1
|
Monsia R, Bhattacharyya S. Efficient and Explainable Virtual Screening of Molecules through Fingerprint-Generating Networks Integrated with Artificial Neural Networks. ACS OMEGA 2025; 10:4896-4911. [PMID: 39959102 PMCID: PMC11822703 DOI: 10.1021/acsomega.4c10289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 02/18/2025]
Abstract
A machine learning-based drug screening technique has been developed and optimized using a novel, stitched neural network architecture with trainable, graph convolution-based fingerprints as a base into an artificial neural network. The architecture is efficient, explainable, and performant as a tool for the binary classification of ligands based on a user-chosen docking score threshold. Assessment using two standardized virtual screening databases substantiated the architecture's ability to learn molecular features and substructures and predict ligand classes based on binding affinity values more effectively than similar contemporary counterparts. Furthermore, to highlight the architecture's utility to groups and laboratories with varying resources, experiments were carried out using randomly sampled small molecules from the ZINC database and their computational docking scores against six drug-design relevant proteins. This new architecture proved to be more efficient in screening molecules that less favorably bind to a specific target thereby retaining top-hit molecules. Compared to similar protocols developed using Morgan fingerprints, the neural fingerprint-based model shows superiority in retaining the best ligands while filtering molecules at a higher relative rate. Lastly, the explainability of the model was investigated; it was revealed that the model accurately emphasized important chemical substructures and atoms through the intermediate fingerprint, which, in turn, contributed heavily to the ultimate prediction of a ligand as binding tightly to a certain protein.
Collapse
Affiliation(s)
| | - Sudeep Bhattacharyya
- Department of Chemistry and
Biochemistry, University of Wisconsin—Eau
Claire, Eau Claire, Wisconsin 54701, United States
| |
Collapse
|
2
|
Suo Y, Qian X, Xiong Z, Liu X, Wang C, Mu B, Wu X, Lu W, Cui M, Liu J, Chen Y, Zheng M, Lu X. Enhancing the Predictive Power of Machine Learning Models through a Chemical Space Complementary DEL Screening Strategy. J Med Chem 2024; 67:18969-18980. [PMID: 39441849 DOI: 10.1021/acs.jmedchem.4c01416] [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: 10/25/2024]
Abstract
DNA-encoded library (DEL) technology is an effective method for small molecule drug discovery, enabling high-throughput screening against target proteins. While DEL screening produces extensive data, it can reveal complex patterns not easily recognized by human analysis. Lead compounds from DEL screens often have higher molecular weights, posing challenges for drug development. This study refines traditional DELs by integrating alternative techniques like photocross-linking screening to enhance chemical diversity. Combining these methods improved predictive performance for small molecule identification models. Using this approach, we predicted active small molecules for BRD4 and p300, achieving hit rates of 26.7 and 35.7%. Notably, the identified compounds exhibit smaller molecular weights and better modification potential compared to traditional DEL molecules. This research demonstrates the synergy between DEL and AI technologies, enhancing drug discovery.
Collapse
Affiliation(s)
- Yanrui Suo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Zhang Jiang Hi-Tech Park, Pudong, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xu Qian
- DEL Department, Suzhou Alphama Biotechnology Co., Ltd., Suzhou 215125,China
| | - Zhaoping Xiong
- Technology Development Department, Suzhou Alphama Biotechnology Co., Ltd., Suzhou 215125,China
| | - Xiaohong Liu
- Technology Development Department, Suzhou Alphama Biotechnology Co., Ltd., Suzhou 215125,China
| | - Chao Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Zhang Jiang Hi-Tech Park, Pudong, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Baiyang Mu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Zhang Jiang Hi-Tech Park, Pudong, Shanghai 201203, China
- Shandong Second Medical University, Weifang 261053, China
| | - Xinyuan Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Zhang Jiang Hi-Tech Park, Pudong, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Weiwei Lu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Zhang Jiang Hi-Tech Park, Pudong, Shanghai 201203, China
| | - Meiying Cui
- DEL Department, Suzhou Alphama Biotechnology Co., Ltd., Suzhou 215125,China
| | - Jiaxiang Liu
- DEL Department, Suzhou Alphama Biotechnology Co., Ltd., Suzhou 215125,China
| | - Yujie Chen
- DEL Department, Suzhou Alphama Biotechnology Co., Ltd., Suzhou 215125,China
| | - Mingyue Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Zhang Jiang Hi-Tech Park, Pudong, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaojie Lu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Zhang Jiang Hi-Tech Park, Pudong, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
3
|
Monsia R, Bhattacharyya S. Virtual Screening of Molecules via Neural Fingerprint-based Deep Learning Technique. RESEARCH SQUARE 2024:rs.3.rs-4355625. [PMID: 38766198 PMCID: PMC11100899 DOI: 10.21203/rs.3.rs-4355625/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
A machine learning-based drug screening technique has been developed and optimized using convolutional neural network-derived fingerprints. The optimization of weights in the neural network-based fingerprinting technique was compared with fixed Morgan fingerprints in regard to binary classification on drug-target binding affinity. The assessment was carried out using six different target proteins using randomly chosen small molecules from the ZINC15 database for training. This new architecture proved to be more efficient in screening molecules that less favorably bind to specific targets and retaining molecules that favorably bind to it. Scientific contribution We have developed a new neural fingerprint-based screening model that has a significant ability to capture hits. Despite using a smaller dataset, this model is capable of mapping chemical space similar to other contemporary algorithms designed for molecular screening. The novelty of the present algorithm lies in the speed with which the models are trained and tuned before testing its predictive capabilities and hence is a significant step forward in the field of machine learning-embedded computational drug discovery.
Collapse
|
4
|
Sadeghi A, Sadeghi M, Fakhar M, Zakariaei Z, Sadeghi M. Scoping Review of Deep Learning Techniques for Diagnosis, Drug Discovery, and Vaccine Development in Leishmaniasis. Transbound Emerg Dis 2024; 2024:6621199. [PMID: 40303156 PMCID: PMC12019899 DOI: 10.1155/2024/6621199] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 10/15/2023] [Accepted: 12/21/2023] [Indexed: 05/02/2025]
Abstract
Leishmania, a single-cell parasite prevalent in tropical and subtropical regions worldwide, can cause varying degrees of leishmaniasis, ranging from self-limiting skin lesions to potentially fatal visceral complications. As such, the parasite has been the subject of much interest in the scientific community. In recent years, advances in diagnostic techniques such as flow cytometry, molecular biology, proteomics, and nanodiagnosis have contributed to progress in the diagnosis of this deadly disease. Additionally, the emergence of artificial intelligence (AI), including its subbranches such as machine learning and deep learning, has revolutionized the field of medicine. The high accuracy of AI and its potential to reduce human and laboratory errors make it an especially promising tool in diagnosis and treatment. Despite the promising potential of deep learning in the medical field, there has been no review study on the applications of this technology in the context of leishmaniasis. To address this gap, we provide a scoping review of deep learning methods in the diagnosis of the disease, drug discovery, and vaccine development. In conducting a thorough search of available literature, we analyzed articles in detail that used deep learning methods for various aspects of the disease, including diagnosis, drug discovery, vaccine development, and related proteins. Each study was individually analyzed, and the methodology and results were presented. As the first and only review study on this topic, this paper serves as a quick and comprehensive resource and guide for the future research in this field.
Collapse
Affiliation(s)
- Alireza Sadeghi
- Intelligent Mobile Robot Lab (IMRL), Department of Mechatronics Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mahdieh Sadeghi
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahdi Fakhar
- Toxoplasmosis Research Center, Iranian National Registry Center for Lophomoniasis and Toxoplasmosis, Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zakaria Zakariaei
- Toxicology and Forensic Medicine Division, Mazandaran Registry Center for Opioids Poisoning, Antimicrobial Resistance Research Center, Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | | |
Collapse
|
5
|
Güner E, Özkan Ö, Yalcin-Ozkat G, Ölgen S. Determination of Novel SARS-CoV-2 Inhibitors by Combination of Machine Learning and Molecular Modeling Methods. Med Chem 2024; 20:153-231. [PMID: 37957860 DOI: 10.2174/0115734064265609231026063624] [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: 06/25/2023] [Revised: 08/30/2023] [Accepted: 09/25/2023] [Indexed: 11/15/2023]
Abstract
INTRODUCTION Within the scope of the project, this study aimed to find novel inhibitors by combining computational methods. In order to design inhibitors, it was aimed to produce molecules similar to the RdRp inhibitor drug Favipiravir by using the deep learning method. METHODS For this purpose, a Trained Neural Network (TNN) was used to produce 75 molecules similar to Favipiravir by using Simplified Molecular Input Line Entry System (SMILES) representations. The binding properties of molecules to Viral RNA-dependent RNA polymerase (RdRp) were studied by using molecular docking studies. To confirm the accuracy of this method, compounds were also tested against 3CL protease (3CLpro), which is another important enzyme for the progression of SARS-CoV-2. Compounds having better binding energies and RMSD values than favipiravir were searched with similarity analysis on the ChEMBL drug database in order to find similar structures with RdRp and 3CLpro inhibitory activities. RESULTS A similarity search found new 200 potential RdRp and 3CLpro inhibitors structurally similar to produced molecules, and these compounds were again evaluated for their receptor interactions with molecular docking studies. Compounds showed better interaction with RdRp protease than 3CLpro. This result presented that artificial intelligence correctly produced structures similar to favipiravir that act more specifically as RdRp inhibitors. In addition, Lipinski's rules were applied to the molecules that showed the best interaction with RdRp, and 7 compounds were determined to be potential drug candidates. Among these compounds, a Molecular Dynamic simulation study was applied for ChEMBL ID:1193133 to better understand the existence and duration of the compound in the receptor site. CONCLUSION The results confirmed that the ChEMBL ID:1193133 compound showed good Root Mean Square Deviation (RMSD), Root Mean Square Fluctuation (RMSF), hydrogen bonding, and remaining time in the active site; therefore, it was considered that it could be active against the virus. This compound was also tested for antiviral activity, and it was determined that it did not delay viral infection, although it was cytotoxic between 5mg/mL-1.25mg/mL concentrations. However, if other compounds could be tested, it might provide a chance to obtain activity, and compounds should also be tested against the enzymes as well as the other types of viruses.
Collapse
Affiliation(s)
- Ersin Güner
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, 34010 Topkapı, İstanbul, Turkey
| | - Özgür Özkan
- Teknokent Arı, Pinticks Software Company, Istanbul Technical University, Reşitpaşa Mah. Katar Street, No:4/B204 Sarıyer, İstanbul, Turkey
| | - Gözde Yalcin-Ozkat
- Bioengineering Department, Faculty of Engineering and Architecture, Recep Tayyip Erdogan University, 53100 Rize, Turkey
- Max Planck Institute for Dynamics of Complex Technical Systems, Molecular Simulations and Design Group, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Süreyya Ölgen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, 34010 Topkapı, İstanbul, Turkey
| |
Collapse
|
6
|
Haneczok J, Delijewski M, Moldzio R. AI molecular property prediction for Parkinson's Disease reveals potential repurposing drug candidates based on the increase of the expression of PINK1. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 241:107731. [PMID: 37544165 DOI: 10.1016/j.cmpb.2023.107731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 05/20/2023] [Accepted: 07/23/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Parkinson's Disease (PD), a common neurodegenerative disorder and one of the major current challenges in neuroscience and pharmacology, may potentially be tackled by the modern AI techniques employed in drug discovery based on molecular property prediction. The aim of our study was to explore the application of a machine learning setup for the identification of the best potential drug candidates among FDA approved drugs, based on their predicted PINK1 expression-enhancing activity. METHODS Our study relies on supervised machine learning paradigm exploiting in vitro data and utilizing the scaffold splits methodology in order to assess model's capability to extract molecular patterns and generalize from them to new, unseen molecular representations. Models' predictions are combined in a meta-ensemble setup for finding new pharmacotherapies based on the predicted expression of PINK1. RESULTS The proposed machine learning setup can be used for discovering new drugs for PD based on the predicted increase of expression of PINK1. Our study identified nitazoxanide as well as representatives of imidazolidines, trifluoromethylbenzenes, anilides, nitriles, stilbenes and steroid esters as the best potential drug candidates for PD with PINK1 expression-enhancing activity on or inside the cell's mitochondria. CONCLUSIONS The applied methodology allows to reveal new potential drug candidates against PD. Next to novel indications, it allows also to confirm the utility of already known antiparkinson drugs, in the new context of PINK1 expression, and indicates the potential for simultaneous utilization of different mechanisms of action.
Collapse
Affiliation(s)
| | - Marcin Delijewski
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland.
| | - Rudolf Moldzio
- Institute of Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
7
|
Han R, Yoon H, Kim G, Lee H, Lee Y. Revolutionizing Medicinal Chemistry: The Application of Artificial Intelligence (AI) in Early Drug Discovery. Pharmaceuticals (Basel) 2023; 16:1259. [PMID: 37765069 PMCID: PMC10537003 DOI: 10.3390/ph16091259] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Artificial intelligence (AI) has permeated various sectors, including the pharmaceutical industry and research, where it has been utilized to efficiently identify new chemical entities with desirable properties. The application of AI algorithms to drug discovery presents both remarkable opportunities and challenges. This review article focuses on the transformative role of AI in medicinal chemistry. We delve into the applications of machine learning and deep learning techniques in drug screening and design, discussing their potential to expedite the early drug discovery process. In particular, we provide a comprehensive overview of the use of AI algorithms in predicting protein structures, drug-target interactions, and molecular properties such as drug toxicity. While AI has accelerated the drug discovery process, data quality issues and technological constraints remain challenges. Nonetheless, new relationships and methods have been unveiled, demonstrating AI's expanding potential in predicting and understanding drug interactions and properties. For its full potential to be realized, interdisciplinary collaboration is essential. This review underscores AI's growing influence on the future trajectory of medicinal chemistry and stresses the importance of ongoing synergies between computational and domain experts.
Collapse
Affiliation(s)
| | | | | | | | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
8
|
Hou R, Xie C, Gui Y, Li G, Li X. Machine-Learning-Based Data Analysis Method for Cell-Based Selection of DNA-Encoded Libraries. ACS OMEGA 2023; 8:19057-19071. [PMID: 37273617 PMCID: PMC10233830 DOI: 10.1021/acsomega.3c02152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
DNA-encoded library (DEL) is a powerful ligand discovery technology that has been widely adopted in the pharmaceutical industry. DEL selections are typically performed with a purified protein target immobilized on a matrix or in solution phase. Recently, DELs have also been used to interrogate the targets in the complex biological environment, such as membrane proteins on live cells. However, due to the complex landscape of the cell surface, the selection inevitably involves significant nonspecific interactions, and the selection data are much noisier than the ones with purified proteins, making reliable hit identification highly challenging. Researchers have developed several approaches to denoise DEL datasets, but it remains unclear whether they are suitable for cell-based DEL selections. Here, we report the proof-of-principle of a new machine-learning (ML)-based approach to process cell-based DEL selection datasets by using a Maximum A Posteriori (MAP) estimation loss function, a probabilistic framework that can account for and quantify uncertainties of noisy data. We applied the approach to a DEL selection dataset, where a library of 7,721,415 compounds was selected against a purified carbonic anhydrase 2 (CA-2) and a cell line expressing the membrane protein carbonic anhydrase 12 (CA-12). The extended-connectivity fingerprint (ECFP)-based regression model using the MAP loss function was able to identify true binders and also reliable structure-activity relationship (SAR) from the noisy cell-based selection datasets. In addition, the regularized enrichment metric (known as MAP enrichment) could also be calculated directly without involving the specific machine-learning model, effectively suppressing low-confidence outliers and enhancing the signal-to-noise ratio. Future applications of this method will focus on de novo ligand discovery from cell-based DEL selections.
Collapse
Affiliation(s)
- Rui Hou
- Department
of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
- Laboratory
for Synthetic Chemistry and Chemical Biology LimitedHealth@InnoHK, Innovation and Technology Commission, Hong Kong SAR, China
| | - Chao Xie
- Department
of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yuhan Gui
- Department
of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Institute
of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Xiaoyu Li
- Department
of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
- Laboratory
for Synthetic Chemistry and Chemical Biology LimitedHealth@InnoHK, Innovation and Technology Commission, Hong Kong SAR, China
| |
Collapse
|
9
|
Gao Y, Chen S, Tong J, Fu X. Topology-enhanced molecular graph representation for anti-breast cancer drug selection. BMC Bioinformatics 2022; 23:382. [PMID: 36123643 PMCID: PMC9484163 DOI: 10.1186/s12859-022-04913-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/24/2022] [Indexed: 12/24/2022] Open
Abstract
Background Breast cancer is currently one of the cancers with a higher mortality rate in the world. The biological research on anti-breast cancer drugs focuses on the activity of estrogen receptors alpha (ER\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\alpha$$\end{document}α), the pharmacokinetic properties and the safety of the compounds, which, however, is an expensive and time-consuming process. Developments of deep learning bring potential to efficiently facilitate the candidate drug selection against breast cancer. Methods In this paper, we propose an Anti-Breast Cancer Drug selection method utilizing Gated Graph Neural Networks (ABCD-GGNN) to topologically enhance the molecular representation of candidate drugs. By constructing atom-level graphs through atomic descriptors for each distinct compound, ABCD-GGNN can topologically learn both the implicit structure and substructure characteristics of a candidate drug and then integrate the representation with explicit discrete molecular descriptors to generate a molecule-level representation. As a result, the representation of ABCD-GGNN can inductively predict the ER\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\alpha$$\end{document}α, the pharmacokinetic properties and the safety of each candidate drug. Finally, we design a ranking operator whose inputs are the predicted properties so as to statistically select the appropriate drugs against breast cancer. Results Extensive experiments conducted on our collected anti-breast cancer candidate drug dataset demonstrate that our proposed method outperform all the other representative methods in the tasks of predicting ER\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\alpha$$\end{document}α, and the pharmacokinetic properties and safety of the compounds. Extended result analysis demonstrates the efficiency and biological rationality of the operator we design to calculate the candidate drug ranking from the predicted properties. Conclusion In this paper, we propose the ABCD-GGNN representation method to efficiently integrate the topological structure and substructure features of the molecules with the discrete molecular descriptors. With a ranking operator applied, the predicted properties efficiently facilitate the candidate drug selection against breast cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04913-6.
Collapse
Affiliation(s)
- Yue Gao
- School of Computer Science (National Pilot Software Engineering School), Beijing University of Posts and Telecommunications, Beijing, China.,Key Laboratory of Trustworthy Distributed Computing and Service (BUPT), Ministry of Education, Beijing, China
| | - Songling Chen
- School of Computer Science (National Pilot Software Engineering School), Beijing University of Posts and Telecommunications, Beijing, China.,Key Laboratory of Trustworthy Distributed Computing and Service (BUPT), Ministry of Education, Beijing, China
| | - Junyi Tong
- School of Science, Beijing University of Posts and Telecommunications, Beijing, China
| | - Xiangling Fu
- School of Computer Science (National Pilot Software Engineering School), Beijing University of Posts and Telecommunications, Beijing, China. .,Key Laboratory of Trustworthy Distributed Computing and Service (BUPT), Ministry of Education, Beijing, China.
| |
Collapse
|
10
|
Adams J, Agyenkwa-Mawuli K, Agyapong O, Wilson MD, Kwofie SK. EBOLApred: A machine learning-based web application for predicting cell entry inhibitors of the Ebola virus. Comput Biol Chem 2022; 101:107766. [DOI: 10.1016/j.compbiolchem.2022.107766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/10/2022] [Accepted: 08/29/2022] [Indexed: 11/03/2022]
|
11
|
Li X, Pan F, Yang Z, Gao F, Li J, Zhang F, Wang T. Construction of QSAR model based on cysteine‐containing dipeptides and screening of natural tyrosinase inhibitors. J Food Biochem 2022; 46:e14338. [DOI: 10.1111/jfbc.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaofang Li
- Biomedical Nanocenter, School of Life Science Inner Mongolia Agricultural University Hohhot China
- Pharmacy Laboratory Inner Mongolia International Mongolian Hospital Hohhot China
| | - Fei Pan
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Oral Disease, Stomatology Hospital, Department of Biomedical Engineering, School of Basic Medical Sciences Guangzhou Medical University Guangzhou China
- Beijing Engineering and Technology Research Center of Food Additives Beijing Technology and Business University Beijing China
| | - Zichen Yang
- Beijing Engineering and Technology Research Center of Food Additives Beijing Technology and Business University Beijing China
| | - Feng Gao
- Biomedical Nanocenter, School of Life Science Inner Mongolia Agricultural University Hohhot China
| | - Jiawei Li
- Pharmacy Laboratory Inner Mongolia International Mongolian Hospital Hohhot China
| | - Feng Zhang
- Pharmacy Laboratory Inner Mongolia International Mongolian Hospital Hohhot China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Oral Disease, Stomatology Hospital, Department of Biomedical Engineering, School of Basic Medical Sciences Guangzhou Medical University Guangzhou China
| | - Tegexibaiyin Wang
- Pharmacy Laboratory Inner Mongolia International Mongolian Hospital Hohhot China
| |
Collapse
|
12
|
Gao K, Wang R, Chen J, Cheng L, Frishcosy J, Huzumi Y, Qiu Y, Schluckbier T, Wei X, Wei GW. Methodology-Centered Review of Molecular Modeling, Simulation, and Prediction of SARS-CoV-2. Chem Rev 2022; 122:11287-11368. [PMID: 35594413 PMCID: PMC9159519 DOI: 10.1021/acs.chemrev.1c00965] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite tremendous efforts in the past two years, our understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), virus-host interactions, immune response, virulence, transmission, and evolution is still very limited. This limitation calls for further in-depth investigation. Computational studies have become an indispensable component in combating coronavirus disease 2019 (COVID-19) due to their low cost, their efficiency, and the fact that they are free from safety and ethical constraints. Additionally, the mechanism that governs the global evolution and transmission of SARS-CoV-2 cannot be revealed from individual experiments and was discovered by integrating genotyping of massive viral sequences, biophysical modeling of protein-protein interactions, deep mutational data, deep learning, and advanced mathematics. There exists a tsunami of literature on the molecular modeling, simulations, and predictions of SARS-CoV-2 and related developments of drugs, vaccines, antibodies, and diagnostics. To provide readers with a quick update about this literature, we present a comprehensive and systematic methodology-centered review. Aspects such as molecular biophysics, bioinformatics, cheminformatics, machine learning, and mathematics are discussed. This review will be beneficial to researchers who are looking for ways to contribute to SARS-CoV-2 studies and those who are interested in the status of the field.
Collapse
Affiliation(s)
- Kaifu Gao
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rui Wang
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jiahui Chen
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Limei Cheng
- Clinical
Pharmacology and Pharmacometrics, Bristol
Myers Squibb, Princeton, New Jersey 08536, United States
| | - Jaclyn Frishcosy
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuta Huzumi
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuchi Qiu
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tom Schluckbier
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xiaoqi Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
13
|
Xu H, Buckeridge DL, Wang F, Tarczy-Hornoch P. Novel informatics approaches to COVID-19 Research: From methods to applications. J Biomed Inform 2022; 129:104028. [PMID: 35181495 PMCID: PMC8847074 DOI: 10.1016/j.jbi.2022.104028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 10/30/2022]
Affiliation(s)
- Hua Xu
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David L Buckeridge
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Fei Wang
- Department of Population Health Sciences, Cornell University, New York, NY, USA
| | - Peter Tarczy-Hornoch
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Chalasani RD, Radhika Y. Prediction of ITK inhibitor kinases activity based on posterior probabilistic weighted average based ensemble voting classification. JOURNAL OF INTELLIGENT & FUZZY SYSTEMS 2022. [DOI: 10.3233/jifs-221412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
ITK inhibitor is used for the treatment of asthma and activity of inhibitor prediction helps to provide better treatment. Few researches were carried out for the analysis and prediction of kinases activity. Existing methods applied for the inhibitor prediction have limitations of imbalance dataset and lower performance. In this research, the Posterior Probabilistic Weighted Average Based Ensemble voting (PPWAE)ensemble method is proposed with various classifier for effective prediction of kinases activity. The PPWAE model selects the most probable class from the classification method for prediction. The co-train model has two advantages: Features are trained together to increases the learning rate of model and probability is measured for each model to select the efficient classifier. Support Vector Machine (SVM), Random Forest (RF), Decision Tree (DT), Classification and Regression Tree (CART), and Nave Bayes were among the classifiers employed. The results suggest that the Probabilistic Co-train ensemble technique performs well in kinase activity prediction. In the prediction of ITK inhibitor activity, the suggested ensemble method has a 74.27 percent accuracy, while the conventional SVM method has a 60% accuracy.
Collapse
Affiliation(s)
- Rama Devi Chalasani
- Department of CSE, GIT, Gitam Deemed to be University. Visakhapatnam, A.P, India
| | - Y. Radhika
- Department of CSE, GIT, Gitam Deemed to be University. Visakhapatnam, A.P, India
| |
Collapse
|
15
|
Data-Driven Analytics Leveraging Artificial Intelligence in the Era of COVID-19: An Insightful Review of Recent Developments. Symmetry (Basel) 2021. [DOI: 10.3390/sym14010016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
This paper presents the role of artificial intelligence (AI) and other latest technologies that were employed to fight the recent pandemic (i.e., novel coronavirus disease-2019 (COVID-19)). These technologies assisted the early detection/diagnosis, trends analysis, intervention planning, healthcare burden forecasting, comorbidity analysis, and mitigation and control, to name a few. The key-enablers of these technologies was data that was obtained from heterogeneous sources (i.e., social networks (SN), internet of (medical) things (IoT/IoMT), cellular networks, transport usage, epidemiological investigations, and other digital/sensing platforms). To this end, we provide an insightful overview of the role of data-driven analytics leveraging AI in the era of COVID-19. Specifically, we discuss major services that AI can provide in the context of COVID-19 pandemic based on six grounds, (i) AI role in seven different epidemic containment strategies (a.k.a non-pharmaceutical interventions (NPIs)), (ii) AI role in data life cycle phases employed to control pandemic via digital solutions, (iii) AI role in performing analytics on heterogeneous types of data stemming from the COVID-19 pandemic, (iv) AI role in the healthcare sector in the context of COVID-19 pandemic, (v) general-purpose applications of AI in COVID-19 era, and (vi) AI role in drug design and repurposing (e.g., iteratively aligning protein spikes and applying three/four-fold symmetry to yield a low-resolution candidate template) against COVID-19. Further, we discuss the challenges involved in applying AI to the available data and privacy issues that can arise from personal data transitioning into cyberspace. We also provide a concise overview of other latest technologies that were increasingly applied to limit the spread of the ongoing pandemic. Finally, we discuss the avenues of future research in the respective area. This insightful review aims to highlight existing AI-based technological developments and future research dynamics in this area.
Collapse
|