1
|
Wang J, He R, Wang X, Li H, Lu Y. MCF-DTI: Multi-Scale Convolutional Local-Global Feature Fusion for Drug-Target Interaction Prediction. Molecules 2025; 30:274. [PMID: 39860144 PMCID: PMC11767603 DOI: 10.3390/molecules30020274] [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: 11/26/2024] [Revised: 12/21/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Predicting drug-target interactions (DTIs) is a crucial step in the development of new drugs and drug repurposing. In this paper, we propose a novel drug-target prediction model called MCF-DTI. The model utilizes the SMILES representation of drugs and the sequence features of targets, employing a multi-scale convolutional neural network (MSCNN) with parallel shared-weight modules to extract features from the drug side. For the target side, it combines MSCNN with Transformer modules to capture both local and global features effectively. The extracted features are then weighted and fused, enabling comprehensive feature representation to enhance the predictive power of the model. Experimental results on the Davis dataset demonstrate that MCF-DTI achieves an AUC of 0.9746 and an AUPR of 0.9542, outperforming other state-of-the-art models. Our case study demonstrates that our model effectively validated several known drug-target relationships in lung cancer and predicted the therapeutic potential of certain preclinical compounds in treating lung cancer. These findings contribute valuable insights for subsequent drug repurposing efforts and novel drug development.
Collapse
Affiliation(s)
- Jihong Wang
- School of Computer, Guangdong University of Education, Guangzhou 510310, China
| | - Ruijia He
- School of Computer, Guangdong University of Education, Guangzhou 510310, China
| | - Xiaodan Wang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Hongjian Li
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Yulei Lu
- School of Computer, Guangdong University of Education, Guangzhou 510310, China
| |
Collapse
|
2
|
de Alencar Morais Lima W, de Souza JG, García-Villén F, Loureiro JL, Raffin FN, Fernandes MAC, Souto EB, Severino P, Barbosa RDM. Next-generation pediatric care: nanotechnology-based and AI-driven solutions for cardiovascular, respiratory, and gastrointestinal disorders. World J Pediatr 2025; 21:8-28. [PMID: 39192003 DOI: 10.1007/s12519-024-00834-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/21/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Global pediatric healthcare reveals significant morbidity and mortality rates linked to respiratory, cardiac, and gastrointestinal disorders in children and newborns, mostly due to the complexity of therapeutic management in pediatrics and neonatology, owing to the lack of suitable dosage forms for these patients, often rendering them "therapeutic orphans". The development and application of pediatric drug formulations encounter numerous challenges, including physiological heterogeneity within age groups, limited profitability for the pharmaceutical industry, and ethical and clinical constraints. Many drugs are used unlicensed or off-label, posing a high risk of toxicity and reduced efficacy. Despite these circumstances, some regulatory changes are being performed, thus thrusting research innovation in this field. DATA SOURCES Up-to-date peer-reviewed journal articles, books, government and institutional reports, data repositories and databases were used as main data sources. RESULTS Among the main strategies proposed to address the current pediatric care situation, nanotechnology is specially promising for pediatric respiratory diseases since they offer a non-invasive, versatile, tunable, site-specific drug release. Tissue engineering is in the spotlight as strategy to address pediatric cardiac diseases, together with theragnostic systems. The integration of nanotechnology and theragnostic stands poised to refine and propel nanomedicine approaches, ushering in an era of innovative and personalized drug delivery for pediatric patients. Finally, the intersection of drug repurposing and artificial intelligence tools in pediatric healthcare holds great potential. This promises not only to enhance efficiency in drug development in general, but also in the pediatric field, hopefully boosting clinical trials for this population. CONCLUSIONS Despite the long road ahead, the deepening of nanotechnology, the evolution of tissue engineering, and the combination of traditional techniques with artificial intelligence are the most recently reported strategies in the specific field of pediatric therapeutics.
Collapse
Affiliation(s)
| | - Jackson G de Souza
- InovAI Lab, nPITI/IMD, Federal University of Rio Grande Do Norte, Natal, RN, 59078-970, Brazil
| | - Fátima García-Villén
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Granada, Campus of Cartuja, 18071, Granada, Spain.
| | - Julia Lira Loureiro
- Laboratory of Galenic Pharmacy, Department of Pharmacy, Federal University of Rio Grande Do Norte, Natal, 59012-570, Brazil
| | - Fernanda Nervo Raffin
- Laboratory of Galenic Pharmacy, Department of Pharmacy, Federal University of Rio Grande Do Norte, Natal, 59012-570, Brazil
| | - Marcelo A C Fernandes
- InovAI Lab, nPITI/IMD, Federal University of Rio Grande Do Norte, Natal, RN, 59078-970, Brazil
- Department of Computer Engineering and Automation, Federal University of Rio Grande Do Norte, Natal, RN, 59078-970, Brazil
| | - Eliana B Souto
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Patricia Severino
- Industrial Biotechnology Program, University of Tiradentes (UNIT), Aracaju, Sergipe, 49032-490, Brazil
| | - Raquel de M Barbosa
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Seville, C/Professor García González, 2, 41012, Seville, Spain.
| |
Collapse
|
3
|
Liu B, Tsoumakas G. Integrating Similarities via Local Interaction Consistency and Optimizing Area Under the Curve Measures via Matrix Factorization for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2212-2225. [PMID: 39226198 DOI: 10.1109/tcbb.2024.3453499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
In drug discovery, identifying drug-target interactions (DTIs) via experimental approaches is a tedious and expensive procedure. Computational methods efficiently predict DTIs and recommend a small part of potential interacting pairs for further experimental confirmation, accelerating the drug discovery process. Although fusing heterogeneous drug and target similarities can improve the prediction ability, the existing similarity combination methods ignore the interaction consistency for neighbour entities. Furthermore, area under the precision-recall curve (AUPR) and area under the receiver operating characteristic curve (AUC) are two widely used evaluation metrics in DTI prediction. However, the two metrics are seldom considered as losses within existing DTI prediction methods. We propose a local interaction consistency (LIC) aware similarity integration method to fuse vital information from diverse views for DTI prediction models. Furthermore, we propose two matrix factorization (MF) methods that optimize AUPR and AUC using convex surrogate losses respectively, and then develop an ensemble MF approach that takes advantage of the two area under the curve metrics by combining the two single metric based MF models. Experimental results under different prediction settings show that the proposed methods outperform various competitors in terms of the metric(s) they optimize and are reliable in discovering potential new DTIs.
Collapse
|
4
|
Chen Z, Zhang L, Li R, Wang J, Chen L, Jin Y, Gao M, Han Z, Zhang K, Wang J, Li X, Yang C. The development and validation of a prognostic prediction modeling study in acute myocardial infarction patients after percutaneous coronary intervention: hemorrhea and major cardiovascular adverse events. J Thorac Dis 2024; 16:6216-6228. [PMID: 39444902 PMCID: PMC11494537 DOI: 10.21037/jtd-24-1362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024]
Abstract
Background Percutaneous coronary intervention (PCI) is one of the most important diagnostic and therapeutic techniques in cardiology. At present, the traditional prediction models for postoperative events after PCI are ineffective, but machine learning has great potential in identification and prediction of risk. Machine learning can reduce overfitting through regularization techniques, cross-validation and ensemble learning, making the model more accurate in predicting large amounts of complex unknown data. This study sought to identify the risk of hemorrhea and major adverse cardiovascular events (MACEs) in patients after PCI through machine learning. Methods The entire study population consisted of 7,931 individual patients who underwent PCI at Jiangsu Provincial Hospital and The Affiliated Wuxi Second People's Hospital from January 2007 to January 2022. The risk of postoperative hemorrhea and MACE (including cardiac death and in-stent restenosis) was predicted by 53 clinical features after admission. The population was assigned to the training set and the validation set in a specific ratio by simple randomization. Different machine learning algorithms, including eXtreme Gradient Boosting (XGBoost), random forest (RF), and deep learning neural network (DNN), were trained to build prediction models. A 5-fold cross-validation was applied to correct errors. Several evaluation indexes, including the area under the receiver operating characteristic (ROC) curve (AUC), accuracy (Acc), sensitivity (Sens), specificity (Spec), and net reclassification improvement (NRI), were used to compare the predictive performance. To improve the interpretability of the model and identify risk factors individually, SHapley Additive exPlanation (SHAP) was introduced. Results In this study, 306 patients (3.9%) experienced hemorrhea, 107 patients (1.3%) experienced cardiac death, and 218 patients (2.7%) developed in-stent restenosis. In the training set and validation set, except for previous PCI and statins, there were no significant differences. XGBoost was observed to be the best predictor of every event, namely hemorrhea [AUC: 0.921, 95% confidence interval (CI): 0.864-0.978, Acc: 0.845, Sens: 0.851, Spec: 0.837 and NRI: 0.140], cardiac death (AUC: 0.939, 95% CI: 0.903-0.975, Acc: 0.914, Sens: 0.950, Spec: 0.800 and NRI: 0.148), and in-stent restenosis (AUC: 0.915; 95% CI: 0.863-0.967, Acc: 0.834, Sens: 0.778, Spec: 0.902 and NRI: 0.077). SHAP showed that the number of stents had the greatest influence on hemorrhea, while age and drug-coated balloon were the main factors in cardiogenic death and stent restenosis (all P<0.05). Conclusions The XGBoost model (machine learning) performed better than the traditional logistic regression model in identifying hemorrhea and MACE after PCI. Machine learning models can be used as a tool for risk prediction. The machine learning model described in this study can personalize the prediction of hemorrhea and MACE after PCI for specific patients, helping clinicians adjust intervenable features.
Collapse
Affiliation(s)
- Zijie Chen
- Department of Cardiology, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Lizhu Zhang
- Department of Cardiology, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Rui Li
- Department of Clinical Research Center, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Jing Wang
- Department of Clinical Research Center, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Liang Chen
- Department of Cardiology, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Yan Jin
- Department of Cardiology, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Mingzhu Gao
- Department of Clinical Research Center, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Zhijun Han
- Department of Clinical Research Center, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Kaixin Zhang
- Department of Clinical Research Center, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Junhong Wang
- Department of Cardiology, Jiangsu Provincial Hospital, Nanjing Medical University, Nanjing, China
| | - Xing Li
- Department of Cardiology, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| | - Chengjian Yang
- Department of Cardiology, The Affiliated Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi, China
| |
Collapse
|
5
|
Wang W, Yu M, Sun B, Li J, Liu D, Zhang H, Wang X, Zhou Y. SMGCN: Multiple Similarity and Multiple Kernel Fusion Based Graph Convolutional Neural Network for Drug-Target Interactions Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:143-154. [PMID: 38051618 DOI: 10.1109/tcbb.2023.3339645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Accurately identifying potential drug-target interactions (DTIs) is a critical step in accelerating drug discovery. Despite many studies that have been conducted over the past decades, detecting DTIs remains a highly challenging and complicated process. Therefore, we propose a novel method called SMGCN, which combines multiple similarity and multiple kernel fusion based on Graph Convolutional Network (GCN) to predict DTIs. In order to capture the features of the network structure and fully explore direct or indirect relationships between nodes, we propose the method of multiple similarity, which combines similarity fusion matrices with Random Walk with Restart (RWR) and cosine similarity. Then, we use GCN to extract multi-layer low-dimensional embedding features. Unlike traditional GCN methods, we incorporate Multiple Kernel Learning (MKL). Finally, we use the Dual Laplace Regularized Least Squares method to predict novel DTIs through combinatorial kernels in drug and target spaces. We conduct experiments on a golden standard dataset, and demonstrate the effectiveness of our proposed model in predicting DTIs through showing significant improvements in Area Under the Curve (AUC) and Area Under the Precision-Recall Curve (AUPR). In addition, our model can also discover some new DTIs, which can be verified by the KEGG BRITE Database and relevant literature.
Collapse
|
6
|
Abdul Raheem AK, Dhannoon BN. Comprehensive Review on Drug-target Interaction Prediction - Latest Developments and Overview. Curr Drug Discov Technol 2024; 21:e010923220652. [PMID: 37680152 DOI: 10.2174/1570163820666230901160043] [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: 01/23/2023] [Revised: 05/29/2023] [Accepted: 07/18/2023] [Indexed: 09/09/2023]
Abstract
Drug-target interactions (DTIs) are an important part of the drug development process. When the drug (a chemical molecule) binds to a target (proteins or nucleic acids), it modulates the biological behavior/function of the target, returning it to its normal state. Predicting DTIs plays a vital role in the drug discovery (DD) process as it has the potential to enhance efficiency and reduce costs. However, DTI prediction poses significant challenges and expenses due to the time-consuming and costly nature of experimental assays. As a result, researchers have increased their efforts to identify the association between medications and targets in the hopes of speeding up drug development and shortening the time to market. This paper provides a detailed discussion of the initial stage in drug discovery, namely drug-target interactions. It focuses on exploring the application of machine learning methods within this step. Additionally, we aim to conduct a comprehensive review of relevant papers and databases utilized in this field. Drug target interaction prediction covers a wide range of applications: drug discovery, prediction of adverse effects and drug repositioning. The prediction of drugtarget interactions can be categorized into three main computational methods: docking simulation approaches, ligand-based methods, and machine-learning techniques.
Collapse
Affiliation(s)
- Ali K Abdul Raheem
- Software Department, College of Information Technology, University of Babylon, Hillah, Babil, Iraq
- University of Warith Al-Anbiyaa, Kerbala, Iraq
| | - Ban N Dhannoon
- Department of Computer Science, College of Science, Al-Nahrain University, Baghdad, Iraq
| |
Collapse
|
7
|
Nath A, Chaube R. Mining Chemogenomic Spaces for Prediction of Drug-Target Interactions. Methods Mol Biol 2024; 2714:155-169. [PMID: 37676598 DOI: 10.1007/978-1-0716-3441-7_9] [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] [Indexed: 09/08/2023]
Abstract
The pipeline of drug discovery consists of a number of processes; drug-target interaction determination is one of the salient steps among them. Computational prediction of drug-target interactions can facilitate in reducing the search space of experimental wet lab-based verifications steps, thus considerably reducing time and other resources dedicated to the drug discovery pipeline. While machine learning-based methods are more widespread for drug-target interaction prediction, network-centric methods are also evolving. In this chapter, we focus on the process of the drug-target interaction prediction from the perspective of using machine learning algorithms and the various stages involved for developing an accurate predictor.
Collapse
Affiliation(s)
- Abhigyan Nath
- Department of Biochemistry, Pt. Jawahar Lal Nehru Memorial Medical College, Raipur, India
| | - Radha Chaube
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
8
|
Samantasinghar A, Ahmed F, Rahim CSA, Kim KH, Kim S, Choi KH. Artificial intelligence-assisted repurposing of lubiprostone alleviates tubulointerstitial fibrosis. Transl Res 2023; 262:75-88. [PMID: 37541485 DOI: 10.1016/j.trsl.2023.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/19/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Tubulointerstitial fibrosis (TIF) is the most prominent cause which leads to chronic kidney disease (CKD) and end-stage renal failure. Despite extensive research, there have been many clinical trial failures, and there is currently no effective treatment to cure renal fibrosis. This demonstrates the necessity of more effective therapies and better preclinical models to screen potential drugs for TIF. In this study, we investigated the antifibrotic effect of the machine learning-based repurposed drug, lubiprostone, validated through an advanced proximal tubule on a chip system and in vivo UUO mice model. Lubiprostone significantly downregulated TIF biomarkers including connective tissue growth factor (CTGF), extracellular matrix deposition (Fibronectin and collagen), transforming growth factor (TGF-β) downstream signaling markers especially, Smad-2/3, matrix metalloproteinase (MMP2/9), plasminogen activator inhibitor-1 (PAI-1), EMT and JAK/STAT-3 pathway expression in the proximal tubule on a chip model and UUO model compared to the conventional 2D culture. These findings suggest that the proximal tubule on a chip model is a more physiologically relevant model for studying and identifying potential biomarkers for fibrosis compared to conventional in vitro 2D culture and alternative of an animal model. In conclusion, the high throughput Proximal tubule-on-chip system shows improved in vivo-like function and indicates the potential utility for renal fibrosis drug screening. Additionally, repurposed Lubiprostone shows an effective potency to treat TIF via inhibiting 3 major profibrotic signaling pathways such as TGFβ/Smad, JAK/STAT, and epithelial-mesenchymal transition (EMT), and restores kidney function.
Collapse
Affiliation(s)
| | - Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| | | | | | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| |
Collapse
|
9
|
Abbasi Mesrabadi H, Faez K, Pirgazi J. Drug-target interaction prediction based on protein features, using wrapper feature selection. Sci Rep 2023; 13:3594. [PMID: 36869062 PMCID: PMC9984486 DOI: 10.1038/s41598-023-30026-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Drug-target interaction prediction is a vital stage in drug development, involving lots of methods. Experimental methods that identify these relationships on the basis of clinical remedies are time-taking, costly, laborious, and complex introducing a lot of challenges. One group of new methods is called computational methods. The development of new computational methods which are more accurate can be preferable to experimental methods, in terms of total cost and time. In this paper, a new computational model to predict drug-target interaction (DTI), consisting of three phases, including feature extraction, feature selection, and classification is proposed. In feature extraction phase, different features such as EAAC, PSSM and etc. would be extracted from sequence of proteins and fingerprint features from drugs. These extracted features would then be combined. In the next step, one of the wrapper feature selection methods named IWSSR, due to the large amount of extracted data, is applied. The selected features are then given to rotation forest classification, to have a more efficient prediction. Actually, the innovation of our work is that we extract different features; and then select features by the use of IWSSR. The accuracy of the rotation forest classifier based on tenfold on the golden standard datasets (enzyme, ion channels, G-protein-coupled receptors, nuclear receptors) is as follows: 98.12, 98.07, 96.82, and 95.64. The results of experiments indicate that the proposed model has an acceptable rate in DTI prediction and is compatible with the proposed methods in other papers.
Collapse
Affiliation(s)
- Hengame Abbasi Mesrabadi
- Faculty of Computer and Information Technology Engineering, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| | - Karim Faez
- Department of Electrical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Jamshid Pirgazi
- Department of Computer Engineering, University of Science and Technology of Mazandaran, Behshahr, Iran
| |
Collapse
|
10
|
Comparative Studies on Resampling Techniques in Machine Learning and Deep Learning Models for Drug-Target Interaction Prediction. Molecules 2023; 28:molecules28041663. [PMID: 36838652 PMCID: PMC9964614 DOI: 10.3390/molecules28041663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
The prediction of drug-target interactions (DTIs) is a vital step in drug discovery. The success of machine learning and deep learning methods in accurately predicting DTIs plays a huge role in drug discovery. However, when dealing with learning algorithms, the datasets used are usually highly dimensional and extremely imbalanced. To solve this issue, the dataset must be resampled accordingly. In this paper, we have compared several data resampling techniques to overcome class imbalance in machine learning methods as well as to study the effectiveness of deep learning methods in overcoming class imbalance in DTI prediction in terms of binary classification using ten (10) cancer-related activity classes from BindingDB. It is found that the use of Random Undersampling (RUS) in predicting DTIs severely affects the performance of a model, especially when the dataset is highly imbalanced, thus, rendering RUS unreliable. It is also found that SVM-SMOTE can be used as a go-to resampling method when paired with the Random Forest and Gaussian Naïve Bayes classifiers, whereby a high F1 score is recorded for all activity classes that are severely and moderately imbalanced. Additionally, the deep learning method called Multilayer Perceptron recorded high F1 scores for all activity classes even when no resampling method was applied.
Collapse
|
11
|
McNair D. Artificial Intelligence and Machine Learning for Lead-to-Candidate Decision-Making and Beyond. Annu Rev Pharmacol Toxicol 2023; 63:77-97. [PMID: 35679624 DOI: 10.1146/annurev-pharmtox-051921-023255] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The use of artificial intelligence (AI) and machine learning (ML) in pharmaceutical research and development has to date focused on research: target identification; docking-, fragment-, and motif-based generation of compound libraries; modeling of synthesis feasibility; rank-ordering likely hits according to structural and chemometric similarity to compounds having known activity and affinity to the target(s); optimizing a smaller library for synthesis and high-throughput screening; and combining evidence from screening to support hit-to-lead decisions. Applying AI/ML methods to lead optimization and lead-to-candidate (L2C) decision-making has shown slower progress, especially regarding predicting absorption, distribution, metabolism, excretion, and toxicology properties. The present review surveys reasons why this is so, reports progress that has occurred in recent years, and summarizes some of the issues that remain. Effective AI/ML tools to derisk L2C and later phases of development are important to accelerate the pharmaceutical development process, ameliorate escalating development costs, and achieve greater success rates.
Collapse
Affiliation(s)
- Douglas McNair
- Global Health, Integrated Development, Bill & Melinda Gates Foundation, Seattle, Washington, USA;
| |
Collapse
|
12
|
Lungu CN, Mangalagiu V, Mangalagiu II, Mehedinti MC. Benzoquinoline Chemical Space: A Helpful Approach in Antibacterial and Anticancer Drug Design. Molecules 2023; 28:molecules28031069. [PMID: 36770739 PMCID: PMC9921191 DOI: 10.3390/molecules28031069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Benzoquinolines are used in many drug design projects as starting molecules subject to derivatization. This computational study aims to characterize e benzoquinone drug space to ease future drug design processes based on these molecules. The drug space is composed of all benzoquinones, which are active on topoisomerase II and ATP synthase. Topological, chemical, and bioactivity spaces are explored using computational methodologies based on virtual screening and scaffold hopping and molecular docking, respectively. Topological space is a geometrical space in which the elements composing it can be defined as a set of neighbors (which satisfy a particular axiom). In such space, a chemical space can be defined as the property space spanned by all possible molecules and chemical compounds adhering to a given set of construction principles and boundary conditions. In this chemical space, the potentially pharmacologically active molecules form the bioactivity space. Results show a poly-morphological chemical space that suggests distinct characteristics. The chemical space is correlated with properties such as steric energy, the number of hydrogen bonds, the presence of halogen atoms, and membrane permeability-related properties. Lastly, novel chemical compounds (such as oxadiazole methybenzamide and floro methylcyclohexane diene) with drug-like potential, active on TOPO II and ATP synthase have been identified.
Collapse
Affiliation(s)
- Claudiu N. Lungu
- Department of Surgery, Emergency Country Clinical Hospital, 800010 Galati, Romania
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
- Department of Morphological and Functional Science, University of Medicine and Pharmacy, Dunarea de Jos, 800017 Galati, Romania
- Correspondence: (C.N.L.); (I.I.M.)
| | - Violeta Mangalagiu
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
- Faculty of Food Engineering, Stefan cel Mare University of Suceava, 13 Universitatii Str., 720229 Suceava, Romania
| | - Ionel I. Mangalagiu
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
- Institute of Interdisciplinary Research-CERNESIM Centre, Alexandru Ioan Cuza University of Iasi, 11 Carol I, 700506 Iasi, Romania
- Correspondence: (C.N.L.); (I.I.M.)
| | - Mihaela C. Mehedinti
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
- Department of Morphological and Functional Science, University of Medicine and Pharmacy, Dunarea de Jos, 800017 Galati, Romania
| |
Collapse
|
13
|
Jamali AA, Kusalik A, Wu FX. NMTF-DTI: A Nonnegative Matrix Tri-factorization Approach With Multiple Kernel Fusion for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:586-594. [PMID: 34914594 DOI: 10.1109/tcbb.2021.3135978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Prediction of drug-target interactions (DTIs) plays a significant role in drug development and drug discovery. Although this task requires a large investment in terms of time and cost, especially when it is performed experimentally, the results are not necessarily significant. Computational DTI prediction is a shortcut to reduce the risks of experimental methods. In this study, we propose an effective approach of nonnegative matrix tri-factorization, referred to as NMTF-DTI, to predict the interaction scores between drugs and targets. NMTF-DTI utilizes multiple kernels (similarity measures) for drugs and targets and Laplacian regularization to boost the prediction performance. The performance of NMTF-DTI is evaluated via cross-validation and is compared with existing DTI prediction methods in terms of the area under the receiver operating characteristic (ROC) curve (AUC) and the area under the precision and recall curve (AUPR). We evaluate our method on four gold standard datasets, comparing to other state-of-the-art methods. Cross-validation and a separate, manually created dataset are used to set parameters. The results show that NMTF-DTI outperforms other competing methods. Moreover, the results of a case study also confirm the superiority of NMTF-DTI.
Collapse
|
14
|
Lei S, Lei X, Liu L. Drug repositioning based on heterogeneous networks and variational graph autoencoders. Front Pharmacol 2022; 13:1056605. [PMID: 36618933 PMCID: PMC9812491 DOI: 10.3389/fphar.2022.1056605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Predicting new therapeutic effects (drug repositioning) of existing drugs plays an important role in drug development. However, traditional wet experimental prediction methods are usually time-consuming and costly. The emergence of more and more artificial intelligence-based drug repositioning methods in the past 2 years has facilitated drug development. In this study we propose a drug repositioning method, VGAEDR, based on a heterogeneous network of multiple drug attributes and a variational graph autoencoder. First, a drug-disease heterogeneous network is established based on three drug attributes, disease semantic information, and known drug-disease associations. Second, low-dimensional feature representations for heterogeneous networks are learned through a variational graph autoencoder module and a multi-layer convolutional module. Finally, the feature representation is fed to a fully connected layer and a Softmax layer to predict new drug-disease associations. Comparative experiments with other baseline methods on three datasets demonstrate the excellent performance of VGAEDR. In the case study, we predicted the top 10 possible anti-COVID-19 drugs on the existing drug and disease data, and six of them were verified by other literatures.
Collapse
|
15
|
Kumar N, Acharya V. Machine intelligence-driven framework for optimized hit selection in virtual screening. J Cheminform 2022; 14:48. [PMID: 35869511 PMCID: PMC9306080 DOI: 10.1186/s13321-022-00630-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractVirtual screening (VS) aids in prioritizing unknown bio-interactions between compounds and protein targets for empirical drug discovery. In standard VS exercise, roughly 10% of top-ranked molecules exhibit activity when examined in biochemical assays, which accounts for many false positive hits, making it an arduous task. Attempts for conquering false-hit rates were developed through either ligand-based or structure-based VS separately; however, nonetheless performed remarkably well. Here, we present an advanced VS framework—automated hit identification and optimization tool (A-HIOT)—comprises chemical space-driven stacked ensemble for identification and protein space-driven deep learning architectures for optimization of an array of specific hits for fixed protein receptors. A-HIOT implements numerous open-source algorithms intending to integrate chemical and protein space leading to a high-quality prediction. The optimized hits are the selective molecules which we retrieve after extreme refinement implying chemical space and protein space modules of A-HIOT. Using CXC chemokine receptor 4, we demonstrated the superior performance of A-HIOT for hit molecule identification and optimization with tenfold cross-validation accuracies of 94.8% and 81.9%, respectively. In comparison with other machine learning algorithms, A-HIOT achieved higher accuracies of 96.2% for hit identification and 89.9% for hit optimization on independent benchmark datasets for CXCR4 and 86.8% for hit identification and 90.2% for hit optimization on independent test dataset for androgen receptor (AR), thus, shows its generalizability and robustness. In conclusion, advantageous features impeded in A-HIOT is making a reliable approach for bridging the long-standing gap between ligand-based and structure-based VS in finding the optimized hits for the desired receptor. The complete resource (framework) code is available at https://gitlab.com/neeraj-24/A-HIOT.
Graphical Abstract
Collapse
|
16
|
Wang H, Guo F, Du M, Wang G, Cao C. A novel method for drug-target interaction prediction based on graph transformers model. BMC Bioinformatics 2022; 23:459. [PMID: 36329406 PMCID: PMC9635108 DOI: 10.1186/s12859-022-04812-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/23/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Drug-target interactions (DTIs) prediction becomes more and more important for accelerating drug research and drug repositioning. Drug-target interaction network is a typical model for DTIs prediction. As many different types of relationships exist between drug and target, drug-target interaction network can be used for modeling drug-target interaction relationship. Recent works on drug-target interaction network are mostly concentrate on drug node or target node and neglecting the relationships between drug-target. RESULTS We propose a novel prediction method for modeling the relationship between drug and target independently. Firstly, we use different level relationships of drugs and targets to construct feature of drug-target interaction. Then, we use line graph to model drug-target interaction. After that, we introduce graph transformer network to predict drug-target interaction. CONCLUSIONS This method introduces a line graph to model the relationship between drug and target. After transforming drug-target interactions from links to nodes, a graph transformer network is used to accomplish the task of predicting drug-target interactions.
Collapse
Affiliation(s)
- Hongmei Wang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun, China
| | - Fang Guo
- College of Computer Science and Engineering, Changchun University of Technology, Changchun, China
| | - Mengyan Du
- College of Computer Science and Engineering, Changchun University of Technology, Changchun, China
| | - Guishen Wang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun, China.
| | - Chen Cao
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China. .,Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada.
| |
Collapse
|
17
|
Liu B, Papadopoulos D, Malliaros FD, Tsoumakas G, Papadopoulos AN. Multiple similarity drug-target interaction prediction with random walks and matrix factorization. Brief Bioinform 2022; 23:6692553. [PMID: 36070659 DOI: 10.1093/bib/bbac353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 11/14/2022] Open
Abstract
The discovery of drug-target interactions (DTIs) is a very promising area of research with great potential. The accurate identification of reliable interactions among drugs and proteins via computational methods, which typically leverage heterogeneous information retrieved from diverse data sources, can boost the development of effective pharmaceuticals. Although random walk and matrix factorization techniques are widely used in DTI prediction, they have several limitations. Random walk-based embedding generation is usually conducted in an unsupervised manner, while the linear similarity combination in matrix factorization distorts individual insights offered by different views. To tackle these issues, we take a multi-layered network approach to handle diverse drug and target similarities, and propose a novel optimization framework, called Multiple similarity DeepWalk-based Matrix Factorization (MDMF), for DTI prediction. The framework unifies embedding generation and interaction prediction, learning vector representations of drugs and targets that not only retain higher order proximity across all hyper-layers and layer-specific local invariance, but also approximate the interactions with their inner product. Furthermore, we develop an ensemble method (MDMF2A) that integrates two instantiations of the MDMF model, optimizing the area under the precision-recall curve (AUPR) and the area under the receiver operating characteristic curve (AUC), respectively. The empirical study on real-world DTI datasets shows that our method achieves statistically significant improvement over current state-of-the-art approaches in four different settings. Moreover, the validation of highly ranked non-interacting pairs also demonstrates the potential of MDMF2A to discover novel DTIs.
Collapse
Affiliation(s)
- Bin Liu
- Key Laboratory of Data Engineering and Visual Computing, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
- School of Informatics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Fragkiskos D Malliaros
- Paris-Saclay University, CentraleSupélec, Inria, Centre for Visual Computing (CVN), 91190 Gif-Sur-Yvette, France
| | - Grigorios Tsoumakas
- School of Informatics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | |
Collapse
|
18
|
Detecting Drug–Target Interactions with Feature Similarity Fusion and Molecular Graphs. BIOLOGY 2022; 11:biology11070967. [PMID: 36101348 PMCID: PMC9312204 DOI: 10.3390/biology11070967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/12/2022] [Accepted: 06/24/2022] [Indexed: 12/03/2022]
Abstract
Simple Summary Accurate identification of potential targets for drugs to interact with can accelerate drug development. The identification of drug–target interactions can provide insights into hidden drug efficacy. This paper presents a prediction model based on feature similarity fusion that can identify crucial features of drugs and targets to help predict drug–target interactions. Abstract The key to drug discovery is the identification of a target and a corresponding drug compound. Effective identification of drug–target interactions facilitates the development of drug discovery. In this paper, drug similarity and target similarity are considered, and graphical representations are used to extract internal structural information and intermolecular interaction information about drugs and targets. First, drug similarity and target similarity are fused using the similarity network fusion (SNF) method. Then, the graph isomorphic network (GIN) is used to extract the features with information about the internal structure of drug molecules. For target proteins, feature extraction is carried out using TextCNN to efficiently capture the features of target protein sequences. Three different divisions (CVD, CVP, CVT) are used on the standard dataset, and experiments are carried out separately to validate the performance of the model for drug–target interaction prediction. The experimental results show that our method achieves better results on AUC and AUPR. The docking results also show the superiority of the proposed model in predicting drug–target interactions.
Collapse
|
19
|
da Mota VHS, Freire de Melo F, de Brito BB, Silva FAFD, Teixeira KN. Molecular docking of DS-3032B, a mouse double minute 2 enzyme antagonist with potential for oncology treatment development. World J Clin Oncol 2022; 13:496-504. [PMID: 35949428 PMCID: PMC9244969 DOI: 10.5306/wjco.v13.i6.496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/16/2021] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It is known that p53 suppression is an important marker of poor prognosis of cancers, especially in solid tumors of the breast, lung, stomach, and esophagus; liposarcomas, glioblastomas, and leukemias. Because p53 has mouse double minute 2 (MDM2) as its primary negative regulator, this molecular docking study seeks to answer the following hypotheses: Is the interaction between DS-3032B and MDM2 stable enough for this drug to be considered as a promising neoplastic inhibitor? AIM To analyze, in silico, the chemical bonds between the antagonist DS-3032B and its binding site in MDM2. METHODS For molecular docking simulations, the file containing structures of MDM2 (receptor) and the drug DS-3032B (ligand) were selected. The three-dimensional structure of MDM2 was obtained from Protein Data Bank, and the one for DS-3032B was obtained from PubChem database. The location and dimensions of the Grid box was determined using AutoDock Tools software. In this case, the dimensions of the Grid encompassed the entire receptor. The ligand DS-3032B interacts with the MDM2 receptor in a physiological environment with pH 7.4; thus, to simulate more reliably, its interaction was made with the calculation for the prediction of its protonation state using the MarvinSketch® software. Both ligands, with and without the protonation, were prepared for molecular docking using the AutoDock Tools software. This software detects the torsion points of the drug and calculates the angle of the torsions. Molecular docking simulations were performed using the tools of the AutoDock platform connected to the Vina software. The analyses of the amino acid residues involved in the interactions between the receptor and the ligand as well as the twists of the ligand, atoms involved in the interactions, and type, strength, and length of the interactions were performed using the PyMol software (pymol.org/2) and Discovery Studio from BIOVIA®. RESULTS The global alignment indicated crystal structure 5SWK was more suitable for docking simulations by presenting the p53 binding site. The three-dimensional structure 5SWK for MDM2 was selected from Protein Data Bank and the three-dimensional structure of DS-3032B was selected from PubChem (Compound CID: 73297272; Milademetan). After molecular docking simulations, the most stable conformer was selected for both protonated and non-protonated DS-3032B. The interaction between MDM2 and DS-3032B occurs with high affinity; no significant difference was observed in the affinity energies between the MDM2/pronated DS-3032B (-9.9 kcal/mol) and MDM2/non-protonated DS-3032B conformers (-10.0 kcal/mol). Sixteen amino acid residues of MDM2 are involved in chemical bonds with the protonated DS-3032B; these 16 residues of MDM2 belong to the p53 biding site region and provide high affinity to interaction and stability to drug-protein complex. CONCLUSION Molecular docking indicated that DS-3032B antagonist binds to the same region of the p53 binding site in the MDM2 with high affinity and stability, and this suggests therapeutic efficiency.
Collapse
Affiliation(s)
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Breno Bittencourt de Brito
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | |
Collapse
|
20
|
DTIP-TC2A: An analytical framework for drug-target interactions prediction methods. Comput Biol Chem 2022; 99:107707. [DOI: 10.1016/j.compbiolchem.2022.107707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 05/01/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022]
|
21
|
Deng C, Han D, Feng M, Lv Z, Li D. Differential diagnostic value of the ResNet50, random forest, and DS ensemble models for papillary thyroid carcinoma and other thyroid nodules. J Int Med Res 2022; 50:3000605221094276. [PMID: 35469474 PMCID: PMC9087260 DOI: 10.1177/03000605221094276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective To explore the differential diagnostic efficiency of the
residual network (ResNet)50, random forest (RF), and DS ensemble models for
papillary thyroid carcinoma (PTC) and other pathological types of thyroid
nodules. Methods This study retrospectively analyzed 559 patients with
thyroid nodules and collected thyroid pathological images and auxiliary
examination results (laboratory and ultrasound results) to construct datasets.
The pathological image dataset was used to train a ResNet50 model, the text
dataset was used to train a random forest (RF) model, and a DS ensemble model
was constructed from the results of the two models. The differential diagnostic
values of the three models for PTC and other types of thyroid nodules were then
compared. Results The DS ensemble model had the highest sensitivity,
specificity, accuracy, and area under the receiver operating characteristic
curve (85.87%, 97.18%, 93.77%, and 0.982, respectively). Conclusions Compared with Resnet50 and the RF models trained only on
imaging data or text information, respectively, the DS ensemble model showed
better diagnostic value for PTC.
Collapse
Affiliation(s)
- Chengwen Deng
- Shanghai Tenth People's Hospital Tongji University, Shanghai, China
| | - Dongyan Han
- Shanghai Tenth People's Hospital Tongji University, Shanghai, China
| | | | - Zhongwei Lv
- Shanghai Tenth People's Hospital Tongji University, Shanghai, China
| | - Dan Li
- Shanghai Tenth People's Hospital Tongji University, Shanghai, China
| |
Collapse
|
22
|
Shao K, Zhang Y, Wen Y, Zhang Z, He S, Bo X. DTI-HETA: prediction of drug-target interactions based on GCN and GAT on heterogeneous graph. Brief Bioinform 2022; 23:6563180. [PMID: 35380622 DOI: 10.1093/bib/bbac109] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/14/2022] [Accepted: 03/03/2022] [Indexed: 12/19/2022] Open
Abstract
Drug-target interaction (DTI) prediction plays an important role in drug repositioning, drug discovery and drug design. However, due to the large size of the chemical and genomic spaces and the complex interactions between drugs and targets, experimental identification of DTIs is costly and time-consuming. In recent years, the emerging graph neural network (GNN) has been applied to DTI prediction because DTIs can be represented effectively using graphs. However, some of these methods are only based on homogeneous graphs, and some consist of two decoupled steps that cannot be trained jointly. To further explore GNN-based DTI prediction by integrating heterogeneous graph information, this study regards DTI prediction as a link prediction problem and proposes an end-to-end model based on HETerogeneous graph with Attention mechanism (DTI-HETA). In this model, a heterogeneous graph is first constructed based on the drug-drug and target-target similarity matrices and the DTI matrix. Then, the graph convolutional neural network is utilized to obtain the embedded representation of the drugs and targets. To highlight the contribution of different neighborhood nodes to the central node in aggregating the graph convolution information, a graph attention mechanism is introduced into the node embedding process. Afterward, an inner product decoder is applied to predict DTIs. To evaluate the performance of DTI-HETA, experiments are conducted on two datasets. The experimental results show that our model is superior to the state-of-the-art methods. Also, the identification of novel DTIs indicates that DTI-HETA can serve as a powerful tool for integrating heterogeneous graph information to predict DTIs.
Collapse
Affiliation(s)
| | | | - Yuqi Wen
- Beijing Institute of Radiation Medicine, Beijing, China
| | | | - Song He
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiaochen Bo
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
23
|
Amiri Souri E, Laddach R, Karagiannis SN, Papageorgiou LG, Tsoka S. Novel drug-target interactions via link prediction and network embedding. BMC Bioinformatics 2022; 23:121. [PMID: 35379165 PMCID: PMC8978405 DOI: 10.1186/s12859-022-04650-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND As many interactions between the chemical and genomic space remain undiscovered, computational methods able to identify potential drug-target interactions (DTIs) are employed to accelerate drug discovery and reduce the required cost. Predicting new DTIs can leverage drug repurposing by identifying new targets for approved drugs. However, developing an accurate computational framework that can efficiently incorporate chemical and genomic spaces remains extremely demanding. A key issue is that most DTI predictions suffer from the lack of experimentally validated negative interactions or limited availability of target 3D structures. RESULTS We report DT2Vec, a pipeline for DTI prediction based on graph embedding and gradient boosted tree classification. It maps drug-drug and protein-protein similarity networks to low-dimensional features and the DTI prediction is formulated as binary classification based on a strategy of concatenating the drug and target embedding vectors as input features. DT2Vec was compared with three top-performing graph similarity-based algorithms on a standard benchmark dataset and achieved competitive results. In order to explore credible novel DTIs, the model was applied to data from the ChEMBL repository that contain experimentally validated positive and negative interactions which yield a strong predictive model. Then, the developed model was applied to all possible unknown DTIs to predict new interactions. The applicability of DT2Vec as an effective method for drug repurposing is discussed through case studies and evaluation of some novel DTI predictions is undertaken using molecular docking. CONCLUSIONS The proposed method was able to integrate and map chemical and genomic space into low-dimensional dense vectors and showed promising results in predicting novel DTIs.
Collapse
Affiliation(s)
- E Amiri Souri
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, Bush House, London, WC2B 4BG, UK
| | - R Laddach
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, Bush House, London, WC2B 4BG, UK
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, SE1 9RT, UK
| | - S N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, SE1 9RT, UK
| | - L G Papageorgiou
- Centre for Process Systems Engineering, Department of Chemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - S Tsoka
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, Bush House, London, WC2B 4BG, UK.
| |
Collapse
|
24
|
A Novel Deep Neural Network Technique for Drug–Target Interaction. Pharmaceutics 2022; 14:pharmaceutics14030625. [PMID: 35336000 PMCID: PMC8954728 DOI: 10.3390/pharmaceutics14030625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 01/20/2023] Open
Abstract
Drug discovery (DD) is a time-consuming and expensive process. Thus, the industry employs strategies such as drug repositioning and drug repurposing, which allows the application of already approved drugs to treat a different disease, as occurred in the first months of 2020, during the COVID-19 pandemic. The prediction of drug–target interactions is an essential part of the DD process because it can accelerate it and reduce the required costs. DTI prediction performed in silico have used approaches based on molecular docking simulations, including similarity-based and network- and graph-based ones. This paper presents MPS2IT-DTI, a DTI prediction model obtained from research conducted in the following steps: the definition of a new method for encoding molecule and protein sequences onto images; the definition of a deep-learning approach based on a convolutional neural network in order to create a new method for DTI prediction. Training results conducted with the Davis and KIBA datasets show that MPS2IT-DTI is viable compared to other state-of-the-art (SOTA) approaches in terms of performance and complexity of the neural network model. With the Davis dataset, we obtained 0.876 for the concordance index and 0.276 for the MSE; with the KIBA dataset, we obtained 0.836 and 0.226 for the concordance index and the MSE, respectively. Moreover, the MPS2IT-DTI model represents molecule and protein sequences as images, instead of treating them as an NLP task, and as such, does not employ an embedding layer, which is present in other models.
Collapse
|
25
|
Drug-target interaction prediction via an ensemble of weighted nearest neighbors with interaction recovery. APPL INTELL 2022. [DOI: 10.1007/s10489-021-02495-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
26
|
Staszak M, Staszak K, Wieszczycka K, Bajek A, Roszkowski K, Tylkowski B. Machine learning in drug design: Use of artificial intelligence to explore the chemical structure–biological activity relationship. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2022. [DOI: 10.1002/wcms.1568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Maciej Staszak
- Institute of Technology and Chemical Engineering Poznan University of Technology Poznan Poland
| | - Katarzyna Staszak
- Institute of Technology and Chemical Engineering Poznan University of Technology Poznan Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering Poznan University of Technology Poznan Poland
| | - Anna Bajek
- Department of Tissue Engineering Collegium Medicum, Nicolaus Copernicus University Bydgoszcz Poland
| | - Krzysztof Roszkowski
- Department of Oncology Collegium Medicum Nicolaus Copernicus University Bydgoszcz Poland
| | - Bartosz Tylkowski
- Department of Chemical Engineering University Rovira i Virgili Tarragona Spain
- Eurecat, Centre Tecnològic de Catalunya Chemical Technologies Unit Tarragona Spain
| |
Collapse
|
27
|
Alakus TB, Turkoglu I. A Comparative Study of Amino Acid Encoding Methods for Predicting Drug-Target Interactions in COVID-19 Disease. MODELING, CONTROL AND DRUG DEVELOPMENT FOR COVID-19 OUTBREAK PREVENTION 2022:619-643. [DOI: 10.1007/978-3-030-72834-2_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
28
|
Machine learning & deep learning in data-driven decision making of drug discovery and challenges in high-quality data acquisition in the pharmaceutical industry. Future Med Chem 2021; 14:245-270. [PMID: 34939433 DOI: 10.4155/fmc-2021-0243] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Predicting novel small molecule bioactivities for the target deconvolution, hit-to-lead optimization in drug discovery research, requires molecular representation. Previous reports have demonstrated that machine learning (ML) and deep learning (DL) have substantial implications in virtual screening, peptide synthesis, drug ADMET screening and biomarker discovery. These strategies can increase the positive outcomes in the drug discovery process without false-positive rates and can be achieved in a cost-effective way with a minimum duration of time by high-quality data acquisition. This review substantially discusses the recent updates in AI tools as cheminformatics application in medicinal chemistry for the data-driven decision making of drug discovery and challenges in high-quality data acquisition in the pharmaceutical industry while improving small-molecule bioactivities and properties.
Collapse
|
29
|
Drug-Target Interaction Prediction Based on Multisource Information Weighted Fusion. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:6044256. [PMID: 34908912 PMCID: PMC8635946 DOI: 10.1155/2021/6044256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/22/2021] [Indexed: 01/08/2023]
Abstract
Recently, in most existing studies, it is assumed that there are no interaction relationships between drugs and targets with unknown interactions. However, unknown interactions mean the relationships between drugs and targets have just not been confirmed. In this paper, samples for which the relationship between drugs and targets has not been determined are considered unlabeled. A weighted fusion method of multisource information is proposed to screen drug-target interactions. Firstly, some drug-target pairs which may have interactions are selected. Secondly, the selected drug-target pairs are added to the positive samples, which are regarded as known to have interaction relationships, and the original interaction relationship matrix is revised. Finally, the revised datasets are used to predict the interaction derived from the bipartite local model with neighbor-based interaction profile inferring (BLM-NII). Experiments demonstrate that the proposed method has greatly improved specificity, sensitivity, precision, and accuracy compared with the BLM-NII method. In addition, compared with several state-of-the-art methods, the area under the receiver operating characteristic curve (AUC) and the area under the precision-recall curve (AUPR) of the proposed method are excellent.
Collapse
|
30
|
Screening of Potential Indonesia Herbal Compounds Based on Multi-Label Classification for 2019 Coronavirus Disease. BIG DATA AND COGNITIVE COMPUTING 2021. [DOI: 10.3390/bdcc5040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 pandemic spreads rapidly and requires an acceleration in the process of drug discovery. Drug repurposing can help accelerate the drug discovery process by identifying new efficacy for approved drugs, and it is considered an efficient and economical approach. Research in drug repurposing can be done by observing the interactions of drug compounds with protein related to a disease (DTI), then predicting the new drug-target interactions. This study conducted multilabel DTI prediction using the stack autoencoder-deep neural network (SAE-DNN) algorithm. Compound features were extracted using PubChem fingerprint, daylight fingerprint, MACCS fingerprint, and circular fingerprint. The results showed that the SAE-DNN model was able to predict DTI in COVID-19 cases with good performance. The SAE-DNN model with a circular fingerprint dataset produced the best average metrics with an accuracy of 0.831, recall of 0.918, precision of 0.888, and F-measure of 0.89. Herbal compounds prediction results using the SAE-DNN model with the circular, daylight, and PubChem fingerprint dataset resulted in 92, 65, and 79 herbal compounds contained in herbal plants in Indonesia respectively.
Collapse
|
31
|
Jung YS, Kim Y, Cho YR. Comparative analysis of network-based approaches and machine learning algorithms for predicting drug-target interactions. Methods 2021; 198:19-31. [PMID: 34737033 DOI: 10.1016/j.ymeth.2021.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 01/06/2023] Open
Abstract
Computational prediction of drug-target interactions (DTIs) is of particular importance in the process of drug repositioning because of its efficiency in selecting potential candidates for DTIs. A variety of computational methods for predicting DTIs have been proposed over the past decade. Our interest is which methods or techniques are the most advantageous for increasing prediction accuracy. This article provides a comprehensive overview of network-based, machine learning, and integrated DTI prediction methods. The network-based methods handle a DTI network along with drug and target similarities in a matrix form and apply graph-theoretic algorithms to identify new DTIs. Machine learning methods use known DTIs and the features of drugs and target proteins as training data to build a predictive model. Integrated methods combine these two techniques. We assessed the prediction performance of the selected state-of-the-art methods using two different benchmark datasets. Our experimental results demonstrate that the integrated methods outperform the others in general. Some previous methods showed low accuracy on predicting interactions of unknown drugs which do not exist in the training dataset. Combining similarity matrices from multiple features by data fusion was not beneficial in increasing prediction accuracy. Finally, we analyzed future directions for further improvements in DTI predictions.
Collapse
Affiliation(s)
- Yi-Sue Jung
- Division of Software, Yonsei University - Mirae Campus, Republic of Korea
| | - Yoonbee Kim
- Division of Software, Yonsei University - Mirae Campus, Republic of Korea
| | - Young-Rae Cho
- Division of Software, Yonsei University - Mirae Campus, Republic of Korea; Division of Digital Healthcare, Yonsei University - Mirae Campus, Republic of Korea.
| |
Collapse
|
32
|
Yue Y, He S. DTI-HeNE: a novel method for drug-target interaction prediction based on heterogeneous network embedding. BMC Bioinformatics 2021; 22:418. [PMID: 34479477 PMCID: PMC8414716 DOI: 10.1186/s12859-021-04327-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Prediction of the drug-target interaction (DTI) is a critical step in the drug repurposing process, which can effectively reduce the following workload for experimental verification of potential drugs' properties. In recent studies, many machine-learning-based methods have been proposed to discover unknown interactions between drugs and protein targets. A recent trend is to use graph-based machine learning, e.g., graph embedding to extract features from drug-target networks and then predict new drug-target interactions. However, most of the graph embedding methods are not specifically designed for DTI predictions; thus, it is difficult for these methods to fully utilize the heterogeneous information of drugs and targets (e.g., the respective vertex features of drugs and targets and path-based interactive features between drugs and targets). RESULTS We propose a DTI prediction method DTI-HeNE (DTI based on Heterogeneous Network Embedding), which is specifically designed to cope with the bipartite DTI relations for generating high-quality embeddings of drug-target pairs. This method splits a heterogeneous DTI network into a bipartite DTI network, multiple drug homogeneous networks and target homogeneous networks, and extracts features from these sub-networks separately to better utilize the characteristics of bipartite DTI relations as well as the auxiliary similarity information related to drugs and targets. The features extracted from each sub-network are integrated using pathway information between these sub-networks to acquire new features, i.e., embedding vectors of drug-target pairs. Finally, these features are fed into a random forest (RF) model to predict novel DTIs. CONCLUSIONS Our experimental results show that, the proposed DTI network embedding method can learn higher-quality features of heterogeneous drug-target interaction networks for novel DTIs discovery.
Collapse
Affiliation(s)
- Yang Yue
- College of Information and Electrical Engineering, China Agricultural University, Beijing, 100083, China
| | - Shan He
- Centre for Computational Biology, School of Computer Science, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
33
|
Miethke M, Pieroni M, Weber T, Brönstrup M, Hammann P, Halby L, Arimondo PB, Glaser P, Aigle B, Bode HB, Moreira R, Li Y, Luzhetskyy A, Medema MH, Pernodet JL, Stadler M, Tormo JR, Genilloud O, Truman AW, Weissman KJ, Takano E, Sabatini S, Stegmann E, Brötz-Oesterhelt H, Wohlleben W, Seemann M, Empting M, Hirsch AKH, Loretz B, Lehr CM, Titz A, Herrmann J, Jaeger T, Alt S, Hesterkamp T, Winterhalter M, Schiefer A, Pfarr K, Hoerauf A, Graz H, Graz M, Lindvall M, Ramurthy S, Karlén A, van Dongen M, Petkovic H, Keller A, Peyrane F, Donadio S, Fraisse L, Piddock LJV, Gilbert IH, Moser HE, Müller R. Towards the sustainable discovery and development of new antibiotics. Nat Rev Chem 2021; 5:726-749. [PMID: 34426795 PMCID: PMC8374425 DOI: 10.1038/s41570-021-00313-1] [Citation(s) in RCA: 575] [Impact Index Per Article: 143.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 02/08/2023]
Abstract
An ever-increasing demand for novel antimicrobials to treat life-threatening infections caused by the global spread of multidrug-resistant bacterial pathogens stands in stark contrast to the current level of investment in their development, particularly in the fields of natural-product-derived and synthetic small molecules. New agents displaying innovative chemistry and modes of action are desperately needed worldwide to tackle the public health menace posed by antimicrobial resistance. Here, our consortium presents a strategic blueprint to substantially improve our ability to discover and develop new antibiotics. We propose both short-term and long-term solutions to overcome the most urgent limitations in the various sectors of research and funding, aiming to bridge the gap between academic, industrial and political stakeholders, and to unite interdisciplinary expertise in order to efficiently fuel the translational pipeline for the benefit of future generations.
Collapse
Affiliation(s)
- Marcus Miethke
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Marco Pieroni
- Food and Drug Department, University of Parma, Parma, Italy
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Mark Brönstrup
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Chemical Biology (CBIO), Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Peter Hammann
- Infectious Diseases & Natural Product Research at EVOTEC, and Justus Liebig University Giessen, Giessen, Germany
| | - Ludovic Halby
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, Paris, France
| | - Paola B. Arimondo
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, Paris, France
| | - Philippe Glaser
- Ecology and Evolution of Antibiotic Resistance Unit, Microbiology Department, Institut Pasteur, CNRS UMR3525, Paris, France
| | | | - Helge B. Bode
- Department of Biosciences, Goethe University Frankfurt, Frankfurt, Germany
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, Marburg, Germany
| | - Rui Moreira
- Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Yanyan Li
- Unit MCAM, CNRS, National Museum of Natural History (MNHN), Paris, France
| | - Andriy Luzhetskyy
- Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | - Marnix H. Medema
- Bioinformatics Group, Wageningen University and Research, Wageningen, Netherlands
| | - Jean-Luc Pernodet
- Institute for Integrative Biology of the Cell (I2BC) & Microbiology Department, University of Paris-Saclay, Gif-sur-Yvette, France
| | - Marc Stadler
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Microbial Drugs (MWIS), Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | | | | | - Andrew W. Truman
- Department of Molecular Microbiology, John Innes Centre, Norwich, United Kingdom
| | - Kira J. Weissman
- Molecular and Structural Enzymology Group, Université de Lorraine, CNRS, IMoPA, Nancy, France
| | - Eriko Takano
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, University of Manchester, Manchester, United Kingdom
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Evi Stegmann
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Wolfgang Wohlleben
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Myriam Seemann
- Institute for Chemistry UMR 7177, University of Strasbourg/CNRS, ITI InnoVec, Strasbourg, France
| | - Martin Empting
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
| | - Alexander Titz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Timo Jaeger
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Silke Alt
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | | | | | - Andrea Schiefer
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Kenneth Pfarr
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Heather Graz
- Biophys Ltd., Usk, Monmouthshire, United Kingdom
| | - Michael Graz
- School of Law, University of Bristol, Bristol, United Kingdom
| | | | | | - Anders Karlén
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Hrvoje Petkovic
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany
| | | | | | - Laurent Fraisse
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Laura J. V. Piddock
- The Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland
| | - Ian H. Gilbert
- Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, United Kingdom
| | - Heinz E. Moser
- Novartis Institutes for BioMedical Research (NIBR), Emeryville, CA USA
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| |
Collapse
|
34
|
Cold-Start Problems in Data-Driven Prediction of Drug-Drug Interaction Effects. Pharmaceuticals (Basel) 2021; 14:ph14050429. [PMID: 34063324 PMCID: PMC8147651 DOI: 10.3390/ph14050429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/02/2023] Open
Abstract
Combining drugs, a phenomenon often referred to as polypharmacy, can induce additional adverse effects. The identification of adverse combinations is a key task in pharmacovigilance. In this context, in silico approaches based on machine learning are promising as they can learn from a limited number of combinations to predict for all. In this work, we identify various subtasks in predicting effects caused by drug–drug interaction. Predicting drug–drug interaction effects for drugs that already exist is very different from predicting outcomes for newly developed drugs, commonly called a cold-start problem. We propose suitable validation schemes for the different subtasks that emerge. These validation schemes are critical to correctly assess the performance. We develop a new model that obtains AUC-ROC =0.843 for the hardest cold-start task up to AUC-ROC =0.957 for the easiest one on the benchmark dataset of Zitnik et al. Finally, we illustrate how our predictions can be used to improve post-market surveillance systems or detect drug–drug interaction effects earlier during drug development.
Collapse
|
35
|
Vatansever S, Schlessinger A, Wacker D, Kaniskan HÜ, Jin J, Zhou M, Zhang B. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: State-of-the-arts and future directions. Med Res Rev 2021; 41:1427-1473. [PMID: 33295676 PMCID: PMC8043990 DOI: 10.1002/med.21764] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 01/11/2023]
Abstract
Neurological disorders significantly outnumber diseases in other therapeutic areas. However, developing drugs for central nervous system (CNS) disorders remains the most challenging area in drug discovery, accompanied with the long timelines and high attrition rates. With the rapid growth of biomedical data enabled by advanced experimental technologies, artificial intelligence (AI) and machine learning (ML) have emerged as an indispensable tool to draw meaningful insights and improve decision making in drug discovery. Thanks to the advancements in AI and ML algorithms, now the AI/ML-driven solutions have an unprecedented potential to accelerate the process of CNS drug discovery with better success rate. In this review, we comprehensively summarize AI/ML-powered pharmaceutical discovery efforts and their implementations in the CNS area. After introducing the AI/ML models as well as the conceptualization and data preparation, we outline the applications of AI/ML technologies to several key procedures in drug discovery, including target identification, compound screening, hit/lead generation and optimization, drug response and synergy prediction, de novo drug design, and drug repurposing. We review the current state-of-the-art of AI/ML-guided CNS drug discovery, focusing on blood-brain barrier permeability prediction and implementation into therapeutic discovery for neurological diseases. Finally, we discuss the major challenges and limitations of current approaches and possible future directions that may provide resolutions to these difficulties.
Collapse
Affiliation(s)
- Sezen Vatansever
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Avner Schlessinger
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Daniel Wacker
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - H. Ümit Kaniskan
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jian Jin
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ming‐Ming Zhou
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
36
|
An Ensemble Learning-Based Method for Inferring Drug-Target Interactions Combining Protein Sequences and Drug Fingerprints. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9933873. [PMID: 33987446 PMCID: PMC8093043 DOI: 10.1155/2021/9933873] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 11/24/2022]
Abstract
Identifying the interactions of the drug-target is central to the cognate areas including drug discovery and drug reposition. Although the high-throughput biotechnologies have made tremendous progress, the indispensable clinical trials remain to be expensive, laborious, and intricate. Therefore, a convenient and reliable computer-aided method has become the focus on inferring drug-target interactions (DTIs). In this research, we propose a novel computational model integrating a pyramid histogram of oriented gradients (PHOG), Position-Specific Scoring Matrix (PSSM), and rotation forest (RF) classifier for identifying DTIs. Specifically, protein primary sequences are first converted into PSSMs to describe the potential biological evolution information. After that, PHOG is employed to mine the highly representative features of PSSM from multiple pyramid levels, and the complete describers of drug-target pairs are generated by combining the molecular substructure fingerprints and PHOG features. Finally, we feed the complete describers into the RF classifier for effective prediction. The experiments of 5-fold Cross-Validations (CV) yield mean accuracies of 88.96%, 86.37%, 82.88%, and 76.92% on four golden standard data sets (enzyme, ion channel, G protein-coupled receptors (GPCRs), and nuclear receptor, respectively). Moreover, the paper also conducts the state-of-art light gradient boosting machine (LGBM) and support vector machine (SVM) to further verify the performance of the proposed model. The experimental outcomes substantiate that the established model is feasible and reliable to predict DTIs. There is an excellent prospect that our model is capable of predicting DTIs as an efficient tool on a large scale.
Collapse
|
37
|
Shen C, Luo J, Lai Z, Ding P. Multiview Joint Learning-Based Method for Identifying Small-Molecule-Associated MiRNAs by Integrating Pharmacological, Genomics, and Network Knowledge. J Chem Inf Model 2020; 60:4085-4097. [DOI: 10.1021/acs.jcim.0c00244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Cong Shen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Zihan Lai
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Pingjian Ding
- School of Computer Science, University of South China, Hengyang 421001, China
| |
Collapse
|