1
|
Ambreen S, Umar M, Noor A, Jain H, Ali R. Advanced AI and ML frameworks for transforming drug discovery and optimization: With innovative insights in polypharmacology, drug repurposing, combination therapy and nanomedicine. Eur J Med Chem 2025; 284:117164. [PMID: 39721292 DOI: 10.1016/j.ejmech.2024.117164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024]
Abstract
Artificial Intelligence (AI) and Machine Learning (ML) are transforming drug discovery by overcoming traditional challenges like high costs, time-consuming, and frequent failures. AI-driven approaches streamline key phases, including target identification, lead optimization, de novo drug design, and drug repurposing. Frameworks such as deep neural networks (DNNs), convolutional neural networks (CNNs), and deep reinforcement learning (DRL) models have shown promise in identifying drug targets, optimizing delivery systems, and accelerating drug repurposing. Generative adversarial networks (GANs) and variational autoencoders (VAEs) aid de novo drug design by creating novel drug-like compounds with desired properties. Case studies, such as DDR1 kinase inhibitors designed using generative models and CDK20 inhibitors developed via structure-based methods, highlight AI's ability to produce highly specific therapeutics. Models like SNF-CVAE and DeepDR further advance drug repurposing by uncovering new therapeutic applications for existing drugs. Advanced ML algorithms enhance precision in predicting drug efficacy, toxicity, and ADME-Tox properties, reducing development costs and improving drug-target interactions. AI also supports polypharmacology by optimizing multi-target drug interactions and enhances combination therapy through predictions of drug synergies and antagonisms. In nanomedicine, AI models like CURATE.AI and the Hartung algorithm optimize personalized treatments by predicting toxicological risks and real-time dosing adjustments with high accuracy. Despite its potential, challenges like data quality, model interpretability, and ethical concerns must be addressed. High-quality datasets, transparent models, and unbiased algorithms are essential for reliable AI applications. As AI continues to evolve, it is poised to revolutionize drug discovery and personalized medicine, advancing therapeutic development and patient care.
Collapse
Affiliation(s)
- Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Aaisha Noor
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Himangini Jain
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
2
|
Liu T, Wang S, Zhang Y, Li Y, Liu Y, Huang S. TIWMFLP: Two-Tier Interactive Weighted Matrix Factorization and Label Propagation Based on Similarity Matrix Fusion for Drug-Disease Association Prediction. J Chem Inf Model 2024; 64:8641-8654. [PMID: 39486090 DOI: 10.1021/acs.jcim.4c01589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
Accurately identifying new therapeutic uses for drugs is crucial for advancing pharmaceutical research and development. Matrix factorization is often used in association prediction due to its simplicity and high interpretability. However, existing matrix factorization models do not enable real-time interaction between molecular feature matrices and similarity matrices, nor do they consider the geometric structure of the matrices. Additionally, efficiently integrating multisource data remains a significant challenge. To address these issues, we propose a two-tier interactive weighted matrix factorization and label propagation model based on similarity matrix fusion (TIWMFLP) to assist in personalized treatment. First, we calculate the Gaussian and Laplace kernel similarities for drugs and diseases using known drug-disease associations. We then introduce a new multisource similarity fusion method, called similarity matrix fusion (SMF), to integrate these drug/disease similarities. SMF not only considers the different contributions represented by each neighbor but also incorporates drug-disease association information to enhance the contextual topological relationships and potential features of each drug/disease node in the network. Second, we innovatively developed a two-tier interactive weighted matrix factorization (TIWMF) method to process three biological networks. This method realizes for the first time the real-time interaction between the drug/disease feature matrix and its similarity matrix, allowing for a better capture of the complex relationships between drugs and diseases. Additionally, the weighted matrix of the drug/disease similarity matrix is introduced to preserve the underlying structure of the similarity matrix. Finally, the label propagation algorithm makes predictions based on the three updated biological networks. Experimental outcomes reveal that TIWMFLP consistently surpasses state-of-the-art models on four drug-disease data sets, two small molecule-miRNA data sets, and one miRNA-disease data set.
Collapse
Affiliation(s)
- Tiyao Liu
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Shudong Wang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Yuanyuan Zhang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao 266525, China
| | - Yunyin Li
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Yingye Liu
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Shiyuan Huang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| |
Collapse
|
3
|
Qureshi S, Iqbal SMZ, Ameer A, Karrila S, Ghadi YY, Shah SA. Enhancing drug-target interaction predictions in context of neurodegenerative diseases using bidirectional long short-term memory in male Swiss albino mice pharmaco-EEG analysis. Heliyon 2024; 10:e39279. [PMID: 39524776 PMCID: PMC11550650 DOI: 10.1016/j.heliyon.2024.e39279] [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: 02/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Background and Objective Emerging diseases like Parkinson or Alzheimer's, which are not curable, endanger human mental health and are challenging to research. Drug target interactions (DTI) are pivotal in the screening of candidate drugs and focus on a small pool of drug targets. Electroencephalogram shows the responses to psychotropic medicines in the brain bioelectric activity. Synaptic activity can be analyzed by using Local Field Potential recordings obtained from micro-electrodes implanted in the brain. The aim is to evaluate the effects of drug on brain bioelectric activity and increase the drug classification accuracy. The ultimate goal is to advance our understanding of how drugs affect synaptic activity and open the door to more focused treatment for neurodegenerative diseases. Methods In this study, Pharmaco-EEG recordings are processed using Advanced neural network models, particularly Convolutional Neural Networks, to assess the effects of medications. The five different medicines used in this study are Ephedrine, Fluoxetine, Kratom, Morphine, and Saline. The signals observed are local field potential signals. To overcome some limits of DTI prediction, we propose Bidirectional Long Short-Term Memory (LSTM) for the categorization of intracranial EEG (i-EEG) data, departing from standard approaches. Similar EEG patterns are presumably caused by drugs that work by homologous pharmacological pathways, producing similar psychotropic effects. To improve accuracy and reduce training loss, our study introduces a bidirectional LSTM model for classification along with Bayesian optimization. Results High recall, precision, and F1-Scores, particularly a 95% F1-Score for morphine, ephedrine, fluoxetine, and saline, suggest good performance in predicting these drug classes. Kratom produces a somewhat lower recall of 94%, but a high F1-Score of 97% and perfect precision of 1.00. The weighted average F1-Score, macro average, and overall accuracy are all consistently high (around 97%), indicating that the model works well throughout the spectrum of drugs. Conclusions Improved model performance was demonstrated by using a diversified dataset with five drug categories and bidirectional LSTM boosted with Bayesian optimization for hyperparameter tuning. From earlier limited-category models, it represents a substantial advancement.
Collapse
Affiliation(s)
- Shahnawaz Qureshi
- Intelligent Biomedical Application Lab, Sino-Pak center for Artificial Intelligence, School of Computing, Pak-Austria Fachhochschule: Institute of Applied Sciences and Technology, Mang Haripur, 22620, Pakistan
| | | | - Asif Ameer
- Department of Computer Science, National University of Computing and Emerging Sciences, Faisalabad, 38000, Pakistan
| | - Seppo Karrila
- Faculty of Science and Industrial Technology, Prince of Songkla University, Surat Thani Campus, Muang, Surat Thani, 84000, Thailand
| | - Yazeed Yasin Ghadi
- Department of Computer Science, Al Ain University Abu Dhab, Al Ain, United Arab Emirates
| | - Syed Aziz Shah
- Healthcare Sensing Technology, Faculty Research Centre for Intelligent Healthcare, Coventry University, Coventry, United Kingdom
| |
Collapse
|
4
|
Gu Y, Xu Z, Yang C. Empowering Graph Neural Network-Based Computational Drug Repositioning with Large Language Model-Inferred Knowledge Representation. Interdiscip Sci 2024:10.1007/s12539-024-00654-7. [PMID: 39325266 DOI: 10.1007/s12539-024-00654-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/27/2024]
Abstract
Computational drug repositioning, through predicting drug-disease associations (DDA), offers significant potential for discovering new drug indications. Current methods incorporate graph neural networks (GNN) on drug-disease heterogeneous networks to predict DDAs, achieving notable performances compared to traditional machine learning and matrix factorization approaches. However, these methods depend heavily on network topology, hampered by incomplete and noisy network data, and overlook the wealth of biomedical knowledge available. Correspondingly, large language models (LLMs) excel in graph search and relational reasoning, which can possibly enhance the integration of comprehensive biomedical knowledge into drug and disease profiles. In this study, we first investigate the contribution of LLM-inferred knowledge representation in drug repositioning and DDA prediction. A zero-shot prompting template was designed for LLM to extract high-quality knowledge descriptions for drug and disease entities, followed by embedding generation from language models to transform the discrete text to continual numerical representation. Then, we proposed LLM-DDA with three different model architectures (LLM-DDANode Feat, LLM-DDADual GNN, LLM-DDAGNN-AE) to investigate the best fusion mode for LLM-based embeddings. Extensive experiments on four DDA benchmarks show that, LLM-DDAGNN-AE achieved the optimal performance compared to 11 baselines with the overall relative improvement in AUPR of 23.22%, F1-Score of 17.20%, and precision of 25.35%. Meanwhile, selected case studies of involving Prednisone and Allergic Rhinitis highlighted the model's capability to identify reliable DDAs and knowledge descriptions, supported by existing literature. This study showcases the utility of LLMs in drug repositioning with its generality and applicability in other biomedical relation prediction tasks.
Collapse
Affiliation(s)
- Yaowen Gu
- Department of Chemistry, New York University, New York, NY, 10003, USA
| | - Zidu Xu
- School of Nursing, Columbia University, 560 W 168th Street, New York, NY, 10032, USA.
| | - Carl Yang
- Department of Computer Science, Emory College of Arts and Sciences, Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
5
|
Wang Q, Hu X, Wei Z, Lu H, Liu H. Reinforcement learning-driven exploration of peptide space: accelerating generation of drug-like peptides. Brief Bioinform 2024; 25:bbae444. [PMID: 39256196 PMCID: PMC11387070 DOI: 10.1093/bib/bbae444] [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: 03/17/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Using amino acid residues in peptide generation has solved several key problems, including precise control of amino acid sequence order, customized peptides for property modification, and large-scale peptide synthesis. Proteins contain unknown amino acid residues. Extracting them for the synthesis of drug-like peptides can create novel structures with unique properties, driving drug development. Computer-aided design of novel peptide drug molecules can solve the high-cost and low-efficiency problems in the traditional drug discovery process. Previous studies faced limitations in enhancing the bioactivity and drug-likeness of polypeptide drugs due to less emphasis on the connection relationships in amino acid structures. Thus, we proposed a reinforcement learning-driven generation model based on graph attention mechanisms for peptide generation. By harnessing the advantages of graph attention mechanisms, this model effectively captured the connectivity structures between amino acid residues in peptides. Simultaneously, leveraging reinforcement learning's strength in guiding optimal sequence searches provided a novel approach to peptide design and optimization. This model introduces an actor-critic framework with real-time feedback loops to achieve dynamic balance between attributes, which can customize the generation of multiple peptides for specific targets and enhance the affinity between peptides and targets. Experimental results demonstrate that the generated drug-like peptides meet specified absorption, distribution, metabolism, excretion, and toxicity properties and bioactivity with a success rate of over 90$\%$, thereby significantly accelerating the process of drug-like peptide generation.
Collapse
Affiliation(s)
- Qian Wang
- College of Computer Science and Technology, Ocean University of China, 238 Songling Rd, 266100 Shandong, China
| | - Xiaotong Hu
- College of Computer Science and Technology, Ocean University of China, 238 Songling Rd, 266100 Shandong, China
| | - Zhiqiang Wei
- College of Computer Science and Technology, Ocean University of China, 238 Songling Rd, 266100 Shandong, China
| | - Hao Lu
- College of Computer Science and Technology, Ocean University of China, 238 Songling Rd, 266100 Shandong, China
| | - Hao Liu
- College of Computer Science and Technology, Ocean University of China, 238 Songling Rd, 266100 Shandong, China
| |
Collapse
|
6
|
Liu F, Patt A, Chen C, Huang R, Xu Y, Mathé EA, Zhu Q. Exploring NCATS in-house biomedical data for evidence-based drug repurposing. PLoS One 2024; 19:e0289518. [PMID: 38271343 PMCID: PMC10810548 DOI: 10.1371/journal.pone.0289518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/08/2023] [Indexed: 01/27/2024] Open
Abstract
Drug repurposing is a strategy for identifying new uses of approved or investigational drugs that are outside the scope of the original medical indication. Even though many repurposed drugs have been found serendipitously in the past, the increasing availability of large volumes of biomedical data has enabled more systemic, data-driven approaches for drug candidate identification. At National Center of Advancing Translational Sciences (NCATS), we invent new methods to generate new data and information publicly available to spur innovation and scientific discovery. In this study, we aimed to explore and demonstrate biomedical data generated and collected via two NCATS research programs, the Toxicology in the 21st Century program (Tox21) and the Biomedical Data Translator (Translator) for the application of drug repurposing. These two programs provide complementary types of biomedical data from uncovering underlying biological mechanisms with bioassay screening data from Tox21 for chemical clustering, to enrich clustered chemicals with scientific evidence mined from the Translator towards drug repurposing. 129 chemical clusters have been generated and three of them have been further investigated for drug repurposing candidate identification, which is detailed as case studies.
Collapse
Affiliation(s)
- Fang Liu
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Andrew Patt
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| | - Chloe Chen
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Ruili Huang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| | - Yanji Xu
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Ewy A. Mathé
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| | - Qian Zhu
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| |
Collapse
|
7
|
Rani P, Dutta K, Kumar V. Performance evaluation of drug synergy datasets using computational intelligence approaches. MULTIMEDIA TOOLS AND APPLICATIONS 2024; 83:8971-8997. [DOI: 10.1007/s11042-023-15723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 04/18/2023] [Indexed: 01/03/2025]
|
8
|
Gao Z, Ding P, Xu R. IUPHAR review - Data-driven computational drug repurposing approaches for opioid use disorder. Pharmacol Res 2024; 199:106960. [PMID: 37832859 DOI: 10.1016/j.phrs.2023.106960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Opioid Use Disorder (OUD) is a chronic and relapsing condition characterized by the misuse of opioid drugs, causing significant morbidity and mortality in the United States. Existing medications for OUD are limited, and there is an immediate need to discover treatments with enhanced safety and efficacy. Drug repurposing aims to find new indications for existing medications, offering a time-saving and cost-efficient alternative strategy to traditional drug discovery. Computational approaches have been developed to further facilitate the drug repurposing process. In this paper, we reviewed state-of-the-art data-driven computational drug repurposing approaches for OUD and discussed their advantages and potential challenges. We also highlighted promising repurposed candidate drugs for OUD that were identified by computational drug repurposing techniques and reviewed studies supporting their potential mechanisms of action in treating OUD.
Collapse
Affiliation(s)
- Zhenxiang Gao
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Pingjian Ding
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
9
|
Ai C, Yang H, Ding Y, Tang J, Guo F. Low Rank Matrix Factorization Algorithm Based on Multi-Graph Regularization for Detecting Drug-Disease Association. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:3033-3043. [PMID: 37159322 DOI: 10.1109/tcbb.2023.3274587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Detecting potential associations between drugs and diseases plays an indispensable role in drug development, which has also become a research hotspot in recent years. Compared with traditional methods, some computational approaches have the advantages of fast speed and low cost, which greatly accelerate the progress of predicting the drug-disease association. In this study, we propose a novel similarity-based method of low-rank matrix decomposition based on multi-graph regularization. On the basis of low-rank matrix factorization with L2 regularization, the multi-graph regularization constraint is constructed by combining a variety of similarity matrices from drugs and diseases respectively. In the experiments, we analyze the difference in the combination of different similarities, resulting that combining all the similarity information on drug space is unnecessary, and only a part of the similarity information can achieve the desired performance. Then our method is compared with other existing models on three data sets (Fdataset, Cdataset and LRSSLdataset) and have a good advantage in the evaluation measurement of AUPR. Besides, a case study experiment is conducted and showing that the superior ability for predicting the potential disease-related drugs of our model. Finally, we compare our model with some methods on six real world datasets, and our model has a good performance in detecting real world data.
Collapse
|
10
|
Ding J, Qiao Y, Zhang L. Plant disease prescription recommendation based on electronic medical records and sentence embedding retrieval. PLANT METHODS 2023; 19:91. [PMID: 37633904 PMCID: PMC10463767 DOI: 10.1186/s13007-023-01070-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/11/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND In the era of Agri 4.0 and the popularity of Plantwise systems, the availability of Plant Electronic Medical Records has provided opportunities to extract valuable disease information and treatment knowledge. However, developing an effective prescription recommendation method based on these records presents unique challenges, such as inadequate labeling data, lack of structural and linguistic specifications, incorporation of new prescriptions, and consideration of multiple factors in practical situations. RESULTS This study proposes a plant disease prescription recommendation method called PRSER, which is based on sentence embedding retrieval. The semantic matching model is created using a pre-trained language model and a sentence embedding method with contrast learning ideas, and the constructed prescription reference database is retrieved for optimal prescription recommendations. A multi-vegetable disease dataset and a multi-fruit disease dataset are constructed to compare three pre-trained language models, four pooling types, and two loss functions. The PRSER model achieves the best semantic matching performance by combining MacBERT, CoSENT, and CLS pooling, resulting in a Pearson coefficient of 86.34% and a Spearman coefficient of 77.67%. The prescription recommendation capability of the model is also verified. PRSER performs well in closed-set testing with Top-1/Top-3/Top-5 accuracy of 88.20%/96.07%/97.70%; and slightly worse in open-set testing with Top-1/Top-3/Top-5 accuracy of 82.04%/91.50%/94.90%. Finally, a plant disease prescription recommendation system for mobile terminals is constructed and its generalization ability with incomplete inputs is verified. When only symptom information is available without environment and plant information, our model shows slightly lower accuracy with Top-1/Top-3/Top-5 accuracy of 75.24%/88.35%/91.99% in closed-set testing and Top-1/Top-3/Top-5 accuracy of 75.08%/87.54%/89.84% in open-set testing. CONCLUSIONS The experiments validate the effectiveness and generalization ability of the proposed approach for recommending plant disease prescriptions. This research has significant potential to facilitate the implementation of artificial intelligence in plant disease treatment, addressing the needs of farmers and advancing scientific plant disease management.
Collapse
Affiliation(s)
- Junqi Ding
- China Agricultural University, Beijing, 100083, China
| | - Yan Qiao
- Beijing Plant Protection Station, Beijing, 100029, China
| | - Lingxian Zhang
- China Agricultural University, Beijing, 100083, China.
- Key Laboratory of Agricultural Informationization Standardization, Ministry of Agriculture and Rural Affairs, Beijing, China.
- College of Information and Electrical Engineering, China Agricultural University, 209# No.17 Qinghua Donglu, Haidian District, Beijing, 100083, China.
| |
Collapse
|
11
|
Liu F, Patt A, Chen C, Huang R, Xu Y, Mathé EA, Zhu Q. Exploring NCATS In-House Biomedical Data for Evidence-based Drug Repurposing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.550045. [PMID: 37546930 PMCID: PMC10401966 DOI: 10.1101/2023.07.21.550045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Drug repurposing is a strategy for identifying new uses of approved or investigational drugs that are outside the scope of the original medical indication. Even though many repurposed drugs have been found serendipitously in the past, the increasing availability of large volumes of biomedical data has enabled more systemic, data-driven approaches for drug candidate identification. At National Center of Advancing Translational Sciences (NCATS), we invent new methods to generate new data and information publicly available to spur innovation and scientific discovery. In this study, we aimed to explore and demonstrate biomedical data generated and collected via two NCATS research programs, the Toxicology in the 21st Century program (Tox21) and the Biomedical Data Translator (Translator) for the application of drug repurposing. These two programs provide complementary types of biomedical data from uncovering underlying biological mechanisms with bioassay screening data from Tox21 for chemical clustering, to enrich clustered chemicals with scientific evidence mined from the Translator towards drug repurposing. 129 chemical clusters have been generated and three of them have been further investigated for drug repurposing candidate identification, which is detailed as case studies.
Collapse
Affiliation(s)
- Fang Liu
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, MD
| | - Andrew Patt
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD
| | - Chloe Chen
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, MD
| | - Ruili Huang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD
| | - Yanji Xu
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, MD
| | - Ewy A Mathé
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD
| | - Qian Zhu
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD
| |
Collapse
|
12
|
Rassil A, Chougrad H, Zouaki H. Deep multi-agent fusion Q-Network for graph generation. Knowl Based Syst 2023. [DOI: 10.1016/j.knosys.2023.110509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
13
|
Zhu Z, Yao Z, Qi G, Mazur N, Yang P, Cong B. Associative learning mechanism for drug‐target interaction prediction. CAAI TRANSACTIONS ON INTELLIGENCE TECHNOLOGY 2023. [DOI: 10.1049/cit2.12194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Affiliation(s)
- Zhiqin Zhu
- College of Automation Chongqing University of Posts and Telecommunications Chongqing China
| | - Zheng Yao
- College of Automation Chongqing University of Posts and Telecommunications Chongqing China
| | - Guanqiu Qi
- Computer Information Systems Department State University of New York at Buffalo State Buffalo New York USA
| | - Neal Mazur
- Computer Information Systems Department State University of New York at Buffalo State Buffalo New York USA
| | - Pan Yang
- Department of Cardiovascular Surgery Chongqing General Hospital University of Chinese Academy of Sciences Chongqing China
- Emergency Department The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Baisen Cong
- Data Scientist Diagnostics Digital DH (Shanghai) Diagnostics Co., Ltd. Danaher Company Shanghai China
| |
Collapse
|
14
|
Choi Y, Li R, Quon G. siVAE: interpretable deep generative models for single-cell transcriptomes. Genome Biol 2023; 24:29. [PMID: 36803416 PMCID: PMC9940350 DOI: 10.1186/s13059-023-02850-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/06/2023] [Indexed: 02/22/2023] Open
Abstract
Neural networks such as variational autoencoders (VAE) perform dimensionality reduction for the visualization and analysis of genomic data, but are limited in their interpretability: it is unknown which data features are represented by each embedding dimension. We present siVAE, a VAE that is interpretable by design, thereby enhancing downstream analysis tasks. Through interpretation, siVAE also identifies gene modules and hubs without explicit gene network inference. We use siVAE to identify gene modules whose connectivity is associated with diverse phenotypes such as iPSC neuronal differentiation efficiency and dementia, showcasing the wide applicability of interpretable generative models for genomic data analysis.
Collapse
Affiliation(s)
- Yongin Choi
- Graduate Group in Biomedical Engineering, University of California, Davis, Davis, CA, USA
- Genome Center, University of California, Davis, Davis, CA, USA
| | - Ruoxin Li
- Genome Center, University of California, Davis, Davis, CA, USA
- Graduate Group in Biostatistics, University of California, Davis, Davis, CA, USA
| | - Gerald Quon
- Graduate Group in Biomedical Engineering, University of California, Davis, Davis, CA, USA.
- Genome Center, University of California, Davis, Davis, CA, USA.
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
15
|
Peng Y, Zhao S, Zeng Z, Hu X, Yin Z. LGBMDF: A cascade forest framework with LightGBM for predicting drug-target interactions. Front Microbiol 2023; 13:1092467. [PMID: 36687573 PMCID: PMC9849804 DOI: 10.3389/fmicb.2022.1092467] [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: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023] Open
Abstract
Prediction of drug-target interactions (DTIs) plays an important role in drug development. However, traditional laboratory methods to determine DTIs require a lot of time and capital costs. In recent years, many studies have shown that using machine learning methods to predict DTIs can speed up the drug development process and reduce capital costs. An excellent DTI prediction method should have both high prediction accuracy and low computational cost. In this study, we noticed that the previous research based on deep forests used XGBoost as the estimator in the cascade, we applied LightGBM instead of XGBoost to the cascade forest as the estimator, then the estimator group was determined experimentally as three LightGBMs and three ExtraTrees, this new model is called LGBMDF. We conducted 5-fold cross-validation on LGBMDF and other state-of-the-art methods using the same dataset, and compared their Sn, Sp, MCC, AUC and AUPR. Finally, we found that our method has better performance and faster calculation speed.
Collapse
|
16
|
Tangmanussukum P, Kawichai T, Suratanee A, Plaimas K. Heterogeneous network propagation with forward similarity integration to enhance drug-target association prediction. PeerJ Comput Sci 2022; 8:e1124. [PMID: 36262151 PMCID: PMC9575853 DOI: 10.7717/peerj-cs.1124] [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: 06/17/2022] [Accepted: 09/14/2022] [Indexed: 06/16/2023]
Abstract
Identification of drug-target interaction (DTI) is a crucial step to reduce time and cost in the drug discovery and development process. Since various biological data are publicly available, DTIs have been identified computationally. To predict DTIs, most existing methods focus on a single similarity measure of drugs and target proteins, whereas some recent methods integrate a particular set of drug and target similarity measures by a single integration function. Therefore, many DTIs are still missing. In this study, we propose heterogeneous network propagation with the forward similarity integration (FSI) algorithm, which systematically selects the optimal integration of multiple similarity measures of drugs and target proteins. Seven drug-drug and nine target-target similarity measures are applied with four distinct integration methods to finally create an optimal heterogeneous network model. Consequently, the optimal model uses the target similarity based on protein sequences and the fused drug similarity, which combines the similarity measures based on chemical structures, the Jaccard scores of drug-disease associations, and the cosine scores of drug-drug interactions. With an accuracy of 99.8%, this model significantly outperforms others that utilize different similarity measures of drugs and target proteins. In addition, the validation of the DTI predictions of this model demonstrates the ability of our method to discover missing potential DTIs.
Collapse
Affiliation(s)
- Piyanut Tangmanussukum
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Thitipong Kawichai
- Department of Mathematics and Computer Science, Academic Division, Chulachomklao Royal Military Academy, Nakhon Nayok, Thailand
| | - Apichat Suratanee
- Department of Mathematics, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
- Intelligent and Nonlinear Dynamics Innovations Research Center, Science and Technology Research Institute, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
| | - Kitiporn Plaimas
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Omics Science and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
An explainable framework for drug repositioning from disease information network. Neurocomputing 2022. [DOI: 10.1016/j.neucom.2022.09.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Zeng Y, Chen X, Peng D, Zhang L, Huang H. Multi-scaled self-attention for drug-target interaction prediction based on multi-granularity representation. BMC Bioinformatics 2022; 23:314. [PMID: 35922768 PMCID: PMC9347097 DOI: 10.1186/s12859-022-04857-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/22/2022] [Indexed: 11/21/2022] Open
Abstract
Background Drug–target interaction (DTI) prediction plays a crucial role in drug discovery. Although the advanced deep learning has shown promising results in predicting DTIs, it still needs improvements in two aspects: (1) encoding method, in which the existing encoding method, character encoding, overlooks chemical textual information of atoms with multiple characters and chemical functional groups; as well as (2) the architecture of deep model, which should focus on multiple chemical patterns in drug and target representations. Results In this paper, we propose a multi-granularity multi-scaled self-attention (SAN) model by alleviating the above problems. Specifically, in process of encoding, we investigate a segmentation method for drug and protein sequences and then label the segmented groups as the multi-granularity representations. Moreover, in order to enhance the various local patterns in these multi-granularity representations, a multi-scaled SAN is built and exploited to generate deep representations of drugs and targets. Finally, our proposed model predicts DTIs based on the fusion of these deep representations. Our proposed model is evaluated on two benchmark datasets, KIBA and Davis. The experimental results reveal that our proposed model yields better prediction accuracy than strong baseline models. Conclusion Our proposed multi-granularity encoding method and multi-scaled SAN model improve DTI prediction by encoding the chemical textual information of drugs and targets and extracting their various local patterns, respectively.
Collapse
Affiliation(s)
- Yuni Zeng
- School of Information Science and Technology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiangru Chen
- College of Computer Science, Sichuan University, Chengdu, China
| | - Dezhong Peng
- College of Computer Science, Sichuan University, Chengdu, China.,Shenzhen Peng Cheng Laboratory, Shenzhen, China.,Chengdu Sobey Digital Technology Co., Ltd, Chengdu, China
| | - Lijun Zhang
- Sichuan Zhiqian Technology Co., Ltd, Chengdu, China.,Chengdu Ruibei Yingte Information Technology Co., Ltd, Chengdu, China
| | - Haixiao Huang
- Sichuan Provincial Commission of Politics and Law, Chengdu, China.
| |
Collapse
|
19
|
Pan X, Lin X, Cao D, Zeng X, Yu PS, He L, Nussinov R, Cheng F. Deep learning for drug repurposing: Methods, databases, and applications. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2022. [DOI: 10.1002/wcms.1597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xiaoqin Pan
- School of Computer Science and Engineering Hunan University Changsha Hunan China
| | - Xuan Lin
- School of Computer Science Xiangtan University Xiangtan China
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education Xiangtan University Xiangtan China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences Central South University Changsha China
| | - Xiangxiang Zeng
- School of Computer Science and Engineering Hunan University Changsha Hunan China
| | - Philip S. Yu
- Department of Computer Science University of Illinois at Chicago Chicago Illinois USA
| | - Lifang He
- Department of Computer Science and Engineering Lehigh University Bethlehem Pennsylvania USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research National Cancer Institute at Frederick Frederick Maryland USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel Aviv University Tel Aviv Israel
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Cleveland Ohio USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine Case Western Reserve University Cleveland Ohio USA
- Case Comprehensive Cancer Center Case Western Reserve University School of Medicine Cleveland Ohio USA
| |
Collapse
|
20
|
Yi T, Zhang S, Bu Z, Du J, Fang C. Link prediction based on higher-order structure extraction and autoencoder learning in directed networks. Knowl Based Syst 2022. [DOI: 10.1016/j.knosys.2022.108241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Zhao J, Shao M, Wang H, Yu X, Li B, Liu X. Cyber threat prediction using dynamic heterogeneous graph learning. Knowl Based Syst 2022. [DOI: 10.1016/j.knosys.2021.108086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
22
|
Identifying Cancer Sub-types from genomic scale data sets using Confidence Based Integration (CBI). J Biomed Inform 2022; 126:103997. [DOI: 10.1016/j.jbi.2022.103997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/03/2022] [Accepted: 01/13/2022] [Indexed: 11/20/2022]
|
23
|
Drug–disease associations prediction via Multiple Kernel-based Dual Graph Regularized Least Squares. Appl Soft Comput 2021. [DOI: 10.1016/j.asoc.2021.107811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
24
|
Yan S, Yang A, Kong S, Bai B, Li X. Predictive intelligence powered attentional stacking matrix factorization algorithm for the computational drug repositioning. Appl Soft Comput 2021. [DOI: 10.1016/j.asoc.2021.107633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Gupta R, Srivastava D, Sahu M, Tiwari S, Ambasta RK, Kumar P. Artificial intelligence to deep learning: machine intelligence approach for drug discovery. Mol Divers 2021; 25:1315-1360. [PMID: 33844136 PMCID: PMC8040371 DOI: 10.1007/s11030-021-10217-3] [Citation(s) in RCA: 407] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Drug designing and development is an important area of research for pharmaceutical companies and chemical scientists. However, low efficacy, off-target delivery, time consumption, and high cost impose a hurdle and challenges that impact drug design and discovery. Further, complex and big data from genomics, proteomics, microarray data, and clinical trials also impose an obstacle in the drug discovery pipeline. Artificial intelligence and machine learning technology play a crucial role in drug discovery and development. In other words, artificial neural networks and deep learning algorithms have modernized the area. Machine learning and deep learning algorithms have been implemented in several drug discovery processes such as peptide synthesis, structure-based virtual screening, ligand-based virtual screening, toxicity prediction, drug monitoring and release, pharmacophore modeling, quantitative structure-activity relationship, drug repositioning, polypharmacology, and physiochemical activity. Evidence from the past strengthens the implementation of artificial intelligence and deep learning in this field. Moreover, novel data mining, curation, and management techniques provided critical support to recently developed modeling algorithms. In summary, artificial intelligence and deep learning advancements provide an excellent opportunity for rational drug design and discovery process, which will eventually impact mankind. The primary concern associated with drug design and development is time consumption and production cost. Further, inefficiency, inaccurate target delivery, and inappropriate dosage are other hurdles that inhibit the process of drug delivery and development. With advancements in technology, computer-aided drug design integrating artificial intelligence algorithms can eliminate the challenges and hurdles of traditional drug design and development. Artificial intelligence is referred to as superset comprising machine learning, whereas machine learning comprises supervised learning, unsupervised learning, and reinforcement learning. Further, deep learning, a subset of machine learning, has been extensively implemented in drug design and development. The artificial neural network, deep neural network, support vector machines, classification and regression, generative adversarial networks, symbolic learning, and meta-learning are examples of the algorithms applied to the drug design and discovery process. Artificial intelligence has been applied to different areas of drug design and development process, such as from peptide synthesis to molecule design, virtual screening to molecular docking, quantitative structure-activity relationship to drug repositioning, protein misfolding to protein-protein interactions, and molecular pathway identification to polypharmacology. Artificial intelligence principles have been applied to the classification of active and inactive, monitoring drug release, pre-clinical and clinical development, primary and secondary drug screening, biomarker development, pharmaceutical manufacturing, bioactivity identification and physiochemical properties, prediction of toxicity, and identification of mode of action.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Swati Tiwari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
26
|
Jarada TN, Rokne JG, Alhajj R. SNF-NN: computational method to predict drug-disease interactions using similarity network fusion and neural networks. BMC Bioinformatics 2021; 22:28. [PMID: 33482713 PMCID: PMC7821180 DOI: 10.1186/s12859-020-03950-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 12/22/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Drug repositioning is an emerging approach in pharmaceutical research for identifying novel therapeutic potentials for approved drugs and discover therapies for untreated diseases. Due to its time and cost efficiency, drug repositioning plays an instrumental role in optimizing the drug development process compared to the traditional de novo drug discovery process. Advances in the genomics, together with the enormous growth of large-scale publicly available data and the availability of high-performance computing capabilities, have further motivated the development of computational drug repositioning approaches. More recently, the rise of machine learning techniques, together with the availability of powerful computers, has made the area of computational drug repositioning an area of intense activities. RESULTS In this study, a novel framework SNF-NN based on deep learning is presented, where novel drug-disease interactions are predicted using drug-related similarity information, disease-related similarity information, and known drug-disease interactions. Heterogeneous similarity information related to drugs and disease is fed to the proposed framework in order to predict novel drug-disease interactions. SNF-NN uses similarity selection, similarity network fusion, and a highly tuned novel neural network model to predict new drug-disease interactions. The robustness of SNF-NN is evaluated by comparing its performance with nine baseline machine learning methods. The proposed framework outperforms all baseline methods ([Formula: see text] = 0.867, and [Formula: see text]=0.876) using stratified 10-fold cross-validation. To further demonstrate the reliability and robustness of SNF-NN, two datasets are used to fairly validate the proposed framework's performance against seven recent state-of-the-art methods for drug-disease interaction prediction. SNF-NN achieves remarkable performance in stratified 10-fold cross-validation with [Formula: see text] ranging from 0.879 to 0.931 and [Formula: see text] from 0.856 to 0.903. Moreover, the efficiency of SNF-NN is verified by validating predicted unknown drug-disease interactions against clinical trials and published studies. CONCLUSION In conclusion, computational drug repositioning research can significantly benefit from integrating similarity measures in heterogeneous networks and deep learning models for predicting novel drug-disease interactions. The data and implementation of SNF-NN are available at http://pages.cpsc.ucalgary.ca/ tnjarada/snf-nn.php .
Collapse
Affiliation(s)
- Tamer N Jarada
- Department of Computer Science, University of Calgary, Calgary, AB, Canada
| | - Jon G Rokne
- Department of Computer Science, University of Calgary, Calgary, AB, Canada
| | - Reda Alhajj
- Department of Computer Science, University of Calgary, Calgary, AB, Canada.
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey.
- Department of Health Informatics, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|