BiGraph-DTA: Predicting drug-target interactions of hepatoprotective agents with graph convolutional networks

Arief Sartono , Bambang Riyanto Trilaksono , Sophi Damayanti , Anto Satriyo Nugroho , Firdayani Firdayani

Quant. Biol. ›› 2026, Vol. 14 ›› Issue (1) : e70022

PDF (1966KB)
Quant. Biol. ›› 2026, Vol. 14 ›› Issue (1) : e70022 DOI: 10.1002/qub2.70022
RESEARCH ARTICLE

BiGraph-DTA: Predicting drug-target interactions of hepatoprotective agents with graph convolutional networks

Author information +
History +
PDF (1966KB)

Abstract

Predicting drug-target affinity (DTA) is critical for discovering and developing hepatoprotective agents that can prevent and treat liver diseases. In this study, we propose BiGraph-DTA, a new predictive model for identifying DTA score prediction for hepatoprotective compounds by combining graph convolutional networks and bidirectional long short-term memory networks. This model is based on powerful frameworks that process both graph representations of molecular structures and sequential information from protein sequences to capture complex dependencies and interactions. Leveraging a curated hepatoprotective dataset (from ChEMBL) consisting of 21,421 interactions, the model outperforms traditional machine learning methods (such as random forest and XGBoost) as well as other deep learning methods (such as DeepDTA and GraphDTA) in terms of predictive performance. The BiGraph-DTA obtained the best mean squared error of 0.7885, R2 of 0.7208, and concordance index of 0.8508. Our proposed architecture holds potential for accelerating the drug discovery process of hepatoprotective therapy by highlighting the framework through which candidate drugs and their corresponding protein targets can be identified based on robust data-driven knowledge. This model, therefore, provides a new opportunity for discovering new hepatoprotective compounds, which may also make it possible to speed up finding new liver disease drugs.

Keywords

BiGraph-DTA / drug discovery / drug-target affinity score prediction / graph convolutional network (GCN) / hepatoprotective agents

Cite this article

Download citation ▾
Arief Sartono, Bambang Riyanto Trilaksono, Sophi Damayanti, Anto Satriyo Nugroho, Firdayani Firdayani. BiGraph-DTA: Predicting drug-target interactions of hepatoprotective agents with graph convolutional networks. Quant. Biol., 2026, 14(1): e70022 DOI:10.1002/qub2.70022

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Chupradit S , Bokov D , Zamanian MY , Heidari M , Hakimizadeh E . Hepatoprotective and therapeutic effects of resveratrol: a focus on anti-inflammatory and antioxidative activities. Fund Clin Pharmacol. 2022; 36 (3): 468- 85.

[2]

Jha K , Saha S , Singh H . Prediction of protein-protein interaction using graph neural networks. Sci Rep. 2022; 12 (1): 8360.

[3]

Jiang M , Li Z , Zhang S , Wang S , Wang X , Yuan Q , et al. Drug-target affinity prediction using graph neural network and contact maps. RSC Adv. 2020; 10 (35): 20701- 12.

[4]

Peng J , Li J , Shang X . learning-based method for drug-target interaction prediction based on feature representation learning and deep neural network. BMC Bioinf. 2020; 21: (S13): 394.

[5]

Ai H , Chen W , Zhang L , Huang L , Yin Z , Hu H , et al. Predicting drug-induced liver injury using ensemble learning methods and molecular fingerprints. Toxicol Sci. 2018; 165 (1): 100- 7.

[6]

Ahn JC , Connell A , Simonetto DA , Hughes C , Shah VH . Application of artificial intelligence for the diagnosis and treatment of liver diseases. Hepatology. 2021; 73 (6): 2546- 63.

[7]

Zhang Z , Chen L , Zhong F , Wang D , Jiang J , Zhang S , et al. Graph neural network approaches for drug-target interactions. Curr Opin Struct Biol. 2022; 73: 102327.

[8]

Zhang S , Tong H , Xu J , Maciejewski R . Graph convolutional networks: a comprehensive review. Computational Social Networks. 2019; 6 (1): 11.

[9]

Zhang X-M , Liang L , Liu L , Tang M-J . Graph neural networks and their current applications in bioinformatics. Front Genet. 2021; 12: 690049.

[10]

Zhang Y , Hu Y , Han N , Yang A , Liu X , Cai H . A survey of drug-target interaction and affinity prediction methods via graph neural networks. Comput Biol Med. 2023; 163: 107136.

[11]

He S , Ye T , Wang R , Zhang C , Zhang X , Sun G , et al. An in silico model for predicting drug-induced hepatotoxicity. Int J Mol Sci. 2019; 20 (8): 1897.

[12]

Gaudelet T , Day B , Jamasb AR , Soman J , Regep C , Liu G , et al. Utilizing graph machine learning within drug discovery and development. Briefings Bioinf. 2021; 22 (6): bbab159.

[13]

Wang H , Liu R , Schyman P , Wallqvist A . Deep neural network models for predicting chemically induced liver toxicity endpoints from transcriptomic responses. Front Pharmacol. 2019; 10: 42.

[14]

Xiong J , Xiong Z , Chen K , Jiang H , Zheng M . Graph neural networks for automated de novo drug design. Drug Discov Today. 2021; 26 (6): 1382- 93.

[15]

Nguyen T , Le H , Quinn TP , Nguyen T , Le TD , Venkatesh S . GraphDTA: predicting drug-target binding affinity with graph neural networks. Bioinformatics. 2021; 37 (8): 1140- 7.

[16]

Zhang L , Song H , Aletras N , Lu H . Graph node-feature convolution for representation learning. 2018. Preprint at arXiv: 1812.00086.

[17]

Viñas R , Zheng X , Hayes J . A graph-based imputation method for sparse medical records. 2023. Preprint at arXiv: 2111.09084.

[18]

Wan N , Zhang Y . Enhancing temporal dynamics and hypergraph convolutions for sequential recommendation. In: 202410th international conference on big data and information analytics (BigDIA). Chiang Mai, Thailand: IEEE; 2024. p. 794- 801.

[19]

Zhang W , Zhang K , Jiang L , Zhang F . Multi-scale multi-step dependency graph neural network for multivariate time-series forecasting. Communications in Computer and Information Science. 2024: 95- 111.

[20]

Zhao Q , Duan G , Zhao H , Zheng K , Li Y , Wang J . GIFDTI: prediction of drug-target interactions based on global molecular and intermolecular interaction representation learning. IEEE ACM Trans Comput Biol Bioinf. 2023; 20 (3): 1943- 52.

[21]

Wang P , Zheng S , Jiang Y , Li C , Liu J , Wen C , et al. X-DPI: a structure-aware multi-modal deep learning model for drug-protein interactions prediction. 2021. Preprint at bioRxiv: 2021.06.17.448780.

[22]

Wang P , Zheng S , Jiang Y , Li C , Liu J , Wen C , et al. Structure-aware multimodal deep learning for drug-protein interaction prediction. J Chem Inf Model. 2022; 62 (5): 1308- 17.

[23]

Xia C-Q , Pan X , Shen H-B . Protein-ligand binding residue prediction enhancement through hybrid deep heterogeneous learning of sequence and structure data. Bioinformatics. 2020; 36 (10): 3018- 27.

[24]

Lim S , Kim Y , Gu J , Lee S , Shin W , Kim S . Supervised chemical graph mining improves drug-induced liver injury prediction. iScience. 2023; 26 (1): 105677.

[25]

Hu K-Q . Cyclooxygenase 2(COX2)-prostanoid pathway and liver diseases. Prostagl Leukot Essent Fat Acids. 2003; 69 (5): 329- 37.

[26]

Wang H , Zhang R , Zhu Y , Teng T , Cheng Y , Chowdhury A , et al. Microsomal prostaglandin E synthase 2 deficiency is resistant to acetaminophen-induced liver injury. Arch Toxicol. 2019; 93 (10): 2863- 78.

[27]

Kumar V , Xin X , Ma J , Tan C , Osna N , Mahato RI . Therapeutic targets, novel drugs, and delivery systems for diabetes associated NAFLD and liver fibrosis. Adv Drug Deliv Rev. 2021; 176: 113888.

[28]

Ahmadi Badi S , Bereimipour A , Rohani P , Khatami S , Siadat SD . Interplay between gut microbiota and the master iron regulator, hepcidin, in the pathogenesis of liver fibrosis. Pathogens and Disease. 2024; 82: ftae005.

[29]

Liu K , Chen X , Ren Y , Liu C , Lv T , Liu Y , et al. Multi-target-based polypharmacology prediction (mTPP): an approach using virtual screening and machine learning for multi-target drug discovery. Chem Biol Interact. 2022; 368: 110239.

[30]

Vernetti LA , Vogt A , Gough A , Taylor DL . Evolution of experimental models of the liver to predict human drug hepatotoxicity and efficacy. Clin Liver Dis. 2017; 21 (1): 197- 214.

[31]

Lim J , Ryu S , Park K , Choe YJ , Ham J , Kim WY . Predicting drug-target interaction using a novel graph neural network with 3D structure-embedded graph representation. J Chem Inf Model. 2019; 59 (9): 3981- 8.

[32]

Tran HNT , Thomas JJ , Ahamed Hassain Malim NH . DeepNC: a framework for drug-target interaction prediction with graph neural networks. PeerJ. 2022; 10: e13163.

[33]

Zhu Z . Systematic optimization of overfitting problem in machine learning. Highlights in Science, Engineering and Technology. 2024; 111: 353- 9.

[34]

Khan SH , Hayat M , Porikli F . Regularization of deep neural networks with spectral dropout. Neural Netw. 2019; 110: 82- 90.

[35]

Salman S , Liu X . Overfitting mechanism and avoidance in deep neural networks. 2019. Preprint at arXiv: 1901.06566.

[36]

Yin W , Kann K . Yu M , Schütze H . Comparative study of CNN and RNN for natural language processing. 2017. Preprint at arXiv: 1702.01923.

[37]

Davis MI , Hunt JP , Herrgard S , Ciceri P , Wodicka LM , Pallares G , et al. Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol. 2011; 29 (11): 1046- 51.

[38]

Tang J , Szwajda A , Shakyawar S , Xu T , Hintsanen P , Wennerberg K , et al. Making sense of large-scale kinase inhibitor bioactivity data sets: a comparative and integrative analysis. J Chem Inf Model. 2014; 54 (3): 735- 43.

[39]

Stretch C , Khan S , Asgarian N , Eisner R , Vaisipour S , Damaraju S , et al. Effects of sample size on differential gene expression, rank order and prediction accuracy of a gene signature. PLoS One. 2013; 8 (6): e65380.

[40]

Yuan H , Liu M , Kang L , Miao C , Wu Y . An empirical study of the effect of background data size on the stability of SHapley Additive exPlanations (SHAP) for deep learning models. 2022. Preprint at arXiv: 2204.11351.

[41]

Aguiar C , Camps I . Molecular docking in drug discovery: techniques, applications, and advancements. Curr Med Chem. 2024; 32 (28): 5924- 38.

[42]

Wang L , He X , Ji B , Han F , Niu T , Cai L , et al. Geometry optimization algorithms in conjunction with the machine learning potential ANI-2x facilitate the structure-based virtual screening and binding mode prediction. Biomolecules. 2024; 14 (6): 648.

[43]

Chin R , Maass AI , Ulapane N , Manzie C , Shames I , Nešić D , et al. Active learning for linear parameter-varying system identification. IFAC-PapersOnLine. 2020; 53 (2): 989- 94.

[44]

Mohamadi S , Amindavar H . Deep bayesian active learning, A Brief survey on recent advances. 2020. Preprint at arXiv: 2012.08044.

[45]

Bolton EE , Wang Y , Thiessen PA , Bryant SH . PubChem: integrated platform of small molecules and biological activities. In: Annual reports in computational chemistry. Amsterdam: Elsevier; 2008. p. 217- 41.

[46]

Domitrović R , Potočnjak I . A comprehensive overview of hepatoprotective natural compounds: mechanism of action and clinical perspectives. Arch Toxicol. 2016; 90 (1): 39- 79.

[47]

He T , Heidemeyer M , Ban F , Cherkasov A , Ester M . SimBoost: a read-across approach for predicting drug-target binding affinities using gradient boosting machines. J Cheminf. 2017; 9 (1): 24.

[48]

Iresha MR , Firdayani F , Sani AW , Karimah N , Listiana S , Tanasa IY , et al. Machine learning model and molecular docking for screening medicinal plants as HIV-1 reverse transcriptase inhibitors. Karbala International Journal of Modern Science. 2024; 10 (1): 7.

[49]

Kipf TN , Welling M . Semi-supervised classification with Graph Convolutional Networks. 2017. Preprint at arXiv: 1609.02907.

[50]

Baker H , Hallowell MR , Tixier AJ-P . AI-based prediction of independent construction safety outcomes from universal attributes. Autom ConStruct. 2020; 118: 103146.

[51]

Öztürk H , Özgür A , Ozkirimli E . DeepDTA: deep drug-target binding affinity prediction. Bioinformatics. 2018; 34 (17): i821- 9.

[52]

Srivastava N , Hinton G , Krizhevsky A , Sutskever I , Salakhutdinov R . Dropout: a simple way to prevent neural networks from overfitting. J Mach Learn Res. 2014; 15 (1): 1929- 58.

[53]

LeCun Y , Bengio Y , Hinton G . Deep learning. Nature. 2015; 521 (7553): 436- 44.

[54]

Breiman L . Random forests. Mach Learn. 2001; 45 (1): 5- 32.

[55]

Sahin EK , Colkesen I , Kavzoglu T . A comparative assessment of canonical correlation forest, random forest, rotation forest and logistic regression methods for landslide susceptibility mapping. Geocarto Int. 2020; 35 (4): 341- 63.

[56]

Barzegar R , Razzagh S , Quilty J , Adamowski J , Kheyrollah Pour H , Booij MJ . Improving GALDIT-based groundwater vulnerability predictive mapping using coupled resampling algorithms and machine learning models. J Hydrol. 2021; 598: 126370.

[57]

Kavzoglu T , Teke A . Predictive performances of ensemble machine learning algorithms in landslide susceptibility mapping using random forest, extreme gradient boosting (XGBoost) and natural gradient boosting (NGBoost). Arabian J Sci Eng. 2022; 47 (6): 7367- 85.

[58]

Guo X , Gui X , Xiong H , Hu X , Li Y , Cui H , et al. Critical role of climate factors for groundwater potential mapping in arid regions: insights from random forest, XGBoost, and LightGBM algorithms. J Hydrol. 2023; 621: 129599.

[59]

Chen T , Guestrin C . XGBoost: a scalable tree boosting system. In: Proceedings of the 22nd ACM SIGKDD international conference on knowledge discovery and data mining. San Francisco, CA, USA: Association for Computing Machinery (ACM); 2016. p. 785- 94.

[60]

Li L , Qiao J , Yu G , Wang L , Li H-Y , Liao C , et al. Interpretable tree-based ensemble model for predicting beach water quality. Water Res. 2022; 211: 118078.

RIGHTS & PERMISSIONS

The Author(s). Quantitative Biology published by John Wiley & Sons Australia, Ltd on behalf of Higher Education Press.

AI Summary AI Mindmap
PDF (1966KB)

43

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/