1
|
Yamane F, Ikemura K, Kondo M, Ueno M, Okuda M. Identification of dequalinium as a potent inhibitor of human organic cation transporter 2 by machine learning based QSAR model. Sci Rep 2025; 15:2581. [PMID: 39833227 DOI: 10.1038/s41598-024-79377-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/08/2024] [Indexed: 01/22/2025] Open
Abstract
Human organic cation transporter 2 (hOCT2/SLC22A2) is a key drug transporter that facilitates the transport of endogenous and exogenous organic cations. Because hOCT2 is responsible for the development of adverse effects caused by platinum-based anti-cancer agents, drugs with OCT2 inhibitory effects may serve as prophylactic agents against the toxicity of platinum-based anti-cancer agents. In the present study, we established a machine learning-based quantitative structure-activity relationship (QSAR) model for hOCT2 inhibitors based on the public ChEMBL database and explored novel hOCT2 inhibitors among the FDA-approved drugs. Using our QSAR model, we identified 162 candidate hOCT2 inhibitors among the FDA-approved drugs registered in the DrugBank database. After manual selection and in vitro assays, we found that dequalinium, a quaternary ammonium cation antimicrobial agent, is a potent hOCT2 inhibitor (IC50 = 88.16 ± 7.14 nM). Moreover, dequalinium inhibited hOCT2-mediated transport of platinum anti-cancer agents (cisplatin and oxaliplatin) in a concentration-dependent manner. Our study is the first to demonstrate the construction of a novel machine learning-based QSAR model for hOCT2 inhibitors and identify a novel hOCT2 inhibitor among FDA-approved drugs using this model.
Collapse
Affiliation(s)
- Fumihiro Yamane
- Department of Hospital Pharmacy, School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kenji Ikemura
- Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Masayoshi Kondo
- Department of Hospital Pharmacy, School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Manami Ueno
- Department of Hospital Pharmacy, School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Masahiro Okuda
- Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
2
|
Hang NT, Duy ND, Anh TDH, Mai LTN, Loan NTB, Cong NT, Phuong NV. Enhanced prediction of beta-secretase inhibitory compounds with mol2vec technique and machine learning algorithms. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:1109-1127. [PMID: 39704060 DOI: 10.1080/1062936x.2024.2440903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
A comprehensive computational strategy that combined QSAR modelling, molecular docking, and ADMET analysis was used to discover potential inhibitors for β-secretase 1 (BACE-1). A dataset of 1,138 compounds with established BACE-1 inhibitory activities was used to build a QSAR model using mol2vec descriptors and support vector regression. The obtained model demonstrated strong predictive performance (training set: r2 = 0.790, RMSE = 0.540, MAE = 0.362; test set: r2 = 0.705, RMSE = 0.641, MAE = 0.495), indicating its reliability in identifying potent BACE-1 inhibitors. By applying this QSAR model together with molecular docking, seven compounds (ZINC8790287, ZINC20464117, ZINC8878274, ZINC96116481, ZINC217682404, ZINC217786309 and ZINC96113994) were identified as promising candidates, exhibiting predicted log IC50 values ranging from 0.361 to 1.993 and binding energies ranging from -10.8 to -10.7 kcal/mol. Further analysis using ADMET studies and molecular dynamics simulations provided further support for the potential of compound 279 (ZINC96116481) and compound 945 (ZINC96113994) as drug candidates. However, since our study is purely theoretical, further experimental validation through in vitro and in vivo studies is essential to confirm these promising findings.
Collapse
Affiliation(s)
- N T Hang
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - N D Duy
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - T D H Anh
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - L T N Mai
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - N T B Loan
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - N T Cong
- Faculty of Pharmacy, Dai Nam University, Hanoi, Vietnam
| | - N V Phuong
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, Vietnam
| |
Collapse
|
3
|
Hang NT, My TTK, Van Anh LT, Van Anh PT, Anh TDH, Van Phuong N. Identification of potential FAK inhibitors using mol2vec molecular descriptor-based QSAR, molecular docking, ADMET study, and molecular dynamics simulation. Mol Divers 2024; 28:2163-2175. [PMID: 38582821 DOI: 10.1007/s11030-024-10839-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/07/2024] [Indexed: 04/08/2024]
Abstract
This study aims to identify potential focal adhesion kinase (FAK) inhibitors through an integrated computational approach, combining mol2vec descriptor-based QSAR, molecular docking, ADMET study, and molecular dynamics simulation. A dataset of 437 compounds with known FAK inhibitory activities was used to develop QSAR models using machine learning algorithms combined with mol2vec descriptors. Subsequently, the most promising compounds were subjected to molecular docking against FAK to evaluate their binding affinities and key interactions. ADMET study and molecular dynamics simulation were also employed to investigate the pharmacokinetic, drug-like properties, and the stability of the protein-ligand complexes. The results showed that the mol2vec descriptor-based QSAR model established by support vector regression demonstrated good predictive performance (R2 = 0.813, RMSE = 0.453, MAE = 0.263 in case of training set, and R2 = 0.729, RMSE = 0.635, MAE = 0.477 in case of test set), indicating their reliability in identifying potent FAK inhibitors. Using this QSAR model and molecular docking, compound 21 (ZINC000004523722) was identified as the most potential compound, with predicted logIC50 value and binding energy of 2.59 and - 9.3 kcal/mol, respectively. The results of molecular dynamics simulation and ADMET study also further suggested its potential as a promising drug candidate. However, because our research was merely theoretical, additional in vitro and in vivo studies are required for the verification of these results.
Collapse
Affiliation(s)
- Nguyen Thu Hang
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam
| | - Than Thi Kieu My
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam
| | - Le Thi Van Anh
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam
| | - Phan Thi Van Anh
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam
| | - Thai Doan Hoang Anh
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam
| | - Nguyen Van Phuong
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam.
| |
Collapse
|
4
|
Hang NT, Anh TDH, Thanh LN, Anh NV, Van Phuong N. In silico screening of Fyn kinase inhibitors using classification-based QSAR model, molecular docking, molecular dynamics and ADME study. Mol Divers 2024; 28:2217-2228. [PMID: 38886315 DOI: 10.1007/s11030-024-10905-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024]
Abstract
This study aimed to use a computational approach that combined the classification-based QSAR model, molecular docking, ADME studies, and molecular dynamics (MD) to identify potential inhibitors of Fyn kinase. First, a robust classification model was developed from a dataset of 1,078 compounds with known Fyn kinase inhibitory activity, using the XGBoost algorithm. After that, molecular docking was performed between potential compounds identified from the QSAR model and Fyn kinase to assess their binding strengths and key interactions, followed by MD simulations. ADME studies were additionally conducted to preliminarily evaluate the pharmacokinetics and drug-like characteristics of these compounds. The results showed that our obtained model exhibited good predictive performance with an accuracy of 0.95 on the test set, affirming its reliability in identifying potent Fyn kinase inhibitors. Through the application of this model in conjunction with molecular docking and ADME studies, nine compounds were identified as potential Fyn kinase inhibitors, including 208 (ZINC70708110), 728 (ZINC8792432), 734 (ZINC8792187), 736 (ZINC8792350), 738 (ZINC8792286), 739 (ZINC8792309), 817 (ZINC33901069), 852 (ZINC20759145), and 1227 (ZINC100006936). MD simulations further demonstrated that the four most promising compounds, 728, 734, 736, and 852 exhibited stable binding with Fyn kinase during the simulation process. Additionally, a web-based platform ( https://fynkinase.streamlit.app/ ) has been developed to streamline the screening process. This platform enables users to predict the activity of their substances of interest on Fyn kinase from their SMILES, using our classification-based QSAR model and molecular docking.
Collapse
Affiliation(s)
- Nguyen Thu Hang
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam
| | - Thai Doan Hoang Anh
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam
| | - Le Nguyen Thanh
- Department of Analytical Chemistry and Standardization, National Institute of Medicinal Materials, 3B Quang Trung, Hanoi, 10000, Vietnam
| | - Nguyen Viet Anh
- Institute of Information Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, 10000, Vietnam
| | - Nguyen Van Phuong
- Department of Pharmacognosy, Faculty of Pharmacognosy and Traditional Medicine, Hanoi University of Pharmacy, Hanoi, 11000, Vietnam.
| |
Collapse
|
5
|
Duan H, Lou C, Gu Y, Wang Y, Li W, Liu G, Tang Y. In Silico prediction of inhibitors for multiple transporters via machine learning methods. Mol Inform 2024; 43:e202300270. [PMID: 38235949 DOI: 10.1002/minf.202300270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/02/2024] [Accepted: 01/17/2024] [Indexed: 01/19/2024]
Abstract
Transporters play an indispensable role in facilitating the transport of nutrients, signaling molecules and the elimination of metabolites and toxins in human cells. Contemporary computational methods have been employed in the prediction of transporter inhibitors. However, these methods often focus on isolated endpoints, overlooking the interactions between transporters and lacking good interpretation. In this study, we integrated a comprehensive dataset and constructed models to assess the inhibitory effects on seven transporters. Both conventional machine learning and multi-task deep learning methods were employed. The results demonstrated that the MLT-GAT model achieved superior performance with an average AUC value of 0.882. It is noteworthy that our model excels not only in prediction performance but also in achieving robust interpretability, aided by GNN-Explainer. It provided valuable insights into transporter inhibition. The reliability of our model's predictions positioned it as a promising and valuable tool in the field of transporter inhibition research. Related data and code are available at https://gitee.com/wutiantian99/transporter_code.git.
Collapse
Affiliation(s)
- Hao Duan
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Chaofeng Lou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yaxin Gu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yimeng Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|