1
|
Aruna AS, Babu KRR, Deepthi K. A deep drug prediction framework for viral infectious diseases using an optimizer-based ensemble of convolutional neural network: COVID-19 as a case study. Mol Divers 2025; 29:2473-2487. [PMID: 39379663 DOI: 10.1007/s11030-024-11003-7] [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: 07/28/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024]
Abstract
The SARS-CoV-2 outbreak highlights the persistent vulnerability of humanity to epidemics and emerging microbial threats, emphasizing the lack of time to develop disease-specific treatments. Therefore, it appears beneficial to utilize existing resources and therapies. Computational drug repositioning is an effective strategy that redirects authorized drugs to new therapeutic purposes. This strategy holds significant promise for newly emerging diseases, as drug discovery is a lengthy and expensive process. Through this study, we present an ensemble method based on the convolutional neural network integrated with genetic algorithm and deep forest classifier for virus-drug association prediction (CGDVDA). We generated feature vectors by combining drug chemical structure and virus genomic sequence-based similarities, and extracted prominent deep features by applying the convolutional neural network. The convoluted features are optimized using the genetic algorithm and classified using the ensemble deep forest classifier to predict novel virus-drug associations. The proposed method predicts drugs for COVID-19 and other viral diseases in the dataset. The model could achieve ROC-AUC scores of 0.9159 on fivefold cross-validation. We compared the performance of the model with state-of-the-art approaches and classifiers. The experimental results and case studies illustrate the efficacy of CGDVDA in predicting drugs against viral infectious diseases.
Collapse
Affiliation(s)
- A S Aruna
- Dept. of Information Technology, Government Engineering College Palakkad, APJ Abdul Kalam Technological University, Palakkad, Kerala, 678633, India.
- Department of Computer Science, College of Engineering Vadakara, Kozhikode, Kerala, 673105, India.
| | - K R Remesh Babu
- Dept. of Information Technology, Government Engineering College Palakkad, APJ Abdul Kalam Technological University, Palakkad, Kerala, 678633, India
| | - K Deepthi
- Department of Computer Science, Central University of Kerala (Govt. of India), Kasaragod, Kerala, 671320, India
| |
Collapse
|
2
|
Akgüller Ö, Balcı MA, Cioca G. A Multi-Modal Graph Neural Network Framework for Parkinson's Disease Therapeutic Discovery. Int J Mol Sci 2025; 26:4453. [PMID: 40362692 PMCID: PMC12072649 DOI: 10.3390/ijms26094453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder lacking effective disease-modifying treatments. In this study, we integrated large-scale protein-protein interaction networks with a multi-modal graph neural network (GNN) to identify and prioritize multi-target drug repurposing candidates for PD. Network analysis and advanced clustering methods delineated functional modules, and a novel Functional Centrality Index was employed to pinpoint key nodes within the PD interactome. The GNN model, incorporating molecular descriptors, network topology, and uncertainty quantification, predicted candidate drugs that simultaneously target critical proteins implicated in lysosomal dysfunction, mitochondrial impairment, synaptic disruption, and neuroinflammation. Among the top hits were compounds such as dithiazanine, ceftolozane, DL-α-tocopherol, bromisoval, imidurea, medronic acid, and modufolin. These findings provide mechanistic insights into PD pathology and demonstrate that a polypharmacology approach can reveal repurposing opportunities for existing drugs. Our results highlight the potential of network-based deep learning frameworks to accelerate the discovery of multi-target therapies for PD and other multifactorial neurodegenerative diseases.
Collapse
Affiliation(s)
- Ömer Akgüller
- Faculty of Science, Department of Mathematics, Mugla Sitki Kocman University, Mugla 48000, Turkey;
| | - Mehmet Ali Balcı
- Faculty of Science, Department of Mathematics, Mugla Sitki Kocman University, Mugla 48000, Turkey;
| | - Gabriela Cioca
- Faculty of Medicine, Preclinical Department, Lucian Blaga University of Sibiu, 550024 Sibiu, Romania;
| |
Collapse
|
3
|
Bäumlin E, Andenmatten D, Luginbühl J, Lalou A, Schwaller N, Karousis ED. The impact of Coronavirus Nsp1 on host mRNA degradation is independent of its role in translation inhibition. Cell Rep 2025; 44:115488. [PMID: 40153437 DOI: 10.1016/j.celrep.2025.115488] [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: 10/15/2024] [Revised: 12/09/2024] [Accepted: 03/07/2025] [Indexed: 03/30/2025] Open
Abstract
When host cells are infected with coronaviruses, the first viral protein produced is non-structural protein 1 (Nsp1). This protein inhibits host protein synthesis and induces host mRNA degradation to enhance viral proliferation. Despite its critical role, the mechanism by which Nsp1 mediates cellular mRNA degradation remains unclear. In this study, we use cell-free translation to address how host mRNA stability is regulated by Nsp1. We reveal that SARS-CoV-2 Nsp1 binding to the ribosome is enough to trigger mRNA degradation independently of ribosome collisions or active translation. MERS-CoV Nsp1 inhibits translation without triggering degradation, highlighting mechanistic differences between the two Nsp1 counterparts. Nsp1 and viral mRNAs appear to co-evolve, rendering viral mRNAs immune to Nsp1-mediated degradation in SARS-CoV-2, MERS-CoV, and Bat-Hp viruses. By providing insights into the mode of action of Nsp1, our study helps to understand the biology of Nsp1 better and find strategies for therapeutic targeting against coronaviral infections.
Collapse
Affiliation(s)
- Emilie Bäumlin
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Dominic Andenmatten
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Jonas Luginbühl
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland; Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Aurélien Lalou
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland; Multidisciplinary Center for Infectious Diseases, University of Bern, 3012 Bern, Switzerland
| | - Nino Schwaller
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Evangelos D Karousis
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland; Multidisciplinary Center for Infectious Diseases, University of Bern, 3012 Bern, Switzerland.
| |
Collapse
|
4
|
Pamidimukkala JV, Parthasarathy BR, Senapati S. Decoding potential host protein targets against Flaviviridae using protein-protein interaction network. Int J Biol Macromol 2025; 310:143217. [PMID: 40250655 DOI: 10.1016/j.ijbiomac.2025.143217] [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: 02/13/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Flaviviridae family comprises some of the most vulnerable viruses known for causing widespread outbreaks, high mortality rates, and severe long-term health complications in humans. Viruses like Dengue (DENV), Zika (ZIKV) and Hepatitis C (HCV) are endemic across the globe, especially in tropical and subtropical regions, infecting multiple tissues and leading to significant health crises. Investigating virus-host interactions across tissues can reveal tissue-specific drug targets and aid antiviral drug repurposing. In this study, we employed a multi-step computational approach to construct a comprehensive virus-human interactome by integrating virus-host protein-protein interactions (PPIs) with tissue-specific gene expression profiles to study key protein targets associated with Flaviviridae infections. Mapping drug-target predictions revealed druggable proteins - CCNA2 in peripheral blood mononuclear cells (PBMC) and EIF2S2, CDK7 and CARS in the liver, with Tamoxifen, Sirolimus, Entrectinib and L-cysteine as potential repurposable drugs, respectively. Further protein-ligand docking and molecular dynamics (MD) simulations were conducted to assess the stability of the complexes. These findings highlight common druggable human targets exploited by DENV, ZIKV and HCV, providing a foundation for broad-spectrum antiviral therapies. By focusing on shared host pathways and targets in viral replication, we propose promising drug candidates, supporting the development of unified therapeutic strategies against Flaviviridae viruses.
Collapse
Affiliation(s)
- Jaya Vasavi Pamidimukkala
- Department of Biotechnology and BJM School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Bharath Raj Parthasarathy
- Department of Biotechnology and BJM School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Sanjib Senapati
- Department of Biotechnology and BJM School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
5
|
Wang J, Niu Q, Yu Y, Liu J, Zhang S, Zong W, Tian S, Wang Z, Li B. Modular-Based Synergetic Mechanisms of Jasminoidin and Ursodeoxycholic Acid in Cerebral Ischemia Therapy. Biomedicines 2025; 13:938. [PMID: 40299522 PMCID: PMC12025273 DOI: 10.3390/biomedicines13040938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives: Jasminoidin (JA) and ursodeoxycholic acid (UA) have been shown to exert synergistic effects on cerebral ischemia (CI) therapy, but the underlying mechanisms remain to be elucidated. Objective: To elucidate the synergistic mechanisms involved in the combined use of JA and UA (JU) for CI therapy using a driver-induced modular screening (DiMS) strategy. Methods: Network proximity and topology-based approaches were used to identify synergistic modules and driver genes from an anti-ischemic microarray dataset (ArrayExpress, E-TABM-662). A middle cerebral artery occlusion/reperfusion (MCAO/R) model was established in 30 Sprague Dawley rats, divided into sham, vehicle, JA (25 mg/mL), UA (7 mg/mL), and JU (JA:UA = 1:1) groups. After 90 minutes of ischemia, infarct volume and neurological deficit scores were evaluated. Western blotting was performed 24 h after administration to validate key protein changes. Results: Six, eleven, and four drug-responsive On_modules were identified for JA, UA, and JU, respectively. Three synergistic modules (Sy-modules, JU-Mod-7, 8, and 10) and 12 driver genes (e.g., NRF1, FN1, CUL3) were identified, mainly involving the PI3K-Akt and MAPK pathways and regulation of the actin cytoskeleton. JA and UA synergistically reduced infarct volume and neurological deficit score (2.5, p < 0.05) in MCAO/R rats. In vivo studies demonstrated that JU suppressed the expression of CUL3, FN1, and ITGA4, while it increased that of NRF1. Conclusions: JU acts synergistically on CI-reperfusion injury by regulating FN1, CUL3, ITGA4, and NRF1 and inducing the PI3K-Akt, MAPK, and actin cytoskeleton pathways. DiMS provides a new approach to uncover mechanisms of combination therapies.
Collapse
Affiliation(s)
- Jingai Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Qikai Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Yanan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.Y.); (J.L.)
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.Y.); (J.L.)
| | - Siqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Siwei Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.Y.); (J.L.)
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| |
Collapse
|
6
|
Saranraj K, Kiran PU. Drug repurposing: Clinical practices and regulatory pathways. Perspect Clin Res 2025; 16:61-68. [PMID: 40322475 PMCID: PMC12048090 DOI: 10.4103/picr.picr_70_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 05/08/2025] Open
Abstract
Drug repurposing, also known as drug repositioning or reprofiling, involves identifying new therapeutic uses for existing drugs beyond their original indications. Historical examples include sildenafil citrate transitioning to an erectile dysfunction treatment and thalidomide shifting from a sedative to an immunomodulatory agent. Advocates tout its potential to address unmet medical needs by expediting development, reducing costs, and using drugs with established safety profiles. However, concerns exist regarding specificity for new indications, safety, and regulatory exploitation. Ethical considerations include equitable access, informed consent when using drugs off-label, and transparency. Recent advancements include artificial intelligence (AI) applications, network pharmacology, and omics technologies. Clinical trials explore repurposed drugs' efficacy, with regulatory agencies facilitating approval. Challenges include intellectual property protection, drug target specificity, trial design complexities, and funding limitations. Ethical challenges encompass patient autonomy, potential conflicts of interest due to financial incentives for industries, and resource allocation. Future directions involve precision medicine, AI, and global collaboration. In conclusion, drug repurposing offers a promising pathway for therapeutic innovation but requires careful consideration of its complexities and ethical implications to maximize benefits and minimize risks.
Collapse
Affiliation(s)
- K. Saranraj
- Department of Pharmacology, Rangaraya Medical College, Kakinada, Andhra Pradesh, India
| | - P. Usha Kiran
- Department of Pharmacology, Rangaraya Medical College, Kakinada, Andhra Pradesh, India
| |
Collapse
|
7
|
Carroll E, Scaber J, Huber KVM, Brennan PE, Thompson AG, Turner MR, Talbot K. Drug repurposing in amyotrophic lateral sclerosis (ALS). Expert Opin Drug Discov 2025; 20:447-464. [PMID: 40029669 PMCID: PMC11974926 DOI: 10.1080/17460441.2025.2474661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Identifying treatments that can alter the natural history of amyotrophic lateral sclerosis (ALS) is challenging. For years, drug discovery in ALS has relied upon traditional approaches with limited success. Drug repurposing, where clinically approved drugs are reevaluated for other indications, offers an alternative strategy that overcomes some of the challenges associated with de novo drug discovery. AREAS COVERED In this review, the authors discuss the challenge of drug discovery in ALS and examine the potential of drug repurposing for the identification of new effective treatments. The authors consider a range of approaches, from screening in experimental models to computational approaches, and outline some general principles for preclinical and clinical research to help bridge the translational gap. Literature was reviewed from original publications, press releases and clinical trials. EXPERT OPINION Despite remaining challenges, drug repurposing offers the opportunity to improve therapeutic options for ALS patients. Nevertheless, stringent preclinical research will be necessary to identify the most promising compounds together with innovative experimental medicine studies to bridge the translational gap. The authors further highlight the importance of combining expertise across academia, industry and wider stakeholders, which will be key in the successful delivery of repurposed therapies to the clinic.
Collapse
Affiliation(s)
- Emily Carroll
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jakub Scaber
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Kilian V. M. Huber
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Paul E. Brennan
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| |
Collapse
|
8
|
Tanoli Z, Fernández-Torras A, Özcan UO, Kushnir A, Nader KM, Gadiya Y, Fiorenza L, Ianevski A, Vähä-Koskela M, Miihkinen M, Seemab U, Leinonen H, Seashore-Ludlow B, Tampere M, Kalman A, Ballante F, Benfenati E, Saunders G, Potdar S, Gómez García I, García-Serna R, Talarico C, Beccari AR, Schaal W, Polo A, Costantini S, Cabri E, Jacobs M, Saarela J, Budillon A, Spjuth O, Östling P, Xhaard H, Quintana J, Mestres J, Gribbon P, Ussi AE, Lo DC, de Kort M, Wennerberg K, Fratelli M, Carreras-Puigvert J, Aittokallio T. Computational drug repurposing: approaches, evaluation of in silico resources and case studies. Nat Rev Drug Discov 2025:10.1038/s41573-025-01164-x. [PMID: 40102635 DOI: 10.1038/s41573-025-01164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Repurposing of existing drugs for new indications has attracted substantial attention owing to its potential to accelerate drug development and reduce costs. Hundreds of computational resources such as databases and predictive platforms have been developed that can be applied for drug repurposing, making it challenging to select the right resource for a specific drug repurposing project. With the aim of helping to address this challenge, here we overview computational approaches to drug repurposing based on a comprehensive survey of available in silico resources using a purpose-built drug repurposing ontology that classifies the resources into hierarchical categories and provides application-specific information. We also present an expert evaluation of selected resources and three drug repurposing case studies implemented within the Horizon Europe REMEDi4ALL project to demonstrate the practical use of the resources. This comprehensive Review with expert evaluations and case studies provides guidelines and recommendations on the best use of various in silico resources for drug repurposing and establishes a basis for a sustainable and extendable drug repurposing web catalogue.
Collapse
Affiliation(s)
- Ziaurrehman Tanoli
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland.
| | | | - Umut Onur Özcan
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aleksandr Kushnir
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kristen Michelle Nader
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Yojana Gadiya
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, Bonn, Germany
| | - Laura Fiorenza
- Dipartimento di Elettronica, Informazione e Bioingegneria (DEIB), Politecnico di Milano, Milan, Italy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mitro Miihkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Umair Seemab
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Brinton Seashore-Ludlow
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Marianna Tampere
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Adelinn Kalman
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Flavio Ballante
- Chemical Biology Consortium Sweden (CBCS), SciLifeLab, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gary Saunders
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | | | | | - Wesley Schaal
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrea Polo
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Susan Costantini
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Enrico Cabri
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marc Jacobs
- Fraunhofer-Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Alfredo Budillon
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Päivi Östling
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Henri Xhaard
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jordi Quintana
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
| | - Jordi Mestres
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
- Institut de Quimica Computacional i Catalisi, Facultat de Ciencies, Universitat de Girona, Girona, Catalonia, Spain
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
| | - Anton E Ussi
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Donald C Lo
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Martin de Kort
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Krister Wennerberg
- Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
9
|
Zhang Y, Sui X, Pan F, Yu K, Li K, Tian S, Erdengasileng A, Han Q, Wang W, Wang J, Wang J, Sun D, Chung H, Zhou J, Zhou E, Lee B, Zhang P, Qiu X, Zhao T, Zhang J. A comprehensive large scale biomedical knowledge graph for AI powered data driven biomedical research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.13.562216. [PMID: 38168218 PMCID: PMC10760044 DOI: 10.1101/2023.10.13.562216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
To address the rapid growth of scientific publications and data in biomedical research, knowledge graphs (KGs) have become a critical tool for integrating large volumes of heterogeneous data to enable efficient information retrieval and automated knowledge discovery (AKD). However, transforming unstructured scientific literature into KGs remains a significant challenge, with previous methods unable to achieve human-level accuracy. In this study, we utilized an information extraction pipeline that won first place in the LitCoin NLP Challenge (2022) to construct a large-scale KG named iKraph using all PubMed abstracts. The extracted information matches human expert annotations and significantly exceeds the content of manually curated public databases. To enhance the KG's comprehensiveness, we integrated relation data from 40 public databases and relation information inferred from high-throughput genomics data. This KG facilitates rigorous performance evaluation of AKD, which was infeasible in previous studies. We designed an interpretable, probabilistic-based inference method to identify indirect causal relations and applied it to real-time COVID-19 drug repurposing from March 2020 to May 2023. Our method identified 600-1400 candidate drugs per month, with one-third of those discovered in the first two months later supported by clinical trials or PubMed publications. These outcomes are very challenging to attain through alternative approaches that lack a thorough understanding of the existing literature. A cloud-based platform (https://biokde.insilicom.com) was developed for academic users to access this rich structured data and associated tools.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Statistics, Florida State University, Tallahassee, FL 32306
- Insilicom LLC, Tallahassee, FL 32303
| | - Xin Sui
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | - Feng Pan
- Insilicom LLC, Tallahassee, FL 32303
| | | | - Keqiao Li
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | - Shubo Tian
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | | | - Qing Han
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | - Wanjing Wang
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | | | - Jian Wang
- 977 Wisteria Ter., Sunnyvale, CA 94086
| | | | | | - Jun Zhou
- Insilicom LLC, Tallahassee, FL 32303
| | - Eric Zhou
- Insilicom LLC, Tallahassee, FL 32303
| | - Ben Lee
- Insilicom LLC, Tallahassee, FL 32303
| | - Peili Zhang
- Forward Informatics, Winchester, Massachusetts, 01890
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY 14642
| | - Tingting Zhao
- Insilicom LLC, Tallahassee, FL 32303
- Department of Geography, Florida State University, Tallahassee, FL 32306
| | - Jinfeng Zhang
- Department of Statistics, Florida State University, Tallahassee, FL 32306
- Insilicom LLC, Tallahassee, FL 32303
| |
Collapse
|
10
|
Tang X, Zhou C, Lu C, Meng Y, Xu J, Hu X, Tian G, Yang J. Enhancing Drug Repositioning Through Local Interactive Learning With Bilinear Attention Networks. IEEE J Biomed Health Inform 2025; 29:1644-1655. [PMID: 37988217 DOI: 10.1109/jbhi.2023.3335275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Drug repositioning has emerged as a promising strategy for identifying new therapeutic applications for existing drugs. In this study, we present DRGBCN, a novel computational method that integrates heterogeneous information through a deep bilinear attention network to infer potential drugs for specific diseases. DRGBCN involves constructing a comprehensive drug-disease network by incorporating multiple similarity networks for drugs and diseases. Firstly, we introduce a layer attention mechanism to effectively learn the embeddings of graph convolutional layers from these networks. Subsequently, a bilinear attention network is constructed to capture pairwise local interactions between drugs and diseases. This combined approach enhances the accuracy and reliability of predictions. Finally, a multi-layer perceptron module is employed to evaluate potential drugs. Through extensive experiments on three publicly available datasets, DRGBCN demonstrates better performance over baseline methods in 10-fold cross-validation, achieving an average area under the receiver operating characteristic curve (AUROC) of 0.9399. Furthermore, case studies on bladder cancer and acute lymphoblastic leukemia confirm the practical application of DRGBCN in real-world drug repositioning scenarios. Importantly, our experimental results from the drug-disease network analysis reveal the successful clustering of similar drugs within the same community, providing valuable insights into drug-disease interactions. In conclusion, DRGBCN holds significant promise for uncovering new therapeutic applications of existing drugs, thereby contributing to the advancement of precision medicine.
Collapse
|
11
|
Gangwal A, Lavecchia A. Artificial Intelligence in Natural Product Drug Discovery: Current Applications and Future Perspectives. J Med Chem 2025; 68:3948-3969. [PMID: 39916476 PMCID: PMC11874025 DOI: 10.1021/acs.jmedchem.4c01257] [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: 06/02/2024] [Revised: 12/01/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025]
Abstract
Drug discovery, a multifaceted process from compound identification to regulatory approval, historically plagued by inefficiencies and time lags due to limited data utilization, now faces urgent demands for accelerated lead compound identification. Innovations in biological data and computational chemistry have spurred a shift from trial-and-error methods to holistic approaches to medicinal chemistry. Computational techniques, particularly artificial intelligence (AI), notably machine learning (ML) and deep learning (DL), have revolutionized drug development, enhancing data analysis and predictive modeling. Natural products (NPs) have long served as rich sources of biologically active compounds, with many successful drugs originating from them. Advances in information science expanded NP-related databases, enabling deeper exploration with AI. Integrating AI into NP drug discovery promises accelerated discoveries, leveraging AI's analytical prowess, including generative AI for data synthesis. This perspective illuminates AI's current landscape in NP drug discovery, addressing strengths, limitations, and future trajectories to advance this vital research domain.
Collapse
Affiliation(s)
- Amit Gangwal
- Department
of Natural Product Chemistry, Shri Vile
Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, 424001 Maharashtra, India
| | - Antonio Lavecchia
- “Drug
Discovery” Laboratory, Department of Pharmacy, University of Naples Federico II, I-80131 Naples, Italy
| |
Collapse
|
12
|
Ambreen S, Umar M, Noor A, Jain H, Ali R. Advanced AI and ML frameworks for transforming drug discovery and optimization: With innovative insights in polypharmacology, drug repurposing, combination therapy and nanomedicine. Eur J Med Chem 2025; 284:117164. [PMID: 39721292 DOI: 10.1016/j.ejmech.2024.117164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024]
Abstract
Artificial Intelligence (AI) and Machine Learning (ML) are transforming drug discovery by overcoming traditional challenges like high costs, time-consuming, and frequent failures. AI-driven approaches streamline key phases, including target identification, lead optimization, de novo drug design, and drug repurposing. Frameworks such as deep neural networks (DNNs), convolutional neural networks (CNNs), and deep reinforcement learning (DRL) models have shown promise in identifying drug targets, optimizing delivery systems, and accelerating drug repurposing. Generative adversarial networks (GANs) and variational autoencoders (VAEs) aid de novo drug design by creating novel drug-like compounds with desired properties. Case studies, such as DDR1 kinase inhibitors designed using generative models and CDK20 inhibitors developed via structure-based methods, highlight AI's ability to produce highly specific therapeutics. Models like SNF-CVAE and DeepDR further advance drug repurposing by uncovering new therapeutic applications for existing drugs. Advanced ML algorithms enhance precision in predicting drug efficacy, toxicity, and ADME-Tox properties, reducing development costs and improving drug-target interactions. AI also supports polypharmacology by optimizing multi-target drug interactions and enhances combination therapy through predictions of drug synergies and antagonisms. In nanomedicine, AI models like CURATE.AI and the Hartung algorithm optimize personalized treatments by predicting toxicological risks and real-time dosing adjustments with high accuracy. Despite its potential, challenges like data quality, model interpretability, and ethical concerns must be addressed. High-quality datasets, transparent models, and unbiased algorithms are essential for reliable AI applications. As AI continues to evolve, it is poised to revolutionize drug discovery and personalized medicine, advancing therapeutic development and patient care.
Collapse
Affiliation(s)
- Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Aaisha Noor
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Himangini Jain
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
13
|
Lal JC, Fang MZ, Hussain M, Abraham A, Tonegawa-Kuji R, Hou Y, Chung MK, Collier P, Cheng F. Discovery of plasma proteins and metabolites associated with left ventricular cardiac dysfunction in pan-cancer patients. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:17. [PMID: 39948601 PMCID: PMC11823021 DOI: 10.1186/s40959-025-00309-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/23/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND Cancer-therapy related cardiac dysfunction (CTRCD) remains a significant cause of morbidity and mortality in cancer survivors. In this study, we aimed to identify differential plasma proteins and metabolites associated with left ventricular dysfunction (LVD) in cancer patients. METHODS We analyzed data from 50 patients referred to the Cleveland Clinic Cardio-Oncology Center for echocardiograph assessment, integrating electronic health records, proteomic, and metabolomic profiles. LVD was defined as an ejection fraction ≤ 55% based on echocardiographic evaluation. Classification-based machine learning models were used to predict LVD using plasma metabolites and proteins as input features. RESULTS We identified 13 plasma proteins (P < 0.05) and 14 plasma metabolites (P < 0.05) associated with LVD. Key proteins included markers of inflammation (ST2, TNFRSF14, OPN, and AXL) and chemotaxis (RARRES2, MMP-2, MEPE, and OPN). Notably, sex-specific associations were observed, such as uridine (P = 0.003) in males. Furthermore, metabolomic features significantly associated with LVD included 1-Methyl-4-imidazoleacetic acid (P = 0.015), COL1A1 (P = 0.009), and MMP-2 (P = 0.016), and pointing to metabolic shifts and heightened inflammation in patients with LVD. CONCLUSION Our findings suggest that circulating metabolites may non-invasively detect clinical and molecular differences in patients with LVD, providing insights into underlying disease pathways and potential therapeutic targets.
Collapse
Affiliation(s)
- Jessica C Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michelle Z Fang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Muzna Hussain
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Abel Abraham
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Reina Tonegawa-Kuji
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mina K Chung
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Patrick Collier
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department Of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart and Vascular Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
14
|
Yousaf MA, Michel M, Khan ATA, Noreen M, Bano S. Repurposing doxycycline for the inhibition of monkeypox virus DNA polymerase: a comprehensive computational study. In Silico Pharmacol 2025; 13:27. [PMID: 39958784 PMCID: PMC11825436 DOI: 10.1007/s40203-025-00307-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/17/2025] [Indexed: 02/18/2025] Open
Abstract
The global spread of monkeypox, caused by the double-stranded DNA monkeypox virus (MPXV), has underscored the urgent need for effective antiviral treatments. In this study, we aim to identify a potent inhibitor for MPXV DNA polymerase (DNAP), a critical enzyme in the virus replication process. Using a computational drug repurposing approach, we performed a virtual screening of 1615 FDA-approved drugs based on drug-likeness and molecular docking against DNAP. Among these, 1430 compounds met Lipinski's rule of five for drug-likeness, with Doxycycline emerging as the most promising competitive inhibitor, binding strongly to the DNAP active site with a binding affinity of - 9.3 kcal/mol. This interaction involved significant hydrogen bonds, electrostatic interactions, and hydrophobic contacts, with Doxycycline demonstrating a stronger affinity than established antivirals for smallpox, including Cidofovir, Brincidofovir, and Tecovirimat. Stability and flexibility analyses through a 200 ns molecular dynamics simulation and normal mode analysis confirmed the robustness of Doxycycline binding to DNAP. Overall, our results suggest Doxycycline as a promising candidate for monkeypox treatment, though additional experimental and clinical studies are needed to confirm its therapeutic potential and clinical utility. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-025-00307-7.
Collapse
Affiliation(s)
- Muhammad Abrar Yousaf
- Section of Biology and Genetics, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Maurice Michel
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Abeedha Tu-Allah Khan
- School of Biological Sciences, Faculty of Life-Sciences, University of the Punjab, Lahore, Pakistan
- Department of Biological Sciences, Faculty of Allied Health Sciences, Superior University, Lahore, Pakistan
| | - Misbah Noreen
- Department of Biological Sciences, Virtual University of Pakistan, Lahore, Pakistan
- Department of Wildlife and Ecology, University of Veterinary and Animal Sciences, Ravi Campus, Pattoki, Pakistan
| | - Saddia Bano
- Department of Biological Sciences, Virtual University of Pakistan, Lahore, Pakistan
| |
Collapse
|
15
|
Pal S, Nance KD, Joshi DR, Kales SC, Ye L, Hu X, Shamim K, Zakharov AV. Applications of Machine Learning Approaches for the Discovery of SARS-CoV-2 PLpro Inhibitors. J Chem Inf Model 2025; 65:1338-1356. [PMID: 39818814 DOI: 10.1021/acs.jcim.4c02126] [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: 01/19/2025]
Abstract
The global impact of SARS-CoV-2 highlights the need for treatments beyond vaccination, given the limited availability of effective medications. While Pfizer introduced Paxlovid, an FDA-approved antiviral targeting the SARS-CoV-2 main protease (Mpro), this study focuses on designing new antivirals against another protease, papain-like protease (PLpro), which is crucial for viral replication and immune suppression. NCATS/NIH performed a high-throughput screen of ∼15,000 molecules from an internal molecular library, identifying initial hits with a 0.5% success rate. To improve the hit rate and identify potent inhibitors, machine learning-based virtual screens were applied to ∼150,000 compounds, yielding 125 top predicted hits. Biochemical evaluation revealed 25 promising compounds, with a 20% hit-rate and IC50 values from 1.75 μM to <36 μM across 13 chemotypes. Further analog screening of those chemotypes, as part of the structure-activity relationships, led to 20 additional hits. Additionally, the hit-to-lead optimization of chemotype 7 produced 10 more analogs. These PLpro inhibitors provide promising templates for antiviral development against COVID-19.
Collapse
Affiliation(s)
- Sourav Pal
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| | - Kellie D Nance
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| | - Dirgha Raj Joshi
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| | - Stephen C Kales
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| | - Lin Ye
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| | - Xin Hu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| | - Khalida Shamim
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| | - Alexey V Zakharov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
16
|
Sekaran R, Munnangi AK, Ramachandran M, Khishe M. Cayley-Purser secured communication and jackknife correlative classification for COVID patient data analysis. Sci Rep 2025; 15:4666. [PMID: 39920299 PMCID: PMC11806013 DOI: 10.1038/s41598-025-88105-1] [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: 09/04/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
Internet of Medical Things (IoMT) is a group of medical devices that connect the healthcare information technology to minimize the redundant hospital visit and healthcare system troubles. IoMT connect the patients to the doctor and transmit the medical data over the network. The spread of corona virus has put the people at high risk. Due to increasing number of cases and its stress on health professionals, IoMT technology is used in many healthcare centers. But, the security level and data classification accuracy was not improved by existing methods during the data communication. In order to solve these issues, Cayley-Purser Cryptographic Secured Communication based Jackknife Correlative Data Classification (CPCSC-JCDC) method is designed. The key objective of CPCSC-JCDC method is to collect the patient information through IoMT devices and send to the doctor in more secured manner. Initially in CPCSC-JCDC method, the patient data is collected. After the data collection process, the data gets encrypted with help of public key of the patient by using cayley-purser cryptosystem. After the encryption process, the data is sent to the doctor. The doctor receives and decrypts the patient data by using their private key. After decryption process, the doctor analyses the patient data and classifies the data as emergency case or normal case by using jackknife correlation function. This helps to minimize the patient readmission rate and increase the patient satisfaction level. Experimental evaluation is carried out by Novel Corona Virus 2019 dataset using different metrics like data classification accuracy, data classification time and security level. The evaluation result shows that CPCSC-JCDC method improves the security level as well as accuracy and minimizes the time consumption during data communication than existing works.
Collapse
Affiliation(s)
- Ramesh Sekaran
- Department of Computer Science and Engineering, JAIN (Deemed-to-be University), Bangalore, Karnataka, 562112, India
| | - Ashok Kumar Munnangi
- Department of Information Technology, Velagapudi Ramakrishna Siddhartha Engineering College (Autonomous), Vijayawada, Andhra Pradesh, India
| | | | - Mohammad Khishe
- Applied Science Research Center, Applied Science Private University, Amman, Jordan.
- Jadara University Research Center, Jadara University, Irbid, Jordan.
- Department of Electrical Engineering, Imam Khomeini Naval Science University, Nowshahr, Iran.
| |
Collapse
|
17
|
Cao Y, Han J, Xiao Y, Wang Z, Zhang H, Fang R, Li J, Dong M, Chen R, Zhu G, Han J, Sun L. Xiao-Er-Kang-Du capsules regulate autophagy against the influenza B virus (Victoria strain) through the mTOR/ULK1/Beclin1/VPS34 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118872. [PMID: 39366496 DOI: 10.1016/j.jep.2024.118872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiao-er-kang-du (XEKD) capsule is a Chinese herbal formula used for treatment of upper respiratory tract infection caused by various viruses in pediatric patients in China. XEKD is used clinically for the treatment of influenza-like symptoms, including fever, chills, cough, stuffy and runny nose, headache, and sore throat, with remarkable efficacy. However, the pharmacologic mechanism of XEKD against influenza B virus (IBV) infection is unclear. AIM OF THE STUDY The main purpose of the present work is to explore the curative effect as well as possible mechanisms of XEKD against influenza B virus (IBV) (Victoria strain). MATERIALS AND METHODS Both in vivo and in vitro experiments were performed to confirm the antiviral properties of XEKD. High-performance liquid chromatography was used to analyze the active components and assess the stability of XEKD. In addition, the mechanism of action of XEKD against IBV (Victoria) was investigated by western blot, immunofluorescence, and immunohistochemical analyses, in addition to confocal fluorescence microscopy. RESULTS The findings revealed that XEKD demonstrated antiviral effects against IBV infection in both in vivo and in vitro via the mTOR/ULK1/Beclin1/VPS34 pathway and promote cellular autophagy to mitigate IBV-induced lung tissue damage. The results of this work are expected to lead to a deeper understanding of the mechanism underlying the effect of the XEKD capsule against IBV infections. CONCLUSIONS IBV infection was found to inhibit autophagy, which exacerbated inflammatory damage. XEKD regulates autophagy through the mTOR/ULK1/Beclin1/VPS34 pathway and exerts antiviral effects, thereby laying a foundation for further development of XEKD as a potential therapeutic against IBV infection.
Collapse
Affiliation(s)
- Yan Cao
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; College of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Jing Han
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China
| | - Yan Xiao
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Zhongtian Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China
| | - Haiyang Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China
| | - Ruikang Fang
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Jingjing Li
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Meiwen Dong
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Rui Chen
- College of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| | - Guangze Zhu
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| | - Jicheng Han
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| | - Liping Sun
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China.
| |
Collapse
|
18
|
Zong W, Tian S, Niu Q, Li X, Wang P, Tong L, Zhang S, Zheng D, Zhang Y, Xiong W, Cai Q, Zeng Z, Wang J, Xu H, Zhang H, Li B. Comparable clinical advantages identification of three formulae on rheumatic disease using a modular-based network proximity approach. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118764. [PMID: 39218127 DOI: 10.1016/j.jep.2024.118764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/09/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herbal formulae have been used in China for thousands of years but have unclear clinical positioning and unknown characteristic indications make it difficult to determine their specific application in various diseases, which seriously hamper their clinical value. Identifying the precise clinical positioning and clinical advantages of similar formulae for related diseases is a critical issue. AIM OF THIS STUDY To develop a methodology based on modular pharmacology to determine the clinical advantages and precise clinical position of similar formulae. MATERIALS AND METHODS In this study, we proposed a modular-based network proximity approach to explore drug repositioning and clinical advantages of three formulae, Shirebi tablets (SRB), Yuxuebi capsules (YXB), and Wangbifukang granules (WBFK), for rheumatic disease. First, we constructed a rheumatology target network, and modules were obtained using the cluster tool molecular complex detection (MCODE). Based on the modular interaction map established by a quantitative approach for inter-module coordination and its transition (IMCC), using a targeting rate (TR) matrix to identify targeted modules of three formulae. Moreover, the network proximity Z-score and Jaccard similarity coefficient were used to identify potential optimal symptomatic indications and related diseases using three formulae. At the same time, the driver genes for SRB and gouty arthritis were identified by flow centrality and shortest distance, and the epresentative driver genes were validated by in vivo experiments. RESULTS 32 modules were obtained using the MCODE method. 4, 4, and 14 characteristic targeted modules of SRB, YXB, and WBFK, respectively, were identified using a targeting rate (TR) matrix. Module 2, 16, and 19 were targeted by both SRB and WBFK. The common effects of SRB and WBFK focused on inflammatory response and innate immune response, YXB was found to be involved in the collagen catabolic process, transmembrane receptor protein serine/threonine kinase signaling pathway. Moreover, potential optimal symptomatic indications and representative related diseases were identified for three formulae: SRB was significantly associated with GA (Z = -20.26); YXB was significantly associated with AS (Z = -4.532), MI (Z = -29.11), RhFv (Z = -6.945), OA (Z = -39.97), and GA (Z = -13.03); and WBFK was significantly associated with MI (Z = -205.5), SLE (Z = -37.65), RhFv (Z = -42.45), and GA (Z = -17.24). Finally, 8 driver genes for SRB and gouty arthritis were identified,the representative driver genes TRAF6 and NFE2L1 were validated by in vivo experiments. CONCLUSIONS The modular-based network proximity approach proposed in this study may provide a new perspective for the precise drug repositioning and clinical advantages of similar formulae in disease treatment.
Collapse
Affiliation(s)
- Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Siwei Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qikai Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xin Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Pengqian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lin Tong
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Siqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Danping Zheng
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanqiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wei Xiong
- Peking Union Medical College Hospital, Beijing, 100005, China
| | - Qiujie Cai
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ziling Zeng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jing'ai Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Huamin Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
19
|
Yang Y, Cheng F. Artificial intelligence streamlines scientific discovery of drug-target interactions. Br J Pharmacol 2025. [PMID: 39843168 DOI: 10.1111/bph.17427] [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: 07/22/2024] [Revised: 10/04/2024] [Accepted: 11/01/2024] [Indexed: 01/24/2025] Open
Abstract
Drug discovery is a complicated process through which new therapeutics are identified to prevent and treat specific diseases. Identification of drug-target interactions (DTIs) stands as a pivotal aspect within the realm of drug discovery and development. The traditional process of drug discovery, especially identification of DTIs, is marked by its high costs of experimental assays and low success rates. Computational methods have emerged as indispensable tools, especially those employing artificial intelligence (AI) methods, which could streamline the process, thereby reducing costs and time consumption and potentially increasing success rates. In this review, we focus on the application of AI techniques in DTI prediction. Specifically, we commence with a comprehensive overview of drug discovery and development, along with systematic prediction and validation of DTIs. We proceed to highlight the prominent databases and toolkits used in developing AI methods for DTI prediction, as well as with methodologies for evaluating their efficacy. We further extend the exploration into three primary types of state-of-the-art AI methods used in DTI prediction, including classical machine learning, deep learning and network-based methods. Finally, we summarize the key findings and outline the current challenges and future directions that AI methods face in scientific drug discovery and development.
Collapse
Affiliation(s)
- Yuxin Yang
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
20
|
Tian S, Xu M, Geng X, Fang J, Xu H, Xue X, Hu H, Zhang Q, Yu D, Guo M, Zhang H, Lu J, Guo C, Wang Q, Liu S, Zhang W. Network Medicine-Based Strategy Identifies Maprotiline as a Repurposable Drug by Inhibiting PD-L1 Expression via Targeting SPOP in Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410285. [PMID: 39499771 PMCID: PMC11714211 DOI: 10.1002/advs.202410285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Indexed: 11/07/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are drugs that inhibit immune checkpoint (ICP) molecules to restore the antitumor activity of immune cells and eliminate tumor cells. Due to the limitations and certain side effects of current ICIs, such as programmed death protein-1, programmed cell death-ligand 1, and cytotoxic T lymphocyte-associated antigen 4 (CTLA4) antibodies, there is an urgent need to find new drugs with ICP inhibitory effects. In this study, a network-based computational framework called multi-network algorithm-driven drug repositioning targeting ICP (Mnet-DRI) is developed to accurately repurpose novel ICIs from ≈3000 Food and Drug Administration-approved or investigational drugs. By applying Mnet-DRI to PD-L1, maprotiline (MAP), an antidepressant drug is repurposed, as a potential PD-L1 modifier for colorectal and lung cancers. Experimental validation revealed that MAP reduced PD-L1 expression by targeting E3 ubiquitin ligase speckle-type zinc finger structural protein (SPOP), and the combination of MAP and anti-CTLA4 in vivo significantly enhanced the antitumor effect, providing a new alternative for the clinical treatment of colorectal and lung cancer.
Collapse
Affiliation(s)
- Saisai Tian
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Mengting Xu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Xiangxin Geng
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Jiansong Fang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Hanchen Xu
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghai200032China
| | - Xinying Xue
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijing100038China
| | - Hongmei Hu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Qing Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Dianping Yu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Mengmeng Guo
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Jinyuan Lu
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Chengyang Guo
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Weidong Zhang
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsInstitute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100193China
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosafetyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| |
Collapse
|
21
|
Xu J, Song W, Xu Z, Danziger MM, Karavani E, Zang C, Chen X, Li Y, Paz IMR, Gohel D, Su C, Zhou Y, Hou Y, Shimoni Y, Pieper AA, Hu J, Wang F, Rosen‐Zvi M, Leverenz JB, Cummings J, Cheng F. Single-microglia transcriptomic transition network-based prediction and real-world patient data validation identifies ketorolac as a repurposable drug for Alzheimer's disease. Alzheimers Dement 2025; 21:e14373. [PMID: 39641322 PMCID: PMC11782846 DOI: 10.1002/alz.14373] [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: 04/15/2024] [Revised: 09/10/2024] [Accepted: 10/02/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION High microglial heterogeneities hinder the development of microglia-targeted treatment for Alzheimer's disease (AD). METHODS We integrated 0.7 million single-nuclei RNA-sequencing transcriptomes from human brains using a variational autoencoder. We predicted AD-relevant microglial subtype-specific transition networks for disease-associated microglia (DAM), tau microglia, and neuroinflammation-like microglia (NIM). We prioritized drugs by specifically targeting microglia-specific transition networks and validated drugs using two independent real-world patient databases. RESULTS We identified putative AD molecular drivers (e.g., SYK, CTSB, and INPP5D) in transition networks of DAM and NIM. Via specifically targeting NIM, we identified that usage of ketorolac was associated with reduced AD incidence in both MarketScan (hazard ratio [HR] = 0.89) and INSIGHT (HR = 0.83) Clinical Research Network databases, mechanistically supported by ketorolac-treated transcriptomic data from AD patient induced pluripotent stem cell-derived microglia. DISCUSSION This study offers insights into the pathobiology of AD-relevant microglial subtypes and identifies ketorolac as a potential anti-inflammatory treatment for AD. HIGHLIGHTS An integrative analysis of ≈ 0.7 million single-nuclei RNA-sequencing transcriptomes from human brains identified Alzheimer's disease (AD)-relevant microglia subtypes. Network-based analysis identified putative molecular drivers (e.g., SYK, CTSB, INPP5D) of transition networks between disease-associated microglia (DAM) and neuroinflammation-like microglia (NIM). Via network-based prediction and population-based validation, we identified that usage of ketorolac (a US Food and Drug Administration-approved anti-inflammatory medicine) was associated with reduced AD incidence in two independent patient databases. Mechanistic observation showed that ketorolac treatment downregulated the Type-I interferon signaling in patient induced pluripotent stem cell-derived microglia, mechanistically supporting its protective effects in real-world patient databases.
Collapse
Affiliation(s)
- Jielin Xu
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Wenqiang Song
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Zhenxing Xu
- Department of Population Health SciencesWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
- Institute of Artificial Intelligence for Digital HealthWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
| | - Michael M. Danziger
- AI for Accelerated Healthcare and Life Sciences DiscoveryIBM Research‐IsraelHaifaIsrael
| | - Ehud Karavani
- AI for Accelerated Healthcare and Life Sciences DiscoveryIBM Research‐IsraelHaifaIsrael
| | - Chengxi Zang
- Department of Population Health SciencesWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
- Institute of Artificial Intelligence for Digital HealthWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
| | - Xin Chen
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Yichen Li
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Isabela M Rivera Paz
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Dhruv Gohel
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Chang Su
- Department of Population Health SciencesWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
- Institute of Artificial Intelligence for Digital HealthWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
| | - Yadi Zhou
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Yuan Hou
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Yishai Shimoni
- AI for Accelerated Healthcare and Life Sciences DiscoveryIBM Research‐IsraelHaifaIsrael
| | - Andrew A. Pieper
- Brain Health Medicines Center, Harrington Discovery InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Department of PsychiatryCase Western Reserve UniversityClevelandOhioUSA
- Geriatric PsychiatryGRECCLouis Stokes Cleveland VA Medical CenterClevelandOhioUSA
- Institute for Transformative Molecular MedicineSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of NeurosciencesCase Western Reserve UniversitySchool of MedicineClevelandOhioUSA
| | - Jianying Hu
- IBM T.J. Watson Research CenterYorktown HeightsNew YorkUSA
| | - Fei Wang
- Department of Population Health SciencesWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
- Institute of Artificial Intelligence for Digital HealthWeill Cornell MedicineCornell UniversityNew YorkNew YorkUSA
| | - Michal Rosen‐Zvi
- AI for Accelerated Healthcare and Life Sciences DiscoveryIBM Research‐IsraelHaifaIsrael
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthNeurological InstituteCleveland ClinicClevelandOhioUSA
| | - Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada Las VegasLas VegasNevadaUSA
| | - Feixiong Cheng
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
22
|
Zuo Z, Wen R, Jing S, Chen X, Liu R, Xue J, Zhang L, Li Q. Ganoderma lucidum (Curtis) P. Karst. Immunomodulatory Protein Has the Potential to Improve the Prognosis of Breast Cancer Through the Regulation of Key Prognosis-Related Genes. Pharmaceuticals (Basel) 2024; 17:1695. [PMID: 39770537 PMCID: PMC11677753 DOI: 10.3390/ph17121695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Breast cancer in women is the most commonly diagnosed and most malignant tumor. Although luminal A breast cancer (LumA) has a relatively better prognosis, it still has a persistent pattern of recurrence. Ganoderma lucidum (Curtis) P. Karst. is a kind of traditional Chinese medicine and has antitumor effects. In this study, we aimed to identify the genes relevant to prognosis, find novel targets, and investigate the function of the bioactive protein from G. lucidum, called FIP-glu, in improving prognosis. Methods: Gene expression data and clinical information of LumA breast cancer patients were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Using bioinformatics methods, a predictive risk model was constructed to predict the prognosis for each patient. The cell counting kit-8 (CCK8) and clone formation assays were used to validate gene function. The ability of FIP-glu to regulate RNA levels of risk genes was validated. Results: Six risk genes (slit-roundabout GTPase-activating protein 2 (SRGAP2), solute carrier family 35 member 2 (SLC35A2), sequence similarity 114 member A1 (FAM114A1), tumor protein P53-inducible protein 11 (TP53I11), transmembrane protein 63C (TMEM63C), and polymeric immunoglobulin receptor (PIGR)) were identified, and a prognostic model was constructed. The prognosis was worse in the high-risk group and better in the low-risk group. The receiver operating characteristic (ROC) curve confirmed the model's accuracy. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that the differentially expressed genes (DEGs) between the high- and low-risk groups were significantly enriched in the immune responses. TMEM63C could promote tumor viability, growth, and proliferation in vitro. FIP-glu significantly regulated these risk genes, and attenuated the promoting effect of TMEM63C in breast cancer cells. Conclusions: SRGAP2, SLC35A2, FAM114A1, TP53I11, TMEM63C, and PIGR were identified as the potential risk genes for predicting the prognosis of patients. TMEM63C could be a potential novel therapeutic target. Moreover, FIP-glu was a promising drug for improving the prognosis of LumA breast cancer.
Collapse
Affiliation(s)
- Zanwen Zuo
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Ruihua Wen
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Shuang Jing
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Xianghui Chen
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Ruisang Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life Science and Health Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Jianping Xue
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Lei Zhang
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Qizhang Li
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| |
Collapse
|
23
|
Ellison ST, Hayman I, Derr K, Derr P, Frebert S, Itkin Z, Shen M, Jones A, Olson W, Corey L, Wald A, Johnston C, Fong Y, Ferrer M, Zhu J. Identification of potent HSV antivirals using 3D bioprinted human skin equivalents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626896. [PMID: 39713402 PMCID: PMC11661117 DOI: 10.1101/2024.12.04.626896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Herpes simplex virus (HSV) infection has worldwide public health concerns and lifelong medical impacts. The standard therapy, acyclovir, has limited efficacy in preventing HSV subclinical virus shedding, and drug resistance occurs in immunocompromised patients, highlighting the need for novel therapeutics. HSV infection manifests in the skin epidermal layer, but current drug discovery utilizes Vero cells and fibroblasts monolayer cultures, capturing neither in vivo relevance nor tissue environment. To bridge the gap, we established 3D bioprinted human skin equivalents that recapitulate skin architecture in a 96-well plate format amenable for antiviral screening and preclinical testing. Screening a library of 738 compounds with broad targets and mechanisms of action, we identified potent antivirals, including most of the known anti-HSV compounds, validating the translational relevance of our assay. Acyclovir was dramatically less potent for inhibiting HSV in keratinocytes compared to donor-matched fibroblasts. In contrast, antivirals against HSV helicase/primase or host replication pathways displayed similar potency across cell types and donor sources in 2D and 3D models. The reduced potency of acyclovir in keratinocytes, the primary cell type encountered by HSV reactivation, helps explain the limited benefit acyclovir and its congeners play in reducing sexual transmission. Finally, we demonstrated that our 3D bioprinted skin platform can integrate patient-derived cells, facilitating the incorporation of variable genetic backgrounds early into drug testing. Thus, these data indicate that the 3D bioprinted human skin equivalent assay platform provides a more physiologically relevant approach to identifying potential antivirals for HSV-directed drug development.
Collapse
Affiliation(s)
- S. Tori Ellison
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health; Rockville, Maryland 20850, USA
| | - Ian Hayman
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA 98195, USA
| | - Kristy Derr
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health; Rockville, Maryland 20850, USA
| | - Paige Derr
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health; Rockville, Maryland 20850, USA
| | - Shayne Frebert
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health; Rockville, Maryland 20850, USA
| | - Zina Itkin
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health; Rockville, Maryland 20850, USA
| | - Min Shen
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health; Rockville, Maryland 20850, USA
| | - Anthony Jones
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA 98195, USA
| | - Wendy Olson
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA 98195, USA
| | - Lawrence Corey
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA 98195, USA
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center; Seattle, WA 98109, USA
| | - Anna Wald
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA 98195, USA
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center; Seattle, WA 98109, USA
- Department of Medicine, University of Washington; Seattle, WA 98195, USA
- Department of Global Health, University of Washington; Seattle, WA 98195, USA
| | - Christine Johnston
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA 98195, USA
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center; Seattle, WA 98109, USA
- Department of Medicine, University of Washington; Seattle, WA 98195, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center; Seattle, WA 98109, USA
| | - Marc Ferrer
- Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health; Rockville, Maryland 20850, USA
| | - Jia Zhu
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA 98195, USA
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center; Seattle, WA 98109, USA
| |
Collapse
|
24
|
Ahmed F, Samantasinghar A, Ali W, Choi KH. Network-based drug repurposing identifies small molecule drugs as immune checkpoint inhibitors for endometrial cancer. Mol Divers 2024; 28:3879-3895. [PMID: 38227161 DOI: 10.1007/s11030-023-10784-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/25/2023] [Indexed: 01/17/2024]
Abstract
Endometrial cancer (EC) is the 6th most common cancer in women around the world. Alone in the United States (US), 66,200 new cases and 13,030 deaths are expected to occur in 2023 which needs the rapid development of potential therapies against EC. Here, a network-based drug-repurposing strategy is developed which led to the identification of 16 FDA-approved drugs potentially repurposable for EC as potential immune checkpoint inhibitors (ICIs). A network of EC-associated immune checkpoint proteins (ICPs)-induced protein interactions (P-ICP) was constructed. As a result of network analysis of P-ICP, top key target genes closely interacting with ICPs were shortlisted followed by network proximity analysis in drug-target interaction (DTI) network and pathway cross-examination which identified 115 distinct pathways of approved drugs as potential immune checkpoint inhibitors. The presented approach predicted 16 drugs to target EC-associated ICPs-induced pathways, three of which have already been used for EC and six of them possess immunomodulatory properties providing evidence of the validity of the strategy. Classification of the predicted pathways indicated that 15 drugs can be divided into two distinct pathway groups, containing 17 immune pathways and 98 metabolic pathways. In addition, drug-drug correlation analysis provided insight into finding useful drug combinations. This fair and verified analysis creates new opportunities for the quick repurposing of FDA-approved medications in clinical trials.
Collapse
Affiliation(s)
- Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| | - Anupama Samantasinghar
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| | - Wajid Ali
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Jeju, Republic of Korea.
| |
Collapse
|
25
|
Huanbutta K, Burapapadh K, Kraisit P, Sriamornsak P, Ganokratanaa T, Suwanpitak K, Sangnim T. Artificial intelligence-driven pharmaceutical industry: A paradigm shift in drug discovery, formulation development, manufacturing, quality control, and post-market surveillance. Eur J Pharm Sci 2024; 203:106938. [PMID: 39419129 DOI: 10.1016/j.ejps.2024.106938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
The advent of artificial intelligence (AI) has catalyzed a profound transformation in the pharmaceutical industry, ushering in a paradigm shift across various domains, including drug discovery, formulation development, manufacturing, quality control, and post-market surveillance. This comprehensive review examines the multifaceted impact of AI-driven technologies on all stages of the pharmaceutical life cycle. It discusses the application of machine learning algorithms, data analytics, and predictive modeling to accelerate drug discovery processes, optimize formulation development, enhance manufacturing efficiency, ensure stringent quality control measures, and revolutionize post-market surveillance methodologies. By describing the advancements, challenges, and future prospects of harnessing AI in the pharmaceutical landscape, this review offers valuable insights into the evolving dynamics of drug development and regulatory practices in the era of AI-driven innovation.
Collapse
Affiliation(s)
- Kampanart Huanbutta
- Department of Manufacturing Pharmacy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Kanokporn Burapapadh
- Department of Manufacturing Pharmacy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Pakorn Kraisit
- Thammasat University Research Unit in Smart Materials and Innovative Technology for Pharmaceutical Applications (SMIT-Pharm), Faculty of Pharmacy, Thammasat University, Pathumthani 12120, Thailand
| | - Pornsak Sriamornsak
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand; Academy of Science, The Royal Society of Thailand, Bangkok, 10300, Thailand
| | - Thittaporn Ganokratanaa
- Applied Computer Science Program, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| | - Kittipat Suwanpitak
- Faculty of Pharmaceutical Sciences, Burapha University, 169, Seansook, Muang, Chonburi, 20131, Thailand
| | - Tanikan Sangnim
- Faculty of Pharmaceutical Sciences, Burapha University, 169, Seansook, Muang, Chonburi, 20131, Thailand.
| |
Collapse
|
26
|
Barghash RF, Gemmati D, Awad AM, Elbakry MMM, Tisato V, Awad K, Singh AV. Navigating the COVID-19 Therapeutic Landscape: Unveiling Novel Perspectives on FDA-Approved Medications, Vaccination Targets, and Emerging Novel Strategies. Molecules 2024; 29:5564. [PMID: 39683724 PMCID: PMC11643501 DOI: 10.3390/molecules29235564] [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: 09/26/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Amidst the ongoing global challenge of the SARS-CoV-2 pandemic, the quest for effective antiviral medications remains paramount. This comprehensive review delves into the dynamic landscape of FDA-approved medications repurposed for COVID-19, categorized as antiviral and non-antiviral agents. Our focus extends beyond conventional narratives, encompassing vaccination targets, repurposing efficacy, clinical studies, innovative treatment modalities, and future outlooks. Unveiling the genomic intricacies of SARS-CoV-2 variants, including the WHO-designated Omicron variant, we explore diverse antiviral categories such as fusion inhibitors, protease inhibitors, transcription inhibitors, neuraminidase inhibitors, nucleoside reverse transcriptase, and non-antiviral interventions like importin α/β1-mediated nuclear import inhibitors, neutralizing antibodies, and convalescent plasma. Notably, Molnupiravir emerges as a pivotal player, now licensed in the UK. This review offers a fresh perspective on the historical evolution of COVID-19 therapeutics, from repurposing endeavors to the latest developments in oral anti-SARS-CoV-2 treatments, ushering in a new era of hope in the battle against the pandemic.
Collapse
Affiliation(s)
- Reham F. Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Cairo 12622, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ahmed M. Awad
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Mustafa M. M. Elbakry
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Veronica Tisato
- Centre Hemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Kareem Awad
- Institute of Pharmaceutical and Drug Industries Research, National Research Center, Dokki, Cairo 12622, Egypt;
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| |
Collapse
|
27
|
Li Y, You ZH, Yuan Y, Mi CG, Huang YA, Yi HC, Hou LX. Integrated Knowledge Graph and Drug Molecular Graph Fusion via Adversarial Networks for Drug-Drug Interaction Prediction. J Chem Inf Model 2024; 64:8361-8372. [PMID: 39475566 DOI: 10.1021/acs.jcim.4c01647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
The Co-administration of multiple drugs can enhance the efficacy of disease treatment by reducing drug resistance and side effects. However, it also raises the risk of adverse drug interactions, presenting a challenging problem in healthcare. Various approaches have been developed to predict drug-drug interactions (DDIs) by leveraging both knowledge graphs and drug attribute information. While these methods have shown promise, they often fail to effectively capture correlations between biomedical information in the knowledge graph and drug properties. This work introduces a novel end-to-end DDI predictor framework based on generative adversarial networks. This framework utilizes a message-passing neural network to capture molecular structure information while employing the knowledge-aware graph attention network to capture the representation of drugs in the knowledge graph through considering the importance of different multihop neighbor nodes and relationships. The dual generative adversarial networks employ two generators and two discriminators in knowledge graph embedding and molecular topology embedding for adversarial training to capture the interrelations and complementary knowledge between molecular structure information and semantic information from the knowledge graph. comprehensive experiments have demonstrated that the proposed method outperforms state-of-the-art algorithms in binary classification, with improvements of 1.0% in accuracy, 0.45% in area under the receiver operating characteristic curve (AUC), 0.24% in area under the precision-recall curve (AUPR), and 0.98% in F1 score. Furthermore, for multiclass classification tasks, improvements were observed across various evaluation metrics, including 0.9% in accuracy, 0.25% in macro precision, 0.2% in macro recall, and 0.28% in macro F1. Additionally, ablation studies were conducted to showcase the effectiveness and robustness of our method in DDI prediction tasks.
Collapse
Affiliation(s)
- Yu Li
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Yang Yuan
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou213164, China
| | - Cheng-Gang Mi
- Foreign Language and Literature Institute, Xi'an International Studies University, Xi'an710129, China
| | - Yu-An Huang
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Hai-Cheng Yi
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Lin-Xuan Hou
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| |
Collapse
|
28
|
Zheng L, Zhang Q, Luo P, Shi F, Zhang Y, He X, An Y, Cheng G, Pan X, Li Z, Zhou B, Wang J. Chemical Proteomics Approaches for Screening Small Molecule Inhibitors Covalently Binding to SARS-Cov-2. Adv Biol (Weinh) 2024; 8:e2300612. [PMID: 39410782 DOI: 10.1002/adbi.202300612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/01/2024] [Indexed: 11/13/2024]
Abstract
Although various strategies have been used to prevent and treat SARS-CoV-2, the spread and evolution of SARS-CoV-2 is still progressing rapidly. The emerging variants Omicron and its sublineage have a greater ability to spread and escape nearly all current monoclonal antibodies treatments, highlighting an urgent need to develop therapeutics targeting current and emerging Omicron variants or recombinants with breadth and potency. Here, some small molecule drugs are rapidly identified that could covalently binding to receptor binding domain (RBD) protein of Omicron through the combined application of artificial intelligence (AI) and activity-based protein profiling (ABPP) technology. The surface plasmon resonance (SPR) and pseudo-virus neutralization experiments further reveal that an FDA-approved drug gallic acid has robust neutralization potency against Omicron pseudo-virus with the IC50 values of 23.56 × 10-6 m. Taken together, a platform combining AI intelligence, biochemical, SPR, molecular docking, and pseudo-virus-based screening for rapid identification and evaluation of potential anti-SARS-CoV-2 small molecule drugs is established and the effectiveness of the platform is validated.
Collapse
Affiliation(s)
- Liuhai Zheng
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Qian Zhang
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Piao Luo
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fei Shi
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Ying Zhang
- State Key Laboratory for Quality Esurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaoxue He
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430207, China
| | - Yehai An
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guangqing Cheng
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Xiaoyan Pan
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Zhijie Li
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Boping Zhou
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Jigang Wang
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory for Quality Esurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China
| |
Collapse
|
29
|
Rabaan AA, Alfaresi M, Alrasheed HA, Al Kaabi NA, Abduljabbar WA, Al Fares MA, Al-Subaie MF, Alissa M. Network-Based Drug Repurposing and Genomic Analysis to Unveil Potential Therapeutics for Monkeypox Virus. Chem Biodivers 2024; 21:e202400895. [PMID: 39082609 DOI: 10.1002/cbdv.202400895] [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: 04/09/2024] [Accepted: 07/22/2024] [Indexed: 10/10/2024]
Abstract
The emergence of the human monkeypox virus (MPXV) and the lack of effective medications have necessitated the exploration of various strategies to combat its infection. This study employs a network-based approach to drug discovery, utilizing the BLASTn and phylogenetic analysis to compare the MPXV genome with those of 18 related orthopoxviruses, revealing over 75 % genomic similarity. Through a literature review, 160 human-host proteins linked to MPXV and its relatives were identified, leading to the construction of a human-host protein interactome. Analysis of this interactome highlighted 39 central hub proteins, which were then examined for potential drug targets. The process successfully revealed 15 targets already approved for use with medications. Additionally, the functional enrichment analysis provided insights into potential pathways and disorders connected with these targets. Four medications, namely Baricitinib, Infliximab, Adalimumab, and Etanercept, have been identified as potential candidates for repurposing to combat MPXV. In addition, the pharmacophore-based screening identified a molecule that is comparable to Baricitinib and has the potential to be effective against MPXV. The findings of the study suggest that ZINC22060520 is a promising medication for treating MPXV infection and proposes these medications as potential options for additional experimental and clinical assessment in the battle against MPXV.
Collapse
Affiliation(s)
- Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan
| | - Mubarak Alfaresi
- Department of Microbiology, National Reference laboratory, Cleveland Clinic Abu Dhabi, Abu Dhabi, 92323, United Arab Emirates
- Department of Pathology, College of Medicine, Mohammed Bin Rashid, University of Medicine and Health Sciences, Dubai, 505055, United Arab Emirates
| | - Hayam A Alrasheed
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Nawal A Al Kaabi
- College of Medicine and Health Science, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
- Sheikh Khalifa Medical City, Abu Dhabi Health Services Company (SEHA), Abu Dhabi, 51900, United Arab Emirates
| | - Wesam A Abduljabbar
- Department of Medical Laboratory Sciences, Fakeeh College for Medical Science, Jeddah, 21134, Saudi Arabia
| | - Mona A Al Fares
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah, 21589, Saudi Arabia
| | - Maha F Al-Subaie
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh, 13328, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| |
Collapse
|
30
|
Wang J, Wang X, Pang Y. StructNet-DDI: Molecular Structure Characterization-Based ResNet for Prediction of Drug-Drug Interactions. Molecules 2024; 29:4829. [PMID: 39459198 PMCID: PMC11510539 DOI: 10.3390/molecules29204829] [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: 09/05/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
This study introduces a deep learning framework based on SMILES representations of chemical structures to predict drug-drug interactions (DDIs). The model extracts Morgan fingerprints and key molecular descriptors, transforming them into raw graphical features for input into a modified ResNet18 architecture. The deep residual network, enhanced with regularization techniques, efficiently addresses training issues such as gradient vanishing and exploding, resulting in superior predictive performance. Experimental results show that StructNet-DDI achieved an AUC of 99.7%, an accuracy of 94.4%, and an AUPR of 99.9%, demonstrating the model's effectiveness and reliability. These findings highlight that StructNet-DDI can effectively extract crucial features from molecular structures, offering a simple yet robust tool for DDI prediction.
Collapse
Affiliation(s)
- Jihong Wang
- School of Computer, Guangdong University of Education, Guangzhou 510310, China
| | - Xiaodan Wang
- School of Pharmaceutical Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Yuyao Pang
- School of Pharmaceutical Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| |
Collapse
|
31
|
Chen X, Zhou B, Jiang X, Zhong H, You A, Zou T, Zhou C, Liu X, Zhang Y. Drug repurposing to tackle parainfluenza 3 based on multi-similarities and network proximity analysis. Front Pharmacol 2024; 15:1428925. [PMID: 39411066 PMCID: PMC11473393 DOI: 10.3389/fphar.2024.1428925] [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: 05/07/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Given that there is currently no clinically approved drug or vaccine for parainfluenza 3 (PIV3), we applied a drug repurposing method based on disease similarity and chemical similarity to screen 2,585 clinically approved chemical drugs using PIV3 potential drugs BCX-2798 and zanamivir as our controls. Twelve candidate drugs were obtained after being screened with good disease similarity and chemical similarity (S > 0.50, T > 0.56). When docking them with the PIV3 target protein, hemagglutinin-neuraminidase (HN), only oseltamivir was docked with a better score than BCX-2798, which indicates that oseltamivir has an inhibitory effect on PIV3. After the distance (Z d c ) between the drug target of 14 drugs and the PIV3 disease target was measured by the network proximity method based on the PIV3 disease module, it was found that theZ d c values of amikacin, oseltamivir, ribavirin, and streptomycin were less than those of the control. Thus, oseltamivir is the best potential drug because it met all the above screening requirements. Additionally, to explore whether oseltamivir binds to HN stably, molecular dynamics simulation of the binding of oseltamivir to HN was carried out, and the results showed that the RMSD value of the complex tended to be stable within 100 ns, and the binding free energy of the complex was low (-10.60 kcal/mol). It was proved that oseltamivir screened by our drug repurposing method had the potential feasibility of treating PIV3.
Collapse
Affiliation(s)
- Xinyue Chen
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Bo Zhou
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyi Jiang
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Huayu Zhong
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Aijing You
- The Second Clinical College of Chongqing Medical University, Chongqing, China
| | - Taiyan Zou
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Medical Data Science Academy, College of Medical Informatics, Chongqing Medical University, Chongqing, China
- Chongqing Engineering Research Center for Clinical Big-Data and Drug Evaluation, Chongqing Medical University, Chongqing, China
| | - Chengcheng Zhou
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Liu
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yonghong Zhang
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Medical Data Science Academy, College of Medical Informatics, Chongqing Medical University, Chongqing, China
- Chongqing Engineering Research Center for Clinical Big-Data and Drug Evaluation, Chongqing Medical University, Chongqing, China
| |
Collapse
|
32
|
Yu TWB. A phenotypic drug discovery approach by latent interaction in deep learning. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240720. [PMID: 40191531 PMCID: PMC11972434 DOI: 10.1098/rsos.240720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 04/09/2025]
Abstract
Contemporary drug discovery paradigms rely heavily on binding assays about the bio-physicochemical processes. However, this dominant approach suffers from overlooked higher-order interactions arising from the intricacies of molecular mechanisms, such as those involving cis-regulatory elements. It introduces potential impairments and restrains the potential development of computational methods. To address this limitation, I developed a deep learning model that leverages an end-to-end approach, relying exclusively on therapeutic information about drugs. By transforming textual representations of drug and virus genetic information into high-dimensional latent representations, this method evades the challenges arising from insufficient information about binding specificities. Its strengths lie in its ability to implicitly consider complexities such as epistasis and chemical-genetic interactions, and to handle the pervasive challenge of data scarcity. Through various modeling skills and data augmentation techniques, the proposed model demonstrates outstanding performance in out-of-sample validations, even in scenarios with unknown complex interactions. Furthermore, the study highlights the importance of chemical diversity for model training. While the method showcases the feasibility of deep learning in data-scarce scenarios, it reveals a promising alternative for drug discovery in situations where knowledge of underlying mechanisms is limited.
Collapse
Affiliation(s)
- Tat Wai Billy Yu
- Macao Polytechnic University, Macau SAR, People’s Republic of China
| |
Collapse
|
33
|
Nashruddin SNABM, Salleh FHM, Yunus RM, Zaman HB. Artificial intelligence-powered electrochemical sensor: Recent advances, challenges, and prospects. Heliyon 2024; 10:e37964. [PMID: 39328566 PMCID: PMC11425101 DOI: 10.1016/j.heliyon.2024.e37964] [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: 08/01/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Integrating artificial intelligence (AI) with electrochemical biosensors is revolutionizing medical treatments by enhancing patient data collection and enabling the development of advanced wearable sensors for health, fitness, and environmental monitoring. Electrochemical biosensors, which detect biomarkers through electrochemical processes, are significantly more effective. The integration of artificial intelligence is adept at identifying, categorizing, characterizing, and projecting intricate data patterns. As the Internet of Things (IoT), big data, and big health technologies move from theory to practice, AI-powered biosensors offer significant opportunities for real-time disease detection and personalized healthcare. Still, they also pose challenges such as data privacy, sensor stability, and algorithmic bias. This paper highlights the critical advances in material innovation, biorecognition elements, signal transduction, data processing, and intelligent decision systems necessary for developing next-generation wearable and implantable devices. Despite existing limitations, the integration of AI into biosensor systems shows immense promise for creating future medical devices that can provide early detection and improved patient outcomes, marking a transformative step forward in healthcare technology.
Collapse
Affiliation(s)
- Siti Nur Ashakirin Binti Mohd Nashruddin
- Institute of Informatics and Computing in Energy (IICE), Department of Computing, College of Computing & Informatics, Universiti Tenaga Nasional, 43000, Kajang, Selangor Darul Ehsan, Malaysia
| | - Faridah Hani Mohamed Salleh
- Institute of Informatics and Computing in Energy (IICE), Department of Computing, College of Computing & Informatics, Universiti Tenaga Nasional, 43000, Kajang, Selangor Darul Ehsan, Malaysia
| | - Rozan Mohamad Yunus
- Fuel Cell Institute, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Halimah Badioze Zaman
- Institute of Informatics and Computing in Energy (IICE), Department of Computing, College of Computing & Informatics, Universiti Tenaga Nasional, 43000, Kajang, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
34
|
Zhang S, Niu Q, Zong W, Song Q, Tian S, Wang J, Liu J, Zhang H, Wang Z, Li B. Endotype-driven Co-module mechanisms of danhong injection in the Co-treatment of cardiovascular and cerebrovascular diseases: A modular-based drug and disease integrated analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118287. [PMID: 38705429 DOI: 10.1016/j.jep.2024.118287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular and cerebrovascular diseases are the leading causes of death worldwide and interact closely with each other. Danhong Injection (DHI) is a widely used preparation for the co-treatment of brain and heart diseases (CTBH). However, the underlying molecular endotype mechanisms of DHI in the CTBH remain unclear. AIM OF THIS STUDY To elucidate the underlying endotype mechanisms of DHI in the CTBH. MATERIALS AND METHODS In this study, we proposed a modular-based disease and drug-integrated analysis (MDDIA) strategy for elucidating the systematic CTBH mechanisms of DHI using high-throughput transcriptome-wide sequencing datasets of DHI in the treatment of patients with stable angina pectoris (SAP) and cerebral infarction (CI). First, we identified drug-targeted modules of DHI and disease modules of SAP and CI based on the gene co-expression networks of DHI therapy and the protein-protein interaction networks of diseases. Moreover, module proximity-based topological analyses were applied to screen CTBH co-module pairs and driver genes of DHI. At the same time, the representative driver genes were validated via in vitro experiments on hypoxia/reoxygenation-related cardiomyocytes and neuronal cell lines of H9C2 and HT22. RESULTS Seven drug-targeted modules of DHI and three disease modules of SAP and CI were identified by co-expression networks. Five modes of modular relationships between the drug and disease modules were distinguished by module proximity-based topological analyses. Moreover, 13 targeted module pairs and 17 driver genes associated with DHI in the CTBH were also screened. Finally, the representative driver genes AKT1, EDN1, and RHO were validated by in vitro experiments. CONCLUSIONS This study, based on clinical sequencing data and modular topological analyses, integrated diseases and drug targets. The CTBH mechanism of DHI may involve the altered expression of certain driver genes (SRC, STAT3, EDN1, CYP1A1, RHO, RELA) through various enriched pathways, including the Wnt signaling pathway.
Collapse
Affiliation(s)
- Siqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qikai Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qi Song
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Siwei Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jingai Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huamin Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
35
|
Nisar H, Wajid B, Anwar F, Ahmad A, Javaid A, Attique SA, Nisar W, Saeed A, Shahid S, Sadaf S. Bioinformatics and systems biology analysis revealed PMID26394986-Compound-10 as potential repurposable drug against covid-19. J Biomol Struct Dyn 2024; 42:7972-7985. [PMID: 37534820 DOI: 10.1080/07391102.2023.2242500] [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: 02/02/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
The global health pandemic known as COVID-19, which stems from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a significant concern worldwide. Several treatment methods exist for COVID-19; however, there is an urgent demand for previously established drugs and vaccines to effectively combat the disease. Since, discovering new drugs poses a significant challenge, making the repurposing of existing drugs can potentially reduce time and costs compared to developing entirely new drugs from scratch. The objective of this study is to identify hub genes and associated repurposed drugs targeting them. We analyzed differentially expressed genes (DEGs) by analyzing RNA-seq transcriptomic datasets and integrated with genes associated with COVID-19 present in different databases. We detected 173 Covid-19 associated genes for the construction of a protein-protein interaction (PPI) network which resulted in the identification of the top 10 hub genes/proteins (STAT1, IRF7, MX1, IRF9, ISG15, OAS3, OAS2, OAS1, IRF3, and IRF1). Hub genes were subjected to GO functional and KEGG pathway enrichment analyses, which indicated some key roles and signaling pathways that were strongly related to SARS-CoV-2 infections. We conducted drug repurposing analysis using CMap, TTD, and DrugBank databases with these 10 hub genes, leading to the identification of Piceatannol, CKD-712, and PMID26394986-Compound-10 as top-ranked candidate drugs. Finally, drug-gene interactions analysis through molecular docking and validated via molecular dynamic simulation for 80 ns suggests PMID26394986-Compound-10 as the only potential drug. Our research demonstrates how in silico analysis might produce repurposing candidates to help respond faster to new disease outbreaks.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Haseeb Nisar
- Department of Life-Sciences, University of Management and Technology, Lahore, Pakistan
| | - Bilal Wajid
- Ibn Sina Research & Development Division, Sabz-Qalam, Lahore, Pakistan
- Department of Electrical Engineering, University of Engineering and Technology, Lahore, Pakistan
| | | | - Ashfaq Ahmad
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Anum Javaid
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Syed Awais Attique
- Bioinformatics Institute, Agency for Science, Technology and Research (A(*)STAR), Singapore, Singapore
| | - Wardah Nisar
- Department of Public Health, University of Health Sciences, Lahore, Pakistan
| | - Amir Saeed
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Samiah Shahid
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Saima Sadaf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
36
|
Liu C, Xue Q, Zhang Y, Zhang D, Li Y. Anti-hypertensive effect and potential mechanism of gastrodia-uncaria granules based on network pharmacology and experimental validation. J Clin Hypertens (Greenwich) 2024; 26:1024-1038. [PMID: 38990083 PMCID: PMC11488320 DOI: 10.1111/jch.14833] [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: 03/04/2024] [Revised: 04/18/2024] [Accepted: 05/05/2024] [Indexed: 07/12/2024]
Abstract
Hypertension has become a major contributor to the morbidity and mortality of cardiovascular diseases worldwide. Despite the evidence of the anti-hypertensive effect of gastrodia-uncaria granules (GUG) in hypertensive patients, little is known about its potential therapeutic targets as well as the underlying mechanism. GUG components were sourced from TCMSP and HERB, with bioactive ingredients screened. Hypertension-related targets were gathered from DisGeNET, OMIM, GeneCards, CTD, and GEO. The STRING database constructed a protein-protein interaction network, visualized by Cytoscape 3.7.1. Core targets were analyzed via GO and KEGG using R package ClusterProfiler. Molecular docking with AutodockVina 1.2.2 revealed favorable binding affinities. In vivo studies on hypertensive mice and rats validated network pharmacology findings. GUG yielded 228 active ingredients and 1190 targets, intersecting with 373 hypertension-related genes. PPI network analysis identified five core genes: AKT1, TNF-α, GAPDH, IL-6, and ALB. Top enriched GO terms and KEGG pathways associated with the anti-hypertensive properties of GUG were documented. Molecular docking indicated stable binding of core components to targets. In vivo study showed that GUG could improve vascular relaxation, alleviate vascular remodeling, and lower blood pressure in hypertensive animal models possibly through inhibiting inflammatory factors such as AKT1, mTOR, and CCND1. Integrated network pharmacology and in vivo experiment showed that GUG may exert anti-hypertensive effects by inhibiting inflammation response, which provides some clues for understanding the effect and mechanisms of GUG in the treatment of hypertension.
Collapse
Affiliation(s)
- Chu‐Hao Liu
- Department of Cardiovascular MedicineShanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Qi‐Qi Xue
- Department of Cardiovascular MedicineShanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Yi‐Qing Zhang
- Department of Cardiovascular MedicineShanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Dong‐Yan Zhang
- Department of Cardiovascular MedicineShanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Yan Li
- Department of Cardiovascular MedicineShanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
37
|
Sikirzhytskaya A, Tyagin I, Sutton SS, Wyatt MD, Safro I, Shtutman M. AI-based mining of biomedical literature: Applications for drug repurposing for the treatment of dementia. RESEARCH SQUARE 2024:rs.3.rs-4750719. [PMID: 39184100 PMCID: PMC11343300 DOI: 10.21203/rs.3.rs-4750719/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Neurodegenerative pathologies such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, Multiple sclerosis, HIV-associated neurocognitive disorder, and others significantly affect individuals, their families, caregivers, and healthcare systems. While there are no cures yet, researchers worldwide are actively working on the development of novel treatments that have the potential to slow disease progression, alleviate symptoms, and ultimately improve the overall health of patients. Huge volumes of new scientific information necessitate new analytical approaches for meaningful hypothesis generation. To enable the automatic analysis of biomedical data we introduced AGATHA, an effective AI-based literature mining tool that can navigate massive scientific literature databases, such as PubMed. The overarching goal of this effort is to adapt AGATHA for drug repurposing by revealing hidden connections between FDA-approved medications and a health condition of interest. Our tool converts the abstracts of peer-reviewed papers from PubMed into multidimensional space where each gene and health condition are represented by specific metrics. We implemented advanced statistical analysis to reveal distinct clusters of scientific terms within the virtual space created using AGATHA-calculated parameters for selected health conditions and genes. Partial Least Squares Discriminant Analysis was employed for categorizing and predicting samples (122 diseases and 20889 genes) fitted to specific classes. Advanced statistics were employed to build a discrimination model and extract lists of genes specific to each disease class. Here we focus on drugs that can be repurposed for dementia treatment as an outcome of neurodegenerative diseases. Therefore, we determined dementia-associated genes statistically highly ranked in other disease classes. Additionally, we report a mechanism for detecting genes common to multiple health conditions. These sets of genes were classified based on their presence in biological pathways, aiding in selecting candidates and biological processes that are exploitable with drug repurposing.
Collapse
|
38
|
Gao K, Cao W, He Z, Liu L, Guo J, Dong L, Song J, Wu Y, Zhao Y. Network medicine analysis for dissecting the therapeutic mechanism of consensus TCM formulae in treating hepatocellular carcinoma with different TCM syndromes. Front Endocrinol (Lausanne) 2024; 15:1373054. [PMID: 39211446 PMCID: PMC11357915 DOI: 10.3389/fendo.2024.1373054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality worldwide. Traditional Chinese Medicine (TCM) is widely utilized as an adjunct therapy, improving patient survival and quality of life. TCM categorizes HCC into five distinct syndromes, each treated with specific herbal formulae. However, the molecular mechanisms underlying these treatments remain unclear. Methods We employed a network medicine approach to explore the therapeutic mechanisms of TCM in HCC. By constructing a protein-protein interaction (PPI) network, we integrated genes associated with TCM syndromes and their corresponding herbal formulae. This allowed for a quantitative analysis of the topological and functional relationships between TCM syndromes, HCC, and the specific formulae used for treatment. Results Our findings revealed that genes related to the five TCM syndromes were closely associated with HCC-related genes within the PPI network. The gene sets corresponding to the five TCM formulae exhibited significant proximity to HCC and its related syndromes, suggesting the efficacy of TCM syndrome differentiation and treatment. Additionally, through a random walk algorithm applied to a heterogeneous network, we prioritized active herbal ingredients, with results confirmed by literature. Discussion The identification of these key compounds underscores the potential of network medicine to unravel the complex pharmacological actions of TCM. This study provides a molecular basis for TCM's therapeutic strategies in HCC and highlights specific herbal ingredients as potential leads for drug development and precision medicine.
Collapse
Affiliation(s)
- Kai Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - WanChen Cao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - ZiHao He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Liu Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - JinCheng Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Lei Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Jini Song
- New York Institute of Technology College of Osteopathic Medicine, Arkansas State University, Jonesboro, AR, United States
| | - Yang Wu
- The Research Center for Ubiquitous Computing Systems (CUbiCS), Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
- The Research Center for Ubiquitous Computing Systems (CUbiCS), Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
39
|
Liu H, Zhang N, Jia Y, Wang J, Ye A, Yang S, Zhou H, Lv Y, Xu C, Wang S. ncStem: a comprehensive resource of curated and predicted ncRNAs in cancer stemness. Database (Oxford) 2024; 2024:baae081. [PMID: 39137906 PMCID: PMC11321241 DOI: 10.1093/database/baae081] [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: 03/25/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024]
Abstract
Cancer stemness plays an important role in cancer initiation and progression, and is the major cause of tumor invasion, metastasis, recurrence, and poor prognosis. Non-coding RNAs (ncRNAs) are a class of RNA transcripts that generally cannot encode proteins and have been demonstrated to play a critical role in regulating cancer stemness. Here, we developed the ncStem database to record manually curated and predicted ncRNAs associated with cancer stemness. In total, ncStem contains 645 experimentally verified entries, including 159 long non-coding RNAs (lncRNAs), 254 microRNAs (miRNAs), 39 circular RNAs (circRNAs), and 5 other ncRNAs. The detailed information of each entry includes the ncRNA name, ncRNA identifier, disease, reference, expression direction, tissue, species, and so on. In addition, ncStem also provides computationally predicted cancer stemness-associated ncRNAs for 33 TCGA cancers, which were prioritized using the random walk with restart (RWR) algorithm based on regulatory and co-expression networks. The total predicted cancer stemness-associated ncRNAs included 11 132 lncRNAs and 972 miRNAs. Moreover, ncStem provides tools for functional enrichment analysis, survival analysis, and cell location interrogation for cancer stemness-associated ncRNAs. In summary, ncStem provides a platform to retrieve cancer stemness-associated ncRNAs, which may facilitate research on cancer stemness and offer potential targets for cancer treatment. Database URL: http://www.nidmarker-db.cn/ncStem/index.html.
Collapse
Affiliation(s)
- Hui Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Nan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yijie Jia
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Jun Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Aokun Ye
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Siru Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Honghan Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yingli Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Chaohan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Shuyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| |
Collapse
|
40
|
Tomić D, Murgić J, Fröbe A, Skala K, Vrljičak A, Medved Rogina B, Kolarek B, Bojović V. Exploring potential therapeutic combinations for castration-sensitive prostate cancer using supercomputers: a proof of concept study. Sci Rep 2024; 14:18824. [PMID: 39138333 PMCID: PMC11322545 DOI: 10.1038/s41598-024-69880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024] Open
Abstract
To address the challenge of finding new combination therapies against castration-sensitive prostate cancer, we introduce Vini, a computational tool that predicts the efficacy of drug combinations at the intracellular level by integrating data from the KEGG, DrugBank, Pubchem, Protein Data Bank, Uniprot, NCI-60 and COSMIC databases. Vini is a computational tool that predicts the efficacy of drugs and their combinations at the intracellular level. It addresses the problem comprehensively by considering all known target genes, proteins and small molecules and their mutual interactions involved in the onset and development of cancer. The results obtained point to new, previously unexplored combination therapies that could theoretically be promising candidates for the treatment of castration-sensitive prostate cancer and could prevent the inevitable progression of the cancer to the incurable castration-resistant stage. Furthermore, after analyzing the obtained triple combinations of drugs and their targets, the most common targets became clear: ALK, BCL-2, mTOR, DNA and androgen axis. These results may help to define future therapies against castration-sensitive prostate cancer. The use of the Vini computer model to explore therapeutic combinations represents an innovative approach in the search for effective treatments for castration-sensitive prostate cancer, which, if clinically validated, could potentially lead to new breakthrough therapies.
Collapse
Affiliation(s)
- Draško Tomić
- Centre for Informatics and Computing, Rudjer Boskovic Institute, 10000, Zagreb, Croatia.
| | - Jure Murgić
- Department of Oncology and Nuclear Medicine, Sisters of Charity Hospital, 10000, Zagreb, Croatia
| | - Ana Fröbe
- Department of Oncology and Nuclear Medicine, Sisters of Charity Hospital, 10000, Zagreb, Croatia
| | - Karolj Skala
- Centre for Informatics and Computing, Rudjer Boskovic Institute, 10000, Zagreb, Croatia
| | - Antonela Vrljičak
- Department of Oncology and Nuclear Medicine, Sisters of Charity Hospital, 10000, Zagreb, Croatia
| | - Branka Medved Rogina
- Centre for Informatics and Computing, Rudjer Boskovic Institute, 10000, Zagreb, Croatia
| | - Branimir Kolarek
- Centre for Informatics and Computing, Rudjer Boskovic Institute, 10000, Zagreb, Croatia
| | - Viktor Bojović
- Centre for Informatics and Computing, Rudjer Boskovic Institute, 10000, Zagreb, Croatia
| |
Collapse
|
41
|
Liao K, Wang F, Xia C, Xu Z, Zhong S, Bi W, Ruan J. The cGAS-STING pathway in COPD: targeting its role and therapeutic potential. Respir Res 2024; 25:302. [PMID: 39113033 PMCID: PMC11308159 DOI: 10.1186/s12931-024-02915-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Chronic obstructive pulmonary disease(COPD) is a gradually worsening and fatal heterogeneous lung disease characterized by airflow limitation and increasingly decline in lung function. Currently, it is one of the leading causes of death worldwide. The consistent feature of COPD is airway inflammation. Several inflammatory factors are known to be involved in COPD pathogenesis; however, anti-inflammatory therapy is not the first-line treatment for COPD. Although bronchodilators, corticosteroids and roflumilast could improve airflow and control symptoms, they could not reverse the disease. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway plays an important novel role in the immune system and has been confirmed to be a key mediator of inflammation during infection, cellular stress, and tissue damage. Recent studies have emphasized that abnormal activation of cGAS-STING contributes to COPD, providing a direction for new treatments that we urgently need to develop. Here, we focused on the cGAS-STING pathway, providing insight into its molecular mechanism and summarizing the current knowledge on the role of the cGAS-STING pathway in COPD. Moreover, we explored antagonists of cGAS and STING to identify potential therapeutic strategies for COPD that target the cGAS-STING pathway.
Collapse
Affiliation(s)
- Kexin Liao
- First Clinical Medical College, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Fengshuo Wang
- College of Pharmacy, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Chenhao Xia
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Ze Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Sen Zhong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Wenqi Bi
- First Clinical Medical College, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Jingjing Ruan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| |
Collapse
|
42
|
Mittal RK, Purohit P, Sankaranarayanan M, Muzaffar-Ur-Rehman M, Taramelli D, Signorini L, Dolci M, Basilico N. In-vitro antiviral activity and in-silico targeted study of quinoline-3-carboxylate derivatives against SARS-Cov-2 isolate. Mol Divers 2024; 28:2651-2665. [PMID: 37480422 DOI: 10.1007/s11030-023-10703-w] [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: 06/07/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
In recent years, the viral outbreak named COVID-19 showed that infectious diseases have a huge impact on both global health and the financial and economic sectors. The lack of efficacious antiviral drugs worsened the health problem. Based on our previous experience, we investigated in vitro and in silico a series of quinoline-3-carboxylate derivatives against a SARS-CoV-2 isolate. In the present study, the in-vitro antiviral activity of a series of quinoline-3-carboxylate compounds and the in silico target-based molecular dynamics (MD) and metabolic studies are reported. The compounds' activity against SARS-CoV-2 was evaluated using plaque assay and RT-qPCR. Moreover, from the docking scores, it appears that the most active compounds (1j and 1o) exhibit stronger binding affinity to the primary viral protease (NSP5) and the exoribonuclease domain of non structural protein 14 (NSP14). Additionally, the in-silico metabolic analysis of 1j and 1o defines CYP2C9 and CYP3A4 as the major P450 enzymes involved in their metabolism.
Collapse
Affiliation(s)
- Ravi Kumar Mittal
- National Institute of Pharmaceutical Education and Research, S A S Nagar Mohali, Punjab, 160062, India
- Galgotias College of Pharmacy, Greater Noida, UttarPradesh, India
| | - Priyank Purohit
- School of Pharmacy, Graphic Era Hill University, Dehradun, Uttarakhand, 248002, India.
| | - Murugesan Sankaranarayanan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, BITS Pilani, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Mohammed Muzaffar-Ur-Rehman
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, BITS Pilani, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Donatella Taramelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| | - Lucia Signorini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| | - Maria Dolci
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| |
Collapse
|
43
|
Cousins HC, Nayar G, Altman RB. Computational Approaches to Drug Repurposing: Methods, Challenges, and Opportunities. Annu Rev Biomed Data Sci 2024; 7:15-29. [PMID: 38598857 DOI: 10.1146/annurev-biodatasci-110123-025333] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Drug repurposing refers to the inference of therapeutic relationships between a clinical indication and existing compounds. As an emerging paradigm in drug development, drug repurposing enables more efficient treatment of rare diseases, stratified patient populations, and urgent threats to public health. However, prioritizing well-suited drug candidates from among a nearly infinite number of repurposing options continues to represent a significant challenge in drug development. Over the past decade, advances in genomic profiling, database curation, and machine learning techniques have enabled more accurate identification of drug repurposing candidates for subsequent clinical evaluation. This review outlines the major methodologic classes that these approaches comprise, which rely on (a) protein structure, (b) genomic signatures, (c) biological networks, and (d) real-world clinical data. We propose that realizing the full impact of drug repurposing methodologies requires a multidisciplinary understanding of each method's advantages and limitations with respect to clinical practice.
Collapse
Affiliation(s)
- Henry C Cousins
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| | - Gowri Nayar
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| | - Russ B Altman
- Departments of Genetics, Medicine, and Bioengineering, Stanford University, Stanford, California, USA
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| |
Collapse
|
44
|
Jena NR, Pant S. Peptide inhibitors derived from the nsp7 and nsp8 cofactors of nsp12 targeting different substrate binding sites of nsp12 of the SARS-CoV-2. J Biomol Struct Dyn 2024; 42:7077-7089. [PMID: 37434315 DOI: 10.1080/07391102.2023.2235012] [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: 03/24/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023]
Abstract
SARS-COV-2 is responsible for the COVID-19 pandemic, which has infected more than 767 million people worldwide including about 7 million deaths till 5 June 2023. Despite the emergency use of certain vaccines, deaths due to COVID-19 have not yet stopped completed. Therefore, it is imperative to design and develop drugs that can be used to treat patients suffering from COVID-19. Here, two peptide inhibitors derived from nsp7 and nsp8 cofactors of nsp12 have been shown to block different substrate binding sites of nsp12 that are mainly responsible for the replication of the viral genome of SARS-CoV-2. By using the docking, molecular dynamics (MD), and MM/GBSA techniques, it is shown that these inhibitors can bind to multiple binding sites of nsp12, such as the interface of nsp7 and nsp12, interface of nsp8 and nsp12, RNA primer entry site, and nucleoside triphosphate (NTP) entry site. The relative binding free energies of the most stable protein-peptide complexes are found to lie between ∼-34.20 ± 10.07 to -59.54 ± 9.96 kcal/mol. Hence, it is likely that these inhibitors may bind to different sites of nsp12 to block the access of its cofactors and the viral genome, thereby affecting the replication. It is thus proposed that these peptide inhibitors may be further developed as potential drug candidates to suppress the viral loads in COVID-19 patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- N R Jena
- Discipline of Natural Sciences, Indian Institute of Information Technology, Design, and Manufacturing, Jabalpur, India
| | - Suyash Pant
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Kolkata, India
| |
Collapse
|
45
|
Su C, Hou Y, Xu J, Xu Z, Zhou M, Ke A, Li H, Xu J, Brendel M, Maasch JRMA, Bai Z, Zhang H, Zhu Y, Cincotta MC, Shi X, Henchcliffe C, Leverenz JB, Cummings J, Okun MS, Bian J, Cheng F, Wang F. Identification of Parkinson's disease PACE subtypes and repurposing treatments through integrative analyses of multimodal data. NPJ Digit Med 2024; 7:184. [PMID: 38982243 PMCID: PMC11233682 DOI: 10.1038/s41746-024-01175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Parkinson's disease (PD) is a serious neurodegenerative disorder marked by significant clinical and progression heterogeneity. This study aimed at addressing heterogeneity of PD through integrative analysis of various data modalities. We analyzed clinical progression data (≥5 years) of individuals with de novo PD using machine learning and deep learning, to characterize individuals' phenotypic progression trajectories for PD subtyping. We discovered three pace subtypes of PD exhibiting distinct progression patterns: the Inching Pace subtype (PD-I) with mild baseline severity and mild progression speed; the Moderate Pace subtype (PD-M) with mild baseline severity but advancing at a moderate progression rate; and the Rapid Pace subtype (PD-R) with the most rapid symptom progression rate. We found cerebrospinal fluid P-tau/α-synuclein ratio and atrophy in certain brain regions as potential markers of these subtypes. Analyses of genetic and transcriptomic profiles with network-based approaches identified molecular modules associated with each subtype. For instance, the PD-R-specific module suggested STAT3, FYN, BECN1, APOA1, NEDD4, and GATA2 as potential driver genes of PD-R. It also suggested neuroinflammation, oxidative stress, metabolism, PI3K/AKT, and angiogenesis pathways as potential drivers for rapid PD progression (i.e., PD-R). Moreover, we identified repurposable drug candidates by targeting these subtype-specific molecular modules using network-based approach and cell line drug-gene signature data. We further estimated their treatment effects using two large-scale real-world patient databases; the real-world evidence we gained highlighted the potential of metformin in ameliorating PD progression. In conclusion, this work helps better understand clinical and pathophysiological complexity of PD progression and accelerate precision medicine.
Collapse
Grants
- R21 AG083003 NIA NIH HHS
- R01 AG082118 NIA NIH HHS
- R56 AG074001 NIA NIH HHS
- R01AG076448 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1AG072449 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- MJFF-023081 Michael J. Fox Foundation for Parkinson's Research (Michael J. Fox Foundation)
- R01AG080991 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P30 AG072959 NIA NIH HHS
- 3R01AG066707-01S1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R21AG083003 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG066707 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R35 AG071476 NIA NIH HHS
- RF1 AG082211 NIA NIH HHS
- R56AG074001 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG082118 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R25 AG083721 NIA NIH HHS
- RF1AG082211 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 NS093334 NINDS NIH HHS
- AG083721-01 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1NS133812 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P20GM109025 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1 NS133812 NINDS NIH HHS
- R35AG71476 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AG073323 NIA NIH HHS
- R01 AG066707 NIA NIH HHS
- R01AG053798 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG076234 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 AG076448 NIA NIH HHS
- R01 AG080991 NIA NIH HHS
- R01 AG076234 NIA NIH HHS
- U01NS093334 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P20 GM109025 NIGMS NIH HHS
- P30AG072959 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1 AG072449 NIA NIH HHS
- R01 AG053798 NIA NIH HHS
- 3R01AG066707-02S1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AG073323 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- ALZDISCOVERY-1051936 Alzheimer's Association
Collapse
Affiliation(s)
- Chang Su
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Yu Hou
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zhenxing Xu
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Manqi Zhou
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Alison Ke
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Haoyang Li
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jie Xu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Matthew Brendel
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jacqueline R M A Maasch
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computer Science, Cornell Tech, Cornell University, New York, NY, USA
| | - Zilong Bai
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Haotan Zhang
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Zhu
- Department of Computer Science, University of Texas at Arlington, Arlington, TX, USA
| | - Molly C Cincotta
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xinghua Shi
- Department of Computer and Information Sciences, Temple University, Philadelphia, PA, USA
| | - Claire Henchcliffe
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Michael S Okun
- Department of Neurology, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
46
|
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
|
47
|
Ma M, Huang M, He Y, Fang J, Li J, Li X, Liu M, Zhou M, Cui G, Fan Q. Network Medicine: A Potential Approach for Virtual Drug Screening. Pharmaceuticals (Basel) 2024; 17:899. [PMID: 39065749 PMCID: PMC11280361 DOI: 10.3390/ph17070899] [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: 04/25/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Traditional drug screening methods typically focus on a single protein target and exhibit limited efficiency due to the multifactorial nature of most diseases, which result from disturbances within complex networks of protein-protein interactions rather than single gene abnormalities. Addressing this limitation requires a comprehensive drug screening strategy. Network medicine is rooted in systems biology and provides a comprehensive framework for understanding disease mechanisms, prevention, and therapeutic innovations. This approach not only explores the associations between various diseases but also quantifies the relationships between disease genes and drug targets within interactome networks, thus facilitating the prediction of drug-disease relationships and enabling the screening of therapeutic drugs for specific complex diseases. An increasing body of research supports the efficiency and utility of network-based strategies in drug screening. This review highlights the transformative potential of network medicine in virtual therapeutic screening for complex diseases, offering novel insights and a robust foundation for future drug discovery endeavors.
Collapse
Affiliation(s)
- Mingxuan Ma
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Mei Huang
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Yinting He
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 570000, China;
| | - Jiachao Li
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Xiaohan Li
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Mengchen Liu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Mei Zhou
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Guozhen Cui
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519000, China; (M.M.); (M.H.); (Y.H.); (J.L.); (M.L.); (M.Z.)
| | - Qing Fan
- Basic Medical Science Department, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China
| |
Collapse
|
48
|
He H, Xie J, Huang D, Zhang M, Zhao X, Ying Y, Wang J. DRTerHGAT: A drug repurposing method based on the ternary heterogeneous graph attention network. J Mol Graph Model 2024; 130:108783. [PMID: 38677034 DOI: 10.1016/j.jmgm.2024.108783] [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: 01/09/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Drug repurposing is an effective method to reduce the time and cost of drug development. Computational drug repurposing can quickly screen out the most likely associations from large biological databases to achieve effective drug repurposing. However, building a comprehensive model that integrates drugs, proteins, and diseases for drug repurposing remains challenging. This study proposes a drug repurposing method based on the ternary heterogeneous graph attention network (DRTerHGAT). DRTerHGAT designs a novel protein feature extraction process consisting of a large-scale protein language model and a multi-task autoencoder, so that protein features can be extracted accurately and efficiently from amino acid sequences. The ternary heterogeneous graph of drug-protein-disease comprehensively considering the relationships among the three types of nodes, including three homogeneous and three heterogeneous relationships. Based on the graph and the extracted protein features, the deep features of the drugs and the diseases are extracted by graph convolutional networks (GCN) and heterogeneous graph node attention networks (HGNA). In the experiments, DRTerHGAT is proven superior to existing advanced methods and DRTerHGAT variants. DRTerHGAT's powerful ability for drug repurposing is also demonstrated in Alzheimer's disease.
Collapse
Affiliation(s)
- Hongjian He
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Jiang Xie
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China.
| | - Dingkai Huang
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Mengfei Zhang
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Xuyu Zhao
- School of Life Sciences,Shanghai University, Shanghai, China
| | - Yiwei Ying
- School of Life Sciences,Shanghai University, Shanghai, China
| | - Jiao Wang
- School of Life Sciences,Shanghai University, Shanghai, China.
| |
Collapse
|
49
|
Carnivali GS, Borges CC. Method to link medicines to diseases using multiplex networks. Comput Methods Biomech Biomed Engin 2024:1-14. [PMID: 38907637 DOI: 10.1080/10255842.2024.2362860] [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: 01/25/2024] [Accepted: 05/28/2024] [Indexed: 06/24/2024]
Abstract
The reuse of well-established medicines using computational modeling has gained a lot of attention due to its tremendous benefits. Based on this perspective, a new method for linking known medicines to diseases is proposed. The creation of a new treatment or medicine can be financially and temporally costly and the reuse of medicines is one possibility to accelerate this process efficiently. The main purpose of the reuse of medicines is to reduce some stages of the development of new medicines, motivating the proposition of several methods nowadays. In this work, a new method is developed aiming to connect known medicines to diseases based on available networks of protein interactions and available lists of medicines that affect protein action. The concepts of multiplex networks are used to connect subgraphs of vertices that represent medicines and proteins. The core of the procedure is determined by a weighting strategy constructed to define precisely the more relevant connections. The method was compared to other network link methods in the literature and a case study was presented and evaluated by the proposed method.
Collapse
|
50
|
Khan S, Partuk EO, Chiaravalli J, Kozer N, Shurrush KA, Elbaz-Alon Y, Scher N, Giraud E, Tran-Rajau J, Agou F, Barr HM, Avinoam O. High-throughput screening identifies broad-spectrum Coronavirus entry inhibitors. iScience 2024; 27:110019. [PMID: 38883823 PMCID: PMC11176637 DOI: 10.1016/j.isci.2024.110019] [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: 12/26/2023] [Revised: 04/04/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
The COVID-19 pandemic highlighted the need for antivirals against emerging coronaviruses (CoV). Inhibiting spike (S) glycoprotein-mediated viral entry is a promising strategy. To identify small molecule inhibitors that block entry downstream of receptor binding, we established a high-throughput screening (HTS) platform based on pseudoviruses. We employed a three-step process to screen nearly 200,000 small molecules. First, we identified hits that inhibit pseudoviruses bearing the SARS-CoV-2 S glycoprotein. Counter-screening against pseudoviruses with the vesicular stomatitis virus glycoprotein (VSV-G), yielded sixty-five SARS-CoV-2 S-specific inhibitors. These were further tested against pseudoviruses bearing the MERS-CoV S glycoprotein, which uses a different receptor. Out of these, five compounds, which included the known broad-spectrum inhibitor Nafamostat, were subjected to further validation and tested against pseudoviruses bearing the S glycoprotein of the Alpha, Delta, and Omicron variants as well as bona fide SARS-CoV-2. This rigorous approach revealed an unreported inhibitor and its derivative as potential broad-spectrum antivirals.
Collapse
Affiliation(s)
- Suman Khan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Efrat Ozer Partuk
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Jeanne Chiaravalli
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Noga Kozer
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Khriesto A Shurrush
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yael Elbaz-Alon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nadav Scher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Emilie Giraud
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Jaouen Tran-Rajau
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Fabrice Agou
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Haim Michael Barr
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|