1
|
Abavisani M, Khoshrou A, Eshaghian S, Karav S, Sahebkar A. Overcoming antibiotic resistance: the potential and pitfalls of drug repurposing. J Drug Target 2025; 33:341-367. [PMID: 39485073 DOI: 10.1080/1061186x.2024.2424895] [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: 06/09/2024] [Revised: 10/18/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Since its emergence shortly after the discovery of penicillin, antibiotic resistance has escalated dramatically, posing a significant health threat and economic burden. Drug repositioning, or drug repurposing, involves identifying new therapeutic applications for existing drugs, utilising their established safety profiles and pharmacological data to swiftly provide effective treatments against resistant pathogens. Several drugs, including otilonium bromide, penfluridol, eltrombopag, ibuprofen, and ceritinib, have demonstrated potent antibacterial activity against multidrug-resistant (MDR) bacteria. These drugs can disrupt biofilms, damage bacterial membranes, and inhibit bacterial growth. The combination of repurposed drugs with conventional antibiotics can reduce the required dosage of individual drugs, mitigate side effects, and delay the development of resistance, making it a promising strategy against MDR bacteria such as Staphylococcus aureus, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Escherichia coli. Despite its promise, drug repurposing faces challenges such as potential off-target effects, toxicity, and regulatory and intellectual property issues, necessitating rigorous evaluations and strategic solutions. This article aims to explore the potential of drug repurposing as a strategy to combat antibiotic resistance, examining its benefits, challenges, and future prospects. We address the legal, economic, and practical challenges associated with repurposing existing drugs, highlight successful examples, and propose solutions to enhance the efficacy and viability of this approach in combating MDR bacterial infections.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Souzan Eshaghian
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Liu H, Yan W, Luo D, Li J, Yan D. A real‑world pharmacovigilance study of raloxifene based on the FDA adverse event reporting system (FAERS). Expert Opin Drug Saf 2024:1-9. [PMID: 39690869 DOI: 10.1080/14740338.2024.2443960] [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: 07/20/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Raloxifene was approved for the treatment of postmenopausal osteoporosis; however, its safety profile remains inadequately understood. This study aimed to evaluate the safety signals associated with raloxifene. RESEARCH DESIGN AND METHODS Adverse events (AEs) related to raloxifene, spanning from the first quarter of 2004 to the fourth quarter of 2023, were extracted from the FDA Adverse Event Reporting System (FAERS) database. A disproportionality analysis was conducted using several methods, including the reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Empirical Bayes Geometric Mean (EBGM). RESULTS The analysis yielded 7 229 reports related to raloxifene across 19 277 AEs. A total of 217 significantly disproportionate signals were identified, including muscle spasms and hot flashes. Notably, the study also uncovered novel AEs, including eye conditions like cataracts and macular degeneration, as well as gynecological issues like uterine polyps and hemorrhage. Additionally, the analysis confirmed that pulmonary embolism and deep vein thrombosis were the two most prevalent thromboembolic AEs. CONCLUSION Our study reaffirmed some existing safety information regarding raloxifene while also unveiling novel risk signals. The findings provided crucial insights to enhance the rational use of the drug and inform safety regulatory strategies.
Collapse
Affiliation(s)
- Hao Liu
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Yan
- Department of Orthopedic Joints, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Luo
- Department of Orthopedic Joints, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinsong Li
- Department of Orthopedic Joints, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dezhi Yan
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Cocetta V, Zorzi M, Bejor S, Cesta MC, De Pizzol M, Theurillat JP, Allegretti M, Alimonti A, Montopoli M, Rugge M. Retrospective Analysis of the Effect of Postmenopausal Women Medications on SARS-CoV-2 Infection Progression. Life (Basel) 2024; 14:1107. [PMID: 39337891 PMCID: PMC11433321 DOI: 10.3390/life14091107] [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: 06/21/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Since the beginning of the COVID-19 pandemic, it has been evident that women and young people were less susceptible to severe infections compared to males. In a previous study, we observed a reduced prevalence of SARS-CoV-2 infections in hormonal-driven breast cancer patients undergoing SERM (selective estrogen receptor modulator) therapy with respect to other treatments inhibiting estrogen synthesis. In addition to being used in anticancer therapy, SERMs are also prescribed for postmenopausal osteoporosis prevention and treatment. Therefore, in this study, a retrospective analysis of the clinical outcomes of SARS-CoV-2 infections in a population of women over 50 years who were treated for the management of menopausal symptoms was performed. SARS-CoV-2 infections, hospitalizations, and death rates were evaluated in women residing in the Italian north-eastern Veneto Region who were undergoing treatment with Estrogen Modulators (EMs); Estrogen or Progestin, and their combination (EPs); Bisphosphonates (BIs); or cholecalciferol (vitamin D3) ± calcium supplementation (CC). The final cohort study included 124,393 women, of whom 6412 were found to be SARS-CoV-2 infected (CoV2+ve). The results indicated that only women treated with vitamin D3 alone or in combination with calcium showed a significant reduction in their SARS-CoV-2 infection risk by 26% (OR 0.74; 95%CI 0.60-0.91). On the other hand, an increased risk of hospitalization (OR 2.69; 95%CI 1.77-4.07) was shown for the same treatments. The results highlighted in this work contribute to shedding some light on the widely debated role of vitamin D in the prevention of SARS-CoV-2 infections and the disease's treatment.
Collapse
Affiliation(s)
- Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
| | - Manuel Zorzi
- Veneto Tumour Registry, Azienda Zero, 35131 Padova, Italy
| | - Stefano Bejor
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
| | | | | | - Jean-Philippe Theurillat
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, 6500 Bellinzona & Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | | | - Andrea Alimonti
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, 6500 Bellinzona & Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
- VIMM-Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy
- Department of Medicine, University of Padova, 35122 Padova, Italy
- Department of Health Sciences and Technology, ETH Zürich, 8092 Zurich, Switzerland
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, 6500 Bellinzona & Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
- VIMM-Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy
| | - Massimo Rugge
- Veneto Tumour Registry, Azienda Zero, 35131 Padova, Italy
- Department of Medicine, University of Padova, 35122 Padova, Italy
| |
Collapse
|
4
|
Shen J, Shentu J, Zhong C, Huang Q, Duan S. RNA splicing factor RBFOX2 is a key factor in the progression of cancer and cardiomyopathy. Clin Transl Med 2024; 14:e1788. [PMID: 39243148 PMCID: PMC11380049 DOI: 10.1002/ctm2.1788] [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: 04/03/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Alternative splicing of pre-mRNA is a fundamental regulatory process in multicellular eukaryotes, significantly contributing to the diversification of the human proteome. RNA-binding fox-1 homologue 2 (RBFOX2), a member of the evolutionarily conserved RBFOX family, has emerged as a critical splicing regulator, playing a pivotal role in the alternative splicing of pre-mRNA. This review provides a comprehensive analysis of RBFOX2, elucidating its splicing activity through direct and indirect binding mechanisms. RBFOX2 exerts substantial influence over the alternative splicing of numerous transcripts, thereby shaping essential cellular processes such as differentiation and development. MAIN BODY OF THE ABSTRACT Dysregulation of RBFOX2-mediated alternative splicing has been closely linked to a spectrum of cardiovascular diseases and malignant tumours, underscoring its potential as a therapeutic target. Despite significant progress, current research faces notable challenges. The complete structural characterisation of RBFOX2 remains elusive, limiting in-depth exploration beyond its RNA-recognition motif. Furthermore, the scarcity of studies focusing on RBFOX2-targeting drugs poses a hindrance to translating research findings into clinical applications. CONCLUSION This review critically assesses the existing body of knowledge on RBFOX2, highlighting research gaps and limitations. By delineating these areas, this analysis not only serves as a foundational reference for future studies but also provides strategic insights for bridging these gaps. Addressing these challenges will be instrumental in unlocking the full therapeutic potential of RBFOX2, paving the way for innovative and effective treatments in various diseases.
Collapse
Affiliation(s)
- Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| | - Jianqiao Shentu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| | - Chenming Zhong
- Medical Genetics Center, School of MedicineNingbo UniversityNingboChina
| | - Qiankai Huang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| |
Collapse
|
5
|
Chauhan D, Maity D, Yadav PK, Vishwakarma S, Agarwal A, Chourasia MK, Gayen JR. Enhanced oral bioavailability of levormeloxifene and raloxifene by nanoemulsion: simultaneous bioanalysis using liquid chromatography-tandem mass spectrometry. Nanomedicine (Lond) 2024; 19:1051-1068. [PMID: 38639565 PMCID: PMC11225398 DOI: 10.2217/nnm-2024-0023] [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: 01/27/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024] Open
Abstract
Aim & objective: Levormeloxifene (L-ORM) and raloxifene (RAL) are selective estrogen receptor modulators used in the treatment of postmenopausal osteoporosis and breast cancer. Here, we developed and validated a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the simultaneous estimation of both drugs. Materials & methods: A quality-by-design (QbD) approach was used for the optimization of the nanoemulsion, and US FDA guidelines were followed for method validation. Results: Multiple reaction monitoring transitions were used for L-ORM (459.05→98.50), RAL (475.00→112.02) and internal standard (180.10→110.2). Analytes were resolved in a C18 column with 80:20 v/v% acetonitrile (ACN), 0.1% formic acid in triple-distilled water as a mobile phase. The developed method was linear over a concentration range of 1-600 ng/ml. Pharmacokinetic results of free L-ORM-RAL and the L-ORM-RAL nanoemulsion showed Cmax of free L-ORM - 70.65 ± 16.64, free RAL 13.53 ± 2.72, L-ORM nanoemulsion 65.07 ± 14.0 and RAL-nanoemulsion 59.27 ± 17.44 ng/ml. Conclusion: Future findings will contribute to the treatment of postmenopausal osteoporosis and breast cancer using L-ORM and RAL.
Collapse
Affiliation(s)
- Divya Chauhan
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debalina Maity
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Pavan K Yadav
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sachin Vishwakarma
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Arun Agarwal
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Manish K Chourasia
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jiaur R Gayen
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
6
|
Waseem T, Rajput TA, Mushtaq MS, Babar MM, Rajadas J. Computational biology approaches for drug repurposing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:91-109. [PMID: 38789189 DOI: 10.1016/bs.pmbts.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
The drug discovery and development (DDD) process greatly relies on the data available in various forms to generate hypotheses for novel drug design. The complex and heterogeneous nature of biological data makes it difficult to utilize or gather meaningful information as such. Computational biology techniques have provided us with opportunities to better understand biological systems through refining and organizing large amounts of data into actionable and systematic purviews. The drug repurposing approach has been utilized to overcome the expansive time periods and costs associated with traditional drug development. It deals with discovering new uses of already approved drugs that have an established safety and efficacy profile, thereby, requiring them to go through fewer development phases. Thus, drug repurposing through computational biology provides a systematic approach to drug development and overcomes the constraints of traditional processes. The current chapter covers the basics, approaches and tools of computational biology that can be employed to effectively develop repurposing profile of already approved drug molecules.
Collapse
Affiliation(s)
- Tanya Waseem
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Tausif Ahmed Rajput
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | | | - Mustafeez Mujtaba Babar
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan; Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute and Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States.
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute and Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
7
|
Umezu Y, Horiyama E, Nakai Y, Ninomiya M, Nishina A, Koketsu M. Synthesis of raloxifene-like quinoxaline derivatives by intramolecular electrophilic cyclization with disulfides. Bioorg Med Chem Lett 2023; 93:129415. [PMID: 37532107 DOI: 10.1016/j.bmcl.2023.129415] [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: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
The intramolecular electrophilic cyclization of alkynes with disulfides to form thieno[2,3-b]quinoxaline structures and to introduce thioether substituents afforded quinoxaline derivatives (7a-7d, 8a-8d). Among obtained eight derivatives, the raloxifene analogues (7c, 8b) showed specifically high cytotoxicity against breast cancer cells (SK-BR-3), and raloxifene analogues (8a) showed the highest cytotoxicity against human leukemia cells (HL-60). None of the raloxifene analogues (7a-7d, 8a-8d) showed cytotoxicity against human lung fibroblasts (WI-38), which are normal cells.
Collapse
Affiliation(s)
- Yuhi Umezu
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Eito Horiyama
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yuto Nakai
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Masayuki Ninomiya
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Division of Instrumental Analysis, Life Science Research Center, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Atsuyoshi Nishina
- Department of Applied Chemistry, College of Science and Technology, Nihon University, 1-5-1 Kandasurugadai, Chiyoda, Tokyo 101-0062, Japan; School of Health and Nutrition, Tokai Gakuen University, 2-901 Nakahira, Tenpaku-ku, Nagoya, Aichi 468-8514, Japan
| | - Mamoru Koketsu
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
8
|
Ali RAH, Altimimi M, Hadi NR. The potential renoprotective effect of Raloxifene in renal ischemia-reperfusion injury in a male rat model. J Med Life 2023; 16:1274-1281. [PMID: 38024816 PMCID: PMC10652674 DOI: 10.25122/jml-2023-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/30/2023] [Indexed: 12/01/2023] Open
Abstract
Renal ischemia-reperfusion injury is caused by a temporary reduction in oxygen-carrying blood flow to the kidney, followed by reperfusion. During ischemia, kidney tissue damage induces overproduction of reactive oxygen species, which produces oxidative stress. The blood flow restoration during the reperfusion period causes further production of reactive oxygen species that ends with apoptosis and cell death. This study aimed to investigate the potential renoprotective effects of Raloxifene on bilateral renal ischemia-reperfusion injury in rats by looking into kidney function biomarkers, urea and creatinine, inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β). Additionally, antioxidant markers such as total antioxidant capacity (TAC) and the pro-apoptotic marker caspase-3 were assessed. Histopathological scores were also employed for evaluation. Our experimental design involved 20 rats divided into four groups: the sham group underwent median laparotomy without ischemia induction, the control group experienced bilateral renal ischemia for 30 minutes followed by 2 hours of reperfusion, the vehicle group received pretreatment with a mixture of corn oil and dimethyl sulfoxide (DMSO) before ischemia induction, and the Raloxifene-treated group was administered Raloxifene at a dose of 10 mg/kg before ischemia induction, followed by ischemia-reperfusion. Urea and creatinine, TNF-α, IL-1β, and caspase-3 in the Raloxifene group were significantly lower compared to the control and vehicle groups. On the other hand, TAC levels in the Raloxifene group were significantly higher than in the control and vehicle groups. This study concluded that Raloxifene had a renoprotective impact via multiple actions as an anti-inflammatory, anti-apoptotic, and antioxidant agent.
Collapse
Affiliation(s)
- Raghad Abdul Hameed Ali
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Murooj Altimimi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| |
Collapse
|
9
|
Wen J, Zhang X, Rush E, Panickan VA, Li X, Cai T, Zhou D, Ho YL, Costa L, Begoli E, Hong C, Gaziano JM, Cho K, Lu J, Liao KP, Zitnik M, Cai T. Multimodal representation learning for predicting molecule-disease relations. Bioinformatics 2023; 39:btad085. [PMID: 36805623 PMCID: PMC9940625 DOI: 10.1093/bioinformatics/btad085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/23/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
MOTIVATION Predicting molecule-disease indications and side effects is important for drug development and pharmacovigilance. Comprehensively mining molecule-molecule, molecule-disease and disease-disease semantic dependencies can potentially improve prediction performance. METHODS We introduce a Multi-Modal REpresentation Mapping Approach to Predicting molecular-disease relations (M2REMAP) by incorporating clinical semantics learned from electronic health records (EHR) of 12.6 million patients. Specifically, M2REMAP first learns a multimodal molecule representation that synthesizes chemical property and clinical semantic information by mapping molecule chemicals via a deep neural network onto the clinical semantic embedding space shared by drugs, diseases and other common clinical concepts. To infer molecule-disease relations, M2REMAP combines multimodal molecule representation and disease semantic embedding to jointly infer indications and side effects. RESULTS We extensively evaluate M2REMAP on molecule indications, side effects and interactions. Results show that incorporating EHR embeddings improves performance significantly, for example, attaining an improvement over the baseline models by 23.6% in PRC-AUC on indications and 23.9% on side effects. Further, M2REMAP overcomes the limitation of existing methods and effectively predicts drugs for novel diseases and emerging pathogens. AVAILABILITY AND IMPLEMENTATION The code is available at https://github.com/celehs/M2REMAP, and prediction results are provided at https://shiny.parse-health.org/drugs-diseases-dev/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jun Wen
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Xiang Zhang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Everett Rush
- Department of Energy, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Vidul A Panickan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Xingyu Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Tianrun Cai
- VA Boston Healthcare System, Boston, MA 02130, USA
- Mass General Brigham, Boston, MA 02130, USA
| | - Doudou Zhou
- Department of Statistics, University of California, Davis, CA 95616, USA
| | - Yuk-Lam Ho
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Lauren Costa
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Edmon Begoli
- Department of Energy, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Chuan Hong
- VA Boston Healthcare System, Boston, MA 02130, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708, USA
| | - J Michael Gaziano
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
- Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Kelly Cho
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
- Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Junwei Lu
- VA Boston Healthcare System, Boston, MA 02130, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Katherine P Liao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Data Science Initiative, Cambridge, MA 02138, USA
| | - Tianxi Cai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
- Mass General Brigham, Boston, MA 02130, USA
| |
Collapse
|
10
|
Jeong K, Chang J, Park SM, Kim J, Jeon S, Kim DH, Kim YE, Lee JC, Im S, Jo Y, Min JY, Lee H, Yeom M, Seok SH, On DI, Noh H, Yun JW, Park JW, Song D, Seong JK, Kim KC, Lee JY, Park HJ, Kim S, Nam TG, Lee W. Rapid discovery and classification of inhibitors of coronavirus infection by pseudovirus screen and amplified luminescence proximity homogeneous assay. Antiviral Res 2023; 209:105473. [PMID: 36435212 PMCID: PMC9682871 DOI: 10.1016/j.antiviral.2022.105473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022]
Abstract
To identify potent antiviral compounds, we introduced a high-throughput screen platform that can rapidly classify hit compounds according to their target. In our platform, we performed a compound screen using a lentivirus-based pseudovirus presenting a spike protein of coronavirus, and we evaluated the hit compounds using an amplified luminescence proximity homogeneous assay (alpha) test with purified host receptor protein and the receptor binding domain of the viral spike. With our screen platform, we were able to identify both spike-specific compounds (class I) and broad-spectrum antiviral compounds (class II). Among the hit compounds, thiosemicarbazide was identified to be selective to the interaction between the viral spike and its host cell receptor, and we further optimized the binding potency of thiosemicarbazide through modification of the pyridine group. Among the class II compounds, we found raloxifene and amiodarone to be highly potent against human coronaviruses including Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2. In particular, using analogs of the benzothiophene moiety, which is also present in raloxifene, we have identified benzothiophene as a novel structural scaffold for broad-spectrum antivirals. This work highlights the strong utility of our screen platform using a pseudovirus assay and an alpha test for rapid identification of potential antiviral compounds and their mechanism of action, which can lead to the accelerated development of therapeutics against newly emerging viral infections.
Collapse
Affiliation(s)
- Kwiwan Jeong
- Bio-center, Gyeonggido Business and Science Accelerator, Suwon, South Korea,Corresponding author
| | - JuOae Chang
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sun-mi Park
- Bio-center, Gyeonggido Business and Science Accelerator, Suwon, South Korea
| | - Jinhee Kim
- Institut Pasteur Korea, Seongnam, South Korea
| | | | - Dong Hwan Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Young-Eui Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Joo Chan Lee
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Somyoung Im
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Yejin Jo
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | | | - Hanbyeul Lee
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Minjoo Yeom
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, South Korea
| | - Da In On
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Hyuna Noh
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, South Korea
| | - Daesub Song
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea,Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, South Korea
| | - Kyung-Chang Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Joo-Yeon Lee
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Hyun-Ju Park
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea,Corresponding author
| | - Seungtaek Kim
- Institut Pasteur Korea, Seongnam, South Korea,Corresponding author
| | - Tae-gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea,Corresponding author
| | - Wonsik Lee
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea,Corresponding author
| |
Collapse
|
11
|
Solis O, Beccari AR, Iaconis D, Talarico C, Ruiz-Bedoya CA, Nwachukwu JC, Cimini A, Castelli V, Bertini R, Montopoli M, Cocetta V, Borocci S, Prandi IG, Flavahan K, Bahr M, Napiorkowski A, Chillemi G, Ooka M, Yang X, Zhang S, Xia M, Zheng W, Bonaventura J, Pomper MG, Hooper JE, Morales M, Rosenberg AZ, Nettles KW, Jain SK, Allegretti M, Michaelides M. The SARS-CoV-2 spike protein binds and modulates estrogen receptors. SCIENCE ADVANCES 2022; 8:eadd4150. [PMID: 36449624 PMCID: PMC9710872 DOI: 10.1126/sciadv.add4150] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein binds angiotensin-converting enzyme 2 as its primary infection mechanism. Interactions between S and endogenous proteins occur after infection but are not well understood. We profiled binding of S against >9000 human proteins and found an interaction between S and human estrogen receptor α (ERα). Using bioinformatics, supercomputing, and experimental assays, we identified a highly conserved and functional nuclear receptor coregulator (NRC) LXD-like motif on the S2 subunit. In cultured cells, S DNA transfection increased ERα cytoplasmic accumulation, and S treatment induced ER-dependent biological effects. Non-invasive imaging in SARS-CoV-2-infected hamsters localized lung pathology with increased ERα lung levels. Postmortem lung experiments from infected hamsters and humans confirmed an increase in cytoplasmic ERα and its colocalization with S in alveolar macrophages. These findings describe the discovery of a S-ERα interaction, imply a role for S as an NRC, and advance knowledge of SARS-CoV-2 biology and coronavirus disease 2019 pathology.
Collapse
Affiliation(s)
- Oscar Solis
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | | | | | | | - Camilo A. Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD 21287, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jerome C. Nwachukwu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, Philadelphia, PA 19122, USA
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | | | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- VIMM- Veneto Institute of Molecular Medicine, Fondazione per la Ricerca Biomedica Avanzata, Padova, Italy
| | - Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Stefano Borocci
- Department for Innovation in Biological, Agro-Food and Forest Systems, DIBAF, University of Tuscia, Viterbo, Italy
| | - Ingrid G. Prandi
- Department for Innovation in Biological, Agro-Food and Forest Systems, DIBAF, University of Tuscia, Viterbo, Italy
| | - Kelly Flavahan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD 21287, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Melissa Bahr
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD 21287, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Anna Napiorkowski
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD 21287, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-Food and Forest Systems, DIBAF, University of Tuscia, Viterbo, Italy
| | - Masato Ooka
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD 20850, USA
| | - Xiaoping Yang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shiliang Zhang
- Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD 20850, USA
| | - Wei Zheng
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD 20850, USA
| | - Jordi Bonaventura
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat, Catalonia, Spain
| | - Martin G. Pomper
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jody E. Hooper
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marisela Morales
- Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kendall W. Nettles
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD 21287, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Marcello Allegretti
- Dompé farmaceutici S.p.A, L’Aquila, Italy
- Corresponding author. (M.M.); (M.A.)
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Corresponding author. (M.M.); (M.A.)
| |
Collapse
|
12
|
Nandi S, Nayak BS, Khede MK, Saxena AK. Repurposing of Chemotherapeutics to Combat COVID-19. Curr Top Med Chem 2022; 22:2660-2694. [PMID: 36453483 DOI: 10.2174/1568026623666221130142517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/16/2022] [Accepted: 10/06/2022] [Indexed: 12/05/2022]
Abstract
Severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) is a novel strain of SARS coronavirus. The COVID-19 disease caused by this virus was declared a pandemic by the World Health Organization (WHO). SARS-CoV-2 mainly spreads through droplets sprayed by coughs or sneezes of the infected to a healthy person within the vicinity of 6 feet. It also spreads through asymptomatic carriers and has negative impact on the global economy, security and lives of people since 2019. Numerous lives have been lost to this viral infection; hence there is an emergency to build up a potent measure to combat SARS-CoV-2. In view of the non-availability of any drugs or vaccines at the time of its eruption, the existing antivirals, antibacterials, antimalarials, mucolytic agents and antipyretic paracetamol were used to treat the COVID-19 patients. Still there are no specific small molecule chemotherapeutics available to combat COVID-19 except for a few vaccines approved for emergency use only. Thus, the repurposing of chemotherapeutics with the potential to treat COVID-19 infected people is being used. The antiviral activity for COVID-19 and biochemical mechanisms of the repurposed drugs are being explored by the biological assay screening and structure-based in silico docking simulations. The present study describes the various US-FDA approved chemotherapeutics repositioned to combat COVID-19 along with their screening for biological activity, pharmacokinetic and pharmacodynamic evaluation.
Collapse
Affiliation(s)
- Sisir Nandi
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research, Affiliated to Uttarakhand Technical University, Kashipur, 244713, India
| | - Bhabani Shankar Nayak
- Department of Pharmaceutics, Institute of Pharmacy and Technology, Salipur, Affiliated to Biju Patnaik University of Technology, Odisha, 754202, India
| | - Mayank Kumar Khede
- Department of Pharmaceutics, Institute of Pharmacy and Technology, Salipur, Affiliated to Biju Patnaik University of Technology, Odisha, 754202, India
| | - Anil Kumar Saxena
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research, Affiliated to Uttarakhand Technical University, Kashipur, 244713, India
| |
Collapse
|
13
|
Jeon H, Lim Y, Lee IG, Kim DI, Kim KP, Hong SH, Kim J, Jung YS, Seo YJ. Inhibition of KIF20A suppresses the replication of influenza A virus by inhibiting viral entry. J Microbiol 2022; 60:1113-1121. [PMID: 36318360 DOI: 10.1007/s12275-022-2436-x] [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: 09/28/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
The influenza A virus (IAV) has caused several pandemics, and therefore there are many ongoing efforts to identify novel antiviral therapeutic strategies including vaccines and antiviral drugs. However, influenza viruses continuously undergo antigenic drift and shift, resulting in the emergence of mutated viruses. In turn, this decreases the efficiency of existing vaccines and antiviral drugs to control IAV infection. Therefore, this study sought to identify alternative therapeutic strategies targeting host cell factors rather than viruses to avoid infection by mutated viruses. Particularly, we investigated the role of KIF20A that is one of kinesin superfamily proteins in the replication of IAV. The KIF20A increased viral protein levels in IAV-infected cells by regulating the initial entry stage during viral infection. Furthermore, the KIF20A inhibitor significantly suppressed viral replication, which protected mice from morbidity and mortality. Therefore, our findings demonstrated that KIF20A is highly involved in the viral replication process and viral propagation both in vitro and in vivo, and could thus be used as a target for the development of novel antiviral drugs.
Collapse
Affiliation(s)
- Hoyeon Jeon
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Younghyun Lim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - In-Gu Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dong-In Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Keun Pil Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea
| | - Jeongkyu Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Youn-Sang Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
14
|
Yang D, Li H, Chen Y, Ren W, Dong M, Li C, Jiao Q. Immunomodulatory mechanisms of abatacept: A therapeutic strategy for COVID-19. Front Med (Lausanne) 2022; 9:951115. [PMID: 35957855 PMCID: PMC9357915 DOI: 10.3389/fmed.2022.951115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by coronavirus-2 (SARS-CoV-2) infection has rapidly spread throughout the world and become a major threat to human beings. Cytokine storm is a major cause of death in severe patients. Abatacept can suppress cytokines used as antirheumatic drugs in clinical applications. This study analyzed the molecular mechanisms of abatacept treatment for COVID-19. Differentially expressed genes (DEGs) were identified by analyzing expression profiling of abatacept treatment for rheumatoid arthritis (RA) patients and SARS-CoV-2 infection patients. We found that 59 DEGs were upregulated in COVID-19 patients and downregulated following abatacept treatment. Gene set enrichment analysis (GSEA) and Gene Ontology (GO) analysis showed that immune and inflammatory responses were potential regulatory mechanisms. Moreover, we verified 8 targeting genes and identified 15 potential drug candidates for the treatment of COVID-19. Our study illustrated that abatacept could be a promising property for preventing severe COVID-19, and we predicted alternative potential drugs for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Dinglong Yang
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Hetong Li
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yujing Chen
- School of Public Health, Xi'an Jiaotong University, Xi'an, China
| | - Weiping Ren
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Mingjie Dong
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Chunjiang Li
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Qiang Jiao
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Qiang Jiao
| |
Collapse
|
15
|
K S NS, Dengale SJ, Mutalik S, Bhat K. Raloxifene HCl – Quercetin Co-amorphous System: Preparation, Characterization, and Investigation of its Behavior in Phosphate Buffer. Drug Dev Ind Pharm 2022; 48:227-238. [DOI: 10.1080/03639045.2022.2104308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Navya Sree K S
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India.
| | - Swapnil J Dengale
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India.
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research (NIPER) Guwahati, Assam-781101, India.
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Krishnamurthy Bhat
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India.
| |
Collapse
|
16
|
Nicastri E, Marinangeli F, Pivetta E, Torri E, Reggiani F, Fiorentino G, Scorzolini L, Vettori S, Marsiglia C, Gavioli EM, Beccari AR, Terpolilli G, De Pizzol M, Goisis G, Mantelli F, Vaia F, Allegretti M. A phase 2 randomized, double-blinded, placebo-controlled, multicenter trial evaluating the efficacy and safety of raloxifene for patients with mild to moderate COVID-19. EClinicalMedicine 2022; 48:101450. [PMID: 35582123 PMCID: PMC9098200 DOI: 10.1016/j.eclinm.2022.101450] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Current available therapeutic options for Coronavirus Disease-2019 (COVID-19) are primarily focused on treating hospitalized patients, and there is a lack of oral therapeutic options to treat mild to moderate outpatient COVID-19 and prevent clinical progression. Raloxifene was found as a promising molecule to treat COVID-19 due to its activity to modulate the replication of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and act as an immunomodulator to decrease proinflammatory cytokines. METHODS This was a phase 2 multicenter, randomized, placebo-controlled trial to evaluate the efficacy and safety of raloxifene in adult patients with mild to moderate COVID-19 between October 2020 to June 2021 in five centers located in Italy. This was a planned 2/3 adaptive study, but due to operational difficulties, the study was discontinued during the phase 2 study segment. Participants were randomized 1:1:1 to receive oral placebo, raloxifene 60 mg, or raloxifene 120 mg by self-administration for a maximum of two weeks. The primary outcomes were the proportion of patients with undetectable SARS-CoV-2 via nasopharyngeal swabs at day 7 and the proportion of patients who did not require supplemental oxygen therapy or mechanical ventilation on day 14. Safety was assessed. The trial is registered (EudraCT 2021-002,476-39, and ClinicalTrials.gov: NCT05172050). FINDINGS A total of 68 participants were enrolled and randomized to placebo (n = 21), raloxifene 60 mg (n = 24), and raloxifene 120 mg (n = 23). The proportion of participants with undetectable SARS-CoV-2 after seven days of treatment with raloxifene 60 mg [36.8%, 7/19 vs. 0.0%, 0/14] and 120 mg [22.2%, 4/18 vs. 0.0%, 0/14] was better compared to placebo, [risk difference (RD) = 0·37 (95% C.I.:0·09-0·59)] and [RD = 0·22 (95% C.I.: -0·03-0·45)], respectively. There was no evidence of effect for requirement of supplemental oxygen and/or mechanical ventilation with effects for raloxifene 60 mg and raloxifene 120 mg over placebo, [RD = 0·09 (95% C.I.: -0·22-0·37)], and [RD = 0·03 (95% C.I.: -0·28-0·33)], respectively. Raloxifene was well tolerated at both doses, and there was no evidence of any difference in the occurrence of serious adverse events. INTERPRETATION Raloxifene showed evidence of effect in the primary virologic endpoint in the treatment of early mild to moderate COVID-19 patients shortening the time of viral shedding. The safety profile was consistent with that reported for other indications. Raloxifene may represent a promising pharmacological option to prevent or mitigate COVID-19 disease progression. FUNDING The study was funded by Dompé Farmaceutici SpA and supported by the funds from the European Commission - Health and Consumers Directorate General, for the Action under the Emergency Support Instrument- Grant to support clinical testing of repurposed medicines to treat SARS-COV-2 patients (PPPA-ESI-CTRM-2020-SI2.837140), and by the COVID-2020-12,371,675 Ricerca finalizzata and line 1 Ricerca Corrente COVID both funded by Italian Ministry of Health.
Collapse
Affiliation(s)
- Emanuele Nicastri
- National Institute for Infectious Diseases, Lazzaro Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy
| | - Franco Marinangeli
- Department of Anesthesiology, Intensive Care, and Pain Therapy, University of L'Aquila, L'Aquila, Italy
| | - Emanuele Pivetta
- Division of Emergency Medicine and High Dependency Unit, Department of General and Specialized Medicine, Città della Salute e della Scienza di Torino University Hospital, Turin, Italy
| | - Elena Torri
- Emergency Department- Emergency Center, Humanitas Gavazzeni, Bergamo, Italy
| | - Francesco Reggiani
- U.O. di Medicina Generale e Nefrologia–IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Giuseppe Fiorentino
- Respiratory Physiopathology and Rehabilitation Unit, AO Ospedali dei Colli, PO Monaldi Hospital, Naples, Italy
| | - Laura Scorzolini
- National Institute for Infectious Diseases, Lazzaro Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy
| | - Serena Vettori
- Respiratory Physiopathology and Rehabilitation Unit, AO Ospedali dei Colli, PO Monaldi Hospital, Naples, Italy
| | | | | | - Andrea R. Beccari
- Dompé farmaceutici SpA, EXSCALATE Labs, Via Tommaso De Amicis, 95, Napoli 80131, Italy
| | | | - Maria De Pizzol
- Dompé farmaceutici SpA, Via Santa Lucia, 6, Milan 20122, Italy
| | - Giovanni Goisis
- Dompé farmaceutici SpA, Via Santa Lucia, 6, Milan 20122, Italy
| | - Flavio Mantelli
- Dompé farmaceutici SpA, Via Campo di Pile, SNC, L'Aquila, Italy
- Corresponding author.
| | - Francesco Vaia
- National Institute for Infectious Diseases, Lazzaro Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy
| | | | | |
Collapse
|
17
|
Iaconis D, Bordi L, Matusali G, Talarico C, Manelfi C, Cesta MC, Zippoli M, Caccuri F, Bugatti A, Zani A, Filippini F, Scorzolini L, Gobbi M, Beeg M, Piotti A, Montopoli M, Cocetta V, Bressan S, Bucci EM, Caruso A, Nicastri E, Allegretti M, Beccari AR. Characterization of raloxifene as a potential pharmacological agent against SARS-CoV-2 and its variants. Cell Death Dis 2022; 13:498. [PMID: 35614039 PMCID: PMC9130985 DOI: 10.1038/s41419-022-04961-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
Abstract
The new coronavirus SARS-CoV-2 is the causative agent of the COVID-19 pandemic, which so far has caused over 6 million deaths in 2 years, despite new vaccines and antiviral medications. Drug repurposing, an approach for the potential application of existing pharmaceutical products to new therapeutic indications, could be an effective strategy to obtain quick answers to medical emergencies. Following a virtual screening campaign on the most relevant viral proteins, we identified the drug raloxifene, a known Selective Estrogen Receptor Modulator (SERM), as a new potential agent to treat mild-to-moderate COVID-19 patients. In this paper we report a comprehensive pharmacological characterization of raloxifene in relevant in vitro models of COVID-19, specifically in Vero E6 and Calu-3 cell lines infected with SARS-CoV-2. A large panel of the most common SARS-CoV-2 variants isolated in Europe, United Kingdom, Brazil, South Africa and India was tested to demonstrate the drug's ability in contrasting the viral cytopathic effect (CPE). Literature data support a beneficial effect by raloxifene against the viral infection due to its ability to interact with viral proteins and activate protective estrogen receptor-mediated mechanisms in the host cells. Mechanistic studies here reported confirm the significant affinity of raloxifene for the Spike protein, as predicted by in silico studies, and show that the drug treatment does not directly affect Spike/ACE2 interaction or viral internalization in infected cell lines. Interestingly, raloxifene can counteract Spike-mediated ADAM17 activation in human pulmonary cells, thus providing new insights on its mechanism of action. A clinical study in mild to moderate COVID-19 patients (NCT05172050) has been recently completed. Our contribution to evaluate raloxifene results on SARS-CoV-2 variants, and the interpretation of the mechanisms of action will be key elements to better understand the trial results, and to design new clinical studies aiming to evaluate the potential development of raloxifene in this indication.
Collapse
Affiliation(s)
| | - Licia Bordi
- grid.419423.90000 0004 1760 4142National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Giulia Matusali
- grid.419423.90000 0004 1760 4142National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | | | | | | | | | - Francesca Caccuri
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Antonella Bugatti
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Alberto Zani
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Federica Filippini
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Laura Scorzolini
- grid.419423.90000 0004 1760 4142National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Marco Gobbi
- grid.4527.40000000106678902Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marten Beeg
- grid.4527.40000000106678902Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Arianna Piotti
- grid.4527.40000000106678902Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Monica Montopoli
- grid.5608.b0000 0004 1757 3470Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, VIMM Veneto Institute Molecular Medicine, Padua, Italy
| | - Veronica Cocetta
- grid.5608.b0000 0004 1757 3470Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, VIMM Veneto Institute Molecular Medicine, Padua, Italy
| | - Silvia Bressan
- grid.5608.b0000 0004 1757 3470Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, VIMM Veneto Institute Molecular Medicine, Padua, Italy
| | - Enrico M. Bucci
- grid.264727.20000 0001 2248 3398Sbarro Health Research Organization, Biology Department CFT, Temple University, Philadelphia, PA USA
| | - Arnaldo Caruso
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Emanuele Nicastri
- grid.419423.90000 0004 1760 4142National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | | | | |
Collapse
|
18
|
Solis O, Beccari AR, Iaconis D, Talarico C, Ruiz-Bedoya CA, Nwachukwu JC, Cimini A, Castelli V, Bertini R, Montopoli M, Cocetta V, Borocci S, Prandi IG, Flavahan K, Bahr M, Napiorkowski A, Chillemi G, Ooka M, Yang X, Zhang S, Xia M, Zheng W, Bonaventura J, Pomper MG, Hooper JE, Morales M, Rosenberg AZ, Nettles KW, Jain SK, Allegretti M, Michaelides M. The SARS-CoV-2 spike protein binds and modulates estrogen receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.05.21.492920. [PMID: 35665018 PMCID: PMC9164441 DOI: 10.1101/2022.05.21.492920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein binds angiotensin-converting enzyme 2 (ACE2) at the cell surface, which constitutes the primary mechanism driving SARS-CoV-2 infection. Molecular interactions between the transduced S and endogenous proteins likely occur post-infection, but such interactions are not well understood. We used an unbiased primary screen to profile the binding of full-length S against >9,000 human proteins and found significant S-host protein interactions, including one between S and human estrogen receptor alpha (ERα). After confirming this interaction in a secondary assay, we used bioinformatics, supercomputing, and experimental assays to identify a highly conserved and functional nuclear receptor coregulator (NRC) LXD-like motif on the S2 subunit and an S-ERα binding mode. In cultured cells, S DNA transfection increased ERα cytoplasmic accumulation, and S treatment induced ER-dependent biological effects and ACE2 expression. Noninvasive multimodal PET/CT imaging in SARS-CoV-2-infected hamsters using [ 18 F]fluoroestradiol (FES) localized lung pathology with increased ERα lung levels. Postmortem experiments in lung tissues from SARS-CoV-2-infected hamsters and humans confirmed an increase in cytoplasmic ERα expression and its colocalization with S protein in alveolar macrophages. These findings describe the discovery and characterization of a novel S-ERα interaction, imply a role for S as an NRC, and are poised to advance knowledge of SARS-CoV-2 biology, COVID-19 pathology, and mechanisms of sex differences in the pathology of infectious disease.
Collapse
Affiliation(s)
- Oscar Solis
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, 21224, MD, USA
| | | | | | | | - Camilo A. Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jerome C. Nwachukwu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, Philadelphia, PA, USA
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | | | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- VIMM- Veneto Institute of Molecular Medicine, Fondazione per la Ricerca Biomedica Avanzata, Padova, Italy
| | - Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Stefano Borocci
- Department for Innovation in Biological, Agro-Food and Forest Systems, DIBAF, University of Tuscia, Viterbo, Italy
| | - Ingrid G. Prandi
- Department for Innovation in Biological, Agro-Food and Forest Systems, DIBAF, University of Tuscia, Viterbo, Italy
| | - Kelly Flavahan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Melissa Bahr
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna Napiorkowski
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-Food and Forest Systems, DIBAF, University of Tuscia, Viterbo, Italy
| | - Masato Ooka
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Xiaoping Yang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shiliang Zhang
- Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, 21224, MD, USA
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Wei Zheng
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Jordi Bonaventura
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat, Catalonia
| | - Martin G. Pomper
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jody E. Hooper
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marisela Morales
- Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, 21224, MD, USA
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kendall W. Nettles
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, 21224, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Hasan MT, Abdulrazak LF, Alam MK, Islam MR, Sathi YH, Al-Zahrani FA, Ahmed K, Bui FM, Moni MA. Discovering Common Pathophysiological Processes between COVID-19 and Cystic Fibrosis by Differential Gene Expression Pattern Analysis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8078259. [PMID: 35528173 PMCID: PMC9076317 DOI: 10.1155/2022/8078259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/04/2022] [Indexed: 12/12/2022]
Abstract
Coronaviruses are a family of viruses that infect mammals and birds. Coronaviruses cause infections of the respiratory system in humans, which can be minor or fatal. A comparative transcriptomic analysis has been performed to establish essential profiles of the gene expression of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) linked to cystic fibrosis (CF). Transcriptomic studies have been carried out in relation to SARS-CoV-2 since a number of people have been diagnosed with CF. The recognition of differentially expressed genes demonstrated 8 concordant genes shared between the SARS-CoV-2 and CF. Extensive gene ontology analysis and the discovery of pathway enrichment demonstrated SARS-CoV-2 response to CF. The gene ontological terms and pathway enrichment mechanisms derived from this research may affect the production of successful drugs, especially for the people with the following disorder. Identification of TF-miRNA association network reveals the interconnection between TF genes and miRNAs, which may be effective to reveal the other influenced disease that occurs for SARS-CoV-2 to CF. The enrichment of pathways reveals SARS-CoV-2-associated CF mostly engaged with the type of innate immune system, Toll-like receptor signaling pathway, pantothenate and CoA biosynthesis, allograft rejection, graft-versus-host disease, intestinal immune network for IgA production, mineral absorption, autoimmune thyroid disease, legionellosis, viral myocarditis, inflammatory bowel disease (IBD), etc. The drug compound identification demonstrates that the drug targets of IMIQUIMOD and raloxifene are the most significant with the significant hub DEGs.
Collapse
Affiliation(s)
- Md. Tanvir Hasan
- Department of Software Engineering, Daffodil International University (DIU), Ashulia, Savar, Dhaka 1341, Bangladesh
| | - Lway Faisal Abdulrazak
- Department of Computer Science, Cihan University Sulaimaniya, Sulaimaniya, 46001 Kurdistan Region, Iraq
| | - Mohammad Khursheed Alam
- Preventive Dentistry Department, College of Dentistry, Jouf University, Sakaka 72345, Saudi Arabia
- Center for Transdisciplinary Research (CFTR), Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Public Health, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Rezwan Islam
- Department of Software Engineering, Daffodil International University (DIU), Ashulia, Savar, Dhaka 1341, Bangladesh
| | - Yeasmin Hena Sathi
- Department of Software Engineering, Daffodil International University (DIU), Ashulia, Savar, Dhaka 1341, Bangladesh
| | | | - Kawsar Ahmed
- Department of Electrical and Computer Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, SK, Canada S7N 5A9
- Group of Bio-photomatix, Department of Information and Communication Technology, Mawlana Bhashani Science and Technology University (MBSTU), Santosh, Tangail 1902, Bangladesh
| | - Francis M. Bui
- Department of Electrical and Computer Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, SK, Canada S7N 5A9
| | - Mohammad Ali Moni
- School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
20
|
Prinelli F, Trevisan C, Noale M, Franchini M, Giacomelli A, Cori L, Jesuthasan N, Incalzi RA, Maggi S, Adorni F. Sex- and gender-related differences linked to SARS-CoV-2 infection among the participants in the web-based EPICOVID19 survey: the hormonal hypothesis. Maturitas 2022; 158:61-69. [PMID: 35241240 PMCID: PMC8642247 DOI: 10.1016/j.maturitas.2021.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/15/2021] [Accepted: 11/30/2021] [Indexed: 01/08/2023]
Abstract
Objective To investigate sex- and gender-based differences linked to SARS-COV-2 infection and to explore the role of hormonal therapy (HT) in females. Study design Data from the self-administered, cross-sectional, web-based EPICOVID19 survey of 198,822 adults living in Italy who completed an online questionnaire during the first wave of the epidemic in Italy (April-May 2020) were analyzed. Main outcomes measures Multivariate binary logistic and multinomial regression models were respectively used to estimate the odds ratios (ORs) with 95% confidence intervals (CIs) for positive nasopharyngeal swab (NPS) test results and severe SARS-CoV-2 infection. Results The data from 6,873 participants (mean age 47.9 ± 14.1 years, 65.8% females) who had a known result from an NPS test were analyzed. According to the multivariate analysis, females had lower odds of a positive result from the NPS test (aOR 0.75, 95%CI 0.66–0.85) and of having a severe infection (aOR 0.46, 95%CI 0.37–0.57) than did their male counterparts. These differences were greater with decreasing age in both sexes. In addition, females aged ≥60 years receiving HT (N = 2,153, 47.6%) had a 46% lower probability of having a positive NPS test (aOR 0.54, 95%CI 0.36–0.80) than their same-aged peers who had never used HT; there were no differences in the younger age groups with respect to HT status. Conclusion Female sex was associated with an age-dependent lower risk of having a severe SARS-CoV-2 infection than their male counterparts. Age seemed to modify the relationship between HT status and infection: while the two were not related among younger participants, it was negative in the older ones. Future prospective studies are needed to elucidate the potential protective role sex hormones may play. Trial registration ClinicalTrials.gov NCT04471701.
Collapse
Affiliation(s)
- Federica Prinelli
- National Research Council, Institute of Biomedical Technologies, Via Fratelli Cervi 93, Segrate, MI 20090, Italy.
| | - Caterina Trevisan
- National Research Council, Neuroscience Institute, Aging Branch, Via Vincenzo Maria Gallucci 16, Padova 35128, Italy
| | - Marianna Noale
- National Research Council, Neuroscience Institute, Aging Branch, Via Vincenzo Maria Gallucci 16, Padova 35128, Italy
| | - Michela Franchini
- National Research Council, Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa, PI 56124, Italy
| | - Andrea Giacomelli
- Infectious Diseases Unit, Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, ASST Fatebenefratelli Sacco, Milan 20157, Italy
| | - Liliana Cori
- National Research Council, Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa, PI 56124, Italy
| | - Nithiya Jesuthasan
- National Research Council, Institute of Biomedical Technologies, Via Fratelli Cervi 93, Segrate, MI 20090, Italy
| | - Raffaele Antonelli Incalzi
- Unit of Geriatrics, Department of Medicine, Biomedical Campus of Rome, via Alvaro del Portillo, 21, Rome 00128, Italy
| | - Stefania Maggi
- National Research Council, Neuroscience Institute, Aging Branch, Via Vincenzo Maria Gallucci 16, Padova 35128, Italy
| | - Fulvio Adorni
- National Research Council, Institute of Biomedical Technologies, Via Fratelli Cervi 93, Segrate, MI 20090, Italy
| |
Collapse
|
21
|
Mechanism of Caspase-1 Inhibition by Four Anti-inflammatory Drugs Used in COVID-19 Treatment. Int J Mol Sci 2022; 23:ijms23031849. [PMID: 35163769 PMCID: PMC8837144 DOI: 10.3390/ijms23031849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/27/2023] Open
Abstract
The inflammatory protease caspase-1 is associated with the release of cytokines. An excessive number of cytokines (a “cytokine storm”) is a dangerous consequence of COVID-19 infection and has been indicated as being among the causes of death by COVID-19. The anti-inflammatory drug colchicine (which is reported in the literature to be a caspase-1 inhibitor) and the corticosteroid drugs, dexamethasone and methylprednisolone, are among the most effective active compounds for COVID-19 treatment. The SERM raloxifene has also been used as a repurposed drug in COVID-19 therapy. In this study, inhibition of caspase-1 by these four compounds was analyzed using computational methods. Our aim was to see if the inhibition of caspase-1, an important biomolecule in the inflammatory response that triggers cytokine release, could shed light on how these drugs help to alleviate excessive cytokine production. We also measured the antioxidant activities of dexamethasone and colchicine when scavenging the superoxide radical using cyclic voltammetry methods. The experimental findings are associated with caspase-1 active site affinity towards these compounds. In evaluating our computational and experimental results, we here formulate a mechanism for caspase-1 inhibition by these drugs, which involves the active site amino acid Cys285 residue and is mediated by a transfer of protons, involving His237 and Ser339. It is proposed that the molecular moiety targeted by all of these drugs is a carbonyl group which establishes a S(Cys285)–C(carbonyl) covalent bond.
Collapse
|
22
|
Santiago C, Jiménez-Aberasturi X, Leicea E, Lete MG, Sotomayor N, Lete E. Microwave-assisted palladium catalysed C-H acylation with aldehydes: synthesis and diversification of 3-acylthiophenes. Org Biomol Chem 2022; 20:852-861. [PMID: 35001098 DOI: 10.1039/d1ob02176a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of MW allows the efficient palladium(II)-catalysed C-3 acylation of thiophenes with aldehydes via C(sp2)-H activation for the synthesis of (cyclo)alkyl/aryl thienyl ketones (43 examples). Compared to standard thermal conditions, the use of MW reduces the reaction time (15 to 30 min vs. 1 to 3 hours), leading to improved yields of the ketones (up to 92%). The control of positional selectivity is achieved by 2-pyridinyl and 2-pyrimidyl ortho-directing groups at C-2 of the thiophene scaffold. To show the synthetic applicability, selected ketones were subjected to further transformations, including intramolecular reactions to directly embed the directing group in the core structure of the new molecule.
Collapse
Affiliation(s)
- Carlos Santiago
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Xabier Jiménez-Aberasturi
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Eztizen Leicea
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Marta G Lete
- CIC bioGUNE, Bizkaia Technology Park, Building 801A, Derio 48170, Spain
| | - Nuria Sotomayor
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Esther Lete
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| |
Collapse
|
23
|
OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1617-1624. [DOI: 10.1093/jac/dkac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/10/2022] [Indexed: 11/12/2022] Open
|
24
|
Hu S, Yin F, Nie L, Wang Y, Qin J, Chen J. Estrogen and Estrogen Receptor Modulators: Potential Therapeutic Strategies for COVID-19 and Breast Cancer. Front Endocrinol (Lausanne) 2022; 13:829879. [PMID: 35399920 PMCID: PMC8985365 DOI: 10.3389/fendo.2022.829879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/21/2022] [Indexed: 01/08/2023] Open
Abstract
Owing to the ongoing coronavirus disease 2019 (COVID-19) pandemic, we need to pay a particular focus on the impact of coronavirus infection on breast cancer patients. Approximately 70% of breast cancer patients express estrogen receptor (ER), and intervention therapy for ER has been the primary treatment strategy to prevent the development and metastasis of breast cancer. Recent studies have suggested that selective estrogen receptor modulators (SERMs) are a potential therapeutic strategy for COVID-19. With its anti-ER and anti-viral combined functions, SERMs may be an effective treatment for COVID-19 in patients with breast cancer. In this review, we explore the latent effect of SERMs, especially tamoxifen, and the mechanism between ER and virus susceptibility.
Collapse
Affiliation(s)
- Shuying Hu
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, China
| | - Feiying Yin
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, China
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Litao Nie
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Yuqin Wang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Jian Qin
- Department of Radiotherapy III, Clinical Oncology Canter, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- *Correspondence: Jian Qin, ; Jian Chen,
| | - Jian Chen
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, China
- Breast Center, the Second Affiliated Hospital of Guilin Medical University, Guilin, China
- *Correspondence: Jian Qin, ; Jian Chen,
| |
Collapse
|
25
|
Allegretti M, Cesta MC, Zippoli M, Beccari A, Talarico C, Mantelli F, Bucci EM, Scorzolini L, Nicastri E. Repurposing the estrogen receptor modulator raloxifene to treat SARS-CoV-2 infection. Cell Death Differ 2022; 29:156-166. [PMID: 34404919 PMCID: PMC8370058 DOI: 10.1038/s41418-021-00844-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/09/2021] [Accepted: 07/25/2021] [Indexed: 12/15/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) necessitates strategies to identify prophylactic and therapeutic drug candidates to enter rapid clinical development. This is particularly true, given the uncertainty about the endurance of the immune memory induced by both previous infections or vaccines, and given the fact that the eradication of SARS-CoV-2 might be challenging to reach, given the attack rate of the virus, which would require unusually high protection by a vaccine. Here, we show how raloxifene, a selective estrogen receptor modulator with anti-inflammatory and antiviral properties, emerges as an attractive candidate entering clinical trials to test its efficacy in early-stage treatment COVID-19 patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Enrico M Bucci
- Sbarro Health Research Organization, Biology Department CFT, Temple University, Philadelphia, PA, USA
| | - Laura Scorzolini
- Lazzaro Spallanzani National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Emanuele Nicastri
- Lazzaro Spallanzani National Institute for Infectious Diseases, IRCCS, Rome, Italy
| |
Collapse
|
26
|
Cromer SJ, Yu EW. Challenges and Opportunities for Osteoporosis Care During the COVID-19 Pandemic. J Clin Endocrinol Metab 2021; 106:e4795-e4808. [PMID: 34343287 PMCID: PMC8385842 DOI: 10.1210/clinem/dgab570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE The coronavirus disease 2019 (COVID-19) has both directly and indirectly affected osteoporosis diagnosis and treatment throughout the world. METHODS This mini-review summarizes the available evidence regarding the effects of COVID-19, its treatment, and the consequences of the pandemic itself on bone health. Additionally, we review evidence and expert recommendations regarding putative effects of osteoporosis medications on COVID-19 outcomes and vaccine efficacy and summarize recommendations for continuation of osteoporosis treatment during the pandemic. RESULTS The use of standard screening procedures to assess for osteoporosis and fracture risk declined dramatically early in the pandemic, while rates of fragility fractures were largely unchanged. COVID-19, its treatments, and public health measures to prevent viral spread are each likely to negatively affect bone health. Osteoporosis treatments are not known to increase risk of adverse events from COVID-19, and preclinical data suggest possible beneficial effects of some therapies. Vitamin D deficiency is clearly associated with adverse outcomes from COVID-19, but it remains unclear whether vitamin D supplementation may improve outcomes. Osteoporosis treatment should be continued whenever possible, and recommendations for substituting therapies, if required, are available. CONCLUSION The COVID-19 pandemic has decreased screening and disrupted treatment for osteoporosis. Osteoporosis medications are safe and effective during the pandemic and should be continued whenever possible. Further studies are needed to fully understand the impact of the COVID-19 pandemic on long-term bone health.
Collapse
Affiliation(s)
- Sara J Cromer
- Division of Endocrinology, Diabetes, and Metabolism; Massachusetts General Hospital; Boston, MA
- Harvard Medical School;Boston, MA
| | - Elaine W Yu
- Division of Endocrinology, Diabetes, and Metabolism; Massachusetts General Hospital; Boston, MA
- Harvard Medical School;Boston, MA
- Corresponding Author: Elaine W. Yu, MD, MGH Endocrine Unit, 50 Blossom Street, Thier 1051, Boston, MA 02114,
| |
Collapse
|
27
|
An JH, Li CY, Chen CY, Wu JB, Shen H. Raloxifene Protects Cisplatin-Induced Renal Injury in Mice via Inhibiting Oxidative Stress. Onco Targets Ther 2021; 14:4879-4890. [PMID: 34588782 PMCID: PMC8473575 DOI: 10.2147/ott.s314810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/04/2021] [Indexed: 12/28/2022] Open
Abstract
Purpose Cisplatin is one of the most widely used antineoplastic drugs but has limited therapeutic effects due to nephrotoxicity. The aim of this study was to determine the possible renoprotective effect of the antioxidant raloxifene on cisplatin-induced nephrotoxicity in mice. Materials and Methods Cisplatin-induced acute renal injury was established in female C57 mice that were treated with saline (normal control) or raloxifene over a 7-day period. The body weight of the mice was recorded. Histopathological examinations of the kidney tissues were performed using H&E, PAS staining and TEM. The histomorphology of liver and other organs was observed by H&E staining. The serum levels of creatinine, blood urea nitrogen (BUN), alanine transaminase (ALT) and glutamic oxalacetic transaminase (AST) were analyzed by specific kits. Superoxide dismutase (SOD) and glutathione (GSH) activity, and the content of malondialdehyde (MDA) in the kidney, liver homogenates and HK-2 cells were measured by WST-8 and thiobarbituric acid colorimetric methods. Moreover, the mitochondrial structures of HK-2 cells were performed using TEM. The viability and proliferation of HK-2 cells were examined by CCK-8 and EdU incorporation assays. The mitochondrial membrane potential was measured by JC-1 fluorescence. Results Raloxifene significantly reduced the levels of serum creatinine, urea, ALT and AST in the cisplatin-treated mice, and alleviated cisplatin-induced renal and hepatic tissue injury. Furthermore, raloxifene also increased the activity of GSH and SOD in the renal tissues and HK-2 cells, and reduced MDA levels, thereby limiting oxidative stress in the kidney. Conclusion Raloxifene protected against cisplatin-induced nephrotoxicity by activating the antioxidant system, along with alleviating liver damage. It should be considered as a potential adjuvant in cisplatin-based chemotherapeutic protocols.
Collapse
Affiliation(s)
- Jian-Hong An
- School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Chun-Yan Li
- School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Chun-Ya Chen
- School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Jian-Bin Wu
- Department of Oncology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Hong Shen
- School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| |
Collapse
|
28
|
Spini A, Giudice V, Brancaleone V, Morgese MG, De Francia S, Filippelli A, Ruggieri A, Ziche M, Ortona E, Cignarella A, Trabace L. Sex-tailored pharmacology and COVID-19: Next steps towards appropriateness and health equity. Pharmacol Res 2021; 173:105848. [PMID: 34454035 PMCID: PMC8387562 DOI: 10.1016/j.phrs.2021.105848] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/21/2021] [Accepted: 08/22/2021] [Indexed: 12/12/2022]
Abstract
Making gender bias visible allows to fill the gaps in knowledge and understand health records and risks of women and men. The coronavirus disease 2019 (COVID-19) pandemic has shown a clear gender difference in health outcomes. The more severe symptoms and higher mortality in men as compared to women are likely due to sex and age differences in immune responses. Age-associated decline in sex steroid hormone levels may mediate proinflammatory reactions in older adults, thereby increasing their risk of adverse outcomes, whereas sex hormones and/or sex hormone receptor modulators may attenuate the inflammatory response and provide benefit to COVID-19 patients. While multiple pharmacological options including anticoagulants, glucocorticoids, antivirals, anti-inflammatory agents and traditional Chinese medicine preparations have been tested to treat COVID-19 patients with varied levels of evidence in terms of efficacy and safety, information on sex-targeted treatment strategies is currently limited. Women may have more benefit from COVID-19 vaccines than men, despite the occurrence of more frequent adverse effects, and long-term safety data with newly developed vectors are eagerly awaited. The prevalent inclusion of men in randomized clinical trials (RCTs) with subsequent extrapolation of results to women needs to be addressed, as reinforcing sex-neutral claims into COVID-19 research may insidiously lead to increased inequities in health care. The huge worldwide effort with over 3000 ongoing RCTs of pharmacological agents should focus on improving knowledge on sex, gender and age as pillars of individual variation in drug responses and enforce appropriateness.
Collapse
Affiliation(s)
- Andrea Spini
- University of Siena, Department of Medicine, Surgery and Neuroscience, 53100 Siena, Italy; University of Bordeaux, Bordeaux Population Health Center, UMR 1219, 33000 Bordeaux, France
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Vincenzo Brancaleone
- Department of Science, University of Basilicata, via Ateneo Lucano, 85100 Potenza, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Silvia De Francia
- Department of Clinical and Biological Sciences, S. Luigi Hospital, University of Turin, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Anna Ruggieri
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Ziche
- University of Siena, Department of Medicine, Surgery and Neuroscience, 53100 Siena, Italy; University of Bordeaux, Bordeaux Population Health Center, UMR 1219, 33000 Bordeaux, France; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Elena Ortona
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Andrea Cignarella
- Department of Medicine, University of Padova, via Giustiniani 2, 35128 Padova, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy.
| |
Collapse
|
29
|
Mslati H, Gentile F, Perez C, Cherkasov A. Comprehensive Consensus Analysis of SARS-CoV-2 Drug Repurposing Campaigns. J Chem Inf Model 2021; 61:3771-3788. [PMID: 34313439 PMCID: PMC8340583 DOI: 10.1021/acs.jcim.1c00384] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Indexed: 01/18/2023]
Abstract
The current COVID-19 pandemic has elicited extensive repurposing efforts (both small and large scale) to rapidly identify COVID-19 treatments among approved drugs. Herein, we provide a literature review of large-scale SARS-CoV-2 antiviral drug repurposing efforts and highlight a marked lack of consistent potency reporting. This variability indicates the importance of standardizing best practices-including the use of relevant cell lines, viral isolates, and validated screening protocols. We further surveyed available biochemical and virtual screening studies against SARS-CoV-2 targets (Spike, ACE2, RdRp, PLpro, and Mpro) and discuss repurposing candidates exhibiting consistent activity across diverse, triaging assays and predictive models. Moreover, we examine repurposed drugs and their efficacy against COVID-19 and the outcomes of representative repurposed drugs in clinical trials. Finally, we propose a drug repurposing pipeline to encourage the implementation of standard methods to fast-track the discovery of candidates and to ensure reproducible results.
Collapse
Affiliation(s)
- Hazem Mslati
- Vancouver Prostate Centre, University of
British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6,
Canada
| | - Francesco Gentile
- Vancouver Prostate Centre, University of
British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6,
Canada
| | - Carl Perez
- Vancouver Prostate Centre, University of
British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6,
Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of
British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6,
Canada
| |
Collapse
|
30
|
Abramenko N, Vellieux F, Tesařová P, Kejík Z, Kaplánek R, Lacina L, Dvořánková B, Rösel D, Brábek J, Tesař A, Jakubek M, Smetana K. Estrogen Receptor Modulators in Viral Infections Such as SARS-CoV-2: Therapeutic Consequences. Int J Mol Sci 2021; 22:6551. [PMID: 34207220 PMCID: PMC8233910 DOI: 10.3390/ijms22126551] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
COVID-19 is a pandemic respiratory disease caused by the SARS-CoV-2 coronavirus. The worldwide epidemiologic data showed higher mortality in males compared to females, suggesting a hypothesis about the protective effect of estrogens against severe disease progression with the ultimate end being patient's death. This article summarizes the current knowledge regarding the potential effect of estrogens and other modulators of estrogen receptors on COVID-19. While estrogen receptor activation shows complex effects on the patient's organism, such as an influence on the cardiovascular/pulmonary/immune system which includes lower production of cytokines responsible for the cytokine storm, the receptor-independent effects directly inhibits viral replication. Furthermore, it inhibits the interaction of IL-6 with its receptor complex. Interestingly, in addition to natural hormones, phytestrogens and even synthetic molecules are able to interact with the estrogen receptor and exhibit some anti-COVID-19 activity. From this point of view, estrogen receptor modulators have the potential to be included in the anti-COVID-19 therapeutic arsenal.
Collapse
Affiliation(s)
- Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Fréderic Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
| | - Petra Tesařová
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic;
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Lukáš Lacina
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Barbora Dvořánková
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
| | - Daniel Rösel
- BIOCEV, Faculty of Sciences, Charles University, 252 50 Vestec, Czech Republic; (D.R.); (J.B.)
| | - Jan Brábek
- BIOCEV, Faculty of Sciences, Charles University, 252 50 Vestec, Czech Republic; (D.R.); (J.B.)
| | - Adam Tesař
- Department of Neurology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic;
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Karel Smetana
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic; (N.A.); (F.V.); (Z.K.); (R.K.); (L.L.); (B.D.); (M.J.)
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
| |
Collapse
|
31
|
Tsourdi E, Yu EW, Jan de Beur SM, Drake MT. Vaccination for Coronavirus Disease 2019 (COVID-19) and Relationship to Osteoporosis Care: Current Evidence and Suggested Approaches. J Bone Miner Res 2021; 36:1042-1047. [PMID: 33831269 PMCID: PMC8249992 DOI: 10.1002/jbmr.4304] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
The development of coronavirus disease 2019 (COVID-19) vaccines has proceeded at an unprecedented pace, with numerous trials conducted simultaneously across the world as a result of massive technological and financial resource expenditures. With multiple vaccines having now received regulatory approval, public health efforts to promote widespread vaccine dissemination are currently underway. There has been particular emphasis placed on vaccination of older populations, the age group in which COVID-19 infection has been most lethal. However, such widespread vaccination approaches have necessarily raised important questions related to potential interactions with underlying diseases and concomitant treatments among persons to be vaccinated. Osteoporosis is a chronic condition marked by reduced bone strength and an associated increased risk for fracture that generally requires sustained medical intervention(s). Osteoporosis is neither associated with a higher risk of COVID-19 infection nor by more pronounced disease severity following infection, such that individuals with osteoporosis need not be more highly prioritized for COVID-19 vaccination. Osteoporosis therapies do not interfere with the efficacy or side effect profiles of COVID-19 vaccines and should not be stopped or indefinitely delayed because of vaccination. Depending on the specific drug profile within an anti-osteoporosis medication category, minor adjustments to the timing of drug administration may be considered with respect to the patient's COVID-19 vaccination schedule. Herein we provide practical recommendations for the care of patients requiring treatment for osteoporosis in the setting of COVID-19 vaccination. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Elena Tsourdi
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany.,Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| | - Elaine W Yu
- Endocrine Unit, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Suzanne M Jan de Beur
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew T Drake
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA.,Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
32
|
Das A, Rana S. The role of human C5a as a non-genomic target in corticosteroid therapy for management of severe COVID19. Comput Biol Chem 2021; 92:107482. [PMID: 33845430 PMCID: PMC8020607 DOI: 10.1016/j.compbiolchem.2021.107482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 01/08/2023]
Abstract
Complement system plays a dual role; physiological as well as pathophysiological. While physiological role protects the host, pathophysiological role can substantially harm the host, by triggering several hyper-inflammatory pathways, referred as "hypercytokinaemia". Emerging clinical evidence suggests that exposure to severe acute respiratory syndrome coronavirus-2 (SARS-CoV2), tricks the complement to aberrantly activate the "hypercytokinaemia" loop, which significantly contributes to the severity of the COVID19. The pathophysiological response of the complement is usually amplified by the over production of potent chemoattractants and inflammatory modulators, like C3a and C5a. Therefore, it is logical that neutralizing the harmful effects of the inflammatory modulators of the complement system can be beneficial for the management of COVID19. While the hunt for safe and efficacious vaccines were underway, polypharmacology based combination therapies were fairly successful in reducing both the morbidity and mortality of COVID19 across the globe. Repurposing of small molecule drugs as "neutraligands" of C5a appears to be an alternative for modulating the hyper-inflammatory signals, triggered by the C5a-C5aR signaling axes. Thus, in the current study, few specific and non-specific immunomodulators (azithromycin, colchicine, famotidine, fluvoxamine, dexamethasone and prednisone) generally prescribed for prophylactic usage for management of COVID19 were subjected to computational and biophysical studies to probe whether any of the above drugs can act as "neutraligands", by selectively binding to C5a over C3a. The data presented in this study indicates that corticosteroids, like prednisone can have potentially better selectively (Kd ∼ 0.38 μM) toward C5a than C3a, suggesting the positive modulatory role of C5a in the general success of the corticosteroid therapy in moderate to severe COVID19.
Collapse
Affiliation(s)
- Aurosikha Das
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, 752050, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, 752050, India.
| |
Collapse
|