1
|
Rosa D, Elya B, Hanafi M, Khatib A, Budiarto E, Nur S, Surya MI. Investigation of alpha-glucosidase inhibition activity of Artabotrys sumatranus leaf extract using metabolomics, machine learning and molecular docking analysis. PLoS One 2025; 20:e0313592. [PMID: 39752479 PMCID: PMC11698457 DOI: 10.1371/journal.pone.0313592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/27/2024] [Indexed: 01/06/2025] Open
Abstract
One way to treat diabetes mellitus type II is by using α-glucosidase inhibitor, that will slow down the postprandial glucose intake. Metabolomics analysis of Artabotrys sumatranus leaf extract was used in this research to predict the active compounds as α-glucosidase inhibitors from this extract. Both multivariate statistical analysis and machine learning approaches were used to improve the confidence of the predictions. After performance comparisons with other machine learning methods, random forest was chosen to make predictive model for the activity of the extract samples. Feature importance analysis (using random feature permutation and Shapley score calculation) was used to identify the predicted active compound as the important features that influenced the activity prediction of the extract samples. The combined analysis of multivariate statistical analysis and machine learning predicted 9 active compounds, where 6 of them were identified as mangiferin, neomangiferin, norisocorydine, apigenin-7-O-galactopyranoside, lirioferine, and 15,16-dihydrotanshinone I. The activities of norisocorydine, apigenin-7-O-galactopyranoside, and lirioferine as α-glucosidase inhibitors have not yet reported before. Molecular docking simulation, both to 3A4A (α-glucosidase enzyme from Saccharomyces cerevisiae, usually used in bioassay test) and 3TOP (a part of α-glucosidase enzyme in human gut) showed strong to very strong binding of the identified predicted active compounds to both receptors, with exception of neomangiferin which only showed strong binding to 3TOP receptor. Isolation based on bioassay guided fractionation further verified the metabolomics prediction by succeeding to isolate mangiferin from the extract, which showed strong α-glucosidase activity when subjected to bioassay test. The correlation analysis also showed a possibility of 3 groups in the predicted active compounds, which might be related to the biosynthesis pathway (need further research for verification). Another result from correlation analysis was that in general the α-glucosidase inhibition activity in the extract had strong correlation to antioxidant activity, which was also reflected in the predicted active compounds. Only one predicted compound had very low positive correlation to antioxidant activity.
Collapse
Affiliation(s)
- Dela Rosa
- Department of Pharmacy, Faculty of Pharmacy, Indonesia University, Depok, Indonesia
- Department of Pharmacy, Faculty of Health Science, Pelita Harapan University, Tangerang, Indonesia
| | - Berna Elya
- Department of Pharmacy, Faculty of Pharmacy, Indonesia University, Depok, Indonesia
| | - Muhammad Hanafi
- Chemistry Research Centre, National Research and Innovation Agency, Science and Technology Research Centre, Serpong, Indonesia
| | - Alfi Khatib
- Department of Pharmaceutical Chemistry, Kulliyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Eka Budiarto
- Department of Information Technology, Faculty of Engineering and Information Technology, Swiss German University, Tangerang, Indonesia
| | - Syamsu Nur
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Almarisah Madani University, Makasar, Indonesia
| | - Muhammad Imam Surya
- Research Centre for Plant Conservation, Botanic Gardens and Forestry, National Research and Innovation Agency, Bogor, Indonesia
| |
Collapse
|
2
|
Karimi-Sani I, Sharifi M, Abolpour N, Lotfi M, Atapour A, Takhshid MA, Sahebkar A. Drug repositioning for Parkinson's disease: an emphasis on artificial intelligence approaches. Ageing Res Rev 2025:102651. [PMID: 39755176 DOI: 10.1016/j.arr.2024.102651] [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: 10/08/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Parkinson's disease (PD) is one of the most incapacitating neurodegenerative diseases (NDDs). PD is the second most common NDD worldwide which affects approximately 1 to 2 percent of people over 65 years. It is an attractive pursuit for artificial intelligence (AI) to contribute to and evolve PD treatments through drug repositioning by repurposing existing drugs, shelved drugs, or even candidates that do not meet the criteria for clinical trials. A search was conducted in three databases Web of Science, Scopus, and PubMed. We reviewed the data related to the last years (1975-present) to identify those drugs currently being proposed for repositioning in PD. Moreover, we reviewed the present status of the computational approach, including AI/Machine Learning (AI/ML)-powered pharmaceutical discovery efforts and their implementation in PD treatment. It was found that the number of drug repositioning studies for PD has increased recently. Repositioning of drugs in PD is taking off, and scientific communities are increasingly interested in communicating its results and finding effective treatment alternatives for PD. A better chance of success in PD drug discovery has been made possible due to AI/ML algorithm advancements. In addition to the experimentation stage of drug discovery, it is also important to leverage AI in the planning stage of clinical trials to make them more effective. New AI-based models or solutions that increase the success rate of drug development are greatly needed.
Collapse
Affiliation(s)
- Iman Karimi-Sani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrdad Sharifi
- Emergency Medicine Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nahid Abolpour
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrzad Lotfi
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran; Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad-Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran; Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Zhou S, Ye L, Huang Y, Valsecchi C, Liu Y, Shao L, Liu J, He T, Liu L, Fan M. Rapid diagnosis and recurrence prediction of choledocholithiasis disease using raw bile with machine learning assisted SERS. Talanta 2025; 282:126979. [PMID: 39383718 DOI: 10.1016/j.talanta.2024.126979] [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: 05/09/2024] [Revised: 09/06/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Surface-enhanced Raman spectroscopy (SERS) analysis based on body fluids has been widely applied in disease diagnose. Choledocholithiasis is a widespread and often recurrent digestive system disease, with limited data on factors predicting its formation and reappearance. Bile contains many components that could provide valuable diagnostic information; however, the current diagnosis of biliary disease by SERS focuses on detecting specific component in the bile, overlooking the complex interplay and correlations among multiple factors that could be crucial for accurate diagnosis. This work directly obtained multi-component SERS spectral information of raw bile from 46 patients. Characteristic information was extracted from bile SERS spectra using Principal Component Analysis (PCA), revealing variations in the content of characteristic components associated with different choledocholithiasis types and their recurrence frequency. Pearson correlation analysis was also introduced to reveal the interactions of primary active substances pertinent to choledocholithiasis diagnosis. The efficacy of PCA and Support Vector Machine (SVM) models in classifying stone types, presented an accuracy of 99.2 %. Furthermore, the interaction patterns among SERS characteristic components in choledocholithiasis recurrence frequency were revealed, and with the support of SVM, the prediction for different recurrence rates reached an accuracy of 95.2 %. Overall, this work demonstrates that integrating SERS with machine learning can support disease diagnosis and the interpretation of correlations among multiple components, facilitating elucidating the disease mechanisms.
Collapse
Affiliation(s)
- Shana Zhou
- School of Environmental Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Liansong Ye
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yuting Huang
- Department of Clinical Laboratory Medicine, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Chiara Valsecchi
- Federal University of Pampa, Campus Alegrete, 97542-160, Alegrete, RS, Brazil
| | - Yingying Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Limei Shao
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Jiao Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Tian He
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Ling Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China.
| | - Meikun Fan
- School of Environmental Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China.
| |
Collapse
|
4
|
Jamir L, P H. Employing Machine Learning Models to Predict Potential α-Glucosidase Inhibitory Plant Secondary Metabolites Targeting Type-2 Diabetes and Their In Vitro Validation. J Chem Inf Model 2024; 64:9150-9162. [PMID: 39352297 DOI: 10.1021/acs.jcim.4c00955] [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: 12/11/2024]
Abstract
The need for new antidiabetic drugs is evident, considering the ongoing global burden of type-2 diabetes mellitus despite notable progress in drug discovery from laboratory research to clinical application. This study aimed to build machine learning (ML) models to predict potential α-glucosidase inhibitors based on the data set comprising over 537 reported plant secondary metabolite (PSM) α-glucosidase inhibitors. We assessed 35 ML models by using seven different fingerprints. The Random forest with the RDKit fingerprint was the best-performing model, with an accuracy (ACC) of 83.74% and an area under the ROC curve (AUC) of 0.803. The resulting robust ML model encompasses all reported α-glucosidase inhibitory PSMs. The model was employed to predict potential α-glucosidase inhibitors from an in-house 5810 PSM database. The model identified 965 PSMs with a prediction activity ≥0.90 for α-glucosidase inhibition. Twenty-four predicted PSMs were subjected to in vitro assay, and 13 were found to inhibit α-glucosidase with IC50 ranging from 0.63 to 7 mg/mL. Among them, seven compounds recorded IC50 values less than the standard drug acarbose and were investigated further to have optimal drug-likeness and medicinal chemistry characteristics. The ML model and in vitro experiments have identified nervonic acid as a promising α-glucosidase inhibitor. This compound should be further investigated for its potential integration into the diabetes treatment system.
Collapse
Affiliation(s)
- Lemnaro Jamir
- Centre for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Hariprasad P
- Centre for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
5
|
Sutcliffe R, Doherty CPA, Morgan HP, Dunne NJ, McCarthy HO. Strategies for the design of biomimetic cell-penetrating peptides using AI-driven in silico tools for drug delivery. BIOMATERIALS ADVANCES 2024; 169:214153. [PMID: 39705787 DOI: 10.1016/j.bioadv.2024.214153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/08/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Cell-penetrating peptides (CPP) have gained rapid attention over the last 25 years; this is attributed to their versatility, customisation, and 'Trojan horse' delivery that evades the immune system. However, the current CPP rational design process is limited, as it requires several rounds of peptide synthesis, prediction and wet-lab validation, which is expensive, time-consuming and requires extensive knowledge in peptide chemistry. Artificial intelligence (AI) has emerged as a promising alternative which can augment the design process, for example by determining physiochemical characteristics, secondary structure, solvent accessibility, disorder and flexibility, as well as predicting in vivo behaviour such as toxicity and peptidase degradation. Other more recent tools utilise supervised machine learning (ML) to predict the penetrative ability of an amino acid sequence. The use of AI in the CPP design process has the potential to reduce development costs and increase the chances of success with respect to delivery. This review provides a survey of in silico tools and AI platforms which can be utilised in the design process, and the key features that should be taken into consideration when designing next generation CPPs.
Collapse
Affiliation(s)
- Rebecca Sutcliffe
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland
| | - Ciaran P A Doherty
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Hugh P Morgan
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Nicholas J Dunne
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
6
|
Yadav MK, Dahiya V, Tripathi MK, Chaturvedi N, Rashmi M, Ghosh A, Raj VS. Unleashing the future: The revolutionary role of machine learning and artificial intelligence in drug discovery. Eur J Pharmacol 2024; 985:177103. [PMID: 39515559 DOI: 10.1016/j.ejphar.2024.177103] [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: 08/01/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Drug discovery is a complex and multifaceted process aimed at identifying new therapeutic compounds with the potential to treat various diseases. Traditional methods of drug discovery are often time-consuming, expensive, and characterized by low success rates. Because of this, there is an urgent need to improve the drug development process using new technologies. The integration of the current state-of-art of artificial intelligence (AI) and machine learning (ML) approaches with conventional methods will enhance the efficiency and effectiveness of pharmaceutical research. This review highlights the transformative impact of AI and ML in drug discovery, discussing current applications, challenges, and future directions in harnessing these technologies to accelerate the development of innovative therapeutics. We have discussed the latest developments in AI and ML technologies to streamline several stages of drug discovery, from target identification and validation to lead optimization and preclinical studies.
Collapse
Affiliation(s)
- Manoj Kumar Yadav
- Department of Biomedical Engineering, SRM University Delhi-NCR, Sonepat, Haryana, India.
| | - Vandana Dahiya
- Department of Biomedical Engineering, SRM University Delhi-NCR, Sonepat, Haryana, India
| | | | - Navaneet Chaturvedi
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Mayank Rashmi
- ICAR-Indian Agricultural Statistics Research Institute, New Delhi, India
| | - Arabinda Ghosh
- Department of Molecular Biology and Bioinformatics, Tripura University, Suryamaninagar, Tripura, India
| | - V Samuel Raj
- Center for Drug Design Discovery and Development (C4D), SRM University Delhi-NCR, Sonepat, Haryana, India.
| |
Collapse
|
7
|
Kanwar RS, Gambhir K, Aggarwal T, Godiwal A, Bhadra K. From Spores to Suffering: Understanding the Role of Anthrax in Bioterrorism. Mil Med 2024:usae535. [PMID: 39656926 DOI: 10.1093/milmed/usae535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/17/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
INTRODUCTION Anthrax, caused by the bacterium Bacillus anthracis, stands as a formidable threat with both natural and bioterrorism-related implications. Its ability to afflict a wide range of hosts, including humans and animals, coupled with its potential use as a bioweapon, underscores the critical importance of understanding and advancing our capabilities to combat this infectious disease. In this context, exploring futuristic approaches becomes imperative, as they hold the promise of not only addressing current challenges but also ushering in a new era in anthrax management. This review delves into strategies to mitigate the impact of anthrax on global health and security, envisioning a future where our arsenal against anthrax is characterized by precision and adaptability. MATERIALS AND METHODS This article highlights the significant potential of anthrax as a bioweapon, while also highlighting current knowledge and strategies developed against this deadly pathogen. We have performed an extensive research and literature review in concordance with the criteria outlined by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. A search strategy was conducted by using numerous keywords on various academic databases, yielding an initial set of 546 records along with 80 supplementary articles. The search included research papers, review papers, perspectives, clinical guidelines, and scientific blogs. The primary and secondary screening of the articles were conducted by 2 independent reviewers along with a third reviewer mitigating any discrepancies and biases encountered during the process. A set of inclusion and exclusion criteria were formulated, and a PICO framework was adapted based on which multiple records were analyzed and considered for the review. RESULTS In total, 53 articles were selected after completing a comprehensive systematic literature review. This review proposes novel approaches and scientific analysis of the complexities surrounding anthrax in the context of bioterrorism, highlighting the emerging technologies and strategies employed for bioterrorism mitigation. CONCLUSIONS The upcoming advancements in anthrax research will be based on cutting-edge technologies and innovative approaches that demonstrate great potential for prevention, detection, and treatment. These advancements may include the incorporation of synthetic biology techniques such as precise manipulation of biological components, nanoscale diagnostics, and Clustered regularly interspaced short palindromic repeats-based technologies, which could revolutionize our ability to combat anthrax on a molecular level. As these progressive methodologies continue to evolve, the integration of these technologies has the potential to redefine our strategies against anthrax, providing more accurate, personalized, and adaptable approaches to address the challenges posed by this infectious threat.
Collapse
Affiliation(s)
- Ratnesh Singh Kanwar
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Kirtida Gambhir
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Tanishka Aggarwal
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Akash Godiwal
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster 3rd Milestone, Faridabad, Haryana 121001, India
| | - Kuntal Bhadra
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| |
Collapse
|
8
|
Lu X, Xie L, Xu L, Mao R, Xu X, Chang S. Multimodal fused deep learning for drug property prediction: Integrating chemical language and molecular graph. Comput Struct Biotechnol J 2024; 23:1666-1679. [PMID: 38680871 PMCID: PMC11046066 DOI: 10.1016/j.csbj.2024.04.030] [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: 01/24/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 05/01/2024] Open
Abstract
Accurately predicting molecular properties is a challenging but essential task in drug discovery. Recently, many mono-modal deep learning methods have been successfully applied to molecular property prediction. However, mono-modal learning is inherently limited as it relies solely on a single modality of molecular representation, which restricts a comprehensive understanding of drug molecules. To overcome the limitations, we propose a multimodal fused deep learning (MMFDL) model to leverage information from different molecular representations. Specifically, we construct a triple-modal learning model by employing Transformer-Encoder, Bidirectional Gated Recurrent Unit (BiGRU), and graph convolutional network (GCN) to process three modalities of information from chemical language and molecular graph: SMILES-encoded vectors, ECFP fingerprints, and molecular graphs, respectively. We evaluate the proposed triple-modal model using five fusion approaches on six molecule datasets, including Delaney, Llinas2020, Lipophilicity, SAMPL, BACE, and pKa from DataWarrior. The results show that the MMFDL model achieves the highest Pearson coefficients, and stable distribution of Pearson coefficients in the random splitting test, outperforming mono-modal models in accuracy and reliability. Furthermore, we validate the generalization ability of our model in the prediction of binding constants for protein-ligand complex molecules, and assess the resilience capability against noise. Through analysis of feature distributions in chemical space and the assigned contribution of each modal model, we demonstrate that the MMFDL model shows the ability to acquire complementary information by using proper models and suitable fusion approaches. By leveraging diverse sources of bioinformatics information, multimodal deep learning models hold the potential for successful drug discovery.
Collapse
Affiliation(s)
- Xiaohua Lu
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Liangxu Xie
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Rongzhi Mao
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Xiaojun Xu
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Shan Chang
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou 213001, China
| |
Collapse
|
9
|
Gu C, Jang WD, Oh KS, Ryu JY. AnoChem: Prediction of chemical structural abnormalities based on machine learning models. Comput Struct Biotechnol J 2024; 23:2116-2121. [PMID: 38808129 PMCID: PMC11130677 DOI: 10.1016/j.csbj.2024.05.017] [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: 02/22/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024] Open
Abstract
De novo drug design aims to rationally discover novel and potent compounds while reducing experimental costs during the drug development stage. Despite the numerous generative models that have been developed, few successful cases of drug design utilizing generative models have been reported. One of the most common challenges is designing compounds that are not synthesizable or realistic. Therefore, methods capable of accurately assessing the chemical structures proposed by generative models for drug design are needed. In this study, we present AnoChem, a computational framework based on deep learning designed to assess the likelihood of a generated molecule being real. AnoChem achieves an area under the receiver operating characteristic curve score of 0.900 for distinguishing between real and generated molecules. We utilized AnoChem to evaluate and compare the performances of several generative models, using other metrics, namely SAscore and Fréschet ChemNet distance (FCD). AnoChem demonstrates a strong correlation with these metrics, validating its effectiveness as a reliable tool for assessing generative models. The source code for AnoChem is available at https://github.com/CSB-L/AnoChem.
Collapse
Affiliation(s)
- Changdai Gu
- Artificial Intelligence Laboratory, Oncocross Co., Ltd., Saechang-ro, Mapo-gu, Seoul 04168, Republic of Korea
- Department of Artificial Intelligence, College of Computing, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Woo Dae Jang
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
- Department of Medicinal and Pharmaceutical Chemistry, University of Science and Technology, Daejeon 34129, Republic of Korea
| | - Kwang-Seok Oh
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
- Department of Medicinal and Pharmaceutical Chemistry, University of Science and Technology, Daejeon 34129, Republic of Korea
| | - Jae Yong Ryu
- Artificial Intelligence Laboratory, Oncocross Co., Ltd., Saechang-ro, Mapo-gu, Seoul 04168, Republic of Korea
- Department of Biotechnology, Duksung Women’s University, 33 Samyang-Ro 144-Gil, Dobong-gu, Seoul 01369, Republic of Korea
| |
Collapse
|
10
|
Lanini J, Huynh MTD, Scebba G, Schneider N, Rodríguez-Pérez R. UNIQUE: A Framework for Uncertainty Quantification Benchmarking. J Chem Inf Model 2024; 64:8379-8386. [PMID: 39542432 PMCID: PMC11600502 DOI: 10.1021/acs.jcim.4c01578] [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: 09/01/2024] [Revised: 10/17/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
Machine learning (ML) models have become key in decision-making for many disciplines, including drug discovery and medicinal chemistry. ML models are generally evaluated prior to their usage in high-stakes decisions, such as compound synthesis or experimental testing. However, no ML model is robust or predictive in all real-world scenarios. Therefore, uncertainty quantification (UQ) in ML predictions has gained importance in recent years. Many investigations have focused on developing methodologies that provide accurate uncertainty estimates for ML-based predictions. Unfortunately, there is no UQ strategy that consistently provides robust estimates about model's applicability on new samples. Depending on the dataset, prediction task, and algorithm, accurate uncertainty estimations might be unfeasible to obtain. Moreover, the optimum UQ metric also varies across applications, and previous investigations have shown a lack of consistency across benchmarks. Herein, the UNIQUE (UNcertaInty QUantification bEnchmarking) framework is introduced to facilitate a comparison of UQ strategies in ML-based predictions. This Python library unifies the benchmarking of multiple UQ metrics, including the calculation of nonstandard UQ metrics (combining information from the dataset and model), and provides a comprehensive evaluation. In this framework, UQ metrics are evaluated for different application scenarios, e.g., eliminating the predictions with the lowest confidence or obtaining a reliable uncertainty estimate for an acquisition function. Taken together, this library will help to standardize UQ investigations and evaluate new methodologies.
Collapse
Affiliation(s)
- Jessica Lanini
- Novartis Biomedical Research, Novartis Campus, 4002 Basel, Switzerland
| | | | - Gaetano Scebba
- Novartis Biomedical Research, Novartis Campus, 4002 Basel, Switzerland
| | - Nadine Schneider
- Novartis Biomedical Research, Novartis Campus, 4002 Basel, Switzerland
| | | |
Collapse
|
11
|
Haga SB. Artificial intelligence, medications, pharmacogenomics, and ethics. Pharmacogenomics 2024; 25:611-622. [PMID: 39545629 DOI: 10.1080/14622416.2024.2428587] [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: 08/15/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
Artificial Intelligence (AI) and Machine Learning (ML) are revolutionizing various scientific and clinical disciplines including pharmacogenomics (PGx) by enabling the analysis of complex datasets and the development of predictive models. The integration of AI and ML with PGx has the potential to provide more precise, data-driven insights into new drug targets, drug efficacy, drug selection, and risk of adverse events. While significant effort to develop and validate these tools remain, ongoing advancements in AI technologies, coupled with improvements in data quality and depth is anticipated to drive the transition of these tools into clinical practice and delivery of individualized treatments and improved patient outcomes. The successful development and integration of AI-assisted PGx tools will require careful consideration of ethical, legal, and social issues (ELSI) in research and clinical practice. This paper explores the intersection of PGx with AI, highlighting current research and potential clinical applications, and ELSI including privacy, oversight, patient and provider knowledge and acceptance, and the impact on patient-provider relationship and new roles.
Collapse
Affiliation(s)
- Susanne B Haga
- Department of Medicine, Division of General Internal Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
12
|
Hudon A, Beaudoin M, Phraxayavong K, Potvin S, Dumais A. Exploring the Intersection of Schizophrenia, Machine Learning, and Genomics: Scoping Review. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2024; 5:e62752. [PMID: 39546776 DOI: 10.2196/62752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/06/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND An increasing body of literature highlights the integration of machine learning with genomic data in psychiatry, particularly for complex mental health disorders such as schizophrenia. These advanced techniques offer promising potential for uncovering various facets of these disorders. A comprehensive review of the current applications of machine learning in conjunction with genomic data within this context can significantly enhance our understanding of the current state of research and its future directions. OBJECTIVE This study aims to conduct a systematic scoping review of the use of machine learning algorithms with genomic data in the field of schizophrenia. METHODS To conduct a systematic scoping review, a search was performed in the electronic databases MEDLINE, Web of Science, PsycNet (PsycINFO), and Google Scholar from 2013 to 2024. Studies at the intersection of schizophrenia, genomic data, and machine learning were evaluated. RESULTS The literature search identified 2437 eligible articles after removing duplicates. Following abstract screening, 143 full-text articles were assessed, and 121 were subsequently excluded. Therefore, 21 studies were thoroughly assessed. Various machine learning algorithms were used in the identified studies, with support vector machines being the most common. The studies notably used genomic data to predict schizophrenia, identify schizophrenia features, discover drugs, classify schizophrenia amongst other mental health disorders, and predict the quality of life of patients. CONCLUSIONS Several high-quality studies were identified. Yet, the application of machine learning with genomic data in the context of schizophrenia remains limited. Future research is essential to further evaluate the portability of these models and to explore their potential clinical applications.
Collapse
Affiliation(s)
- Alexandre Hudon
- Department of psychiatry and addictology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Montréal, QC, Canada
- Institut universitaire en santé mentale de Montréal, Montréal, QC, Canada
| | - Mélissa Beaudoin
- Department of psychiatry and addictology, Université de Montréal, Montréal, QC, Canada
- Faculty of Medicine, McGill University, Montréal, QC, Canada
| | | | - Stéphane Potvin
- Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Montréal, QC, Canada
- Department of psychiatry and addictology, Université de Montréal, Montréal, QC, Canada
| | - Alexandre Dumais
- Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Montréal, QC, Canada
- Department of psychiatry and addictology, Université de Montréal, Montréal, QC, Canada
- Services et Recherches Psychiatriques AD, Montréal, QC, Canada
- Institut nationale de psychiatrie légale Philippe-Pinel, Montréal, QC, Canada
| |
Collapse
|
13
|
Patne AY, Dhulipala SM, Lawless W, Prakash S, Mohapatra SS, Mohapatra S. Drug Discovery in the Age of Artificial Intelligence: Transformative Target-Based Approaches. Int J Mol Sci 2024; 25:12233. [PMID: 39596300 PMCID: PMC11594879 DOI: 10.3390/ijms252212233] [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: 09/20/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
The complexities inherent in drug development are multi-faceted and often hamper accuracy, speed and efficiency, thereby limiting success. This review explores how recent developments in machine learning (ML) are significantly impacting target-based drug discovery, particularly in small-molecule approaches. The Simplified Molecular Input Line Entry System (SMILES), which translates a chemical compound's three-dimensional structure into a string of symbols, is now widely used in drug design, mining, and repurposing. Utilizing ML and natural language processing techniques, SMILES has revolutionized lead identification, high-throughput screening and virtual screening. ML models enhance the accuracy of predicting binding affinity and selectivity, reducing the need for extensive experimental screening. Additionally, deep learning, with its strengths in analyzing spatial and sequential data through convolutional neural networks (CNNs) and recurrent neural networks (RNNs), shows promise for virtual screening, target identification, and de novo drug design. Fragment-based approaches also benefit from ML algorithms and techniques like generative adversarial networks (GANs), which predict fragment properties and binding affinities, aiding in hit selection and design optimization. Structure-based drug design, which relies on high-resolution protein structures, leverages ML models for accurate predictions of binding interactions. While challenges such as interpretability and data quality remain, ML's transformative impact accelerates target-based drug discovery, increasing efficiency and innovation. Its potential to deliver new and improved treatments for various diseases is significant.
Collapse
Affiliation(s)
- Akshata Yashwant Patne
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- Taneja College of Pharmacy Graduate Programs, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
| | - Sai Madhav Dhulipala
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (S.M.D.); (W.L.)
| | - William Lawless
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (S.M.D.); (W.L.)
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada;
| | - Shyam S. Mohapatra
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- Taneja College of Pharmacy Graduate Programs, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- Taneja College of Pharmacy Graduate Programs, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (S.M.D.); (W.L.)
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA
| |
Collapse
|
14
|
Singh N, Singh AK. Screening of phytoconstituents from Bacopa monnieri (L.) Pennell and Mucuna pruriens (L.) DC. to identify potential inhibitors against Cerebroside sulfotransferase. PLoS One 2024; 19:e0307374. [PMID: 39446901 PMCID: PMC11500956 DOI: 10.1371/journal.pone.0307374] [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: 05/17/2024] [Accepted: 07/01/2024] [Indexed: 10/26/2024] Open
Abstract
Cerebroside sulfotransferase (CST) is considered a target protein in developing substrate reduction therapy for metachromatic leukodystrophy. This study employed a multistep virtual screening approach for getting a specific and potent inhibitor against CST from 35 phytoconstituents of Bacopa monnieri (L.) Pennell and 31 phytoconstituents of Mucuna pruriens (L.) DC. from the IMPPAT 2.0 database. Using a binding score cutoff of -8.0 kcal/mol with ADME and toxicity screening, four phytoconstituents IMPHY009537 (Stigmastenol), IMPHY004141 (alpha-Amyrenyl acetate), IMPHY014836 (beta-Sitosterol), and IMPHY001534 (jujubogenin) were considered for in-depth analysis. In the binding pocket of CST, the major amino acid residues that decide the orientation and interaction of compounds are Lys85, His84, His141, Phe170, Tyr176, and Phe177. The molecular dynamics simulation with a 100ns time span further validated the stability and rigidity of the docked complexes of the four hits by exploring the structural deviation and compactness, hydrogen bond interaction, solvent accessible surface area, principal component analysis, and free energy landscape analysis. Stigmastenol from Bacopa monnieri with no potential cross targets was found to be the most potent and selective CST inhibitor followed by alpha-Amyrenyl acetate from Mucuna pruriens as the second-best performing inhibitor against CST. Our computational drug screening approach may contribute to the development of oral drugs against metachromatic leukodystrophy.
Collapse
Affiliation(s)
- Nivedita Singh
- Department of Dravyaguna, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Anil Kumar Singh
- Department of Dravyaguna, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
15
|
Singh N, Singh AK. Phytoconstituents of Withania somnifera (L.) Dunal (Ashwagandha) unveiled potential cerebroside sulfotransferase inhibitors: insight through virtual screening, molecular dynamics, toxicity, and reverse pharmacophore analysis. J Biol Eng 2024; 18:59. [PMID: 39444022 PMCID: PMC11515467 DOI: 10.1186/s13036-024-00456-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
Cerebroside sulfotransferase (CST) is considered as therapeutic target for substrate reduction therapy (SRT) for metachromatic leukodystrophy (MLD). The present study evaluates the therapeutic potential of 57 phytoconstituents of Withania somnifera against CST. Using binding score cutoff ≤-7.0 kcal/mol, top 10 compounds were screened and after ADME and toxicity-based screening, Withasomidienone, 2,4-methylene-cholesterol, and 2,3-Didehydrosomnifericin were identified as safe and potent drug candidates for CST inhibition. Key substrate binding site residues involved in interaction were LYS82, LYS85, SER89, TYR176, PHE170, PHE177. Four steroidal Lactone-based withanolide backbone of these compounds played a critical role in stabilizing their position in the active site pocket. 100 ns molecular dynamics simulation and subsequent trajectory analysis through structural deviation and compactness, principal components, free energy landscape and correlation matrix confirmed the stability of CST-2,3-Didehydrosomnifericin complex throughout the simulation and therefore is considered as the most potent drug candidate for CST inhibition and Withasomidienone as the second most potent drug candidate. The reverse pharmacophore analysis further confirmed the specificity of these two compounds towards CST as no major cross targets were identified. Thus, identified compounds in this study strongly present their candidature for oral drug and provide route for further development of more specific CST inhibitors.
Collapse
Affiliation(s)
- Nivedita Singh
- Department of Dravyaguna, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Anil Kumar Singh
- Department of Dravyaguna, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| |
Collapse
|
16
|
Vicidomini C, Fontanella F, D’Alessandro T, Roviello GN. A Survey on Computational Methods in Drug Discovery for Neurodegenerative Diseases. Biomolecules 2024; 14:1330. [PMID: 39456263 PMCID: PMC11506269 DOI: 10.3390/biom14101330] [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: 09/13/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Currently, the age structure of the world population is changing due to declining birth rates and increasing life expectancy. As a result, physicians worldwide have to treat an increasing number of age-related diseases, of which neurological disorders represent a significant part. In this context, there is an urgent need to discover new therapeutic approaches to counteract the effects of neurodegeneration on human health, and computational science can be of pivotal importance for more effective neurodrug discovery. The knowledge of the molecular structure of the receptors and other biomolecules involved in neurological pathogenesis facilitates the design of new molecules as potential drugs to be used in the fight against diseases of high social relevance such as dementia, Alzheimer's disease (AD) and Parkinson's disease (PD), to cite only a few. However, the absence of comprehensive guidelines regarding the strengths and weaknesses of alternative approaches creates a fragmented and disconnected field, resulting in missed opportunities to enhance performance and achieve successful applications. This review aims to summarize some of the most innovative strategies based on computational methods used for neurodrug development. In particular, recent applications and the state-of-the-art of molecular docking and artificial intelligence for ligand- and target-based approaches in novel drug design were reviewed, highlighting the crucial role of in silico methods in the context of neurodrug discovery for neurodegenerative diseases.
Collapse
Affiliation(s)
- Caterina Vicidomini
- Institute of Biostructures and Bioimaging-Italian National Council for Research (IBB-CNR), Via De Amicis 95, 80145 Naples, Italy
| | - Francesco Fontanella
- Department of Electrical and Information Engineering “Maurizio Scarano”, University of Cassino and Southern Lazio, 03043 Cassino, Italy
| | - Tiziana D’Alessandro
- Department of Electrical and Information Engineering “Maurizio Scarano”, University of Cassino and Southern Lazio, 03043 Cassino, Italy
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging-Italian National Council for Research (IBB-CNR), Via De Amicis 95, 80145 Naples, Italy
| |
Collapse
|
17
|
Luo X, Ding Y, Cao Y, Liu Z, Zhang W, Zeng S, Cheng SH, Li H, Haggarty SJ, Wang X, Zhang J, Shi P. Few-shot meta-learning applied to whole brain activity maps improves systems neuropharmacology and drug discovery. iScience 2024; 27:110875. [PMID: 39319265 PMCID: PMC11419810 DOI: 10.1016/j.isci.2024.110875] [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: 03/11/2024] [Revised: 06/10/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
In this study, we present an approach to neuropharmacological research by integrating few-shot meta-learning algorithms with brain activity mapping (BAMing) to enhance the discovery of central nervous system (CNS) therapeutics. By utilizing patterns from previously validated CNS drugs, our approach facilitates the rapid identification and prediction of potential drug candidates from limited datasets, thereby accelerating the drug discovery process. The application of few-shot meta-learning algorithms allows us to adeptly navigate the challenges of limited sample sizes prevalent in neuropharmacology. The study reveals that our meta-learning-based convolutional neural network (Meta-CNN) models demonstrate enhanced stability and improved prediction accuracy over traditional machine-learning methods. Moreover, our BAM library proves instrumental in classifying CNS drugs and aiding in pharmaceutical repurposing and repositioning. Overall, this research not only demonstrates the effectiveness in overcoming data limitations but also highlights the significant potential of combining BAM with advanced meta-learning techniques in CNS drug discovery.
Collapse
Affiliation(s)
- Xuan Luo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
- National Center for Applied Mathematics Shenzhen, Shenzhen 518000, China
- Department of Mathematics, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yanyun Ding
- National Center for Applied Mathematics Shenzhen, Shenzhen 518000, China
- Institute of Applied Mathematics, Shenzhen Polytechnic University, Shenzhen 518055, China
| | - Yi Cao
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
| | - Zhen Liu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
| | - Wenchong Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
| | - Shangzhi Zeng
- National Center for Applied Mathematics Shenzhen, Shenzhen 518000, China
| | - Shuk Han Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
| | - Honglin Li
- Innovation Center for AI and Drug Discovery, East China Normal University, Shanghai 200062, China
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston, MA 02114, USA
| | - Xin Wang
- Department of Surgery, Chinese University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
| | - Jin Zhang
- National Center for Applied Mathematics Shenzhen, Shenzhen 518000, China
- Department of Mathematics, Southern University of Science and Technology, Shenzhen 518055, China
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
- National Center for Applied Mathematics Shenzhen, Shenzhen 518000, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
18
|
Serrano DR, Luciano FC, Anaya BJ, Ongoren B, Kara A, Molina G, Ramirez BI, Sánchez-Guirales SA, Simon JA, Tomietto G, Rapti C, Ruiz HK, Rawat S, Kumar D, Lalatsa A. Artificial Intelligence (AI) Applications in Drug Discovery and Drug Delivery: Revolutionizing Personalized Medicine. Pharmaceutics 2024; 16:1328. [PMID: 39458657 PMCID: PMC11510778 DOI: 10.3390/pharmaceutics16101328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Artificial intelligence (AI) encompasses a broad spectrum of techniques that have been utilized by pharmaceutical companies for decades, including machine learning, deep learning, and other advanced computational methods. These innovations have unlocked unprecedented opportunities for the acceleration of drug discovery and delivery, the optimization of treatment regimens, and the improvement of patient outcomes. AI is swiftly transforming the pharmaceutical industry, revolutionizing everything from drug development and discovery to personalized medicine, including target identification and validation, selection of excipients, prediction of the synthetic route, supply chain optimization, monitoring during continuous manufacturing processes, or predictive maintenance, among others. While the integration of AI promises to enhance efficiency, reduce costs, and improve both medicines and patient health, it also raises important questions from a regulatory point of view. In this review article, we will present a comprehensive overview of AI's applications in the pharmaceutical industry, covering areas such as drug discovery, target optimization, personalized medicine, drug safety, and more. By analyzing current research trends and case studies, we aim to shed light on AI's transformative impact on the pharmaceutical industry and its broader implications for healthcare.
Collapse
Affiliation(s)
- Dolores R. Serrano
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
- Instituto Universitario de Farmacia Industrial, 28040 Madrid, Spain
| | - Francis C. Luciano
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Brayan J. Anaya
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Baris Ongoren
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Aytug Kara
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Gracia Molina
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Bianca I. Ramirez
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Sergio A. Sánchez-Guirales
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Jesus A. Simon
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Greta Tomietto
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Chrysi Rapti
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Helga K. Ruiz
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Satyavati Rawat
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (S.R.); (D.K.)
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (S.R.); (D.K.)
| | - Aikaterini Lalatsa
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
- CRUK Formulation Unit, School of Pharmacy and Biomedical Sciences, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
19
|
Ivanov J, Tenchov R, Ralhan K, Iyer KA, Agarwal S, Zhou QA. In Silico Insights: QSAR Modeling of TBK1 Kinase Inhibitors for Enhanced Drug Discovery. J Chem Inf Model 2024; 64:7488-7502. [PMID: 39289178 PMCID: PMC11480986 DOI: 10.1021/acs.jcim.4c00864] [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: 06/11/2024] [Revised: 08/17/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
TBK1, or TANK-binding kinase 1, is an enzyme that functions as a serine/threonine protein kinase. It plays a crucial role in various cellular processes, including the innate immune response to viruses, cell proliferation, apoptosis, autophagy, and antitumor immunity. Dysregulation of TBK1 activity can lead to autoimmune diseases, neurodegenerative disorders, and cancer. Due to its central role in these critical pathways, TBK1 is a significant focus of research for therapeutic drug development. In this paper, we explore data from the CAS Content Collection regarding TBK1 and its implication in a large assortment of diseases and disorders. With the demand for developing efficient TBK1 inhibitors being outlined, we focus on utilizing a machine learning approach for developing predictive models for TBK1 inhibition, derived from the fragment-functional analysis descriptors. Using the extensive CAS Content Collection, we assembled a training set of TBK1 inhibitors with experimentally measured IC50 values. We explored several machine learning techniques combined with various molecular descriptors to derive and select the best TBK1 inhibitor QSAR models. Certain significant structural alerts that potentially contribute to inhibition of TBK1 are outlined and discussed. The merit of the article stems from identifying the most adequate TBK1 QSAR models and subsequent successful development of advanced positive training data to facilitate and enhance drug discovery for an important therapeutic target such as TBK1 inhibitors, based on an extensive, wide-ranging set of scientific information provided by the CAS Content Collection.
Collapse
Affiliation(s)
- Julian
M. Ivanov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | | | | |
Collapse
|
20
|
Cheng AH, Ser CT, Skreta M, Guzmán-Cordero A, Thiede L, Burger A, Aldossary A, Leong SX, Pablo-García S, Strieth-Kalthoff F, Aspuru-Guzik A. Spiers Memorial Lecture: How to do impactful research in artificial intelligence for chemistry and materials science. Faraday Discuss 2024. [PMID: 39400305 DOI: 10.1039/d4fd00153b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Machine learning has been pervasively touching many fields of science. Chemistry and materials science are no exception. While machine learning has been making a great impact, it is still not reaching its full potential or maturity. In this perspective, we first outline current applications across a diversity of problems in chemistry. Then, we discuss how machine learning researchers view and approach problems in the field. Finally, we provide our considerations for maximizing impact when researching machine learning for chemistry.
Collapse
Affiliation(s)
- Austin H Cheng
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Cher Tian Ser
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Marta Skreta
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Andrés Guzmán-Cordero
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
- Tinbergen Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Luca Thiede
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Andreas Burger
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | | | - Shi Xuan Leong
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 63737, Singapore
| | | | | | - Alán Aspuru-Guzik
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
- Acceleration Consortium, Toronto, Ontario M5G 1X6, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada
- Department of Materials Science and Engineering, University of Toronto, Canada
- Lebovic Fellow, Canadian Institute for Advanced Research (CIFAR), Canada
| |
Collapse
|
21
|
Ghantasala GSP, Dilip K, Vidyullatha P, Allabun S, Alqahtani MS, Othman M, Abbas M, Soufiene BO. Enhanced ovarian cancer survival prediction using temporal analysis and graph neural networks. BMC Med Inform Decis Mak 2024; 24:299. [PMID: 39390514 PMCID: PMC11468212 DOI: 10.1186/s12911-024-02665-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Ovarian cancer is a formidable health challenge that demands accurate and timely survival predictions to guide clinical interventions. Existing methods, while commendable, suffer from limitations in harnessing the temporal evolution of patient data and capturing intricate interdependencies among different data elements. In this paper, we present a novel methodology which combines Temporal Analysis and Graph Neural Networks (GNNs) to significantly enhance ovarian cancer survival rate predictions. The shortcomings of current processes originate from their disability to correctly seize the complex interactions amongst diverse scientific information units in addition to the dynamic modifications that arise in a affected person`s nation over time. By combining temporal information evaluation and GNNs, our cautioned approach overcomes those drawbacks and, whilst as compared to preceding methods, yields a noteworthy 8.3% benefit in precision, 4.9% more accuracy, 5.5% more advantageous recall, and a considerable 2.9% reduction in prediction latency. Our method's Temporal Analysis factor uses longitudinal affected person information to perceive good-sized styles and tendencies that offer precious insights into the direction of ovarian cancer. Through the combination of GNNs, we offer a robust framework able to shoot complicated interactions among exclusive capabilities of scientific data, permitting the version to realize diffused dependencies that would affect survival results. Our paintings have tremendous implications for scientific practice. Prompt and correct estimation of the survival price of ovarian most cancers allows scientific experts to customize remedy regimens, manipulate assets efficiently, and provide individualized care to patients. Additionally, the interpretability of our version`s predictions promotes a collaborative method for affected person care via way of means of strengthening agreement among scientific employees and the AI-driven selection help system. The proposed approach not only outperforms existing methods but also has the possible to develop ovarian cancer treatment by providing clinicians through a reliable tool for informed decision-making. Through a fusion of Temporal Analysis and Graph Neural Networks, we conduit the gap among data-driven insights and clinical practice, proposing a capable opportunity for refining patient outcomes in ovarian cancer management operations.
Collapse
Affiliation(s)
| | - Kumar Dilip
- Department of Computer Science and Engineering, Alliance University, Bengaluru, India
| | - Pellakuri Vidyullatha
- Department of Computer Science and Engineering, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Guntur, Andhra Pradesh, India
| | - Sarah Allabun
- Department of Medical Education, College of Medicine, Princess Nourah bint Abdulrahman University, P.O.Box 84428, Riyadh, 11671, Saudi Arabia
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia
- BioImaging Unit, Space Research Centre, University of Leicester, Michael Atiyah Building, Leicester, LE1 7RH, UK
| | - Manal Othman
- Department of Medical Education, College of Medicine, Princess Nourah bint Abdulrahman University, P.O.Box 84428, Riyadh, 11671, Saudi Arabia
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Ben Othman Soufiene
- PRINCE Laboratory Research, ISITcom, Hammam Sousse, University of Sousse, Sousse, Tunisia.
| |
Collapse
|
22
|
da Silva Duarte V, Treu L, Campanaro S, Fioravante Guerra A, Giacomini A, Mas A, Corich V, Lemos Junior WJF. Investigating biological mechanisms of colour changes in sustainable food systems: The role of Starmerella bacillaris in white wine colouration using a combination of genomic and biostatistics strategies. Food Res Int 2024; 193:114862. [PMID: 39160049 DOI: 10.1016/j.foodres.2024.114862] [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: 05/27/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
This study explores the biological mechanisms behind colour changes in white wine fermentation using different strains of Starmerella bacillaris. We combined food engineering, genomics, machine learning, and physicochemical analyses to examine interactions between S. bacillaris and Saccharomyces cerevisiae. Significant differences in total polyphenol content were observed, with S. bacillaris fermentation yielding 6 % higher polyphenol content compared to S. cerevisiae EC1118. Genomic analysis identified 12 genes in S. bacillaris with high variant counts that could impact phenotypic properties related to wine color. Notably, SNP analysis revealed numerous missense and synonymous variants, as well as stop-gained and start-lost variants between PAS13 and FRI751, suggesting changes in metabolic pathways affecting pigment production. Besides that, high upstream gene variants in SSK1 and HIP1R indicated potential regulatory changes influencing gene expression. Fermentation trials revealed FRI751 consistently showed high antioxidant activity and polyphenol content (Total Polyphenol: 299.33 ± 3.51 mg GAE/L, DPPH: 1.09 ± 0.01 mmol TE/L, FRAP: 0.95 ± 0.02 mmol TE/L). PAS13 exhibited a balanced profile, while EC1118 had lower values, indicating moderate antioxidant activity. The Weibull model effectively captured nitrogen consumption dynamics, with EC1118 serving as a reliable benchmark. The scale parameter delta for EC1118 was 23.04 ± 2.63, indicating moderate variability in event times. These findings highlight S. bacillaris as a valuable component in sustainable winemaking, offering an alternative to chemical additives for maintaining wine quality and enhancing colours profiles. This study provides insights into the biotechnological and fermented food systems applications of yeast strains in improving food sustainability and supply chain, opening new avenues in food engineering and microbiology.
Collapse
Affiliation(s)
- Vinicius da Silva Duarte
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Laura Treu
- Department Department of Biology, University of Padova, Padova, Italy
| | - Stefano Campanaro
- Department Department of Biology, University of Padova, Padova, Italy.
| | - André Fioravante Guerra
- Centro Federal de Educação Tecnológica Celso Suckow da Fonseca (CEFET/RJ), Valença, Rio de Janeiro, Brazil
| | - Alessio Giacomini
- Department of Agronomy Food Natural Resources Animals and Environment (DAFNAE), University of Padova, Legnaro, Italy; Interdepartmental Centre for Research in Viticulture and Enology (CIRVE), University of Padova, Conegliano, Italy
| | - Albert Mas
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Grup de Biotecnologia Enològica, Facultat d'Enologia, Tarragona, Catalonia, Spain
| | - Viviana Corich
- Department of Agronomy Food Natural Resources Animals and Environment (DAFNAE), University of Padova, Legnaro, Italy; Interdepartmental Centre for Research in Viticulture and Enology (CIRVE), University of Padova, Conegliano, Italy; Department of Land, Environment, Agriculture and Forestry - TeSAF Legnaro, Padova, Italy.
| | | |
Collapse
|
23
|
Kokudeva M, Vichev M, Naseva E, Miteva DG, Velikova T. Artificial intelligence as a tool in drug discovery and development. World J Exp Med 2024; 14:96042. [PMID: 39312699 PMCID: PMC11372739 DOI: 10.5493/wjem.v14.i3.96042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
The rapidly advancing field of artificial intelligence (AI) has garnered substantial attention for its potential application in drug discovery and development. This opinion review critically examined the feasibility and prospects of integrating AI as a transformative tool in the pharmaceutical industry. AI, encompassing machine learning algorithms, deep learning, and data analytics, offers unprecedented opportunities to streamline and enhance various stages of drug development. This opinion review delved into the current landscape of AI-driven approaches, discussing their utilization in target identification, lead optimization, and predictive modeling of pharmacokinetics and toxicity. We aimed to scrutinize the integration of large-scale omics data, electronic health records, and chemical informatics, highlighting the power of AI in uncovering novel therapeutic targets and accelerating drug repurposing strategies. Despite the considerable potential of AI, the review also addressed inherent challenges, including data privacy concerns, interpretability of AI models, and the need for robust validation in real-world clinical settings. Additionally, we explored ethical considerations surrounding AI-driven decision-making in drug development. This opinion review provided a nuanced perspective on the transformative role of AI in drug discovery by discussing the existing literature and emerging trends, presenting critical insights and addressing potential hurdles. In conclusion, this study aimed to stimulate discourse within the scientific community and guide future endeavors to harness the full potential of AI in drug development.
Collapse
Affiliation(s)
- Maria Kokudeva
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia 1000, Bulgaria
| | | | - Emilia Naseva
- Faculty of Public Health, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Dimitrina Georgieva Miteva
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia 1164, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
24
|
Chakraborty C, Bhattacharya M, Lee SS, Wen ZH, Lo YH. The changing scenario of drug discovery using AI to deep learning: Recent advancement, success stories, collaborations, and challenges. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102295. [PMID: 39257717 PMCID: PMC11386122 DOI: 10.1016/j.omtn.2024.102295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Due to the transformation of artificial intelligence (AI) tools and technologies, AI-driven drug discovery has come to the forefront. It reduces the time and expenditure. Due to these advantages, pharmaceutical industries are concentrating on AI-driven drug discovery. Several drug molecules have been discovered using AI-based techniques and tools, and several newly AI-discovered drug molecules have already entered clinical trials. In this review, we first present the data and their resources in the pharmaceutical sector for AI-driven drug discovery and illustrated some significant algorithms or techniques used for AI and ML which are used in this field. We gave an overview of the deep neural network (NN) models and compared them with artificial NNs. Then, we illustrate the recent advancement of the landscape of drug discovery using AI to deep learning, such as the identification of drug targets, prediction of their structure, estimation of drug-target interaction, estimation of drug-target binding affinity, design of de novo drug, prediction of drug toxicity, estimation of absorption, distribution, metabolism, excretion, toxicity; and estimation of drug-drug interaction. Moreover, we highlighted the success stories of AI-driven drug discovery and discussed several collaboration and the challenges in this area. The discussions in the article will enrich the pharmaceutical industry.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, Gangwon-Do 24252, Republic of Korea
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 813204, Taiwan
- Shu-Zen Junior College of Medicine and Management, Kaohsiung 821004, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| |
Collapse
|
25
|
Beccaria R, Lazzeri A, Tiana G. Predicting the Binding of Small Molecules to Proteins through Invariant Representation of the Molecular Structure. J Chem Inf Model 2024; 64:6758-6767. [PMID: 39197011 DOI: 10.1021/acs.jcim.4c00752] [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: 08/30/2024]
Abstract
We present a computational scheme for predicting the ligands that bind to a pocket of a known structure. It is based on the generation of a general abstract representation of the molecules, which is invariant to rotations, translations, and permutations of atoms, and has some degree of isometry with the space of conformations. We use these representations to train a nondeep machine learning algorithm to classify the binding between pockets and molecule pairs and show that this approach has a better generalization capability than existing methods.
Collapse
Affiliation(s)
- R Beccaria
- Department of Physics, University of Milano, via Celoria 16, 20133 Milano, Italy
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
- Faculty of Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| | - A Lazzeri
- Department of Physics, University of Milano, via Celoria 16, 20133 Milano, Italy
| | - G Tiana
- Department of Physics, University of Milano, via Celoria 16, 20133 Milano, Italy
- INFN, via Celoria 16, 20133 Milano, Italy
| |
Collapse
|
26
|
Torrik A, Zarif M. Machine learning assisted sorting of active microswimmers. J Chem Phys 2024; 161:094907. [PMID: 39225539 DOI: 10.1063/5.0216862] [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: 05/01/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Active matter systems, being in a non-equilibrium state, exhibit complex behaviors, such as self-organization, giving rise to emergent phenomena. There are many examples of active particles with biological origins, including bacteria and spermatozoa, or with artificial origins, such as self-propelled swimmers and Janus particles. The ability to manipulate active particles is vital for their effective application, e.g., separating motile spermatozoa from nonmotile and dead ones, to increase fertilization chance. In this study, we proposed a mechanism-an apparatus-to sort and demix active particles based on their motility values (Péclet number). Initially, using Brownian simulations, we demonstrated the feasibility of sorting self-propelled particles. Following this, we employed machine learning methods, supplemented with data from comprehensive simulations that we conducted for this study, to model the complex behavior of active particles. This enabled us to sort them based on their Péclet number. Finally, we evaluated the performance of the developed models and showed their effectiveness in demixing and sorting the active particles. Our findings can find applications in various fields, including physics, biology, and biomedical science, where the sorting and manipulation of active particles play a pivotal role.
Collapse
Affiliation(s)
- Abdolhalim Torrik
- Department of Physical and Computational Chemistry, Shahid Beheshti University, Tehran 19839-9411, Iran
| | - Mahdi Zarif
- Department of Physical and Computational Chemistry, Shahid Beheshti University, Tehran 19839-9411, Iran
| |
Collapse
|
27
|
Pham TD, Teh MT, Chatzopoulou D, Holmes S, Coulthard P. Artificial Intelligence in Head and Neck Cancer: Innovations, Applications, and Future Directions. Curr Oncol 2024; 31:5255-5290. [PMID: 39330017 PMCID: PMC11430806 DOI: 10.3390/curroncol31090389] [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: 08/07/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
Artificial intelligence (AI) is revolutionizing head and neck cancer (HNC) care by providing innovative tools that enhance diagnostic accuracy and personalize treatment strategies. This review highlights the advancements in AI technologies, including deep learning and natural language processing, and their applications in HNC. The integration of AI with imaging techniques, genomics, and electronic health records is explored, emphasizing its role in early detection, biomarker discovery, and treatment planning. Despite noticeable progress, challenges such as data quality, algorithmic bias, and the need for interdisciplinary collaboration remain. Emerging innovations like explainable AI, AI-powered robotics, and real-time monitoring systems are poised to further advance the field. Addressing these challenges and fostering collaboration among AI experts, clinicians, and researchers is crucial for developing equitable and effective AI applications. The future of AI in HNC holds significant promise, offering potential breakthroughs in diagnostics, personalized therapies, and improved patient outcomes.
Collapse
Affiliation(s)
- Tuan D. Pham
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (M.-T.T.); (D.C.); (S.H.); (P.C.)
| | | | | | | | | |
Collapse
|
28
|
Fernández-Díaz R, Cossio-Pérez R, Agoni C, Lam HT, Lopez V, Shields DC. AutoPeptideML: a study on how to build more trustworthy peptide bioactivity predictors. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 40:btae555. [PMID: 39292535 PMCID: PMC11438549 DOI: 10.1093/bioinformatics/btae555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/08/2024] [Accepted: 09/17/2024] [Indexed: 09/20/2024]
Abstract
MOTIVATION Automated machine learning (AutoML) solutions can bridge the gap between new computational advances and their real-world applications by enabling experimental scientists to build their own custom models. We examine different steps in the development life-cycle of peptide bioactivity binary predictors and identify key steps where automation cannot only result in a more accessible method, but also more robust and interpretable evaluation leading to more trustworthy models. RESULTS We present a new automated method for drawing negative peptides that achieves better balance between specificity and generalization than current alternatives. We study the effect of homology-based partitioning for generating the training and testing data subsets and demonstrate that model performance is overestimated when no such homology correction is used, which indicates that prior studies may have overestimated their performance when applied to new peptide sequences. We also conduct a systematic analysis of different protein language models as peptide representation methods and find that they can serve as better descriptors than a naive alternative, but that there is no significant difference across models with different sizes or algorithms. Finally, we demonstrate that an ensemble of optimized traditional machine learning algorithms can compete with more complex neural network models, while being more computationally efficient. We integrate these findings into AutoPeptideML, an easy-to-use AutoML tool to allow researchers without a computational background to build new predictive models for peptide bioactivity in a matter of minutes. AVAILABILITY AND IMPLEMENTATION Source code, documentation, and data are available at https://github.com/IBM/AutoPeptideML and a dedicated web-server at http://peptide.ucd.ie/AutoPeptideML. A static version of the software to ensure the reproduction of the results is available at https://zenodo.org/records/13363975.
Collapse
Affiliation(s)
- Raúl Fernández-Díaz
- IBM Research, Dublin, Dublin D15 HN66, Ireland
- School of Medicine, University College Dublin, Dublin D04 C1P1, Ireland
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin D04 C1P, Ireland
- The SFI Centre for Research Training in Genomics Data Science, Ireland
| | - Rodrigo Cossio-Pérez
- School of Medicine, University College Dublin, Dublin D04 C1P1, Ireland
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin D04 C1P, Ireland
- Department of Science and Technology, National University of Quilmes, Bernal B1876, Provincia de Buenos Aires, Argentina
| | - Clement Agoni
- School of Medicine, University College Dublin, Dublin D04 C1P1, Ireland
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin D04 C1P, Ireland
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | | | | | - Denis C Shields
- School of Medicine, University College Dublin, Dublin D04 C1P1, Ireland
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin D04 C1P, Ireland
| |
Collapse
|
29
|
Manen-Freixa L, Antolin AA. Polypharmacology prediction: the long road toward comprehensively anticipating small-molecule selectivity to de-risk drug discovery. Expert Opin Drug Discov 2024; 19:1043-1069. [PMID: 39004919 DOI: 10.1080/17460441.2024.2376643] [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: 03/15/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Small molecules often bind to multiple targets, a behavior termed polypharmacology. Anticipating polypharmacology is essential for drug discovery since unknown off-targets can modulate safety and efficacy - profoundly affecting drug discovery success. Unfortunately, experimental methods to assess selectivity present significant limitations and drugs still fail in the clinic due to unanticipated off-targets. Computational methods are a cost-effective, complementary approach to predict polypharmacology. AREAS COVERED This review aims to provide a comprehensive overview of the state of polypharmacology prediction and discuss its strengths and limitations, covering both classical cheminformatics methods and bioinformatic approaches. The authors review available data sources, paying close attention to their different coverage. The authors then discuss major algorithms grouped by the types of data that they exploit using selected examples. EXPERT OPINION Polypharmacology prediction has made impressive progress over the last decades and contributed to identify many off-targets. However, data incompleteness currently limits most approaches to comprehensively predict selectivity. Moreover, our limited agreement on model assessment challenges the identification of the best algorithms - which at present show modest performance in prospective real-world applications. Despite these limitations, the exponential increase of multidisciplinary Big Data and AI hold much potential to better polypharmacology prediction and de-risk drug discovery.
Collapse
Affiliation(s)
- Leticia Manen-Freixa
- Oncobell Division, Bellvitge Biomedical Research Institute (IDIBELL) and ProCURE Department, Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Albert A Antolin
- Oncobell Division, Bellvitge Biomedical Research Institute (IDIBELL) and ProCURE Department, Catalan Institute of Oncology (ICO), Barcelona, Spain
- Center for Cancer Drug Discovery, The Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| |
Collapse
|
30
|
Gharibshahian M, Torkashvand M, Bavisi M, Aldaghi N, Alizadeh A. Recent advances in artificial intelligent strategies for tissue engineering and regenerative medicine. Skin Res Technol 2024; 30:e70016. [PMID: 39189880 PMCID: PMC11348508 DOI: 10.1111/srt.70016] [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: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Tissue engineering and regenerative medicine (TERM) aim to repair or replace damaged or lost tissues or organs due to accidents, diseases, or aging, by applying different sciences. For this purpose, an essential part of TERM is the designing, manufacturing, and evaluating of scaffolds, cells, tissues, and organs. Artificial intelligence (AI) or the intelligence of machines or software can be effective in all areas where computers play a role. METHODS The "artificial intelligence," "machine learning," "tissue engineering," "clinical evaluation," and "scaffold" keywords used for searching in various databases and articles published from 2000 to 2024 were evaluated. RESULTS The combination of tissue engineering and AI has created a new generation of technological advancement in the biomedical industry. Experience in TERM has been refined using advanced design and manufacturing techniques. Advances in AI, particularly deep learning, offer an opportunity to improve scientific understanding and clinical outcomes in TERM. CONCLUSION The findings of this research show the high potential of AI, machine learning, and robots in the selection, design, and fabrication of scaffolds, cells, tissues, or organs, and their analysis, characterization, and evaluation after their implantation. AI can be a tool to accelerate the introduction of tissue engineering products to the bedside. HIGHLIGHTS The capabilities of artificial intelligence (AI) can be used in different ways in all the different stages of TERM and not only solve the existing limitations, but also accelerate the processes, increase efficiency and precision, reduce costs, and complications after transplantation. ML predicts which technologies have the most efficient and easiest path to enter the market and clinic. The use of AI along with these imaging techniques can lead to the improvement of diagnostic information, the reduction of operator errors when reading images, and the improvement of image analysis (such as classification, localization, regression, and segmentation).
Collapse
Affiliation(s)
- Maliheh Gharibshahian
- Nervous System Stem Cells Research CenterSemnan University of Medical SciencesSemnanIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineSemnan University of Medical SciencesSemnanIran
| | | | - Mahya Bavisi
- Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced Technologies in MedicineIran University of Medical SciencesTehranIran
| | - Niloofar Aldaghi
- Student Research CommitteeSchool of MedicineShahroud University of Medical SciencesShahroudIran
| | - Akram Alizadeh
- Nervous System Stem Cells Research CenterSemnan University of Medical SciencesSemnanIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineSemnan University of Medical SciencesSemnanIran
| |
Collapse
|
31
|
Periwal N, Arora P, Thakur A, Agrawal L, Goyal Y, Rathore AS, Anand HS, Kaur B, Sood V. Antiprotozoal peptide prediction using machine learning with effective feature selection techniques. Heliyon 2024; 10:e36163. [PMID: 39247292 PMCID: PMC11380031 DOI: 10.1016/j.heliyon.2024.e36163] [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: 06/14/2023] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background Protozoal pathogens pose a considerable threat, leading to notable mortality rates and the ongoing challenge of developing resistance to drugs. This situation underscores the urgent need for alternative therapeutic approaches. Antimicrobial peptides stand out as promising candidates for drug development. However, there is a lack of published research focusing on predicting antimicrobial peptides specifically targeting protozoal pathogens. In this study, we introduce a successful machine learning-based framework designed to predict potential antiprotozoal peptides effective against protozoal pathogens. Objective The primary objective of this study is to classify and predict antiprotozoal peptides using diverse negative datasets. Methods A comprehensive literature review was conducted to gather experimentally validated antiprotozoal peptides, forming the positive dataset for our study. To construct a robust machine learning classifier, multiple negative datasets were incorporated, including (i) non-antimicrobial, (ii) antiviral, (iii) antibacterial, (iv) antifungal, and (v) antimicrobial peptides excluding those targeting protozoal pathogens. Various compositional features of the peptides were extracted using the pfeature algorithm. Two feature selection methods, SVC-L1 and mRMR, were employed to identify highly relevant features crucial for distinguishing between the positive and negative datasets. Additionally, five popular classifiers i.e. Decision Tree, Random Forest, Support Vector Machine, Logistic Regression, and XGBoost were used to build efficient decision models. Results XGBoost was the most effective in classifying antiprotozoal peptides from each negative dataset based on the features selected by the mRMR feature selection method. The proposed machine learning framework efficiently differentiate the antiprotozoal peptides from (i) non-antimicrobial (ii) antiviral (iii) antibacterial (iv) antifungal and (v) antimicrobial with accuracy of 97.27 %, 93.64 %, 86.36 %, 90.91 %, and 89.09 % respectively on the validation dataset. Conclusion The models are incorporated in a user-friendly web server (www.soodlab.com/appred) to predict the antiprotozoal activity of given peptides.
Collapse
Affiliation(s)
- Neha Periwal
- Department of Biochemistry, Jamia Hamdard, India
| | - Pooja Arora
- Department of Zoology, Hansraj College, University of Delhi, India
| | | | | | - Yash Goyal
- Department of Computer Science, Hansraj College, University of Delhi, India
| | - Anand S Rathore
- Department of Zoology, Hansraj College, University of Delhi, India
| | | | - Baljeet Kaur
- Department of Computer Science, Hansraj College, University of Delhi, India
| | - Vikas Sood
- Department of Biochemistry, Jamia Hamdard, India
| |
Collapse
|
32
|
Tom G, Schmid SP, Baird SG, Cao Y, Darvish K, Hao H, Lo S, Pablo-García S, Rajaonson EM, Skreta M, Yoshikawa N, Corapi S, Akkoc GD, Strieth-Kalthoff F, Seifrid M, Aspuru-Guzik A. Self-Driving Laboratories for Chemistry and Materials Science. Chem Rev 2024; 124:9633-9732. [PMID: 39137296 PMCID: PMC11363023 DOI: 10.1021/acs.chemrev.4c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Self-driving laboratories (SDLs) promise an accelerated application of the scientific method. Through the automation of experimental workflows, along with autonomous experimental planning, SDLs hold the potential to greatly accelerate research in chemistry and materials discovery. This review provides an in-depth analysis of the state-of-the-art in SDL technology, its applications across various scientific disciplines, and the potential implications for research and industry. This review additionally provides an overview of the enabling technologies for SDLs, including their hardware, software, and integration with laboratory infrastructure. Most importantly, this review explores the diverse range of scientific domains where SDLs have made significant contributions, from drug discovery and materials science to genomics and chemistry. We provide a comprehensive review of existing real-world examples of SDLs, their different levels of automation, and the challenges and limitations associated with each domain.
Collapse
Affiliation(s)
- Gary Tom
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Stefan P. Schmid
- Department
of Chemistry and Applied Biosciences, ETH
Zurich, Vladimir-Prelog-Weg 1, CH-8093 Zurich, Switzerland
| | - Sterling G. Baird
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Yang Cao
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Kourosh Darvish
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Han Hao
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Stanley Lo
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
| | - Sergio Pablo-García
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
| | - Ella M. Rajaonson
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Marta Skreta
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Naruki Yoshikawa
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Samantha Corapi
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
| | - Gun Deniz Akkoc
- Forschungszentrum
Jülich GmbH, Helmholtz Institute
for Renewable Energy Erlangen-Nürnberg, Cauerstr. 1, 91058 Erlangen, Germany
- Department
of Chemical and Biological Engineering, Friedrich-Alexander Universität Erlangen-Nürnberg, Egerlandstr. 3, 91058 Erlangen, Germany
| | - Felix Strieth-Kalthoff
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- School of
Mathematics and Natural Sciences, University
of Wuppertal, Gaußstraße
20, 42119 Wuppertal, Germany
| | - Martin Seifrid
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Department
of Materials Science and Engineering, North
Carolina State University, Raleigh, North Carolina 27695, United States of America
| | - Alán Aspuru-Guzik
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
- Department
of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
- Department
of Materials Science & Engineering, University of Toronto, Toronto, Ontario M5S 3E4, Canada
- Lebovic
Fellow, Canadian Institute for Advanced
Research (CIFAR), 661
University Ave, Toronto, Ontario M5G 1M1, Canada
| |
Collapse
|
33
|
Yang Y, Gan W, Lin L, Wang L, Wu J, Luo J. Identification of Active Molecules against Thrombocytopenia through Machine Learning. J Chem Inf Model 2024; 64:6506-6520. [PMID: 39109515 DOI: 10.1021/acs.jcim.4c00718] [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: 08/27/2024]
Abstract
Thrombocytopenia, which is associated with thrombopoietin (TPO) deficiency, presents very limited treatment options and can lead to life-threatening complications. Discovering new therapeutic agents against thrombocytopenia has proven to be a challenging task using traditional screening approaches. Fortunately, machine learning (ML) techniques offer a rapid avenue for exploring chemical space, thereby increasing the likelihood of uncovering new drug candidates. In this study, we focused on computational modeling for drug-induced megakaryocyte differentiation and platelet production using ML methods, aiming to gain insights into the structural characteristics of hematopoietic activity. We developed 112 different classifiers by combining eight ML algorithms with 14 molecule features. The top-performing model achieved good results on both 5-fold cross-validation (with an accuracy of 81.6% and MCC value of 0.589) and external validation (with an accuracy of 83.1% and MCC value of 0.642). Additionally, by leveraging the Shapley additive explanations method, the best model provided quantitative assessments of molecular properties and structures that significantly contributed to the predictions. Furthermore, we employed an ensemble strategy to integrate predictions from multiple models and performed in silico predictions for new molecules with potential activity against thrombocytopenia, sourced from traditional Chinese medicine and the Drug Repurposing Hub. The findings of this study could offer valuable insights into the structural characteristics and computational prediction of thrombopoiesis inducers.
Collapse
Affiliation(s)
- Youyou Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wenli Gan
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Lei Lin
- School of Public Health, Southwest Medical University, Luzhou 646000, China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jianming Wu
- Basic Medical College, Southwest Medical University, Luzhou 646000, China
| | - Jiesi Luo
- Basic Medical College, Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
34
|
Crivelli-Decker J, Beckwith Z, Tom G, Le L, Khuttan S, Salomon-Ferrer R, Beall J, Gómez-Bombarelli R, Bortolato A. Machine Learning Guided AQFEP: A Fast and Efficient Absolute Free Energy Perturbation Solution for Virtual Screening. J Chem Theory Comput 2024; 20. [PMID: 39146234 PMCID: PMC11360131 DOI: 10.1021/acs.jctc.4c00399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
Structure-based methods in drug discovery have become an integral part of the modern drug discovery process. The power of virtual screening lies in its ability to rapidly and cost-effectively explore enormous chemical spaces to select promising ligands for further experimental investigation. Relative free energy perturbation (RFEP) and similar methods are the gold standard for binding affinity prediction in drug discovery hit-to-lead and lead optimization phases, but have high computational cost and the requirement of a structural analog with a known activity. Without a reference molecule requirement, absolute FEP (AFEP) has, in theory, better accuracy for hit ID, but in practice, the slow throughput is not compatible with VS, where fast docking and unreliable scoring functions are still the standard. Here, we present an integrated workflow to virtually screen large and diverse chemical libraries efficiently, combining active learning with a physics-based scoring function based on a fast absolute free energy perturbation method. We validated the performance of the approach in the ranking of structurally related ligands, virtual screening hit rate enrichment, and active learning chemical space exploration; disclosing the largest reported collection of free energy simulations to date.
Collapse
Affiliation(s)
| | - Zane Beckwith
- SandboxAQ, Palo Alto, California 94301, United States
| | - Gary Tom
- SandboxAQ, Palo Alto, California 94301, United States
- Department
of Chemistry and Department of Computer Science, University of Toronto, Toronto, ON M5S 3H6, Canada
- Vector
Institute for Artificial Intelligence, Toronto, ON M5S
3H6, Canada
| | - Ly Le
- SandboxAQ, Palo Alto, California 94301, United States
| | - Sheenam Khuttan
- SandboxAQ, Palo Alto, California 94301, United States
- Department
of Chemistry, Brooklyn College of the City
University of New York, Brooklyn, New York 11367, United States
| | | | - Jackson Beall
- SandboxAQ, Palo Alto, California 94301, United States
| | - Rafael Gómez-Bombarelli
- Department
of Materials Science and Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | | |
Collapse
|
35
|
Suriyaamporn P, Pamornpathomkul B, Patrojanasophon P, Ngawhirunpat T, Rojanarata T, Opanasopit P. The Artificial Intelligence-Powered New Era in Pharmaceutical Research and Development: A Review. AAPS PharmSciTech 2024; 25:188. [PMID: 39147952 DOI: 10.1208/s12249-024-02901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
Currently, artificial intelligence (AI), machine learning (ML), and deep learning (DL) are gaining increased interest in many fields, particularly in pharmaceutical research and development, where they assist in decision-making in complex situations. Numerous research studies and advancements have demonstrated how these computational technologies are used in various pharmaceutical research and development aspects, including drug discovery, personalized medicine, drug formulation, optimization, predictions, drug interactions, pharmacokinetics/ pharmacodynamics, quality control/quality assurance, and manufacturing processes. Using advanced modeling techniques, these computational technologies can enhance efficiency and accuracy, handle complex data, and facilitate novel discoveries within minutes. Furthermore, these technologies offer several advantages over conventional statistics. They allow for pattern recognition from complex datasets, and the models, typically developed from data-driven algorithms, can predict a given outcome (model output) from a set of features (model inputs). Additionally, this review discusses emerging trends and provides perspectives on the application of AI with quality by design (QbD) and the future role of AI in this field. Ethical and regulatory considerations associated with integrating AI into pharmaceutical technology were also examined. This review aims to offer insights to researchers, professionals, and others on the current state of AI applications in pharmaceutical research and development and their potential role in the future of research and the era of pharmaceutical Industry 4.0 and 5.0.
Collapse
Affiliation(s)
- Phuvamin Suriyaamporn
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Boonnada Pamornpathomkul
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Theerasak Rojanarata
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
| |
Collapse
|
36
|
Zhang WY, Zheng XL, Coghi PS, Chen JH, Dong BJ, Fan XX. Revolutionizing adjuvant development: harnessing AI for next-generation cancer vaccines. Front Immunol 2024; 15:1438030. [PMID: 39206192 PMCID: PMC11349682 DOI: 10.3389/fimmu.2024.1438030] [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: 05/24/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
With the COVID-19 pandemic, the importance of vaccines has been widely recognized and has led to increased research and development efforts. Vaccines also play a crucial role in cancer treatment by activating the immune system to target and destroy cancer cells. However, enhancing the efficacy of cancer vaccines remains a challenge. Adjuvants, which enhance the immune response to antigens and improve vaccine effectiveness, have faced limitations in recent years, resulting in few novel adjuvants being identified. The advancement of artificial intelligence (AI) technology in drug development has provided a foundation for adjuvant screening and application, leading to a diversification of adjuvants. This article reviews the significant role of tumor vaccines in basic research and clinical treatment and explores the use of AI technology to screen novel adjuvants from databases. The findings of this review offer valuable insights for the development of new adjuvants for next-generation vaccines.
Collapse
Affiliation(s)
- Wan-Ying Zhang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Xiao-Li Zheng
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Paolo Saul Coghi
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jun-Hui Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bing-Jun Dong
- Gynecology Department, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Xing-Xing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
37
|
Heo JI, Ryu J. Natural Products in the Treatment of Retinopathy of Prematurity: Exploring Therapeutic Potentials. Int J Mol Sci 2024; 25:8461. [PMID: 39126030 PMCID: PMC11313229 DOI: 10.3390/ijms25158461] [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: 05/26/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a vascular disorder affecting the retinas of preterm infants. This condition arises when preterm infants in incubators are exposed to high oxygen levels, leading to oxidative stress, inflammatory responses, and a downregulation of vascular endothelial growth factors, which causes the loss of retinal microvascular capillaries. Upon returning to room air, the upregulation of vascular growth factors results in abnormal vascular growth of retinal endothelial cells. Without appropriate intervention, ROP can progress to blindness. The prevalence of ROP has risen, making it a significant cause of childhood blindness. Current treatments, such as laser therapy and various pharmacologic approaches, are limited by their potential for severe adverse effects. Therefore, a deeper understanding of ROP's pathophysiology and the development of innovative treatments are imperative. Natural products from plants, fungi, bacteria, and marine organisms have shown promise in treating various diseases and have gained attention in ROP research due to their minimal side effects and wide-ranging beneficial properties. This review discusses the roles and mechanisms of natural products that hold potential as therapeutic agents in ROP management.
Collapse
Affiliation(s)
| | - Juhee Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
38
|
Shafiq M, Sherwani ZA, Mushtaq M, Nur-E-Alam M, Ahmad A, Ul-Haq Z. A deep learning-based theoretical protocol to identify potentially isoform-selective PI3Kα inhibitors. Mol Divers 2024; 28:1907-1924. [PMID: 38305819 DOI: 10.1007/s11030-023-10799-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/22/2023] [Indexed: 02/03/2024]
Abstract
Phosphoinositide 3-kinase alpha (PI3Kα) is one of the most frequently dysregulated kinases known for their pivotal role in many oncogenic diseases. While the side effects linked to existing drugs against PI3Kα-induced cancers provide an avenue for further research, the significant structural conservation among PI3Ks makes it extremely difficult to develop new isoform-selective PI3Kα inhibitors. Embracing this challenge, we herein designed a hybrid protocol by integrating machine learning (ML) with in silico drug-designing strategies. A deep learning classification model was developed and trained on the physicochemical descriptors data of known PI3Kα inhibitors and used as a screening filter for a database of small molecules. This approach led us to the prediction of 662 compounds showcasing appropriate features to be considered as PI3Kα inhibitors. Subsequently, a multiphase molecular docking was applied to further characterize the predicted hits in terms of their binding affinities and binding modes in the targeted cavity of the PI3Kα. As a result, a total of 12 compounds were identified whereas the best poses highlighted the efficiency of these ligands in maintaining interactions with the crucial residues of the protein to be targeted for the inhibition of associated activity. Notably, potential activity of compound 12 in counteracting PI3Kα function was found in a previous in vitro study. Following the drug-likeness and pharmacokinetic characterizations, six compounds (compounds 1, 2, 3, 6, 7, and 11) with suitable ADME-T profiles and promising bioavailability were selected. The mechanistic studies in dynamic mode further endorsed the potential of identified hits in blocking the ATP-binding site of the receptor with higher binding affinities than the native inhibitor, alpelisib (BYL-719), particularly the compounds 1, 2, and 11. These outcomes support the reliability of the developed classification model and the devised computational strategy for identifying new isoform-selective drug candidates for PI3Kα inhibition.
Collapse
Affiliation(s)
- Muhammad Shafiq
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Zaid Anis Sherwani
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Mamona Mushtaq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Mohammad Nur-E-Alam
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box. 2457, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Aftab Ahmad
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
39
|
Duo L, Liu Y, Ren J, Tang B, Hirst JD. Artificial intelligence for small molecule anticancer drug discovery. Expert Opin Drug Discov 2024; 19:933-948. [PMID: 39074493 DOI: 10.1080/17460441.2024.2367014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/07/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION The transition from conventional cytotoxic chemotherapy to targeted cancer therapy with small-molecule anticancer drugs has enhanced treatment outcomes. This approach, which now dominates cancer treatment, has its advantages. Despite the regulatory approval of several targeted molecules for clinical use, challenges such as low response rates and drug resistance still persist. Conventional drug discovery methods are costly and time-consuming, necessitating more efficient approaches. The rise of artificial intelligence (AI) and access to large-scale datasets have revolutionized the field of small-molecule cancer drug discovery. Machine learning (ML), particularly deep learning (DL) techniques, enables the rapid identification and development of novel anticancer agents by analyzing vast amounts of genomic, proteomic, and imaging data to uncover hidden patterns and relationships. AREA COVERED In this review, the authors explore the important landmarks in the history of AI-driven drug discovery. They also highlight various applications in small-molecule cancer drug discovery, outline the challenges faced, and provide insights for future research. EXPERT OPINION The advent of big data has allowed AI to penetrate and enable innovations in almost every stage of medicine discovery, transforming the landscape of oncology research through the development of state-of-the-art algorithms and models. Despite challenges in data quality, model interpretability, and technical limitations, advancements promise breakthroughs in personalized and precision oncology, revolutionizing future cancer management.
Collapse
Affiliation(s)
- Lihui Duo
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Yu Liu
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Jianfeng Ren
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Bencan Tang
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Jonathan D Hirst
- School of Chemistry, University of Nottingham University Park, Nottingham, UK
| |
Collapse
|
40
|
Feng C, Qiao C, Ji W, Pang H, Wang L, Feng Q, Ge Y, Rui M. In silico screening and in vivo experimental validation of 15-PGDH inhibitors from traditional Chinese medicine promoting liver regeneration. Int J Biol Macromol 2024; 274:133263. [PMID: 38901515 DOI: 10.1016/j.ijbiomac.2024.133263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/25/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
The enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH), which acts as a negative regulator of prostaglandin E2 (PGE2) levels and activity, represents a promising pharmacological target for promoting liver regeneration. In this study, we collected data on 15-PGDH homologous family proteins, their inhibitors, and traditional Chinese medicine (TCM) compounds. Leveraging machine learning and molecular docking techniques, we constructed a prediction model for virtual screening of 15-PGDH inhibitors from TCM compound library and successfully screened genistein as a potential 15-PGDH inhibitor. Through further validation, it was discovered that genistein considerably enhances liver regeneration by inhibiting 15-PGDH, resulting in a significant increase in the PGE2 level. Genistein's effectiveness suggests its potential as a novel therapeutic agent for liver diseases, highlighting this study's contribution to expanding the clinical applications of TCM.
Collapse
Affiliation(s)
- Chunlai Feng
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Chunxue Qiao
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Wei Ji
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Hui Pang
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Li Wang
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Qiuqi Feng
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Yingying Ge
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Mengjie Rui
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China.
| |
Collapse
|
41
|
Guzman-Pando A, Ramirez-Alonso G, Arzate-Quintana C, Camarillo-Cisneros J. Deep learning algorithms applied to computational chemistry. Mol Divers 2024; 28:2375-2410. [PMID: 38151697 DOI: 10.1007/s11030-023-10771-y] [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: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 12/29/2023]
Abstract
Recently, there has been a significant increase in the use of deep learning techniques in the molecular sciences, which have shown high performance on datasets and the ability to generalize across data. However, no model has achieved perfect performance in solving all problems, and the pros and cons of each approach remain unclear to those new to the field. Therefore, this paper aims to review deep learning algorithms that have been applied to solve molecular challenges in computational chemistry. We proposed a comprehensive categorization that encompasses two primary approaches; conventional deep learning and geometric deep learning models. This classification takes into account the distinct techniques employed by the algorithms within each approach. We present an up-to-date analysis of these algorithms, emphasizing their key features and open issues. This includes details of input descriptors, datasets used, open-source code availability, task solutions, and actual research applications, focusing on general applications rather than specific ones such as drug discovery. Furthermore, our report discusses trends and future directions in molecular algorithm design, including the input descriptors used for each deep learning model, GPU usage, training and forward processing time, model parameters, the most commonly used datasets, libraries, and optimization schemes. This information aids in identifying the most suitable algorithms for a given task. It also serves as a reference for the datasets and input data frequently used for each algorithm technique. In addition, it provides insights into the benefits and open issues of each technique, and supports the development of novel computational chemistry systems.
Collapse
Affiliation(s)
- Abimael Guzman-Pando
- Computational Chemistry Physics Laboratory, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Campus II, 31125, Chihuahua, Mexico
| | - Graciela Ramirez-Alonso
- Faculty of Engineering, Universidad Autónoma de Chihuahua, Campus II, 31125, Chihuahua, Mexico
| | - Carlos Arzate-Quintana
- Computational Chemistry Physics Laboratory, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Campus II, 31125, Chihuahua, Mexico
| | - Javier Camarillo-Cisneros
- Computational Chemistry Physics Laboratory, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Campus II, 31125, Chihuahua, Mexico.
| |
Collapse
|
42
|
Gholap AD, Uddin MJ, Faiyazuddin M, Omri A, Gowri S, Khalid M. Advances in artificial intelligence for drug delivery and development: A comprehensive review. Comput Biol Med 2024; 178:108702. [PMID: 38878397 DOI: 10.1016/j.compbiomed.2024.108702] [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: 01/03/2024] [Revised: 05/12/2024] [Accepted: 06/01/2024] [Indexed: 07/24/2024]
Abstract
Artificial intelligence (AI) has emerged as a powerful tool to revolutionize the healthcare sector, including drug delivery and development. This review explores the current and future applications of AI in the pharmaceutical industry, focusing on drug delivery and development. It covers various aspects such as smart drug delivery networks, sensors, drug repurposing, statistical modeling, and simulation of biotechnological and biological systems. The integration of AI with nanotechnologies and nanomedicines is also examined. AI offers significant advancements in drug discovery by efficiently identifying compounds, validating drug targets, streamlining drug structures, and prioritizing response templates. Techniques like data mining, multitask learning, and high-throughput screening contribute to better drug discovery and development innovations. The review discusses AI applications in drug formulation and delivery, clinical trials, drug safety, and pharmacovigilance. It addresses regulatory considerations and challenges associated with AI in pharmaceuticals, including privacy, data security, and interpretability of AI models. The review concludes with future perspectives, highlighting emerging trends, addressing limitations and biases in AI models, and emphasizing the importance of collaboration and knowledge sharing. It provides a comprehensive overview of AI's potential to transform the pharmaceutical industry and improve patient care while identifying further research and development areas.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401404, India.
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Md Faiyazuddin
- School of Pharmacy, Al-Karim University, Katihar, Bihar, 854106, India; Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Tamil Nadu, India.
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON, P3E 2C6, Canada.
| | - S Gowri
- PG & Research, Department of Physics, Cauvery College for Women, Tiruchirapalli, Tamil Nadu, 620018, India
| | - Mohammad Khalid
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; Sunway Centre for Electrochemical Energy and Sustainable Technology (SCEEST), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia; University Centre for Research and Development, Chandigarh University, Mohali, Punjab, 140413, India.
| |
Collapse
|
43
|
Hao Y, Li B, Huang D, Wu S, Wang T, Fu L, Liu X. Developing a Semi-Supervised Approach Using a PU-Learning-Based Data Augmentation Strategy for Multitarget Drug Discovery. Int J Mol Sci 2024; 25:8239. [PMID: 39125808 PMCID: PMC11312053 DOI: 10.3390/ijms25158239] [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: 06/21/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Multifactorial diseases demand therapeutics that can modulate multiple targets for enhanced safety and efficacy, yet the clinical approval of multitarget drugs remains rare. The integration of machine learning (ML) and deep learning (DL) in drug discovery has revolutionized virtual screening. This study investigates the synergy between ML/DL methodologies, molecular representations, and data augmentation strategies. Notably, we found that SVM can match or even surpass the performance of state-of-the-art DL methods. However, conventional data augmentation often involves a trade-off between the true positive rate and false positive rate. To address this, we introduce Negative-Augmented PU-bagging (NAPU-bagging) SVM, a novel semi-supervised learning framework. By leveraging ensemble SVM classifiers trained on resampled bags containing positive, negative, and unlabeled data, our approach is capable of managing false positive rates while maintaining high recall rates. We applied this method to the identification of multitarget-directed ligands (MTDLs), where high recall rates are critical for compiling a list of interaction candidate compounds. Case studies demonstrate that NAPU-bagging SVM can identify structurally novel MTDL hits for ALK-EGFR with favorable docking scores and binding modes, as well as pan-agonists for dopamine receptors. The NAPU-bagging SVM methodology should serve as a promising avenue to virtual screening, especially for the discovery of MTDLs.
Collapse
Affiliation(s)
- Yang Hao
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Bo Li
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Daiyun Huang
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- School of Life Sciences, Fudan University, Shanghai 200092, China
| | - Sijin Wu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
| | - Tianjun Wang
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Lei Fu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
| | - Xin Liu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
| |
Collapse
|
44
|
Li W, Yin Z, Li X, Ma D, Yi S, Zhang Z, Zou C, Bu K, Dai M, Yue J, Chen Y, Zhang X, Zhang S. A hybrid quantum computing pipeline for real world drug discovery. Sci Rep 2024; 14:16942. [PMID: 39043787 PMCID: PMC11266395 DOI: 10.1038/s41598-024-67897-8] [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: 04/01/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
Quantum computing, with its superior computational capabilities compared to classical approaches, holds the potential to revolutionize numerous scientific domains, including pharmaceuticals. However, the application of quantum computing for drug discovery has primarily been limited to proof-of-concept studies, which often fail to capture the intricacies of real-world drug development challenges. In this study, we diverge from conventional investigations by developing a hybrid quantum computing pipeline tailored to address genuine drug design problems. Our approach underscores the application of quantum computation in drug discovery and propels it towards more scalable system. We specifically construct our versatile quantum computing pipeline to address two critical tasks in drug discovery: the precise determination of Gibbs free energy profiles for prodrug activation involving covalent bond cleavage, and the accurate simulation of covalent bond interactions. This work serves as a pioneering effort in benchmarking quantum computing against veritable scenarios encountered in drug design, especially the covalent bonding issue present in both of the case studies, thereby transitioning from theoretical models to tangible applications. Our results demonstrate the potential of a quantum computing pipeline for integration into real world drug design workflows.
Collapse
Affiliation(s)
- Weitang Li
- Tencent Quantum Lab, Shenzhen, 518057, China
| | - Zhi Yin
- AceMapAI Biotechnology, Suzhou, 215000, China.
- School of Science, Ningbo University of Technology, Ningbo, 315211, China.
| | - Xiaoran Li
- AceMapAI Biotechnology, Suzhou, 215000, China
| | | | - Shuang Yi
- AceMapAI Biotechnology, Suzhou, 215000, China
| | | | - Chenji Zou
- Tencent Quantum Lab, Shenzhen, 518057, China
| | - Kunliang Bu
- Tencent Quantum Lab, Shenzhen, 518057, China
| | - Maochun Dai
- Tencent Quantum Lab, Shenzhen, 518057, China
| | - Jie Yue
- Tencent Quantum Lab, Shenzhen, 518057, China
| | - Yuzong Chen
- AceMapAI Joint Lab, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaojin Zhang
- AceMapAI Joint Lab, China Pharmaceutical University, Nanjing, 211198, China.
| | | |
Collapse
|
45
|
Mervin L, Voronov A, Kabeshov M, Engkvist O. QSARtuna: An Automated QSAR Modeling Platform for Molecular Property Prediction in Drug Design. J Chem Inf Model 2024; 64:5365-5374. [PMID: 38950185 DOI: 10.1021/acs.jcim.4c00457] [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: 07/03/2024]
Abstract
Machine-learning (ML) and deep-learning (DL) approaches to predict the molecular properties of small molecules are increasingly deployed within the design-make-test-analyze (DMTA) drug design cycle to predict molecular properties of interest. Despite this uptake, there are only a few automated packages to aid their development and deployment that also support uncertainty estimation, model explainability, and other key aspects of model usage. This represents a key unmet need within the field, and the large number of molecular representations and algorithms (and associated parameters) means it is nontrivial to robustly optimize, evaluate, reproduce, and deploy models. Here, we present QSARtuna, a molecule property prediction modeling pipeline, written in Python and utilizing the Optuna, Scikit-learn, RDKit, and ChemProp packages, which enables the efficient and automated comparison between molecular representations and machine learning models. The platform was developed by considering the increasingly important aspect of model uncertainty quantification and explainability by design. We provide details for our framework and provide illustrative examples to demonstrate the capability of the software when applied to simple molecular property, reaction/reactivity prediction, and DNA encoded library enrichment classification. We hope that the release of QSARtuna will further spur innovation in automatic ML modeling and provide a platform for education of best practices in molecular property modeling. The code for the QSARtuna framework is made freely available via GitHub.
Collapse
Affiliation(s)
- Lewis Mervin
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, United Kingdom
| | - Alexey Voronov
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg 412 96, Sweden
| | - Mikhail Kabeshov
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg 412 96, Sweden
| | - Ola Engkvist
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg 412 96, Sweden
- Department of Computer Science and Engineering, University of Gothenburg, Chalmers University of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
46
|
Gale-Day Z, Shub L, Chuang KV, Keiser MJ. Proximity Graph Networks: Predicting Ligand Affinity with Message Passing Neural Networks. J Chem Inf Model 2024; 64:5439-5450. [PMID: 38953560 PMCID: PMC11267574 DOI: 10.1021/acs.jcim.4c00311] [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: 02/22/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
Message passing neural networks (MPNNs) on molecular graphs generate continuous and differentiable encodings of small molecules with state-of-the-art performance on protein-ligand complex scoring tasks. Here, we describe the proximity graph network (PGN) package, an open-source toolkit that constructs ligand-receptor graphs based on atom proximity and allows users to rapidly apply and evaluate MPNN architectures for a broad range of tasks. We demonstrate the utility of PGN by introducing benchmarks for affinity and docking score prediction tasks. Graph networks generalize better than fingerprint-based models and perform strongly for the docking score prediction task. Overall, MPNNs with proximity graph data structures augment the prediction of ligand-receptor complex properties when ligand-receptor data are available.
Collapse
Affiliation(s)
- Zachary
J. Gale-Day
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, San
Francisco, California 94158, United States
- Institute
for Neurodegenerative Diseases, University
of California, San Francisco, San
Francisco, California 94158, United States
| | - Laura Shub
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, San
Francisco, California 94158, United States
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
- Institute
for Neurodegenerative Diseases, University
of California, San Francisco, San
Francisco, California 94158, United States
- Bakar
Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California 94158, United States
| | - Kangway V. Chuang
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, San
Francisco, California 94158, United States
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
- Institute
for Neurodegenerative Diseases, University
of California, San Francisco, San
Francisco, California 94158, United States
- Bakar
Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California 94158, United States
| | - Michael J. Keiser
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, San
Francisco, California 94158, United States
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
- Institute
for Neurodegenerative Diseases, University
of California, San Francisco, San
Francisco, California 94158, United States
- Bakar
Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
47
|
Abbas MKG, Rassam A, Karamshahi F, Abunora R, Abouseada M. The Role of AI in Drug Discovery. Chembiochem 2024; 25:e202300816. [PMID: 38735845 DOI: 10.1002/cbic.202300816] [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: 12/03/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
The emergence of Artificial Intelligence (AI) in drug discovery marks a pivotal shift in pharmaceutical research, blending sophisticated computational techniques with conventional scientific exploration to break through enduring obstacles. This review paper elucidates the multifaceted applications of AI across various stages of drug development, highlighting significant advancements and methodologies. It delves into AI's instrumental role in drug design, polypharmacology, chemical synthesis, drug repurposing, and the prediction of drug properties such as toxicity, bioactivity, and physicochemical characteristics. Despite AI's promising advancements, the paper also addresses the challenges and limitations encountered in the field, including data quality, generalizability, computational demands, and ethical considerations. By offering a comprehensive overview of AI's role in drug discovery, this paper underscores the technology's potential to significantly enhance drug development, while also acknowledging the hurdles that must be overcome to fully realize its benefits.
Collapse
Affiliation(s)
- M K G Abbas
- Center for Advanced Materials, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Abrar Rassam
- Secondary Education, Educational Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Fatima Karamshahi
- Department of Chemistry and Earth Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Rehab Abunora
- Faculty of Medicine, General Medicine and Surgery, Helwan University, Cairo, Egypt
| | - Maha Abouseada
- Department of Chemistry and Earth Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| |
Collapse
|
48
|
Gupta YD, Bhandary S. Artificial Intelligence for Understanding Mechanisms of Antimicrobial Resistance and Antimicrobial Discovery. ARTIFICIAL INTELLIGENCE AND MACHINE LEARNING IN DRUG DESIGN AND DEVELOPMENT 2024:117-156. [DOI: 10.1002/9781394234196.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
49
|
Ahmed F, Samantasinghar A, Bae MA, Choi KH. Integrated ML-Based Strategy Identifies Drug Repurposing for Idiopathic Pulmonary Fibrosis. ACS OMEGA 2024; 9:29870-29883. [PMID: 39005763 PMCID: PMC11238209 DOI: 10.1021/acsomega.4c03796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/30/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) affects an estimated global population of around 3 million individuals. IPF is a medical condition with an unknown cause characterized by the formation of scar tissue in the lungs, leading to progressive respiratory disease. Currently, there are only two FDA-approved small molecule drugs specifically for the treatment of IPF and this has created a demand for the rapid development of drugs for IPF treatment. Moreover, denovo drug development is time and cost-intensive with less than a 10% success rate. Drug repurposing currently is the most feasible option for rapidly making the drugs to market for a rare and sporadic disease. Normally, the repurposing of drugs begins with a screening of FDA-approved drugs using computational tools, which results in a low hit rate. Here, an integrated machine learning-based drug repurposing strategy is developed to significantly reduce the false positive outcomes by introducing the predock machine-learning-based predictions followed by literature and GSEA-assisted validation and drug pathway prediction. The developed strategy is deployed to 1480 FDA-approved drugs and to drugs currently in a clinical trial for IPF to screen them against "TGFB1", "TGFB2", "PDGFR-a", "SMAD-2/3", "FGF-2", and more proteins resulting in 247 total and 27 potentially repurposable drugs. The literature and GSEA validation suggested that 72 of 247 (29.14%) drugs have been tried for IPF, 13 of 247 (5.2%) drugs have already been used for lung fibrosis, and 20 of 247 (8%) drugs have been tested for other fibrotic conditions such as cystic fibrosis and renal fibrosis. Pathway prediction of the remaining 142 drugs was carried out resulting in 118 distinct pathways. Furthermore, the analysis revealed that 29 of 118 pathways were directly or indirectly involved in IPF and 11 of 29 pathways were directly involved. Moreover, 15 potential drug combinations are suggested for showing a strong synergistic effect in IPF. The drug repurposing strategy reported here will be useful for rapidly developing drugs for treating IPF and other related conditions.
Collapse
Affiliation(s)
- Faheem Ahmed
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Anupama Samantasinghar
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Myung Ae Bae
- Therapeutics
and Biotechnology Division, Korea Research
Institute of Chemical Technology, Daejeon 34114, Korea
| | - Kyung Hyun Choi
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| |
Collapse
|
50
|
Abou Hajal A, Bryce RA, Amor BB, Atatreh N, Ghattas MA. Boosting the Accuracy and Chemical Space Coverage of the Detection of Small Colloidal Aggregating Molecules Using the BAD Molecule Filter. J Chem Inf Model 2024; 64:4991-5005. [PMID: 38920403 DOI: 10.1021/acs.jcim.4c00363] [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: 06/27/2024]
Abstract
The ability to conduct effective high throughput screening (HTS) campaigns in drug discovery is often hampered by the detection of false positives in these assays due to small colloidally aggregating molecules (SCAMs). SCAMs can produce artifactual hits in HTS by nonspecific inhibition of the protein target. In this work, we present a new computational prediction tool for detecting SCAMs based on their 2D chemical structure. The tool, called the boosted aggregation detection (BAD) molecule filter, employs decision tree ensemble methods, namely, the CatBoost classifier and the light gradient-boosting machine, to significantly improve the detection of SCAMs. In developing the filter, we explore models trained on individual data sets, a consensus approach using these models, and, third, a merged data set approach, each tailored for specific drug discovery needs. The individual data set method emerged as most effective, achieving 93% sensitivity and 90% specificity, outperforming existing state-of-the-art models by 20 and 5%, respectively. The consensus models offer broader chemical space coverage, exceeding 90% for all testing sets. This feature is an important aspect particularly for early stage medicinal chemistry projects, and provides information on applicability domain. Meanwhile, the merged data set models demonstrated robust performance, with a notable sensitivity of 79% in the comprehensive 10-fold cross-validation test set. A SHAP analysis of model features indicates the importance of hydrophobicity and molecular complexity as primary factors influencing the aggregation propensity. The BAD molecule filter is readily accessible for the public usage on https://molmodlab-aau.com/Tools.html. This filter provides a new, more robust tool for aggregate prediction in the early stages of drug discovery to optimize hit rates and reduce associated testing and validation overheads.
Collapse
Affiliation(s)
- Abdallah Abou Hajal
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| | - Richard A Bryce
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Boulbaba Ben Amor
- Core42, Inception/G42, Abu Dhabi 2282, United Arab Emirates
- IMT Nord Europe, Villeneuve D'Ascq 59650 France
| | - Noor Atatreh
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| | - Mohammad A Ghattas
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| |
Collapse
|