine drug-drug interactions (DDIs). Recently, deep learning algorithms have become popular to design DDI prediction models. However, most deep learning-based models need several types of drug properties, inducing the application problems for drugs without these properties. In this study, a new deep learning-based model was designed to predict DDIs. For wide applications, drugs were first represented by commonly used properties, referred to as fingerprint features. Then, these features were perfectly fused with the drug interaction network by a type of graph convolutional network method, GraphSAGE, yielding high-level drug features. The inner product was adopted to score the strength of drug pairs. The model was evaluated by 10-fold cross-validation, resulting in an AUROC of 0.9704 and AUPR of 0.9727. Such performance was better than the previous model which directly used drug fingerprint features and was competitive compared with some other previous models that used more drug properties. Furthermore, the ablation tests indicated the importance of the main parts of the model, and we analyzed the strengths and limitations of a model for drugs with different degrees in the network. This model identified some novel DDIs that may bring expected benefits, such as the combination of PEA and cannabinol that may produce better effects. DDIs that may cause unexpected side effects have also been discovered, such as the combined use of WIN 55,212-2 and cannabinol. These DDIs can provide novel insights for treating complex diseases or avoiding adverse drug events.

    Citation: Bo Zhou, Bing Ran, Lei Chen. A GraphSAGE-based model with fingerprints only to predict drug-drug interactions[J]. Mathematical Biosciences and Engineering, 2024, 21(2): 2922-2942. doi: 10.3934/mbe.2024130

    Related Papers:

  • Drugs are an effective way to treat various diseases. Some diseases are so complicated that the effect of a single drug for such diseases is limited, which has led to the emergence of combination drug therapy. The use multiple drugs to treat these diseases can improve the drug efficacy, but it can also bring adverse effects. Thus, it is essential to determine drug-drug interactions (DDIs). Recently, deep learning algorithms have become popular to design DDI prediction models. However, most deep learning-based models need several types of drug properties, inducing the application problems for drugs without these properties. In this study, a new deep learning-based model was designed to predict DDIs. For wide applications, drugs were first represented by commonly used properties, referred to as fingerprint features. Then, these features were perfectly fused with the drug interaction network by a type of graph convolutional network method, GraphSAGE, yielding high-level drug features. The inner product was adopted to score the strength of drug pairs. The model was evaluated by 10-fold cross-validation, resulting in an AUROC of 0.9704 and AUPR of 0.9727. Such performance was better than the previous model which directly used drug fingerprint features and was competitive compared with some other previous models that used more drug properties. Furthermore, the ablation tests indicated the importance of the main parts of the model, and we analyzed the strengths and limitations of a model for drugs with different degrees in the network. This model identified some novel DDIs that may bring expected benefits, such as the combination of PEA and cannabinol that may produce better effects. DDIs that may cause unexpected side effects have also been discovered, such as the combined use of WIN 55,212-2 and cannabinol. These DDIs can provide novel insights for treating complex diseases or avoiding adverse drug events.



    加载中


    [1] G. Lee, C. Park, J. Ahn, Novel deep learning model for more accurate prediction of drug-drug interaction effects, BMC Bioinf., 20 (2019), 415. https://doi.org/10.1186/s12859-019-3013-0 doi: 10.1186/s12859-019-3013-0
    [2] Y. Deng, X. Xu, Y. Qiu, J. Xia, W. Zhang, S. Liu, A multimodal deep learning framework for predicting drug-drug interaction events, Bioinformatics, 36 (2020), 4316–4322. https://doi.org/10.1093/bioinformatics/btaa501 doi: 10.1093/bioinformatics/btaa501
    [3] L. Chen, C. Chu, Y. H. Zhang, M. Zheng, L. Zhu, X. Kong, et al., Identification of drug-drug interactions using chemical interactions, Curr. Bioinf., 12 (2017), 526–534. https://doi.org/10.2174/1574893611666160618094219 doi: 10.2174/1574893611666160618094219
    [4] F. Cheng, Z. Zhao, Machine learning-based prediction of drug–drug interactions by integrating drug phenotypic, therapeutic, chemical, and genomic properties, J. Am. Med. Inf. Assoc., 21 (2014), e278–e286. https://doi.org/10.1136/amiajnl-2013-002512 doi: 10.1136/amiajnl-2013-002512
    [5] B. Ran, L. Chen, M. Li, Y. Han, Q. Dai, Drug-drug interactions prediction using fingerprint only, Comput. Math. Methods Med., 2022 (2022), 7818480. https://doi.org/10.1155/2022/7818480 doi: 10.1155/2022/7818480
    [6] C. Yan, G. Duan, Y. Pan, F. X. Wu, J. Wang, DDIGIP: Predicting drug-drug interactions based on Gaussian interaction profile kernels, BMC Bioinf., 20 (2019), 538. https://doi.org/10.1186/s12859-019-3093-x doi: 10.1186/s12859-019-3093-x
    [7] N. Rohani, C. Eslahchi, Drug-drug interaction predicting by neural network using integrated similarity, Sci. Rep., 9 (2019), 13645. https://doi.org/10.1038/s41598-019-50121-3 doi: 10.1038/s41598-019-50121-3
    [8] A. M. Roy, J. Bhaduri, DenseSPH-YOLOv5: An automated damage detection model based on DenseNet and Swin-Transformer prediction head-enabled YOLOv5 with attention mechanism, Adv. Eng. Inf., 56 (2023), 102007. https://doi.org/10.1016/j.aei.2023.102007 doi: 10.1016/j.aei.2023.102007
    [9] S. Jamil, A. M. Roy, An efficient and robust Phonocardiography (PCG)-based Valvular Heart Diseases (VHD) detection framework using Vision Transformer (ViT), Comput. Biol. Med., 158 (2023), 106734. https://doi.org/10.1016/j.compbiomed.2023.106734 doi: 10.1016/j.compbiomed.2023.106734
    [10] A. M. Roy, J. Bhaduri, T. Kumar, K. Raj, WilDect-YOLO: An efficient and robust computer vision-based accurate object localization model for automated endangered wildlife detection, Ecol. Inf., 75 (2023), 101919. https://doi.org/10.1016/j.ecoinf.2022.101919 doi: 10.1016/j.ecoinf.2022.101919
    [11] B. Jiang, S. Chen, B. Wang, B. Luo, MGLNN: Semi-supervised learning via multiple graph cooperative learning neural networks, Neural Networks, 153 (2022), 204–214. https://doi.org/10.1016/j.neunet.2022.05.024 doi: 10.1016/j.neunet.2022.05.024
    [12] C. He, Y. Liu, H. Li, H. Zhang, Y. Mao, X. Qin, et al., Multi-type feature fusion based on graph neural network for drug-drug interaction prediction, BMC Bioinf., 23 (2022), 224. https://doi.org/10.1186/s12859-022-04763-2 doi: 10.1186/s12859-022-04763-2
    [13] X. Y. Yan, P. W. Yin, X. M. Wu, J. X. Han, Prediction of the drug-drug interaction types with the unified embedding features from drug similarity networks, Front. Pharmacol., 12 (2021), 794205. https://doi.org/10.3389/fphar.2021.794205 doi: 10.3389/fphar.2021.794205
    [14] S. Lin, Y. Wang, L. Zhang, Y. Chu, Y. Liu, Y. Fang, et al., MDF-SA-DDI: Predicting drug-drug interaction events based on multi-source drug fusion, multi-source feature fusion and transformer self-attention mechanism, Briefings Bioinf., 23 (2022), bbab421. https://doi.org/10.1093/bib/bbab421 doi: 10.1093/bib/bbab421
    [15] Y. H. Feng, S. W. Zhang, J. Y. Shi, DPDDI: A deep predictor for drug-drug interactions, BMC Bioinf., 21 (2020), 419. https://doi.org/10.1186/s12859-020-03724-x doi: 10.1186/s12859-020-03724-x
    [16] S. Liu, Y. Zhang, Y. Cui, Y. Qiu, Y. Deng, Z. Zhang, et al., Enhancing drug-drug interaction prediction using deep attention neural networks, IEEE/ACM Trans. Comput. Biol. Bioinf., 20 (2023), 976–985. https://doi.org/10.1109/TCBB.2022.3172421 doi: 10.1109/TCBB.2022.3172421
    [17] D. S. Wishart, C. Knox, A. C. Guo, D. Cheng, S. Shrivastava, D. Tzur, et al., DrugBank: A knowledgebase for drugs, drug actions and drug targets, Nucleic Acids Res., 36 (2008), D901–D906. https://doi.org/10.1093/nar/gkm958 doi: 10.1093/nar/gkm958
    [18] D. S. Wishart, C. Knox, A. C. Guo, S. Shrivastava, M. Hassanali, P. Stothard, et al., DrugBank: A comprehensive resource for in silico drug discovery and exploration, Nucleic Acids Res., 34 (2006), D668–D672. https://doi.org/10.1093/nar/gkj067 doi: 10.1093/nar/gkj067
    [19] W. Hamilton, Z. Ying, J. Leskovec, Inductive representation learning on large graphs, in NIPS'17: Proceedings of the 31st International Conference on Neural Information Processing Systems, (2017), 1024–1034.
    [20] X. Pan, L. Chen, I. Liu, Z. Niu, T. Huang, Y. D. Cai, Identifying protein subcellular locations with embeddings-based node2loc, IEEE/ACM Trans. Comput. Biol. Bioinf., 19 (2022), 666–675. https://doi.org/10.1109/TCBB.2021.3080386 doi: 10.1109/TCBB.2021.3080386
    [21] J. P. Zhou, L. Chen, Z. H. Guo, iATC-NRAKEL: An efficient multi-label classifier for recognizing anatomical therapeutic chemical classes of drugs, Bioinformatics, 36 (2020), 1391–1396. https://doi.org/10.1093/bioinformatics/btz757 doi: 10.1093/bioinformatics/btz757
    [22] C. Wu, L. Chen, A model with deep analysis on a large drug network for drug classification, Math. Biosci. Eng., 20 (2023), 383–401. https://doi.org/10.3934/mbe.2023018 doi: 10.3934/mbe.2023018
    [23] L. Chen, K. Chen, B. Zhou, Inferring drug-disease associations by a deep analysis on drug and disease networks, Math. Biosci. Eng., 20 (2023), 14136–14157. https://doi.org/10.3934/mbe.2023632 doi: 10.3934/mbe.2023632
    [24] L. Chen, J. Xu, Y. Zhou, PDATC-NCPMKL: Predicting drug's Anatomical Therapeutic Chemical (ATC) codes based on network consistency projection and multiple kernel learning, Comput. Biol. Med., 169 (2024), 107862. https://doi.org/10.1016/j.compbiomed.2023.107862 doi: 10.1016/j.compbiomed.2023.107862
    [25] M. A. Alsmirat, F. Al-Alem, M. Al-Ayyoub, Y. Jararweh, B. Gupta, Impact of digital fingerprint image quality on the fingerprint recognition accuracy, Multimedia Tools Appl., 78 (2019), 3649–3688. https://doi.org/10.1007/s11042-017-5537-5 doi: 10.1007/s11042-017-5537-5
    [26] N. Nedjah, R. S. Wyant, L. M. Mourelle, B. B. Gupta, Efficient fingerprint matching on smart cards for high security and privacy in smart systems, Inf. Sci., 479 (2019), 622–639. https://doi.org/10.1016/j.ins.2017.12.038 doi: 10.1016/j.ins.2017.12.038
    [27] B. D. Christie, B. A. Leland, J. G. Nourse, Structure searching in chemical databases by direct lookup methods, J. Chem. Inf. Comput. Sci., 33 (1993), 545–547. https://doi.org/10.1021/ci00014a004 doi: 10.1021/ci00014a004
    [28] D. Rogers, M. Hahn, Extended-connectivity fingerprints, J. Chem. Inf. Model., 50 (2010), 742–754. https://doi.org/10.1021/ci100050t doi: 10.1021/ci100050t
    [29] T. N. Kipf, M. Welling, Semi-supervised classification with graph convolutional networks, arXiv preprint, (2016), arXiv: 160902907. https://doi.org/10.48550/arXiv.1609.02907
    [30] X. Wang, M. Liu, Y. Zhang, S. He, C. Qin, Y. Li, et al., Deep fusion learning facilitates anatomical therapeutic chemical recognition in drug repurposing and discovery, Briefings Bioinf., 22 (2021), bbab289. https://doi.org/10.1093/bib/bbab289 doi: 10.1093/bib/bbab289
    [31] H. Zhao, Y. Li, J. Wang, A convolutional neural network and graph convolutional network-based method for predicting the classification of anatomical therapeutic chemicals, Bioinformatics, 37 (2021), 2841–2847. https://doi.org/10.1093/bioinformatics/btab204 doi: 10.1093/bioinformatics/btab204
    [32] X. Y. Pan, H. B. Shen, Inferring disease-associated microRNAs using semi-supervised multi-label graph convolutional networks, Iscience, 20 (2019), 265–277. https://doi.org/10.1016/j.isci.2019.09.013 doi: 10.1016/j.isci.2019.09.013
    [33] M. Tsubaki, K. Tomii, J. Sese, Compound-protein interaction prediction with end-to-end learning of neural networks for graphs and sequences, Bioinformatics, 35 (2019), 309–318. https://doi.org/10.1093/bioinformatics/bty535 doi: 10.1093/bioinformatics/bty535
    [34] D. P. Kingma, J. Ba, Adam: A method for stochastic optimization, in the 3rd International Conference for Learning Representations, Louisiana, USA, (2019).
    [35] R. Kohavi, A study of cross-validation and bootstrap for accuracy estimation and model selection, in IJCAI'95: Proceedings of the 14th International Joint Conference on Artificial Intelligence-Volume 2, (1995), 1137–1145.
    [36] D. Powers, Evaluation: From precision, recall and F-measure to ROC, informedness, markedness & correlation, J. Mach. Learn. Technol., 2 (2011), 37–63.
    [37] F. Huang, Q. Ma, J. Ren, J. Li, F. Wang, T. Huang, et al., Identification of smoking associated transcriptome aberration in blood with machine learning methods, BioMed Res. Int., 2023 (2023), 5333361. https://doi.org/10.1155/2023/5333361 doi: 10.1155/2023/5333361
    [38] H. Wang, L. Chen, PMPTCE-HNEA: Predicting metabolic pathway types of chemicals and enzymes with a heterogeneous network embedding algorithm, Curr. Bioinf., 18 (2023), 748–759. https://doi.org/10.2174/1574893618666230224121633 doi: 10.2174/1574893618666230224121633
    [39] J. Ren, Y. Zhang, W. Guo, K. Feng, Y. Yuang, T. Huang, et al., Identification of genes associated with the impairment of olfactory and gustatory functions in COVID-19 via machine-learning methods, Life, 13 (2023), 798. https://doi.org/10.3390/life13030798 doi: 10.3390/life13030798
    [40] L. Chen, X. Zhao, PCDA-HNMP: Predicting circRNA-disease association using heterogeneous network and meta-path, Math. Biosci. Eng., 20 (2023), 20553–20575. https://doi.org/10.3934/mbe.2023909 doi: 10.3934/mbe.2023909
    [41] L. Chen, Y. Chen, RMTLysPTM: Recognizing multiple types of lysine PTM sites by deep analysis on sequences, Briefings Bioinf., 25 (2024), bbad450. https://doi.org/10.1093/bib/bbad450 doi: 10.1093/bib/bbad450
    [42] L. Chen, R. Qu, X. Liu, Improved multi-label classifiers for predicting protein subcellular localization, Math. Biosci. Eng., 21 (2024), 214–236. https://doi.org/10.3934/mbe.2024010 doi: 10.3934/mbe.2024010
    [43] B. Matthews, Comparison of the predicted and observed secondary structure of T4 phage lysozyme, Biochim. Biophys. Acta Protein Struct., 405 (1975), 442–451. https://doi.org/10.1016/0005-2795(75)90109-9 doi: 10.1016/0005-2795(75)90109-9
    [44] K. B. Walsh, A. E. McKinney, A. E. Holmes, Minor cannabinoids: Biosynthesis, molecular pharmacology and potential therapeutic uses, Front. Pharmacol., 12 (2021), 777804. https://doi.org/10.3389/fphar.2021.777804 doi: 10.3389/fphar.2021.777804
    [45] B. Rezende, A. K. N. Alencar, G. F. de Bem, F. L. Fontes-Dantas, G. C. Montes, Endocannabinoid system: Chemical characteristics and biological activity, Pharmaceuticals (Basel), 16 (2023), 148. https://doi.org/10.3390/ph16020148 doi: 10.3390/ph16020148
    [46] P. B. Sampson, Phytocannabinoid pharmacology: Medicinal properties of cannabis sativa constituents aside from the "Big Two", J. Nat. Prod., 84 (2021), 142–160. https://doi.org/10.1021/acs.jnatprod.0c00965 doi: 10.1021/acs.jnatprod.0c00965
    [47] InMed pharmaceuticals announces commencement of phase 2 clinical trial investigating cannabinol (CBN), a rare cannabinoid, in the treatment of epidermolysis bullosa, InMed Pharmaceuticals Inc., 2021.
    [48] P. Clayton, M. Hill, N. Bogoda, S. Subah, R. Venkatesh, Palmitoylethanolamide: A natural compound for health management, Int. J. Mol. Sci., 22 (2021), 5305. https://doi.org/10.3390/ijms22105305 doi: 10.3390/ijms22105305
    [49] P. Clayton, S. Subah, R. Venkatesh, M. Hill, N. Bogoda, Palmitoylethanolamide: A potential alternative to cannabidiol, J. Diet. Suppl., 20 (2023), 505–530. https://doi.org/10.1080/19390211.2021.2005733 doi: 10.1080/19390211.2021.2005733
    [50] E. B. Russo, Taming THC: Potential cannabis synergy and phytocannabinoid-terpenoid entourage effects, Br. J. Pharmacol., 163 (2011), 1344–1364. https://doi.org/10.1111/j.1476-5381.2011.01238.x doi: 10.1111/j.1476-5381.2011.01238.x
    [51] W. S. Ho, D. A. Barrett, M. D. Randall, 'Entourage' effects of N-palmitoylethanolamide and N-oleoylethanolamide on vasorelaxation to anandamide occur through TRPV1 receptors, Br. J. Pharmacol., 155 (2008), 837–846. https://doi.org/10.1038/bjp.2008.324 doi: 10.1038/bjp.2008.324
    [52] A. Mabou Tagne, Y. Fotio, L. Lin, E. Squire, F. Ahmed, T. I. Rashid, et al., Palmitoylethanolamide and hemp oil extract exert synergistic anti-nociceptive effects in mouse models of acute and chronic pain, Pharmacol. Res., 167 (2021), 105545. https://doi.org/10.1016/j.phrs.2021.105545 doi: 10.1016/j.phrs.2021.105545
    [53] M. H. Bloch, A. Landeros-Weisenberger, J. A. Johnson, J. F. Leckman, A phase-2 pilot study of a therapeutic combination of Delta (9)-Tetrahydracannabinol and Palmitoylethanolamide for adults with Tourette's syndrome, J. Neuropsychiatry Clin. Neurosci., 33 (2021), 328–336. https://doi.org/10.1176/appi.neuropsych.19080178 doi: 10.1176/appi.neuropsych.19080178
    [54] J. Lott, E. M. Jutkiewicz, M. A. Puthenveedu, The synthetic cannabinoid WIN55,212-2 can disrupt the Golgi apparatus independent of cannabinoid receptor-1, Mol. Pharmacol., 101 (2022), 371–380. https://doi.org/10.1124/molpharm.121.000377 doi: 10.1124/molpharm.121.000377
    [55] J. E. Kuster, J. I. Stevenson, S. J. Ward, T. E. D'Ambra, D. A. Haycock, Aminoalkylindole binding in rat cerebellum: Selective displacement by natural and synthetic cannabinoids, J. Pharmacol. Exp. Ther., 264 (1993), 1352–1363.
    [56] L. Ferraro, M. C. Tomasini, G. L. Gessa, B. W. Bebe, S. Tanganelli, T. Antonelli, The cannabinoid receptor agonist WIN 55,212-2 regulates glutamate transmission in rat cerebral cortex: An in vivo and in vitro study, Cereb. Cortex, 11 (2001), 728–733.
    [57] S. E. O'Sullivan, An update on PPAR activation by cannabinoids, Br. J. Pharmacol., 173 (2016), 1899–1910. https://doi.org/10.1111/bph.13497 doi: 10.1111/bph.13497
    [58] J. A. Fields, M. K. Swinton, P. Montilla-Perez, E. Ricciardelli, F. Telese, The cannabinoid receptor agonist, WIN-55212-2, suppresses the activation of proinflammatory genes induced by interleukin 1 beta in human astrocytes, Cannabis Cannabinoid Res., 7 (2022), 78–92. https://doi.org/10.1089/can.2020.0128 doi: 10.1089/can.2020.0128
    [59] G. T. Carter, S. P. Javaher, M. H. Nguyen, S. Garret, B. H. Carlini, Re-branding cannabis: The next generation of chronic pain medicine, Pain Manage., 5 (2015), 13–21. https://doi.org/10.2217/pmt.14.49 doi: 10.2217/pmt.14.49
    [60] J. P. Szaflarski, E. M. Bebin, Cannabis, cannabidiol, and epilepsy--from receptors to clinical response, Epilepsy Behav., 41 (2014), 277–282. https://doi.org/10.1016/j.yebeh.2014.08.135 doi: 10.1016/j.yebeh.2014.08.135
    [61] J. K. Fitzpatrick, E. J. Downer, Toll-like receptor signalling as a cannabinoid target in multiple sclerosis, Neuropharmacology, 113 (2017), 618–626. https://doi.org/10.1016/j.neuropharm.2016.04.009 doi: 10.1016/j.neuropharm.2016.04.009
    [62] S. G. Fagan, V. A. Campbell, The influence of cannabinoids on generic traits of neurodegeneration, Br. J. Pharmacol., 171 (2014), 1347–1360. https://doi.org/10.1016/j.neuropharm.2016.04.009 doi: 10.1016/j.neuropharm.2016.04.009
    [63] D. An, S. Peigneur, J. Tytgat, WIN55,212-2, a dual modulator of cannabinoid receptors and G protein-coupled inward rectifier potassium channels, Biomedicines, 9 (2021), 484. https://doi.org/10.3390/biomedicines9050484 doi: 10.3390/biomedicines9050484
    [64] C. J. Wenthur, B. Zhou, K. D. Janda, Vaccine-driven pharmacodynamic dissection and mitigation of fenethylline psychoactivity, Nature, 548 (2017), 476–479. https://doi.org/10.1038/nature23464 doi: 10.1038/nature23464
    [65] I. M. Johnson, H. Prakash, J. Prathiba, R. Raghunathan, R. Malathi, Spectral analysis of naturally occurring methylxanthines (theophylline, theobromine and caffeine) binding with DNA, PLoS One, 7 (2012), e50019. https://doi.org/10.1371/journal.pone.0050019 doi: 10.1371/journal.pone.0050019
    [66] M. Wepler, J. M. Preuss, T. Merz, O. McCook, P. Radermacher, J. P. Tuckermann, et al., Impact of downstream effects of glucocorticoid receptor dysfunction on organ function in critical illness-associated systemic inflammation, Intensive. Care Med. Exp., 8 (2020), 37. https://doi.org/10.1186/s40635-020-00325-z doi: 10.1186/s40635-020-00325-z
    [67] A. E. Coutinho, K. E. Chapman, The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights, Mol. Cell. Endocrinol., 335 (2011), 2–13. https://doi.org/10.1016/j.mce.2010.04.005 doi: 10.1016/j.mce.2010.04.005
  • Reader Comments
  • © 2024 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(601) PDF downloads(65) Cited by(0)

Article outline

Figures and Tables

Figures(4)  /  Tables(6)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog