1
|
Martina MG, Rubini D, Radi M, Cagno V. Targeting PI4KB and Src/Abl host kinases as broad-spectrum antiviral strategy: Myth or real opportunity? Antiviral Res 2025; 235:106100. [PMID: 39922541 DOI: 10.1016/j.antiviral.2025.106100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Viruses pose a continuous threat to human health. Limited treatment options exist for current viruses, and the risk of infections with newly emerging or re-emerging viruses is increasing. In a pandemic scenario, having a broad-spectrum antiviral to limit viral spread while developing specific antivirals and vaccines is crucial. Targeting host kinases represents a valuable strategy due to the higher barrier to resistance and the broad-spectrum activity it offers. While cells have redundant kinases for the same biological function, viruses rely on specific kinases for their replication cycle, enabling targeted antiviral action with limited toxicity. This review focuses on two extensively studied kinase targets: the lipid kinase phosphatidylinositol 4-kinase IIIβ (PI4KB) and the tyrosine kinase proteins Src and Abl. Compounds active against these targets are reviewed in terms of the viruses they inhibit, their mechanisms of action and their stage of development. While PI4KB inhibitors have reached clinical trials, those targeting Src and Abl remain largely in the preclinical phase. Nevertheless, opportunities exist to improve potency and further understand the specific roles of these kinases in the life cycle of multiple viruses.
Collapse
Affiliation(s)
- Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Daniele Rubini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy.
| | - Valeria Cagno
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
2
|
Liu J, Hu D, Wang Y, Zhou X, Jiang L, Wang P, Lai H, Wang Y, Xiao H. Exploration of a Predictive Model for Keloid and Potential Therapeutic Drugs Based on Immune Infiltration and Cuproptosis-Related Genes. J Burn Care Res 2024; 45:1217-1231. [PMID: 38334429 DOI: 10.1093/jbcr/irae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Indexed: 02/10/2024]
Abstract
The aim of this study was to investigate the correlation between cuproptosis-related genes and immunoinfiltration in keloid, develop a predictive model for keloid occurrence, and explore potential therapeutic drugs. The microarray datasets (GSE7890 and GSE145725) were obtained from Gene Expression Omnibus database to identify the differentially expressed genes (DEGs) between keloid and nonkeloid samples. Key genes were identified through immunoinfiltration analysis and DEGs and then analyzed for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, followed by the identification of protein-protein interaction networks, transcription factors, and miRNAs associated with key genes. Additionally, a logistic regression analysis was performed to develop a predictive model for keloid occurrence, and potential candidate drugs for keloid treatment were identified. Three key genes (FDX1, PDHB, and DBT) were identified, showing involvement in acetyl-CoA biosynthesis, mitochondrial matrix, oxidoreductase activity, and the tricarboxylic acid cycle. Immune infiltration analysis suggested the involvement of B cells, Th1 cells, dendritic cells, T helper cells, antigen-presenting cell coinhibition, and T cell coinhibition in keloid. These genes were used to develop a logistic regression-based nomogram for predicting keloid occurrence with an area under the curve of 0.859 and good calibration. We identified 32 potential drug molecules and extracted the top 10 compounds based on their P-values, showing promise in targeting key genes and potentially effective against keloid. Our study identified some genes in keloid pathogenesis and potential therapeutic drugs. The predictive model enhances early diagnosis and management. Further research is needed to validate and explore clinical implications.
Collapse
Affiliation(s)
- Jiaming Liu
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| | - Ding Hu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Yaojun Wang
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| | - Xiaoqian Zhou
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| | - Liyuan Jiang
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| | - Peng Wang
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| | - Haijing Lai
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| | - Yu Wang
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| | - Houan Xiao
- Department of Burns and Plastic and Cosmetic Surgery, Xi' an Ninth Hospital, Xi'an, Shaanxi Province, 710054, China
| |
Collapse
|
3
|
Wang Y, Su Y, Zhao K, Huo D, Du Z, Wang Z, Xie H, Liu L, Jin Q, Ren X, Chen X, Zhang D. A deep learning drug screening framework for integrating local-global characteristics: A novel attempt for limited data. Heliyon 2024; 10:e34244. [PMID: 39130417 PMCID: PMC11315141 DOI: 10.1016/j.heliyon.2024.e34244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 07/05/2024] [Indexed: 08/13/2024] Open
Abstract
At the beginning of the "Disease X" outbreak, drug discovery and development are often challenged by insufficient and unbalanced data. To address this problem and maximize the information value of limited data, we propose a drug screening model, LGCNN, based on convolutional neural network (CNN), which enables rapid drug screening by integrating features of drug molecular structures and drug-target interactions at both local and global (LG) levels. Experimental results show that LGCNN exhibits better performance compared to other state-of-the-art classification methods under limited data. In addition, LGCNN was applied to anti-SARS-CoV-2 drug screening to realize therapeutic drug mining against COVID-19. LGCNN transcends the limitations of traditional models for predicting interactions between single drug targets and shows new advantages in predicting multi-target drug-target interactions. Notably, the cross-coronavirus generalizability of the model is also implied by the analysis of targets, drugs, and mechanisms in the prediction results. In conclusion, LGCNN provides new ideas and methods for rapid drug screening in emergency situations where data are scarce.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yangguang Su
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Kairui Zhao
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Diwei Huo
- The Fourth Hospital of Harbin Medical University, No.37 Yiyuan Street, Harbin, Heilongjiang, 150001, China
| | - Zhenshun Du
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Zhiju Wang
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Hongbo Xie
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Lei Liu
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Qing Jin
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xuekun Ren
- College of Mathematics of Harbin Institute of Technology, No.92 Xidazhi Street, Harbin, Heilongjiang, 150001, China
| | - Xiujie Chen
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Denan Zhang
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| |
Collapse
|
4
|
Ng HJ, Alata MK, Nguyen QT, Huynh Duc Vinh P, Tan JY, Wong CL. Managing and treating COVID-19 in patients with hematological malignancies: a narrative review and expert insights. Clin Exp Med 2024; 24:119. [PMID: 38833206 PMCID: PMC11150206 DOI: 10.1007/s10238-024-01381-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/06/2024]
Abstract
Patients with hematologic malignancies (HMs) are at a significantly higher risk of contracting COVID-19 and experiencing severe outcomes compared to individuals without HMs. This heightened risk is influenced by various factors, including the underlying malignancy, immunosuppressive treatments, and patient-related factors. Notably, immunosuppressive regimens commonly used for HM treatment can lead to the depletion of B cells and T cells, which is associated with increased COVID-19-related complications and mortality in these patients. As the pandemic transitions into an endemic state, it remains crucial to acknowledge and address the ongoing risk for individuals with HMs. In this review, we aim to summarize the current evidence to enhance our understanding of the impact of HMs on COVID-19 risks and outcomes, identify particularly vulnerable individuals, and emphasize the need for specialized clinical attention and management. Furthermore, the impaired immune response to COVID-19 vaccination observed in these patients underscores the importance of implementing additional mitigation strategies. This may include targeted prophylaxis and treatment with antivirals and monoclonal antibodies as indicated. To provide practical guidance and considerations, we present two illustrative cases to highlight the real-life challenges faced by physicians caring for patients with HMs, emphasizing the need for individualized management based on disease severity, type, and the unique circumstances of each patient.
Collapse
Affiliation(s)
- Heng Joo Ng
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | | | - Quang The Nguyen
- Stem Cell Transplantation Department, Blood Transfusion Hematology Hospital, Ho Chi Minh, Vietnam
| | - Phu Huynh Duc Vinh
- Stem Cell Transplantation Department, Blood Transfusion Hematology Hospital, Ho Chi Minh, Vietnam
| | - Jing Yuan Tan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Chieh Lee Wong
- Department of Haematology, Sunway Medical Centre, Bandar Sunway, Selangor, Malaysia.
- School of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
5
|
El-Ashwah S, Salmanton-García J, Bilgin YM, Itri F, Žák P, Weinbergerová B, Verga L, Omrani AS, Silva MGD, Szotkowski T, Marchetti M, Buquicchio C, Nucci M, Schönlein M, Farina F, Besson C, Prezioso L, Nizamuddin S, Dávila-Valls J, Martín-Pérez S, Bonuomo V, Van Doesum J, Tisi MC, Passamonti F, Méndez GA, Meers S, Maertens J, López-García A, Glenthøj A, Bonnani M, Rinaldi I, Ormazabal-Vélez I, Labrador J, Kulasekararaj A, Espigado I, Demirkan F, De Jonge N, Collins GP, Calbacho M, Blennow O, Al-Khabori M, Adžić-Vukičević T, Arellano E, Mišković B, Mladenović M, Nordlander A, Ráčil Z, Ammatuna E, Cordoba R, Hersby DS, Gräfe S, Emarah Z, Hanakova M, Sacchi MV, Ijaz M, Rahimli L, Nunes Rodrigues R, Zambrotta GPM, Marchesi F, Cornely OA, Pagano L. The mortality of COVID-19 in CML patients from 2020 until 2022: results from the EPICOVIDEHA survey. Leuk Lymphoma 2024; 65:199-208. [PMID: 37966980 DOI: 10.1080/10428194.2023.2280886] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/10/2023] [Indexed: 11/17/2023]
Abstract
Since the beginning of the COVID-19 pandemic, there has been an overall improvement in patient mortality. However, haematological malignancy patients continue to experience significant impacts from COVID-19, including high rates of hospitalization, intensive care unit (ICU) admissions, and mortality. In comparison to other haematological malignancy patients, individuals with chronic myeloid leukemia (CML) generally have better prognosis. This study, conducted using a large haematological malignancy patient database (EPICOVIDEHA), demonstrated that the majority of CML patients experienced mild infections. The decline in severe and critical infections over the years can largely be attributed to the widespread administration of vaccinations and the positive response they elicited. Notably, the mortality rate among CML patients was low and exhibited a downward trend in subsequent years. Importantly, our analysis provided confirmation of the effectiveness of vaccinations in CML patients.
Collapse
Affiliation(s)
| | - Jon Salmanton-García
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), University of Cologne, University Hospital Cologne, Cologne, Germany
| | - Yavuz M Bilgin
- Department of Internal Medicine, ADRZ, Goes, Netherlands
| | - Federico Itri
- San Luigi Gonzaga Hospital - Orbassano, Orbassano, Italy
| | - Pavel Žák
- University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Barbora Weinbergerová
- Department of Internal Medicine - Hematology and Oncology, Masaryk University Hospital Brno, Brno, Czech Republic
| | - Luisa Verga
- Azienda Ospedaliera San Gerardo - Monza, Monza, Italy
- Università Milano-Bicocca, Milan, Italy
| | - Ali S Omrani
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | | | | | - Monia Marchetti
- Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | | | - Marcio Nucci
- Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martin Schönlein
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Caroline Besson
- Centre Hospitalier de Versailles, Le Chesnay, France
- Université Paris-Saclay, UVSQ, Inserm, Équipe "Exposome et Hérédité", CESP, Villejuif, France
| | - Lucia Prezioso
- Hospital University of Parma - Hematology and Bone Marrow Unit, Parma, Italy
| | | | | | | | - Valentina Bonuomo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | | | - Francesco Passamonti
- Department of Medicine and Surgery, University of Insubria and ASST Sette Laghi, Ospedale di Circolo of Varese, Varese, Italy
| | | | | | - Johan Maertens
- Department of Microbiology, Immunology, and Transplantation, KULeuven, Leuven, Belgium
- Department of Hematology, UZ Leuven, Leuven, Belgium
| | - Alberto López-García
- Fundacion Jimenez Diaz University Hospital, Health Research Institute IIS-FJD, Madrid, Spain
| | - Andreas Glenthøj
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Matteo Bonnani
- Hematology Unit, Fondazione Policlinico Universitario Agostino Gemelli - IRCCS, Rome, Italy
| | - Ikhwan Rinaldi
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | | | - Jorge Labrador
- Department of Hematology, Research Unit, Hospital Universitario de Burgos, Burgos, Spain
| | | | - Ildefonso Espigado
- Department of Hematology, University Hospital Virgen Macarena - University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla (Departamento de Medicina), Seville, Spain
| | - Fatih Demirkan
- Division of Hematology, Dokuz Eylul University, Izmir, Turkey
| | - Nick De Jonge
- Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Graham P Collins
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | | | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Elena Arellano
- Department of Hematology, University Hospital Virgen Macarena - University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla (Departamento de Medicina), Seville, Spain
| | - Bojana Mišković
- COVID-19 Hospital "Batajnica", Belgrade, Serbia
- Clinic for Orthopedic Surgery and Traumatology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Miloš Mladenović
- COVID-19 Hospital "Batajnica", Belgrade, Serbia
- Clinic for Orthopedic Surgery and Traumatology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Anna Nordlander
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Zdeněk Ráčil
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | - Raul Cordoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute IIS-FJD, Madrid, Spain
| | - Ditte Stampe Hersby
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Stefanie Gräfe
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ziad Emarah
- Oncology Center, Mansoura University, Mansoura, Egypt
| | - Michaela Hanakova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Maria Vittoria Sacchi
- Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Marriyam Ijaz
- Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Laman Rahimli
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
| | | | | | - Francesco Marchesi
- Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
- Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine, and University Hospital Cologne, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Livio Pagano
- Hematology Unit, Fondazione Policlinico Universitario Agostino Gemelli - IRCCS, Rome, Italy
- Hematology Unit, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
6
|
Mansour HM. The interference between SARS-COV-2 and Alzheimer's disease: Potential immunological and neurobiological crosstalk from a kinase perspective reveals a delayed pandemic. Ageing Res Rev 2024; 94:102195. [PMID: 38244862 DOI: 10.1016/j.arr.2024.102195] [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: 11/14/2022] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Coronavirus disease 2019 (COVID-19) has infected over 700 million people, with up to 30% developing neurological manifestations, including dementias. However, there is a lack of understanding of common molecular brain markers causing Alzheimer's disease (AD). COVID-19 has etiological cofactors with AD, making patients with AD a vulnerable population at high risk of experiencing more severe symptoms and worse consequences. Both AD and COVID-19 have upregulated several shared kinases, leading to the repositioning of kinase inhibitors (KIs) for the treatment of both diseases. This review provides an overview of the interactions between the immune system and the nervous system in relation to receptor tyrosine kinases, including epidermal growth factor receptors, vascular growth factor receptors, and non-receptor tyrosine kinases such as Bruton tyrosine kinase, spleen tyrosine kinase, c-ABL, and JAK/STAT. We will discuss the promising results of kinase inhibitors in pre-clinical and clinical studies for both COVID-19 and Alzheimer's disease (AD), as well as the challenges in repositioning KIs for these diseases. Understanding the shared kinases between AD and COVID-19 could help in developing therapeutic approaches for both.
Collapse
Affiliation(s)
- Heba M Mansour
- General Administration of Innovative Products, Central Administration of Biological, Innovative Products, and Clinical Studies (Bio-INN), Egyptian Drug Authority (EDA), Giza, Egypt.
| |
Collapse
|
7
|
Ali EA, Al-Sadi A, Al-maharmeh Q, Subahi EA, Bellamkonda A, Kalavar M, Panigrahi K, Alshurafa A, Yassin MA. SARS-CoV-2 and chronic myeloid leukemia: a systematic review. Front Med (Lausanne) 2024; 10:1280271. [PMID: 38327268 PMCID: PMC10847560 DOI: 10.3389/fmed.2023.1280271] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/21/2023] [Indexed: 02/09/2024] Open
Abstract
Introduction Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus causing the coronavirus disease of 2019. The disease has caused millions of deaths since the first pandemic at the end of 2019. Immunocompromised individuals are more likely to develop severe infections. Numerous mutations had developed in SARS-CoV-2, resulting in strains (Alfa Beta Delta Omicron) with varying degrees of virulence disease severity. In CML (chronic myeloid leukemia) patients, there is a lot of controversy regarding the effect of the treatment on the patient outcome. Some reports suggested potential better outcomes among patients with CML, likely due to the use of TKI; other reports showed no significant effects. Additionally, it is unknown how much protection immunization provides for cancer patients. Method In accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standards, we conducted a systematic review. Retrospective, prospective studies, reviews, case series, and case reports of chronic myeloid leukemia patients aged above 18 years who had SARS-CoV-2 infection were included. English literature was screened using PubMed, SCOPUS, and Google Scholar. Search terms include chronic myeloid leukemia, chronic myelogenous leukemia, and SARS-CoV-2 and Coronavirus disease 2019 (COVID-19). We searched the reference lists of the included studies for any new articles. The search included all articles published up to April 20, 2023. The review is registered in PROSPERO (registration number CRD42022326674). Results We reviewed 33 articles of available published literature up to April 2023 and collected data from a total of 682 CML patients with COVID-19. Most patients were in the chronic phase, seven were in the accelerated phase, and eight were in the blast phase. Disease severity was classified according to WHO criteria. Mortality was seen in 45 patients, and there were no reports of thrombotic events. Two hundred seventy-seven patients were in the era before vaccination; among them, eight were in the intensive care unit (ICU), and mortality was 30 (11%). There were 405 patients after the era of vaccination; among them, death was reported in 15 (4%) patients and ICU in 13 patients. Limitations and conclusion The major limitation of this review is the lack of details about the use or hold of TKIs during SARS-CoV-2 infection. Additionally, after the appearance of the different variants of the SARS-CoV-2 virus, few studies mentioned the variant of the virus, which makes it difficult to compare the outcome of the other variants of the SARS-CoV-2 virus in patients with CML. Despite the limitations of the study, CML patients with COVID-19 have no significant increase in mortality compared to other hematological malignancy. Hematological cancers are associated with an increased risk of thrombosis, which is expected to increase in patients with COVID-19. However, patient with CML has not been reported to have a significant increase in thrombosis risk. The available data indicates that COVID-19's effect on patients with chronic myeloid leukemia (CML) still needs to be better understood due to the limited data. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php? RecordID:326674.
Collapse
Affiliation(s)
- Elrazi A. Ali
- Internal Medicine Department, Interfaith Medical Center/One Brooklyn Health, Brooklyn, NY, United States
| | - Anas Al-Sadi
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Qusai Al-maharmeh
- Internal Medicine Department, Saint Michael's Medical Center, Newark, CA, United States
| | - Eihab A. Subahi
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Amulya Bellamkonda
- Internal Medicine Department, Interfaith Medical Center/One Brooklyn Health, Brooklyn, NY, United States
| | - Madhumati Kalavar
- Internal Medicine Department, Interfaith Medical Center/One Brooklyn Health, Brooklyn, NY, United States
| | - Kalpana Panigrahi
- Internal Medicine Department, Interfaith Medical Center/One Brooklyn Health, Brooklyn, NY, United States
| | - Awni Alshurafa
- Department of Oncology-Hematology, National Center for Cancer Care and Research – Hamad Medical Corporation, Doha, Qatar
| | - Mohamed A. Yassin
- Department of Oncology-Hematology, National Center for Cancer Care and Research – Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
8
|
Lee JD, Menasche BL, Mavrikaki M, Uyemura MM, Hong SM, Kozlova N, Wei J, Alfajaro MM, Filler RB, Müller A, Saxena T, Posey RR, Cheung P, Muranen T, Heng YJ, Paulo JA, Wilen CB, Slack FJ. Differences in syncytia formation by SARS-CoV-2 variants modify host chromatin accessibility and cellular senescence via TP53. Cell Rep 2023; 42:113478. [PMID: 37991919 PMCID: PMC10785701 DOI: 10.1016/j.celrep.2023.113478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/13/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) remains a significant public health threat due to the ability of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants to evade the immune system and cause breakthrough infections. Although pathogenic coronaviruses such as SARS-CoV-2 and Middle East respiratory syndrome (MERS)-CoV lead to severe respiratory infections, how these viruses affect the chromatin proteomic composition upon infection remains largely uncharacterized. Here, we use our recently developed integrative DNA And Protein Tagging methodology to identify changes in host chromatin accessibility states and chromatin proteomic composition upon infection with pathogenic coronaviruses. SARS-CoV-2 infection induces TP53 stabilization on chromatin, which contributes to its host cytopathic effect. We mapped this TP53 stabilization to the SARS-CoV-2 spike and its propensity to form syncytia, a consequence of cell-cell fusion. Differences in SARS-CoV-2 spike variant-induced syncytia formation modify chromatin accessibility, cellular senescence, and inflammatory cytokine release via TP53. Our findings suggest that differences in syncytia formation alter senescence-associated inflammation, which varies among SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Jonathan D Lee
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Bridget L Menasche
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Maria Mavrikaki
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Madison M Uyemura
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Su Min Hong
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Nina Kozlova
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Wei
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mia M Alfajaro
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Renata B Filler
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Arne Müller
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Tanvi Saxena
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan R Posey
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Priscilla Cheung
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Taru Muranen
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Yujing J Heng
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Craig B Wilen
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Frank J Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Qi F, Bao M, Gao H, Zhang X, Zhao S, Wang C, Li W, Jiang Q. Patients with chronic myeloid leukemia and coronavirus disease 2019 in the Omicron era. Ann Hematol 2023; 102:2707-2716. [PMID: 37578540 DOI: 10.1007/s00277-023-05413-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
To explore the prevalence and severity of COVID-19 and the mental health during the Omicron pandemic in patients with chronic myeloid leukemia (CML), a cross-sectional survey from 2609 respondents with CML was performed. A total of 1725 (66%) reported that they had COVID-19 during this period. Among them, 1621 (94%) were mild; 97 (6%), moderate; 7 (0.4%), severe; and 0, critical or death. Four hundred three (15%), 199 (8%), and 532 (20%) had moderate to severe depression, anxiety, and distress, respectively. Eight hundred ninety (34%), 667 (26%), and 573 (22%), avoidance, intrusion, and hyper-arousal, respectively. In multivariate analyses, longer TKI-therapy duration was significantly associated with a lower prevalence of COVID-19 (odds ratio [OR] = 0.98; 95% confidence interval [CI], 0.95, 0.99; p = 0.043); however, living in urban areas (OR = 1.6 [1.3, 2.0]; p < 0.001) and having family members with COVID-19 (OR = 18.6 [15.1, 22.8]; p < 0.001), a higher prevalence of COVID-19. Increasing age (OR = 1.2 [1.1, 1.4]; p = 0.009), comorbidity(ies) (OR = 1.7 [1.1, 2.7]; p = 0.010), and multi-TKI-resistant patients receiving 3rd-generation TKIs or investigational agents (OR = 2.2 [1.2, 4.2]; p = 0.010) were significantly associated with moderate or severe COVID-19. Female, comorbidity(ies), unvaccinated, and moderate or severe COVID-19 were significantly associated with almost all adverse mental health consequences; increasing age or forced TKI dose reduction because of various restriction during the pandemic, moderate to severe distress, avoidance, or intrusion; however, mild COVID-19, none or mild anxiety, distress, avoidance, or intrusion. In conclusion, shorter TKI-therapy duration, increasing age, comorbidity(ies), or multi-TKI-resistant patients receiving 3rd-generation TKIs or investigational agents had a higher prevalence of COVID-19 or higher risk of moderate or severe disease in patients with CML; increasing age, female, comorbidity(ies), forced TKI dose reduction due to the pandemic, moderate or severe COVID-19, unvaccinated, a higher likelihood of worse mental health.
Collapse
Affiliation(s)
- Feiyang Qi
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South St, Beijing, 100044, China
| | - Mei Bao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South St, Beijing, 100044, China
| | - Hanlin Gao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South St, Beijing, 100044, China
| | - Xiaoshuai Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South St, Beijing, 100044, China
| | - Shasha Zhao
- Peking University People's Hospital, Qingdao, China
| | | | - Wenwen Li
- Peking University People's Hospital, Qingdao, China
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South St, Beijing, 100044, China.
- Peking University People's Hospital, Qingdao, China.
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
10
|
Lee JD, Menasche BL, Mavrikaki M, Uyemura MM, Hong SM, Kozlova N, Wei J, Alfajaro MM, Filler RB, Müller A, Saxena T, Posey RR, Cheung P, Muranen T, Heng YJ, Paulo JA, Wilen CB, Slack FJ. Differences in syncytia formation by SARS-CoV-2 variants modify host chromatin accessibility and cellular senescence via TP53. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.31.555625. [PMID: 37693555 PMCID: PMC10491142 DOI: 10.1101/2023.08.31.555625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
COVID-19 remains a significant public health threat due to the ability of SARS-CoV-2 variants to evade the immune system and cause breakthrough infections. Although pathogenic coronaviruses such as SARS-CoV-2 and MERS-CoV lead to severe respiratory infections, how these viruses affect the chromatin proteomic composition upon infection remains largely uncharacterized. Here we used our recently developed integrative DNA And Protein Tagging (iDAPT) methodology to identify changes in host chromatin accessibility states and chromatin proteomic composition upon infection with pathogenic coronaviruses. SARS-CoV-2 infection induces TP53 stabilization on chromatin, which contributes to its host cytopathic effect. We mapped this TP53 stabilization to the SARS-CoV-2 spike and its propensity to form syncytia, a consequence of cell-cell fusion. Differences in SARS-CoV-2 spike variant-induced syncytia formation modify chromatin accessibility, cellular senescence, and inflammatory cytokine release via TP53. Our findings suggest that differences in syncytia formation alter senescence-associated inflammation, which varies among SARS-CoV-2 variants.
Collapse
|
11
|
Ćojbašić I, Golubović I, Ćojbašić Ž. Clinical Outcomes of Patients with Chronic Myeloid Leukemia and COVID-19 Infection-A Single Center Survey. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1564. [PMID: 37763683 PMCID: PMC10533142 DOI: 10.3390/medicina59091564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
Background: Previous research has shown different effects of hematological malignancies on the outcome of patients with COVID-19 infection depending on the type of disease and the treatment received. This research was aimed at examining the clinical outcome of COVID-19 infection in positive patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. Methods: We collected retrospective information on chronic myeloid leukemia patients who were treated and monitored in our institution during the pandemic period. Within this cohort, we recorded COVID-19 positive symptomatic patients and analyzed their basic characteristics, symptoms, severity, and outcome. Results: In the study cohort when COVID-19 was diagnosed, 86.7% of patients were on first-generation tyrosine kinase inhibitors therapy-imatinib. At the time of infection, 70% of patients were in molecular remission, 23.4% in complete cytogenetic remission, and 3.3% in complete hematological response. Most patients had symptomatic disease. Within the analyzed group, 56.7% of patients had asymptomatic/mild COVID-19 infection, 23.3% of patients had moderate symptoms which did not require hospitalization, and 20% of patients had severe/critical symptoms that required admission to the intensive care unit. More than half of the patients interrupted treatment with tyrosine kinase inhibitors temporarily during COVID-19. There were no deaths due to COVID-19 infection. Conclusions: In compliance with other larger clinical studies, analysis of the clinical outcome of COVID-19 infection in patients with chronic myeloid leukemia on tyrosine kinase inhibitors therapy in this study showed that they do not have an increased risk for COVID-19 infection and that they have a mild course of the disease with recovery.
Collapse
Affiliation(s)
- Irena Ćojbašić
- Faculty of Medicine, University of Niš, Blvd. Zorana Đinđića 81, 18000 Niš, Serbia;
- Clinic of Hematology, Allergology and Clinical Immunology, University Clinical Centre Niš, Blvd. Zorana Đinđića 48, 18000 Niš, Serbia;
| | - Ivana Golubović
- Clinic of Hematology, Allergology and Clinical Immunology, University Clinical Centre Niš, Blvd. Zorana Đinđića 48, 18000 Niš, Serbia;
| | - Žarko Ćojbašić
- Faculty of Mechanical Engineering, University of Niš, Aleksandra Medvedeva 14, 18000 Niš, Serbia
| |
Collapse
|
12
|
Choi JC, Jung SW, Choi IY, Kang YL, Lee DH, Lee SW, Park SY, Song CS, Choi IS, Lee JB, Oh C. Rottlerin-Liposome Inhibits the Endocytosis of Feline Coronavirus Infection. Vet Sci 2023; 10:380. [PMID: 37368766 DOI: 10.3390/vetsci10060380] [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: 03/24/2023] [Revised: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Rottlerin (R) is a natural extract from Mallotus philippensis with antiviral properties. Feline infectious peritonitis (FIP) is a fatal disease caused by feline coronavirus (FCoV) that is characterized by systemic granulomatous inflammation and high mortality. We investigated the antiviral effect of liposome-loaded R, i.e., rottlerin-liposome (RL), against FCoV. We demonstrated that RL inhibited FCoV replication in a dose-dependent manner, not only in the early endocytosis stage but also in the late stage of replication. RL resolved the low solubility issue of rottlerin and improved its inhibition efficacy at the cellular level. Based on these findings, we suggest that RL is worth further investigation as a potential treatment for FCoV.
Collapse
Affiliation(s)
- Jong-Chul Choi
- Qvet Co., Ltd., 606, Alumini Association Building of Konkuk University, 5 Achasan-ro 36-gil, Gwangjin-gu, Seoul 05066, Republic of Korea
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sung-Won Jung
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - In-Yeong Choi
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yeong-Lim Kang
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong-Hun Lee
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- KU Research Center for Zoonosis, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sang-Won Lee
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- KU Research Center for Zoonosis, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seung-Yong Park
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- KU Research Center for Zoonosis, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Chang-Seon Song
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- KU Research Center for Zoonosis, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - In-Soo Choi
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- KU Research Center for Zoonosis, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Joong-Bok Lee
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- KU Research Center for Zoonosis, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Changin Oh
- Department of Genetics, Yale School of Medicine, P.O. Box 208005, New Haven, CT 06520-8005, USA
| |
Collapse
|
13
|
Fan S, Shen Y, Li S, Xiang X, Li N, Li Y, Xu J, Cui M, Han X, Xia J, Huang Y. The S2 Subunit of Infectious Bronchitis Virus Affects Abl2-Mediated Syncytium Formation. Viruses 2023; 15:1246. [PMID: 37376546 DOI: 10.3390/v15061246] [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: 04/26/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The S2 subunit serves a crucial role in infectious bronchitis virus (IBV) infection, particularly in facilitating membrane fusion. Using reverse genetic techniques, mutant strains of the S2 locus exhibited substantially different syncytium-forming abilities in chick embryonic kidney cells. To determine the precise formation mechanism of syncytium, we demonstrated the co-ordinated role of Abl2 and its mediated cytoskeletal regulatory pathway within the S2 subunit. Using a combination of fluorescence quantification, RNA silencing, and protein profiling techniques, the functional role of S2 subunits in IBV-infected cells was exhaustively determined. Our findings imply that Abl2 is not the primary cytoskeletal regulator, the viral S2 component is involved in indirect regulation, and the three different viral strains activate various cytoskeletal regulatory pathways through Abl2. CRK, CRKL, ABI1, NCKAP1, and ENAH also play a role in cytoskeleton regulation. Our research provides a point of reference for the development of an intracellular regulatory network for the S2 subunit and a foundation for the rational design of antiviral drug targets against Abl2.
Collapse
Affiliation(s)
- Shunyi Fan
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Yuxi Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Shuyun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Xuelian Xiang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Nianling Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Yongxin Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Jing Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Min Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Xinfeng Han
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Jing Xia
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| |
Collapse
|
14
|
Mukherjee S, Manna S, Som N, Dhara S. Organic-Inorganic Hybrid Nanocomposites for Nanotheranostics: Special Focus on Preventing Emerging Variants of SARS-COV-2. BIOMEDICAL MATERIALS & DEVICES (NEW YORK, N.Y.) 2023:1-15. [PMID: 37363138 PMCID: PMC10187951 DOI: 10.1007/s44174-023-00077-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/27/2023] [Indexed: 06/28/2023]
Abstract
The worldwide emerging cases of various respiratory viral diseases and the current escalation of novel coronavirus disease (COVID-19) make people considerably attentive to controlling these viruses through innovative methods. Most re-emerging respiratory diseases envelop RNA viruses that employ attachment between the virus and host cell to get an entry form using the host cell machinery. Emerging variants of COVD-19 also bring about a constant threat to public health as it has wide infectivity and can quickly spread to infect humans. This review focuses on insights into the current investigations to prevent the progression of incipient variants of Severe Acute Respiratory Syndrome Coronavirus (SARS-COV-2) along with similar enveloped RNA viruses that cause respiratory illness in humans and animals. Nanotheranostics is a trailblazing arena of nanomedicine that simultaneously helps prevent or treat diseases and diagnoses. Nanoparticle coating and nanofibers were extensively explored, preventing viral contaminations. Several studies have proven the virucidal activities of metal nanoparticles like copper, silver, and titanium against respiratory viral pathogens. Worldwide many researchers have shown surfaces coated with ionic nanoparticles like zinc or titanium act as potent antiviral agents against RNA viruses. Carbon nanotubes, quantum dots, silica nanoparticles (NPs), polymeric and metallic nanoparticles have also been explored in the field of nanotheranostics in viral detection. In this review, we have comprehensively discussed different types of metallic, ionic, organic nanoparticles and their hybrids showing substantial antiviral properties to stop the progression of the novel coronavirus disease focused on three key classes: prevention, diagnostics, and treatment.
Collapse
Affiliation(s)
- Sayan Mukherjee
- Biomaterials and Tissue Engineering Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Souvik Manna
- Clinical Microbiology & Antibiotic Research Laboratory, CSIR - Institute of Microbial Technology, Chandigarh, India
| | - Nivedita Som
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| | - Santanu Dhara
- Biomaterials and Tissue Engineering Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
15
|
Alizadehmohajer N, Zahedifar S, Sohrabi E, Shaddel Basir S, Nourigheimasi S, Falak R, Nedaeinia R, A Ferns G, Emami Nejad A, Manian M. Using In Silico Bioinformatics Algorithms for the Accurate Prediction of the Impact of Spike Protein Mutations on the Pathogenicity, Stability, and Functionality of the SARS-CoV-2 Virus and Analysis of Potential Therapeutic Targets. Biochem Genet 2023; 61:778-808. [PMID: 36173498 PMCID: PMC9521556 DOI: 10.1007/s10528-022-10282-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 09/01/2022] [Indexed: 11/02/2022]
Abstract
Coronavirus disease 2019 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We have used bioinformatics to investigate seventeen mutations in the spike protein of SARS-CoV-2, as this mediates infection of human cells and is the target of most vaccine strategies and antibody-based therapies. Two mutations, H146Y and S221W, were identified as being most pathogenic. Mutations at positions D614G, A829T, and P1263L might also have deleterious effects on protein function. We hypothesized that candidate small molecules may be repurposed to combat viral infection. We investigated changes in binding energies of the ligands and the mutant proteins by assessing molecular docking. For an understanding of cellular function and organization, protein-protein interactions are also critical. Protein-protein docking for naïve and mutated structures of SARS-CoV-2 S protein was evaluated for their binding energy with the angiotensin-converting enzyme 2 (ACE2). These interactions might limit the binding of the SARS-CoV-2 spike protein to the ACE2 receptor or may have a deleterious effect on protein function that may limit infection. These results may have important implications for the transmission of SARS-CoV-2, its pathogenesis, and the potential for drug repurposing and immune therapies.
Collapse
Affiliation(s)
- Negin Alizadehmohajer
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133, Milan, Italy
| | - Shahrzad Zahedifar
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ehsan Sohrabi
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sedighe Shaddel Basir
- Department of Microbiology, Faculty of New Sciences and Technologies Branch, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, BN1 9PH, Sussex, UK
| | - Asieh Emami Nejad
- Department of Biology, Payame Noor University (PNU), P.O.Box 19395-3697, Tehran, Iran.
| | - Mostafa Manian
- Department of Medical Laboratory Science, Faculty of Medical Science, Kermanshah Branch, Imam Khomeini Campus, Islamic Azad University, Farhikhtegan Bld., Shahid J'afari St., 6718997551, Kermanshah, Iran.
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
16
|
Kandeel M. An overview of the recent progress in Middle East Respiratory Syndrome Coronavirus (MERS-CoV) drug discovery. Expert Opin Drug Discov 2023; 18:385-400. [PMID: 36971501 DOI: 10.1080/17460441.2023.2192921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
INTRODUCTION The Middle East respiratory syndrome coronavirus (MERS-CoV) has remained a public health concern since it first emerged in 2012. Although many potential treatments for MERS-CoV have been developed and tested, none have had complete success in stopping the spread of this deadly disease. MERS-CoV replication comprises attachment, entry, fusion and replication steps. Targeting these events may lead to the creation of medications that effectively treat MERS-CoV infection. AREAS COVERED This review updates the research on the development of inhibitors of MERS-CoV. The main topics are MERS-CoV‒related proteins and host cell proteins that are involved in viral protein activation and infection. EXPERT OPINION Research on discovering drugs that can inhibit MERS-CoV started at a slow pace, and although efforts have steadily increased, clinical trials for new drugs specifically targeting MERS-CoV have not been extensive enough. The explosion in efforts to find new medications for the SARS-CoV-2 virus indirectly enhanced the volume of data on MERS-CoV inhibition by including MERS-CoV in drug assays. The appearance of COVID-19 completely transformed the data available on MERS-CoV inhibition. Despite the fact that new infected cases are constantly being diagnosed, there are currently no approved vaccines for or inhibitors of MERS-CoV.
Collapse
|
17
|
Targeting Human Proteins for Antiviral Drug Discovery and Repurposing Efforts: A Focus on Protein Kinases. Viruses 2023; 15:v15020568. [PMID: 36851782 PMCID: PMC9966946 DOI: 10.3390/v15020568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Despite the great technological and medical advances in fighting viral diseases, new therapies for most of them are still lacking, and existing antivirals suffer from major limitations regarding drug resistance and a limited spectrum of activity. In fact, most approved antivirals are directly acting antiviral (DAA) drugs, which interfere with viral proteins and confer great selectivity towards their viral targets but suffer from resistance and limited spectrum. Nowadays, host-targeted antivirals (HTAs) are on the rise, in the drug discovery and development pipelines, in academia and in the pharmaceutical industry. These drugs target host proteins involved in the virus life cycle and are considered promising alternatives to DAAs due to their broader spectrum and lower potential for resistance. Herein, we discuss an important class of HTAs that modulate signal transduction pathways by targeting host kinases. Kinases are considered key enzymes that control virus-host interactions. We also provide a synopsis of the antiviral drug discovery and development pipeline detailing antiviral kinase targets, drug types, therapeutic classes for repurposed drugs, and top developing organizations. Furthermore, we detail the drug design and repurposing considerations, as well as the limitations and challenges, for kinase-targeted antivirals, including the choice of the binding sites, physicochemical properties, and drug combinations.
Collapse
|
18
|
Morales-Ortega A, Farfán-Sedano AI, San Martín-López JV, Escribá-Bárcena A, Jaenes-Barrios B, Madroñal-Cerezo E, Llarena-Barroso C, Mesa-Plaza N, Frutos-Pérez B, Ruiz-Giardín JM, Duarte-Millán MÁ, Piedrabuena-García SI, Carpintero-García L, Canalejo-Castrillero E, Mora-Hernández B, García-Parra CJ, Magro-García HA, Algaba-García A, Hernández-Muniesa B, Nasarre-López B, Ontañón-Nasarre A, Domínguez-García MJ, Gómez-Santos D, Prieto-Menchero S, García de Tena J, Bermejo F, García-Gil M, Gonzalo-Pascua S, Bernal-Bello D. Baricitinib or imatinib in hospitalized COVID-19 patients: Results from COVINIB, an exploratory randomized clinical trial. J Med Virol 2023; 95:e28495. [PMID: 36639911 DOI: 10.1002/jmv.28495] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Baricitinib and imatinib are considered therapies for coronavirus disease 2019 (COVID-19), but their ultimate clinical impact remains to be elucidated, so our objective is to determine whether these kinase inhibitors provide benefit when added to standard care in hospitalized COVID-19 patients. Phase-2, open-label, randomized trial with a pick-the-winner design conducted from September 2020 to June 2021 in a single Spanish center. Hospitalized adults with COVID-19 pneumonia and a symptom duration ≤10 days were assigned to 3 arms: imatinib (400 mg qd, 7 days) plus standard-care, baricitinib (4 mg qd, 7 days) plus standard-care, or standard-care alone. Primary outcome was time to clinical improvement (discharge alive or a reduction of 2 points in an ordinal scale of clinical status) compared on a day-by-day basis to identify differences ≥15% between the most and least favorable groups. Secondary outcomes included oxygenation and ventilatory support requirements, additional therapies administered, all-cause mortality, and safety. One hundred and sixty-five patients analyzed. Predefined criteria for selection of the most advantageous arm were met for baricitinib, but not for imatinib. However, no statistically significant differences were observed in formal analysis, but a trend toward better results in patients receiving baricitinib was found compared to standard care alone (hazard ratio [HR] for clinical improvement: 1.41, 95% confidence intervals [CI]: 0.96-2.06; HR for discontinuing oxygen: 1.46, 95% CI: 0.94-2.28). No differences were found regarding additional therapies administered or safety. Baricitinib plus standard care showed better results for hospitalized COVID-19 patients, being the most advantageous therapeutic strategy among those proposed in this exploratory clinical trial.
Collapse
Affiliation(s)
- Alejandro Morales-Ortega
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain.,Department of Medicine, Universidad de Alcalá, Madrid, Spain
| | | | | | | | | | - Elena Madroñal-Cerezo
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Cristina Llarena-Barroso
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Nieves Mesa-Plaza
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Begoña Frutos-Pérez
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | | | | | | | - Eduardo Canalejo-Castrillero
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain.,Department of Medical Specialties and Public Health, Universidad Rey Juan Carlos, Madrid, Spain
| | - Belén Mora-Hernández
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | | | - Alicia Algaba-García
- Department of Gastroenterology, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | - Berta Nasarre-López
- Clinical Research Program, Spanish National Cancer Research Centre (CNIO)-Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Ana Ontañón-Nasarre
- Department of Hospital Pharmacy, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | - Dulce Gómez-Santos
- Department of Radiology, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | | | - Fernando Bermejo
- Department of Medical Specialties and Public Health, Universidad Rey Juan Carlos, Madrid, Spain.,Department of Gastroenterology, Hospital Universitario de Fuenlabrada, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital La Paz (IdiPAZ), Madrid, Spain
| | - Mario García-Gil
- Department of Hospital Pharmacy, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Sonia Gonzalo-Pascua
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain.,Department of Medical Specialties and Public Health, Universidad Rey Juan Carlos, Madrid, Spain
| | - David Bernal-Bello
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | |
Collapse
|
19
|
Song C, Li D, Zhang J, Zhao X. Berberine hydrochloride alleviates imatinib mesylate - induced cardiotoxicity through the inhibition of Nrf2-dependent ferroptosis. Food Funct 2023; 14:1087-1098. [PMID: 36594456 DOI: 10.1039/d2fo03331c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Imatinib mesylate (IMA) belonging to the selective tyrosine kinase inhibitor family has been proven to induce cardiotoxic effects along with therapeutic strategies. Nrf2-dependent ferroptosis has been implicated in the cardiotoxicity induced by IMA. The present study was designed to investigate the protective effects of berberine hydrochloride (Ber) on cardiac injuries induced by IMA and to explore its potential mechanisms. In H9c2 cells, cell viability, the generation of reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and labile iron pool (LIP) levels were measured. In a mouse model of IMA-induced cardiomyopathy, serum biomarkers and cardiac tissues were examined. A western blot assay was performed to evaluate the expression of ferroptosis-related proteins in vitro and in vivo. Our results indicated that Ber increased cell viability and MMP and decreased cellular ROS and iron levels in comparison to the IMA group of H9c2 cells. In mice, Ber significantly improved cardiac status and attenuated the level of ferroptosis biomarkers including malonaldehyde (MDA) and iron content. Additionally, Ber downregulated the expression of transferrin receptor (TfR) and P53 and upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), ferritin heavy chain-1 (FTH1), and glutathione peroxidase 4 (GPX4) in H9c2 cells and mice. The present data indicated that Ber has the potential to protect against IMA-induced cardiotoxicity, partly via inhibiting Nrf2-dependent ferroptosis.
Collapse
Affiliation(s)
- Chengzhu Song
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Dongning Li
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China.
| | - Xiaoyan Zhao
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China. .,Engineering Research Center of Coptis Development & Utilization, Ministry of Education
| |
Collapse
|
20
|
Graf I, Herndlhofer S, Kundi M, Greiner G, Sperr M, Hadzijusufovic E, Valent P, Sperr WR. Incidence of symptomatic Covid-19 infections in patients with mastocytosis and chronic myeloid leukemia: A comparison with the general Austrian population. Eur J Haematol 2023; 110:67-76. [PMID: 36193973 PMCID: PMC9874474 DOI: 10.1111/ejh.13875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 04/23/2023]
Abstract
BACKGROUND The SARS-COV-2 (Covid-19) pandemic has impacted the management of patients with hematologic disorders. In some entities, an increased risk for Covid-19 infections was reported, whereas others including chronic myeloid leukemia (CML) had a lower mortality. We have analyzed the prevalence of Covid-19 infections in patients with mastocytosis during the Covid-19 pandemic in comparison to data from CML patients and the general Austrian population. MATERIALS AND METHODS The prevalence of infections and PCR-proven Covid-19 infections was analyzed in 92 patients with mastocytosis. As controls, we used 113 patients with CML and the expected prevalence of Covid-19 in the general Austrian population. RESULTS In 25% of the patients with mastocytosis (23/92) signs and symptoms of infection, including fever (n = 11), dry cough (n = 10), sore throat (n = 12), pneumonia (n = 1), and dyspnea (n = 3) were recorded. Two (8.7%) of these symptomatic patients had a PCR-proven Covid-19 infection. Thus, the prevalence of Covid-19 infections in mastocytosis was 2.2%. The number of comorbidities, subtype of mastocytosis, regular exercise, smoking habits, age, or duration of disease at the time of interview did not differ significantly between patients with and without Covid-19 infections. In the CML cohort, 23.9% (27/113) of patients reported signs and symptoms of infection (fever, n = 8; dry cough, n = 17; sore throat, n = 11; dyspnea, n = 5). Six (22.2%) of the symptomatic patients had a PCR-proven Covid-19 infection. The prevalence of Covid-19 in all CML patients was 5.3%. The observed number of Covid-19 infections neither in mastocytosis nor in CML patients differed significantly from the expected number of Covid-19 infections in the Austrian population. CONCLUSIONS Our data show no significant difference in the prevalence of Covid-19 infections among patients with mastocytosis, CML, and the general Austrian population and thus, in mastocytosis, the risk of a Covid-19 infection was not increased compared to the general population.
Collapse
Affiliation(s)
- Irene Graf
- Division of Hematology and Hemostaseology, Department of Internal Medicine IMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute of Hematology and OncologyMedical University of ViennaViennaAustria
| | - Susanne Herndlhofer
- Division of Hematology and Hemostaseology, Department of Internal Medicine IMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute of Hematology and OncologyMedical University of ViennaViennaAustria
| | - Michael Kundi
- Institute of Environmental HealthMedical University of ViennaViennaAustria
| | - Georg Greiner
- Ihr Labor, Medical Diagnostic LaboratoriesViennaAustria
| | - Martina Sperr
- Division of Hematology and Hemostaseology, Department of Internal Medicine IMedical University of ViennaViennaAustria
| | - Emir Hadzijusufovic
- Division of Hematology and Hemostaseology, Department of Internal Medicine IMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute of Hematology and OncologyMedical University of ViennaViennaAustria
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine IMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute of Hematology and OncologyMedical University of ViennaViennaAustria
| | - Wolfgang R. Sperr
- Division of Hematology and Hemostaseology, Department of Internal Medicine IMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute of Hematology and OncologyMedical University of ViennaViennaAustria
| |
Collapse
|
21
|
Alipoor R, Ranjbar R. Small-molecule metabolites in SARS-CoV-2 treatment: a comprehensive review. Biol Chem 2022; 404:569-584. [PMID: 36490203 DOI: 10.1515/hsz-2022-0323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022]
Abstract
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has quickly spread all over the world. In this respect, traditional medicinal chemistry, repurposing, and computational approaches have been exploited to develop novel medicines for treating this condition. The effectiveness of chemicals and testing methods in the identification of new promising therapies, and the extent of preparedness for future pandemics, have been further highly advantaged by recent breakthroughs in introducing noble small compounds for clinical testing purposes. Currently, numerous studies are developing small-molecule (SM) therapeutic products for inhibiting SARS-CoV-2 infection and replication, as well as managing the disease-related outcomes. Transmembrane serine protease (TMPRSS2)-inhibiting medicinal products can thus prevent the entry of the SARS-CoV-2 into the cells, and constrain its spreading along with the morbidity and mortality due to the coronavirus disease 2019 (COVID-19), particularly when co-administered with inhibitors such as chloroquine (CQ) and dihydroorotate dehydrogenase (DHODH). The present review demonstrates that the clinical-stage therapeutic agents, targeting additional viral proteins, might improve the effectiveness of COVID-19 treatment if applied as an adjuvant therapy side-by-side with RNA-dependent RNA polymerase (RdRp) inhibitors.
Collapse
Affiliation(s)
- Reza Alipoor
- Student Research Committee , Hormozgan University of Medical Sciences , Bandar Abbas , Iran
| | - Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
22
|
Kabbara A, Vialet B, Marquevielle J, Bonnafous P, Mackereth CD, Amrane S. RNA G-quadruplex forming regions from SARS-2, SARS-1 and MERS coronoviruses. Front Chem 2022; 10:1014663. [PMID: 36479439 PMCID: PMC9719988 DOI: 10.3389/fchem.2022.1014663] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/26/2022] [Indexed: 11/14/2023] Open
Abstract
COVID-19 (Corona Virus Disease 2019), SARS (Severe Acute Respiratory Syndrome) and MERS (Middle East Respiratory Syndrome) are infectious diseases each caused by coronavirus outbreaks. Small molecules and other therapeutics are rapidly being developed to treat these diseases, but the threat of new variants and outbreaks argue for the identification of additional viral targets. Here we identify regions in each of the three coronavirus genomes that are able to form G-quadruplex (G4) structures. G4s are structures formed by DNA or RNA with a core of two or more stacked planes of guanosine tetrads. In recent years, numerous DNA and RNA G4s have emerged as promising pharmacological targets for the treatment of cancer and viral infection. We use a combination of bioinformatics and biophysical approaches to identify conserved RNA G4 regions from the ORF1A and S sequences of SARS-CoV, SARS-CoV-2 and MERS-CoV. Although a general depletion of G4-forming regions is observed in coronaviridae, the preservation of these selected G4 sequences support a significance in viral replication. Targeting these RNA structures may represent a new antiviral strategy against these viruses distinct from current approaches that target viral proteins.
Collapse
Affiliation(s)
| | | | | | | | | | - Samir Amrane
- Université de Bordeaux, INSERM U1212, CNRS UMR 5320, ARNA Laboratory, IECB, Bordeaux, France
| |
Collapse
|
23
|
Risner KH, Tieu KV, Wang Y, Getz M, Bakovic A, Bhalla N, Nathan SD, Conway DE, Macklin P, Narayanan A, Alem F. Maraviroc inhibits SARS-CoV-2 multiplication and s-protein mediated cell fusion in cell culture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2020.08.12.246389. [PMID: 32817953 PMCID: PMC7430595 DOI: 10.1101/2020.08.12.246389] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In an effort to identify therapeutic intervention strategies for the treatment of COVID-19, we have investigated a selection of FDA-approved small molecules and biologics that are commonly used to treat other human diseases. A investigation into 18 small molecules and 3 biologics was conducted in cell culture and the impact of treatment on viral titer was quantified by plaque assay. The investigation identified 4 FDA-approved small molecules, Maraviroc, FTY720 (Fingolimod), Atorvastatin and Nitazoxanide that were able to inhibit SARS-CoV-2 infection. Confocal microscopy with over expressed S-protein demonstrated that Maraviroc reduced the extent of S-protein mediated cell fusion as observed by fewer multinucleate cells in the context of drug-treatment. Mathematical modeling of drug-dependent viral multiplication dynamics revealed that prolonged drug treatment will exert an exponential decrease in viral load in a multicellular/tissue environment. Taken together, the data demonstrate that Maraviroc, Fingolimod, Atorvastatin and Nitazoxanide inhibit SARS-CoV-2 in cell culture.
Collapse
Affiliation(s)
- Kenneth H. Risner
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Katie V. Tieu
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Yafei Wang
- Intellegent Systems Engineering, Indiana University, Bloomington, Indiana, United States of America
| | - Michael Getz
- Intellegent Systems Engineering, Indiana University, Bloomington, Indiana, United States of America
| | - Allison Bakovic
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Nishank Bhalla
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Steven D. Nathan
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia, United States of America
| | - Daniel E. Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Paul Macklin
- Intellegent Systems Engineering, Indiana University, Bloomington, Indiana, United States of America
| | - Aarthi Narayanan
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
- American Type Culture Collection, Manassas, Virginia, United States of America
| | - Farhang Alem
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| |
Collapse
|
24
|
Pipitò L, Rujan R, Reynolds CA, Deganutti G. Molecular dynamics studies reveal structural and functional features of the SARS-CoV-2 spike protein. Bioessays 2022; 44:e2200060. [PMID: 35843871 PMCID: PMC9350306 DOI: 10.1002/bies.202200060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/21/2022] [Accepted: 07/01/2022] [Indexed: 12/23/2022]
Abstract
The SARS-CoV-2 virus is responsible for the COVID-19 pandemic the world experience since 2019. The protein responsible for the first steps of cell invasion, the spike protein, has probably received the most attention in light of its central role during infection. Computational approaches are among the tools employed by the scientific community in the enormous effort to study this new affliction. One of these methods, namely molecular dynamics (MD), has been used to characterize the function of the spike protein at the atomic level and unveil its structural features from a dynamic perspective. In this review, we focus on these main findings, including spike protein flexibility, rare S protein conformational changes, cryptic epitopes, the role of glycans, drug repurposing, and the effect of spike protein variants.
Collapse
Affiliation(s)
- Ludovico Pipitò
- Centre for Sport, Exercise and Life Sciences (CSELS)Faculty of Health and Life SciencesCoventry UniversityCoventryUK
| | - Roxana‐Maria Rujan
- Centre for Sport, Exercise and Life Sciences (CSELS)Faculty of Health and Life SciencesCoventry UniversityCoventryUK
| | - Christopher A. Reynolds
- Centre for Sport, Exercise and Life Sciences (CSELS)Faculty of Health and Life SciencesCoventry UniversityCoventryUK
| | - Giuseppe Deganutti
- Centre for Sport, Exercise and Life Sciences (CSELS)Faculty of Health and Life SciencesCoventry UniversityCoventryUK
| |
Collapse
|
25
|
Asif M, Amir M, Hussain A, Achakzai NM, Natesan Pushparaj P, Rasool M. Role of tyrosine kinase inhibitor in chronic myeloid leukemia patients with SARS-CoV-2 infection: A narrative Review. Medicine (Baltimore) 2022; 101:e29660. [PMID: 35777011 PMCID: PMC9239670 DOI: 10.1097/md.0000000000029660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) caused by a novel coronavirus-2 (CoV-2), also known as COVID-19, has spread rapidly worldwide since it is recognized as a public health emergency and has now been declared a pandemic on March 11, 2020, by the World Health Organization. The genome of SARS-CoV-2 comprises a single-stranded positive-sense RNA approximately 27 to 30 kb in size. The virus is transmitted through droplets from humans to humans. Infection with the SARS virus varies from asymptomatic to lethal, such as fever, cough, sore throat, and headache, but in severe cases, pneumonia and acute respiratory distress syndrome. Recently, no specific and effective treatment has been recommended for patients infected with the SARS virus. However, several options can be investigated to control SARS-CoV-2 infection, including monoclonal antibodies, interferons, therapeutic vaccines, and molecular-based targeted drugs. In the current review, we focus on tyrosine kinase inhibitor management and their protective role in SARS-CoV-2 patients with chronic myelogenous leukemia.
Collapse
Affiliation(s)
- Muhammad Asif
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
- Office of Research Innovation and Commercialization, BUITEMS, Quetta, Pakistan
| | - Muhammad Amir
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
| | - Abrar Hussain
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
| | - Niaz M. Achakzai
- Department of Molecular Biology, City Medical Complex, Kabul, Afghanistan
- Department of Molecular Biology, DNA section, Legal Medicine Directorate, Ministry of Public Health, Kabul, Afghanistan
- *Correspondence: Niaz M. Achakzai, Senior forensic DNA specialist, Department of Molecular Biology, DNA section, Legal Medicine Directorate, Ministry of Public Health, Kabul, Afghanistan (e-mail: ),
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
26
|
Weng W, Liu Q, Xue W, Wang H, Fang S, Sun Y, Tan L, Song C, Qiu X, Liu W, Ding C, Liao Y. Characterization of the Protective Efficacy Against QX Strain of a Recombinant Infectious Bronchitis Virus With H120 Backbone and QX Spike Gene. Front Microbiol 2022; 13:883642. [PMID: 35783402 PMCID: PMC9247577 DOI: 10.3389/fmicb.2022.883642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022] Open
Abstract
Infectious bronchitis virus (IBV) has been prevalent in chicken farms for many years, and its control relies on extensive vaccine administration. The continuous emergence of new variants and the low cross-protection efficiency prompt the development of new vaccines. In this study, we develop a reverse genetics technique based on the classical vaccine strain H120 genome, via in vitro ligation method. Using the H120 genome as the backbone, we constructed the recombinant virus rH120-QX(S) by replacing the H120 S gene with the QX S gene, a prevalent strain in China. Biological characteristics of the rH120-QX(S) virus, such as 50% egg lethal dose (ELD50), 50% egg infectious dose (EID50), dwarf embryo, growth curve, and genetic stability, are measured, which are comparable to the parental virus H120. There are no clinical symptoms and tissue lesions in the trachea and kidney in the rH120-QX(S)-infected specific-pathogen-free (SPF) chickens, demonstrating that this recombinant virus does not confer pathogenicity. Furthermore, protection studies show that there is 100% homologous protection of rH120-QX(S) to the virulent QX strain, as shown by the absence of clinical signs and no lethality. Taken together, our results demonstrate that swapping the S gene onto the H120 genetic backbone is a precise and effective way to produce genetically defined IBV vaccine candidates.
Collapse
Affiliation(s)
- Wenlian Weng
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Qingyan Liu
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- College of Agriculture, College of Animal Sciences, Yangtze University, Jingzhou, China
| | - Wenxiang Xue
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Huan Wang
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Shouguo Fang
- College of Agriculture, College of Animal Sciences, Yangtze University, Jingzhou, China
| | - Yingjie Sun
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lei Tan
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Cuiping Song
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xusheng Qiu
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Weiwei Liu
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chan Ding
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Ying Liao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- *Correspondence: Ying Liao
| |
Collapse
|
27
|
Amani B, Zareei S, Amani B, Zareei M, Zareei N, Shabestan R, Akbarzadeh A. Artesunate, imatinib, and infliximab in COVID‐19: A rapid review and meta‐analysis of current evidence. Immun Inflamm Dis 2022; 10:e628. [PMID: 35634954 PMCID: PMC9092000 DOI: 10.1002/iid3.628] [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: 12/01/2021] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 11/26/2022] Open
Abstract
Background and Objective Despite the pervasive vaccination program against coronavirus disease 2019 (COVID‐19), people who got fully vaccinated are still contaminated by severe acute respiratory syndrome coronavirus 2, making an effective and safe therapeutic intervention a crucial need for the patients' survival. The purpose of the present study is to seek available evidence for the efficacy and safety of three promising medications artesunate, imatinib, and infliximab against COVID‐19. Methods A literature search was conducted in PubMed, Cochrane Library, medRxive, and Google Scholar, and the relevant articles published up to January 2022 were found. Furthermore, the clinical trial databases were screened for finding more citations. Data analysis was carried out applying The Cochrane Collaboration tool and Newcastle–Ottawa scale to assess the included studies. Meta‐analysis was performed using RevMan 5.4.1. Results Five published studies were identified as eligible. Meta‐analysis showed that there was no significant difference between the infliximab and control groups in terms of mortality rate (risk ratio [RR]: 0.65; confidence interval [CI] 95%: 0.40–1.07; p = .09). However, a significant difference was observed between the two groups for the hospital discharge (RR: 1.37; CI 95%: 1.04–1.80; p = .03). No remarkable clinical benefit was observed for using imatinib in COVID‐19 patients. Artesunate showed significant improvement in patients with COVID‐19. Conclusion In the present, limited evidence exists for the efficacy and safety of artesunate, imatinib, and infliximab in patients with COVID‐19. The findings of WHO's Solidarity international trial will provide further information regarding these therapeutic interventions.
Collapse
Affiliation(s)
- Bahman Amani
- Department of Health Management and Economics, School of Public Health Tehran University of Medical Sciences Tehran Iran
| | - Sara Zareei
- Department of Cell & Molecular Biology, Faculty of Biological Sciences Kharazmi University Tehran Iran
| | - Behnam Amani
- Department of Health Management and Economics, School of Public Health Tehran University of Medical Sciences Tehran Iran
| | - Mahsa Zareei
- Department of Health Services Management, School of Health Management and Information Sciences Iran University of Medical Sciences Tehran Iran
| | - Neda Zareei
- Shiraz Transplant Research Center Shiraz University of Medical Sciences Shiraz Iran
| | - Rouhollah Shabestan
- Department of Biostatistics and Epidemiology, School of Public Health Tehran University of Medical Sciences Tehran Iran
| | - Arash Akbarzadeh
- Department of Biostatistics and Epidemiology, School of Public Health Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
28
|
Figueiredo DLA, Ximenez JPB, Seiva FRF, Panis C, Bezerra RDS, Ferrasa A, Cecchini AL, de Medeiros AI, Almeida AMF, Ramão A, Boldt ABW, Moya CF, Chin CM, de Paula D, Rech D, Gradia DF, Malheiros D, Venturini D, Tavares ER, Carraro E, Ribeiro EMDSF, Pereira EM, Tuon FF, Follador FAC, Fernandes GSA, Volpato H, Cólus IMDS, de Oliveira JC, Rodrigues JHDS, dos Santos JL, Visentainer JEL, Brandi JC, Serpeloni JM, Bonini JS, de Oliveira KB, Fiorentin K, Lucio LC, Faccin-Galhardi LC, Ferreto LED, Lioni LMY, Consolaro MEL, Vicari MR, Arbex MA, Pileggi M, Watanabe MAE, Costa MAR, Giannini MJSM, Amarante MK, Khalil NM, de Lima QA, Herai RH, Guembarovski RL, Shinsato RN, Mainardes RM, Giuliatti S, Yamada-Ogatta SF, Gerber VKDQ, Pavanelli WR, da Silva WC, Petzl-Erler ML, Valente V, Soares CP, Cavalli LR, Silva WA. COVID-19: The question of genetic diversity and therapeutic intervention approaches. Genet Mol Biol 2022; 44:e20200452. [PMID: 35421211 PMCID: PMC9075701 DOI: 10.1590/1678-4685-gmb-2020-0452] [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: 12/17/2020] [Accepted: 12/24/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2), is the largest pandemic in modern history with very high infection rates and considerable mortality. The disease, which emerged in China's Wuhan province, had its first reported case on December 29, 2019, and spread rapidly worldwide. On March 11, 2020, the World Health Organization (WHO) declared the COVID-19 outbreak a pandemic and global health emergency. Since the outbreak, efforts to develop COVID-19 vaccines, engineer new drugs, and evaluate existing ones for drug repurposing have been intensively undertaken to find ways to control this pandemic. COVID-19 therapeutic strategies aim to impair molecular pathways involved in the virus entrance and replication or interfere in the patients' overreaction and immunopathology. Moreover, nanotechnology could be an approach to boost the activity of new drugs. Several COVID-19 vaccine candidates have received emergency-use or full authorization in one or more countries, and others are being developed and tested. This review assesses the different strategies currently proposed to control COVID-19 and the issues or limitations imposed on some approaches by the human and viral genetic variability.
Collapse
Affiliation(s)
- David Livingstone Alves Figueiredo
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Medicina, Guarapuava, PR, Brazil
- Instituto para Pesquisa do Câncer (IPEC), Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - João Paulo Bianchi Ximenez
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Análises Clínicas, Toxicologia e Ciência de Alimentos, Ribeirão Preto, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Universidade Estadual do Norte do Paraná (UENP), Centro de Ciências Biológicas, Bandeirantes, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Carolina Panis
- Universidade Estadual do Oeste do Paraná, Francisco Beltrão, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Rafael dos Santos Bezerra
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Hemocentro Regional de Ribeirão Preto, Ribeirão Preto, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Adriano Ferrasa
- Universidade Estadual de Ponta Grossa, Ponta Grossa, Programa de Pós Graduação em Computação Aplicada, Ponta Grossa, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Alessandra Lourenço Cecchini
- Universidade Estadual de Londrina, Departamento de Patologia Geral, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Alexandra Ivo de Medeiros
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Ana Marisa Fusco Almeida
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Anelisa Ramão
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Ciências Biológicas, Guarapuava, PR, Brazil
| | - Angelica Beate Winter Boldt
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Carla Fredrichsen Moya
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Medicina Veterinária, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Chung Man Chin
- Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Fármacos e Medicamentos, Araraquara, SP, Brazil
- União das Faculdades dos Grandes Lagos (UNILAGO), Centro de Pesquisa Avançada em Medicina, São José do Rio Preto, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Daniel de Paula
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Farmácia, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Daniel Rech
- Universidade Estadual do Oeste do Paraná (UNIOESTE), Hospital do Câncer Francisco Beltrão, Laboratório de Biologia de Tumores, Francisco Beltrão, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Daniela Fiori Gradia
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Danielle Malheiros
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Danielle Venturini
- Universidade Estadual de Londrina, Centro de Ciências da Saúde, Departamento de patologia, clínica e toxicologia, Laboratório de bioquímica clínica, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Eliandro Reis Tavares
- Universidade Estadual de Londrina, Departamento de Microbiologia, Centro de Ciências Biológicas, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Emerson Carraro
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Laboratório de Virologia Clínica, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Enilze Maria de Souza Fonseca Ribeiro
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Evani Marques Pereira
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Enfermagem, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Felipe Francisco Tuon
- Universidade Católica do Paraná, Laboratório de Doenças Infecciosas Emergentes, Pontifícia Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Franciele Aní Caovilla Follador
- Universidade Estadual do Oeste do Paraná, Departamento de Ciências da Vida, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Francisco Beltrão, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Glaura Scantamburlo Alves Fernandes
- Universidade Estadual de Londrina, Departamento de Biologia Geral, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Hélito Volpato
- Universidade Estadual do Paraná (UNESPAR), Faculdade de Ciências Biológicas, Centro de Ciências Humanas e Educação, Paranavaí, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Ilce Mara de Syllos Cólus
- Universidade Estadual de Londrina, Departamento de Biologia Geral, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Jaqueline Carvalho de Oliveira
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Jean Henrique da Silva Rodrigues
- Universidade do Estado de São Paulo (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Fármacos e Medicamentos, Araraquara, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Jean Leandro dos Santos
- Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Fármacos e Medicamentos, Araraquara, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Jeane Eliete Laguila Visentainer
- Universidade Estadual de Maringá, Laboratório de Imunogenética, Maringá, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Juliana Cristina Brandi
- Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Juliana Mara Serpeloni
- Universidade Estadual de Londrina, Departamento de Biologia Geral, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Juliana Sartori Bonini
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Laboratório de Neuropsicofarmacologia, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Karen Brajão de Oliveira
- Universidade Estadual de Londrina, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Laboratório de Genética Molecular e Imunologia, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Karine Fiorentin
- Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Léia Carolina Lucio
- Universidade Estadual do Oeste do Paraná, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Centro de Ciências da Saúde, Francisco Beltrão, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Ligia Carla Faccin-Galhardi
- Universidade Estadual de Londrina, Departamento de Microbiologia, Centro de Ciências Biológicas, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Lirane Elize Defante Ferreto
- Universidade Estadual do Oeste do Paraná, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Centro de Ciências da Saúde, Francisco Beltrão, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Lucy Megumi Yamauchi Lioni
- Universidade Estadual do Norte do Paraná (UENP), Centro de Ciências Biológicas, Bandeirantes, PR, Brazil
- Universidade Estadual de Londrina, Departamento de Microbiologia, Centro de Ciências Biológicas, Londrina, PR, Brazil
| | - Marcia Edilaine Lopes Consolaro
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Maringá, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Marcelo Ricardo Vicari
- Universidade Estadual de Ponta Grossa, Departamento de Biologia e Genética Estrutural e Molecular, Ponta Grossa, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Marcos Abdo Arbex
- Universidade de Araraquara, Faculdade de Medicina, Área temática de Pneumologia, Araraquara, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Marcos Pileggi
- Universidade Estadual de Ponta Grossa, Departamento de Biologia e Genética Estrutural e Molecular, Ponta Grossa, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Maria Angelica Ehara Watanabe
- Universidade Estadual de Londrina, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Laboratório de Imunologia, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Maria Antônia Ramos Costa
- Universidade do Estado do Paraná, Colegiada de Enfermagem, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Maria José S. Mendes Giannini
- Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Marla Karine Amarante
- Universidade Estadual de Londrina, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Laboratório de Imunologia, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Najeh Maissar Khalil
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Farmácia, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Quirino Alves de Lima
- Universidade Estadual de Maringá, Laboratório de Imunogenética, Maringá, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Roberto H. Herai
- Universidade Católica do Paraná (PUCPR), Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde, Laboratório Experimental Multiusuário, Curitiba, PR, Brazil
- Universitário Católico Salesiano Auxilium (UNISALESIANO), Faculdade de Medicina, Centro Araçatuba, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Roberta Losi Guembarovski
- Universidade Estadual de Londrina, Departamento de Biologia Geral, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Rogério N. Shinsato
- Universidade Católica do Paraná (PUCPR), Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde, Laboratório Experimental Multiusuário, Curitiba, PR, Brazil
- Universitário Católico Salesiano Auxilium (UNISALESIANO), Faculdade de Medicina, Centro Araçatuba, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Rubiana Mara Mainardes
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Farmácia, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Silvana Giuliatti
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Hemocentro Regional de Ribeirão Preto, Ribeirão Preto, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Sueli Fumie Yamada-Ogatta
- Universidade Estadual de Londrina, Departamento de Microbiologia, Centro de Ciências Biológicas, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Viviane Knuppel de Quadros Gerber
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Enfermagem, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Wander Rogério Pavanelli
- Universidade Estadual de Londrina, Laboratório de Imunoparasitologia de Doenças Negligenciadas e Câncer, Londrina, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Weber Claudio da Silva
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Departamento de Farmácia, Guarapuava, PR, Brazil
- Universidade Estadual do Centro-Oeste do Paraná (UNICENTRO), Laboratório de Neuropsicofarmacologia, Guarapuava, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Maria Luiza Petzl-Erler
- Universidade Federal do Paraná, Programa de Pós-Graduação em Genética, Departamento de Genética, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Valeria Valente
- Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil
- Faculdade de Medicina de Ribeirão Preto, Centro de Terapia Celular (CEPID/FAPESP), Ribeirão Preto, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Christiane Pienna Soares
- Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Luciane Regina Cavalli
- Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| | - Wilson Araujo Silva
- Instituto para Pesquisa do Câncer (IPEC), Guarapuava, PR, Brazil
- Faculdade de Medicina de Ribeirão Preto, Centro de Terapia Celular (CEPID/FAPESP), Ribeirão Preto, SP, Brazil
- Instituto Nacional de Ciência e Tecnologia em Células-Tronco e Terapia Celular (INCT/CNPq), Ribeirão Preto, SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Genética, Ribeirão Preto, SP, Brazil
- Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Fundação Araucária, PR, Brazil
| |
Collapse
|
29
|
Kodchakorn K, Chokepaichitkool T, Kongtawelert P. Mutational scanning of spike RBD protein for enhanced ACE2 affinity emerging Southeast Asia in the late transmission phase. Sci Rep 2022; 12:5896. [PMID: 35393512 PMCID: PMC8989122 DOI: 10.1038/s41598-022-09999-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 03/31/2022] [Indexed: 12/11/2022] Open
Abstract
The COVID-19 pandemic has changed the quality of life and economic systems all over the world, as the virus can be transmitted from human to human via air-droplets. Since the SARS-CoV-2 virus was first identified in 2019, the virus has naturally mutated over time. Southeast Asia is one of the areas in the world that has implemented various procedures and measures to slow down the disease outbreaks. The first cluster of COVID-19 was identified from the tourist-travel history, and then the diversity of coronavirus victims has posed a serious issue of human security on a massive scale. To evaluate whether or not naturally occurring mutations have strengthened the infectivity of SARS-CoV-2, we computed in silico the structural dynamics of the RBD-spike protein mutation enhancing ACE2-binding. When considering emerging variations in Southeast Asia, 14 dominant mutations were analyzed by applying the structural and energetic characterization using MD simulations. The ones in the RBD region displayed higher affinity to ACE2 due to the improved interfacial stability of the RBD β-strand surrounding the ACE2 across salt bridge hotspots. The binding hotspots and structurally conserved conformational-epitopes have been identified, which are deleterious for RBD mutation and ACE2 binding. We present an interactive visualization to facilitate the development of effective neutralizing agents for vaccination, prevention and treatment.
Collapse
Affiliation(s)
- Kanchanok Kodchakorn
- Department of Biochemistry, Faculty of Medicine, Thailand Excellence Center for Tissue Engineering and Stem Cells, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Tawan Chokepaichitkool
- Department of Biochemistry, Faculty of Medicine, Thailand Excellence Center for Tissue Engineering and Stem Cells, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Prachya Kongtawelert
- Department of Biochemistry, Faculty of Medicine, Thailand Excellence Center for Tissue Engineering and Stem Cells, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
30
|
Strobelt R, Adler J, Paran N, Yahalom-Ronen Y, Melamed S, Politi B, Shulman Z, Schmiedel D, Shaul Y. Imatinib inhibits SARS-CoV-2 infection by an off-target-mechanism. Sci Rep 2022; 12:5758. [PMID: 35388061 PMCID: PMC8984672 DOI: 10.1038/s41598-022-09664-1] [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] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causal agent of the COVID-19 pandemic. More than 274 million individuals have suffered from COVID-19 and over five million people have died from this disease so far. Therefore, there is an urgent need for therapeutic drugs. Repurposing FDA approved drugs should be favored since evaluation of safety and efficacy of de-novo drug design are both costly and time consuming. We report that imatinib, an Abl tyrosine kinase inhibitor, robustly decreases SARS-CoV-2 infection and uncover a mechanism of action. We show that imatinib inhibits the infection of SARS-CoV-2 and its surrogate lentivector pseudotype. In latter, imatinib inhibited both routes of viral entry, endocytosis and membrane-fusion. We utilized a system to quantify in real-time cell-cell membrane fusion mediated by the SARS-CoV-2 surface protein, Spike, and its receptor, hACE2, to demonstrate that imatinib inhibits this process in an Abl1 and Abl2 independent manner. Furthermore, cellular thermal shift assay revealed a direct imatinib-Spike interaction that affects Spike susceptibility to trypsin digest. Collectively, our data suggest that imatinib inhibits Spike mediated viral entry by an off-target mechanism. These findings mark imatinib as a promising therapeutic drug in inhibiting the early steps of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Romano Strobelt
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Julia Adler
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Yfat Yahalom-Ronen
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Boaz Politi
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dominik Schmiedel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
31
|
Naik RR, Shakya AK, Aladwan SM, El-Tanani M. Kinase Inhibitors as Potential Therapeutic Agents in the Treatment of COVID-19. Front Pharmacol 2022; 13:806568. [PMID: 35444538 PMCID: PMC9014181 DOI: 10.3389/fphar.2022.806568] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Corona virus is quickly spreading around the world. The goal of viral management is to disrupt the virus's life cycle, minimize lung damage, and alleviate severe symptoms. Numerous strategies have been used, including repurposing existing antivirals or drugs used in previous viral outbreaks. One such strategy is to repurpose FDA-approved kinase inhibitors that are potential chemotherapeutic agents and have demonstrated antiviral activity against a variety of viruses, including MERS, SARS-CoV-1, and others, by inhibiting the viral life cycle and the inflammatory response associated with COVID-19. The purpose of this article is to identify licensed kinase inhibitors that have the ability to reduce the virus's life cycle, from entrance through viral propagation from cell to cell. Several of these inhibitors, including imatinib, ruxolitinib, silmitasertib, and tofacitinib (alone and in conjunction with hydroxychloroquine), are now undergoing clinical studies to determine their efficacy as a possible treatment drug. The FDA approved baricitinib (a Janus kinase inhibitor) in combination with remdesivir for the treatment of COVID-19 patients receiving hospital care in November 2020. While in vitro trials with gilteritinib, fedratinib, and osimertinib are encouraging, further research is necessary before these inhibitors may be used to treat COVID-19 patients.
Collapse
Affiliation(s)
- Rajashri R. Naik
- Department of Biopharmaceutics and Clinical Pharmacy, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Ashok K. Shakya
- Faculty of Pharmacy, Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Safwan M. Aladwan
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohamed El-Tanani
- Department of Biopharmaceutics and Clinical Pharmacy, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
- Faculty of Pharmacy, Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| |
Collapse
|
32
|
Mulgaonkar N, Wang H, Mallawarachchi S, Růžek D, Martina B, Fernando S. In silico and in vitro evaluation of imatinib as an inhibitor for SARS-CoV-2. J Biomol Struct Dyn 2022; 41:3052-3061. [PMID: 35220926 DOI: 10.1080/07391102.2022.2045221] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The rapid geographic expansion of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the infectious agent of Coronavirus Disease 2019 (COVID-19) pandemic, poses an immediate need for potent drugs. Enveloped viruses infect the host cell by cellular membrane fusion, a crucial mechanism required for virus replication. The SARS-CoV-2 spike glycoprotein, due to its primary interaction with the human angiotensin-converting enzyme 2 (ACE2) cell-surface receptor, is considered a potential target for drug development. In this study, around 5,800 molecules were virtually screened using molecular docking. Five molecules were selected for in vitro experiments from those that reported docking scores lower than -6 kcal/mol. Imatinib, a Bcr-Abl tyrosine kinase inhibitor, showed maximum antiviral activity in Vero cells. We further investigated the interaction of imatinib, a compound under clinical trials for the treatment of COVID-19, with SARS-CoV-2 RBD, using in silico methods. Molecular dynamics simulations verified that imatinib interacts with RBD residues that are critical for ACE2 binding. This study also provides significant molecular insights on potential repurposable small-molecule drugs and chemical scaffolds for the development of novel drugs targeting the SARS-CoV-2 spike RBD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nirmitee Mulgaonkar
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX, USA
| | - Haoqi Wang
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX, USA
| | - Samavath Mallawarachchi
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX, USA
| | - Daniel Růžek
- Veterinary Research Institute, Brno, and Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Byron Martina
- Artemis One Health Research Institute, Delft, The Netherlands
| | - Sandun Fernando
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX, USA
| |
Collapse
|
33
|
Alizadehmohajer N, Behmardi A, Najafgholian S, Moradi S, Mohammadi F, Nedaeinia R, Haghjooy Javanmard S, Sohrabi E, Salehi R, Ferns GA, Emami Nejad A, Manian M. Screening of potential inhibitors of COVID-19 with repurposing approach via molecular docking. NETWORK MODELING AND ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2022; 11:11. [PMID: 35136710 PMCID: PMC8814570 DOI: 10.1007/s13721-021-00341-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/14/2021] [Accepted: 10/01/2021] [Indexed: 01/20/2023]
Abstract
SARS-CoV-2 (COVID-19) is the causative organism for a pandemic disease with a high rate of infectivity and mortality. In this study, we aimed to assess the affinity between several available small molecule and proteins, including Abl kinase inhibitors, Janus kinase inhibitor, dipeptidyl peptidase 4 inhibitors, RNA-dependent RNA polymerase inhibitors, and Papain-like protease inhibitors, using binding simulation, to test whether they may be effective in inhibiting COVID-19 infection through several mechanisms. The efficiency of inhibitors was evaluated based on docking scores using AutoDock Vina software. Strong ligand-protein interactions were predicted among some of these drugs, that included: Imatinib, Remdesivir, and Telaprevir, and this may render these compounds promising candidates. Some candidate drugs might be efficient in disease control as potential inhibitors or lead compounds against the SARS-CoV-2. It is also worth highlighting the powerful immunomodulatory role of other drugs, such as Abivertinib that inhibits pro-inflammatory cytokine production associated with cytokine release syndrome (CRS) and the progression of COVID-19 infection. The potential role of other Abl kinase inhibitors, including Imatinib in reducing SARS-CoV and MERS-CoV viral titers, immune regulatory function and the development of acute respiratory distress syndrome (ARDS), indicate that this drug may be useful for COVID-19, as the SARS-CoV-2 genome is similar to SARS-CoV.
Collapse
Affiliation(s)
- Negin Alizadehmohajer
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milano, Italy
| | - Abtin Behmardi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Simin Najafgholian
- Department of Emergency Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shabnam Moradi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Forogh Mohammadi
- Department of Veterinary, Agriculture Faculty, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Ehsan Sohrabi
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A. Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, BN1 9PH Sussex UK
| | - Asieh Emami Nejad
- Department of Biology, Payame Noor University (PNU), P.O. Box 19395-3697, Tehran, Iran
| | - Mostafa Manian
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
34
|
Abstract
The global coronavirus disease-19 (COVID-19) has affected more than 140 million and killed more than 3 million people worldwide as of April 20, 2021. The novel human severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been identified as an etiological agent for COVID-19. Several kinases have been proposed as possible mediators of multiple viral infections, including life-threatening coronaviruses like SARS-CoV-1, Middle East syndrome coronavirus (MERS-CoV), and SARS-CoV-2. Viral infections hijack abundant cell signaling pathways, resulting in drastic phosphorylation rewiring in the host and viral proteins. Some kinases play a significant role throughout the viral infection cycle (entry, replication, assembly, and egress), and several of them are involved in the virus-induced hyperinflammatory response that leads to cytokine storm, acute respiratory distress syndrome (ARDS), organ injury, and death. Here, we highlight kinases that are associated with coronavirus infections and their inhibitors with antiviral and potentially anti-inflammatory, cytokine-suppressive, or antifibrotic activity.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry
and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University
Tübingen, Auf der Morgenstelle 8, 72076 Tübingen,
Germany
| | - Stefan Laufer
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry
and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University
Tübingen, Auf der Morgenstelle 8, 72076 Tübingen,
Germany
| |
Collapse
|
35
|
Song C, Li D, Zhang J, Zhao X. Role of ferroptosis in promoting cardiotoxicity induced by Imatinib Mesylate via down-regulating Nrf2 pathways in vitro and in vivo. Toxicol Appl Pharmacol 2022; 435:115852. [PMID: 34973290 DOI: 10.1016/j.taap.2021.115852] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/19/2021] [Accepted: 12/23/2021] [Indexed: 01/20/2023]
Abstract
Imatinib Mesylate (IMA) has been widely used to treat with chronic myeloid leukemia (CML). However, cardiotoxicity associated with IMA is included among the therapeutic strategies. The present study was aimed to discover whether ferroptosis, a programmed iron-dependent cell death, is involved in IMA-induced cardiotoxicity. In vivo, mouse model was established after treated with 25 mg/kg, 50 mg/kg and 100 mg/kg IMA. Serum CK, LDH, AST activities were determined. Cardiac tissues were examined by H&E and Oil Red O staining. MDA was measured to assess production of lipid peroxide. Tissue iron and GSH content were measured. In vitro, cell viability, mitochondria membrane potential, generation of reactive oxygen species (ROS) and cellular iron levels were performed to explore the mechanism of IMA. The in vivo results revealed that IMA treatment significantly increased serum CK, LDH and AST. H&E staining showed that IMA caused cardiac structural injuries. The dose-dependent decrease of GSH and increase of tissue iron and MDA were observed in IMA-treated groups. Oil Red O staining suggested obvious cardiac lipid accumulation after treated with IMA. In H9c2 cardiomyocytes, IMA significantly inhibited cell proliferation in a dose-dependent manner. Mitochondria membrane potential assay showed that IMA destroyed the mitochondrial function. Additionally, IMA increased the cellular ROS and iron levels. Furthermore, IMA down-regulated the expression of Nrf2 and up-regulated the expression of P53 and TfR. These results provided compelling evidence that ferroptosis participates in IMA-induced cardiotoxicity. Ferroptosis could be regarded as a target to protect against cardiotoxicity in IMA-exposed patients.
Collapse
Affiliation(s)
- Chengzhu Song
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Dongning Li
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China.
| | - Xiaoyan Zhao
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| |
Collapse
|
36
|
Serra A, Fratello M, Federico A, Ojha R, Provenzani R, Tasnadi E, Cattelani L, Del Giudice G, Kinaret PAS, Saarimäki LA, Pavel A, Kuivanen S, Cerullo V, Vapalahti O, Horvath P, Lieto AD, Yli-Kauhaluoma J, Balistreri G, Greco D. Computationally prioritized drugs inhibit SARS-CoV-2 infection and syncytia formation. Brief Bioinform 2021; 23:6484515. [PMID: 34962256 PMCID: PMC8769897 DOI: 10.1093/bib/bbab507] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022] Open
Abstract
The pharmacological arsenal against the COVID-19 pandemic is largely based on generic anti-inflammatory strategies or poorly scalable solutions. Moreover, as the ongoing vaccination campaign is rolling slower than wished, affordable and effective therapeutics are needed. To this end, there is increasing attention toward computational methods for drug repositioning and de novo drug design. Here, multiple data-driven computational approaches are systematically integrated to perform a virtual screening and prioritize candidate drugs for the treatment of COVID-19. From the list of prioritized drugs, a subset of representative candidates to test in human cells is selected. Two compounds, 7-hydroxystaurosporine and bafetinib, show synergistic antiviral effects in vitro and strongly inhibit viral-induced syncytia formation. Moreover, since existing drug repositioning methods provide limited usable information for de novo drug design, the relevant chemical substructures of the identified drugs are extracted to provide a chemical vocabulary that may help to design new effective drugs.
Collapse
Affiliation(s)
- Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Michele Fratello
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Ravi Ojha
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riccardo Provenzani
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ervin Tasnadi
- Synthetic and Systems Biology Unit, Biological Research Centre, Eotvos Lorand Research Network, Szeged, Hungary
| | - Luca Cattelani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Giusy Del Giudice
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Pia A S Kinaret
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Laura A Saarimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Suvi Kuivanen
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland.,Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Peter Horvath
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.,Synthetic and Systems Biology Unit, Biological Research Centre, Eotvos Lorand Research Network, Szeged, Hungary
| | - Antonio Di Lieto
- Department of Forensic Psychiatry, Aarhus University, Aarhus, Denmark
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Giuseppe Balistreri
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
37
|
Peralta-Garcia A, Torrens-Fontanals M, Stepniewski TM, Grau-Expósito J, Perea D, Ayinampudi V, Waldhoer M, Zimmermann M, Buzón MJ, Genescà M, Selent J. Entrectinib-A SARS-CoV-2 Inhibitor in Human Lung Tissue (HLT) Cells. Int J Mol Sci 2021; 22:13592. [PMID: 34948390 PMCID: PMC8707862 DOI: 10.3390/ijms222413592] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Since the start of the COVID-19 outbreak, pharmaceutical companies and research groups have focused on the development of vaccines and antiviral drugs against SARS-CoV-2. Here, we apply a drug repurposing strategy to identify drug candidates that are able to block the entrance of the virus into human cells. By combining virtual screening with in vitro pseudovirus assays and antiviral assays in Human Lung Tissue (HLT) cells, we identify entrectinib as a potential antiviral drug.
Collapse
Affiliation(s)
- Alejandro Peralta-Garcia
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences, Hospital del Mar Medical Research Institute (IMIM), Pompeu Fabra University (UPF), 08003 Barcelona, Spain; (A.P.-G.); (M.T.-F.); (T.M.S.)
| | - Mariona Torrens-Fontanals
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences, Hospital del Mar Medical Research Institute (IMIM), Pompeu Fabra University (UPF), 08003 Barcelona, Spain; (A.P.-G.); (M.T.-F.); (T.M.S.)
| | - Tomasz Maciej Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences, Hospital del Mar Medical Research Institute (IMIM), Pompeu Fabra University (UPF), 08003 Barcelona, Spain; (A.P.-G.); (M.T.-F.); (T.M.S.)
- InterAx Biotech AG, PARK InnovAARE, 5234 Villigen, Switzerland; (V.A.); (M.W.); (M.Z.)
| | - Judith Grau-Expósito
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain; (J.G.-E.); (D.P.); (M.J.B.); (M.G.)
| | - David Perea
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain; (J.G.-E.); (D.P.); (M.J.B.); (M.G.)
| | - Vikram Ayinampudi
- InterAx Biotech AG, PARK InnovAARE, 5234 Villigen, Switzerland; (V.A.); (M.W.); (M.Z.)
| | - Maria Waldhoer
- InterAx Biotech AG, PARK InnovAARE, 5234 Villigen, Switzerland; (V.A.); (M.W.); (M.Z.)
| | - Mirjam Zimmermann
- InterAx Biotech AG, PARK InnovAARE, 5234 Villigen, Switzerland; (V.A.); (M.W.); (M.Z.)
| | - María J. Buzón
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain; (J.G.-E.); (D.P.); (M.J.B.); (M.G.)
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain; (J.G.-E.); (D.P.); (M.J.B.); (M.G.)
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences, Hospital del Mar Medical Research Institute (IMIM), Pompeu Fabra University (UPF), 08003 Barcelona, Spain; (A.P.-G.); (M.T.-F.); (T.M.S.)
| |
Collapse
|
38
|
Nesr G, Saleem Z, Arami S. Outcome of COVID-19 in patients with chronic myeloid leukemia: A single centre UK experience. J Med Virol 2021; 94:1274-1276. [PMID: 34914847 DOI: 10.1002/jmv.27527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 11/10/2022]
Affiliation(s)
- George Nesr
- Haematology Department, London North West University Healthcare NHS Trust, London, UK
| | - Zahbia Saleem
- Haematology Department, London North West University Healthcare NHS Trust, London, UK
| | - Siamak Arami
- Haematology Department, London North West University Healthcare NHS Trust, London, UK
| |
Collapse
|
39
|
Ribone SR, Paz SA, Abrams CF, Villarreal MA. Target identification for repurposed drugs active against SARS-CoV-2 via high-throughput inverse docking. J Comput Aided Mol Des 2021; 36:25-37. [PMID: 34825285 PMCID: PMC8616721 DOI: 10.1007/s10822-021-00432-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022]
Abstract
Screening already approved drugs for activity against a novel pathogen can be an important part of global rapid-response strategies in pandemics. Such high-throughput repurposing screens have already identified several existing drugs with potential to combat SARS-CoV-2. However, moving these hits forward for possible development into drugs specifically against this pathogen requires unambiguous identification of their corresponding targets, something the high-throughput screens are not typically designed to reveal. We present here a new computational inverse-docking protocol that uses all-atom protein structures and a combination of docking methods to rank-order targets for each of several existing drugs for which a plurality of recent high-throughput screens detected anti-SARS-CoV-2 activity. We demonstrate validation of this method with known drug-target pairs, including both non-antiviral and antiviral compounds. We subjected 152 distinct drugs potentially suitable for repurposing to the inverse docking procedure. The most common preferential targets were the human enzymes TMPRSS2 and PIKfyve, followed by the viral enzymes Helicase and PLpro. All compounds that selected TMPRSS2 are known serine protease inhibitors, and those that selected PIKfyve are known tyrosine kinase inhibitors. Detailed structural analysis of the docking poses revealed important insights into why these selections arose, and could potentially lead to more rational design of new drugs against these targets.
Collapse
Affiliation(s)
- Sergio R Ribone
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), X5000HUA, Córdoba, Argentina
| | - S Alexis Paz
- Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , X5000HUA, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Fisicoquímica de Córdoba (INFIQC), X5000HUA, Córdoba, Argentina
| | - Cameron F Abrams
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Marcos A Villarreal
- Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , X5000HUA, Córdoba, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Fisicoquímica de Córdoba (INFIQC), X5000HUA, Córdoba, Argentina.
| |
Collapse
|
40
|
Hu S, Jiang S, Qi X, Bai R, Ye XY, Xie T. Races of small molecule clinical trials for the treatment of COVID-19: An up-to-date comprehensive review. Drug Dev Res 2021; 83:16-54. [PMID: 34762760 PMCID: PMC8653368 DOI: 10.1002/ddr.21895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
The coronavirus disease‐19 (COVID‐19) pandemic has become a global threat since its first outbreak at the end of 2019. Several review articles have been published recently, focusing on the aspects of target biology, drug repurposing, and mechanisms of action (MOAs) for potential treatment. This review gathers all small molecules currently in active clinical trials, categorizes them into six sub‐classes, and summarizes their clinical progress. The aim is to provide the researchers from both pharmaceutical industries and academic institutes with the handful information and dataset to accelerate their research programs in searching effective small molecule therapy for treatment of COVID‐19.
Collapse
Affiliation(s)
- Suwen Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China.,Hangzhou Huadong Medicine Group, Pharmaceutical Research Institute Co. Ltd., Hangzhou, China.,Department of Chemistry and Biochemistry Los Angeles, University of California, Los Angeles, California, USA
| | - Songwei Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Xiang Qi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| |
Collapse
|
41
|
Ali N, Prasad K, AlAsmari AF, Alharbi M, Rashid S, Kumar V. Genomics-guided targeting of stress granule proteins G3BP1/2 to inhibit SARS-CoV-2 propagation. Int J Biol Macromol 2021; 190:636-648. [PMID: 34517025 PMCID: PMC8431879 DOI: 10.1016/j.ijbiomac.2021.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 08/07/2021] [Accepted: 09/03/2021] [Indexed: 12/27/2022]
Abstract
SARS-CoV-2 nucleocapsid (N) protein undergoes RNA-induced phase separation (LLPS) and sequesters the host key stress granule (SG) proteins, Ras-GTPase-activating protein SH3-domain-binding protein 1 and 2 (G3BP1 and G3BP2) to inhibit SG formation. This will allow viral packaging and propagation in host cells. Based on a genomic-guided meta-analysis, here we identify upstream regulatory elements modulating the expression of G3BP1 and G3BP2 (collectively called G3BP1/2). Using this strategy, we have identified FOXA1, YY1, SYK, E2F-1, and TGFBR2 as activators and SIN3A, SRF, and AKT-1 as repressors of G3BP1/2 genes. Panels of the activators and repressors were then used to identify drugs that change their gene expression signatures. Two drugs, imatinib, and decitabine have been identified as putative modulators of G3BP1/2 genes and their regulators, suggesting their role as COVID-19 mitigation agents. Molecular docking analysis suggests that both drugs bind to G3BP1/2 with a much higher affinity than the SARS-CoV-2 N protein. This study reports imatinib and decitabine as candidate drugs against N protein and G3BP1/2 protein.
Collapse
Affiliation(s)
- Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Kartikay Prasad
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, UP 201303, India
| | - Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy Girls Section, Prince Sattam Bin AbdulAziz University, Al kharj 11942, Saudi Arabia
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, UP 201303, India.
| |
Collapse
|
42
|
Abdurrahman L, Fang X, Zhang Y. Molecular Insights of SARS-CoV-2 Infection and Molecular Treatments. Curr Mol Med 2021; 22:621-639. [PMID: 34645374 DOI: 10.2174/1566524021666211013121831] [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: 01/08/2021] [Revised: 06/15/2021] [Accepted: 07/23/2021] [Indexed: 01/18/2023]
Abstract
The coronavirus disease emerged in December 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome-related coronavirus 2 (SARS-CoV-2) and its rapid global spread has brought an international health emergency and urgent responses for seeking efficient prevention and therapeutic treatment. This has led to imperative needs for illustration of the molecular pathogenesis of SARS-CoV-2, identification of molecular targets or receptors, and development of antiviral drugs, antibodies, and vaccines. In this study, we investigated the current research progress in combating SARS-CoV-2 infection. Based on the published research findings, we first elucidated, at the molecular level, SARS-CoV-2 viral structures, potential viral host-cell-invasion and pathogenic mechanisms, main virus-induced immune responses, and emerging SARS-CoV-2 variants. We then focused on the main virus- and host-based potential targets, summarized and categorized effective inhibitory molecules based on drug development strategies for COVID-19, that can guide efforts for the identification of new drugs and treatment for this problematic disease. Current research and development of antibodies and vaccines were also introduced and discussed. We concluded that the main virus entry route- SARS-CoV-2 spike protein interaction with ACE2 receptors has played a key role in guiding the development of therapeutic treatments against COVID-19, four main therapeutic strategies may be considered in developing molecular therapeutics, and drug repurposing is likely to be an easy, fast and low-cost approach in such a short period of time with urgent need of antiviral drugs. Additionally, the quick development of antibody and vaccine candidates has yielded promising results, but the wide-scale deployment of safe and effective COVID-19 vaccines remains paramount in solving the pandemic crisis. As new variants of the virus begun to emerge, the efficacy of these vaccines and treatments must be closely evaluated. Finally, we discussed the possible challenges of developing molecular therapeutics for COVID-19 and suggested some potential future efforts. Despite the limited availability of literatures, our attempt in this work to provide a relatively comprehensive overview of current SARS-CoV-2 studies can be helpful for quickly acquiring the key information of COVID-19 and further promoting this important research to control and diminish the pandemic.
Collapse
Affiliation(s)
- Lama Abdurrahman
- Department of Chemistry, The University of Texas Rio Grande Valley, Edinburg, Texas 78539. United States
| | - Xiaoqian Fang
- Department of Molecular Science, School of Medicine, The University of Texas Rio Grande Valley, Edinburg, Texas 78539. United States
| | - Yonghong Zhang
- Department of Chemistry, The University of Texas Rio Grande Valley, Edinburg, Texas 78539. United States
| |
Collapse
|
43
|
Breccia M, Abruzzese E, Accurso V, Attolico I, Barulli S, Bergamaschi M, Binotto G, Bocchia M, Bonifacio M, Caocci G, Capodanno I, Castagnetti F, Cavazzini F, Crisà E, Crugnola M, Stella De Candia M, Elena C, Fava C, Galimberti S, Gozzini A, Gugliotta G, Intermesoli T, Iurlo A, La Barba G, Latagliata R, Leonetti Crescenzi S, Levato L, Loglisci G, Lucchesi A, Luciano L, Lunghi F, Luzi D, Malato A, Cristina Miggiano M, Pizzuti M, Pregno P, Rapezzi D, Rege-Cambrin G, Rosti G, Russo S, Sancetta R, Rita Scortechini A, Sorà F, Sportoletti P, Stagno F, Tafuri A, Tiribelli M, Foà R, Saglio G. COVID-19 infection in chronic myeloid leukaemia after one year of the pandemic in Italy. A Campus CML report. Br J Haematol 2021; 196:559-565. [PMID: 34636033 PMCID: PMC8652631 DOI: 10.1111/bjh.17890] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 11/30/2022]
Abstract
Limited information is available on the impact of the COVID-19 pandemic on the management of chronic myeloid leukaemia (CML). The Campus CML network collected retrospective information on 8 665 CML patients followed at 46 centres throughout Italy during the pandemic between February 2020 and January 2021. Within this cohort, we recorded 217 SARS-CoV-2-positive patients (2·5%). Most patients (57%) were diagnosed as having SARS-CoV-2 infection during the second peak of the pandemic (September 2020 to January 2021). The majority (35%) was aged between 50 and 65 years with a male prevalence (73%). Fifty-six percent of patients presented concomitant comorbidities. The median time from CML diagnosis to SARS-CoV-2 infection was six years (three months to 18 years). Twenty-one patients (9·6%) required hospitalization without the need of respiratory assistance, 18 (8·2%) were hospitalized for respiratory assistance, 8 (3·6%) were admitted to an intensive care unit, while 170 (78%) were only quarantined. Twenty-three percent of patients discontinued tyrosine kinase inhibitor (TKI) therapy during the infection. Twelve patients died due to COVID-19 with a mortality rate of 5·5% in the positive cohort and of 0·13% in the whole cohort. We could also document sequelae caused by the SARS-CoV-2 infection and an impact of the pandemic on the overall management of CML patients.
Collapse
Affiliation(s)
- Massimo Breccia
- Department of Translational and Precision Medicine, Sapienza University, Roma, Italy
| | | | - Vincenzo Accurso
- UO di Ematologia con Trapianto, AU Policlinico Paolo Giaccone, Palermo, Italy
| | - Immacolata Attolico
- UO Ematologia con Trapianto, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Sara Barulli
- Divisione di Ematologia di Muraglia, CTMO Ospedale San Salvatore, Pesaro, Italy
| | | | - Gianni Binotto
- Ematologia ed Immunologia Clinica, Università degli Studi di Padova, Padova, Italy
| | - Monica Bocchia
- U.O.C. Ematologia e Trapianti, A.O. Senese - Policlinico 'Le Scotte', Siena, Italy
| | - Massimiliano Bonifacio
- Divisione di Ematologia, Istituti Ospitalieri di Verona, Policlinico G.B. Rossi, Verona, Italy
| | | | | | - Fausto Castagnetti
- Istituto di Ematologia 'Lorenzo e A. Seragnoli', Policlinico S. Orsola-Malpighi, Bologna, Italy
| | | | - Elena Crisà
- S.C.D.U. Ematologia - DIMECS e Dipartimento Oncologico, Università del Piemonte Orientale Amedeo Avogadro, Novara, Italy
| | | | | | - Chiara Elena
- Ematologia, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Carmen Fava
- A.S.O. Ordine Mauriziano, P.O. Umberto I, Struttura Complessa a Direzione Universitaria-Ematologia e Terapie Cellulari, Torino, Italy
| | - Sara Galimberti
- Ematologia, Università di Pisa - Azienda Ospedaliera Pisana, Pisa, Italy
| | - Antonella Gozzini
- Unità di Ricerca e di Malattie del Sangue, Ematologia San Luca Vecchio, Firenze, Italy
| | - Gabriele Gugliotta
- Istituto di Ematologia 'Lorenzo e A. Seragnoli', Policlinico S. Orsola-Malpighi, Bologna, Italy
| | | | - Alessandra Iurlo
- UOC Ematologia, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | | | | | | | | | | | - Alessandro Lucchesi
- Ematologia, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - Luigiana Luciano
- Ematologia, Azienda Ospedaliera Universitaria - Università degli Studi di Napoli 'Federico II', Napoli, Italy
| | | | - Debora Luzi
- Ematologia, A.O. Santa Maria - Terni S.C Oncoematologia, Terni, Italy
| | | | | | | | - Patrizia Pregno
- Dipartimento di Oncologia ed Ematologia S.C. Ematologia 2, A.O. Città della Salute e della Scienza di Torino S. G. Battista, Torino, Italy
| | | | - Giovanna Rege-Cambrin
- Dipartimento di Scienze Cliniche e Biologiche, Ospedale S. Luigi Gonzaga-Medicina Interna 2, Orbassano, Italy
| | - Gianantonio Rosti
- Ematologia, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - Sabina Russo
- Dipartimento di Medicina Interna, Azienda Ospedaliera Universitaria - Policlinico G. Martino, Messina, Italy
| | | | | | - Federica Sorà
- Ematologia, Policlinico Gemelli - Università Cattolica del Sacro Cuore, Roma, Italy
| | | | - Fabio Stagno
- Hematology Section and BMT Unit, AOU Policlinico 'Rodolico-San Marco', Catania, Italy
| | | | - Mario Tiribelli
- Clinica Ematologica-Centro Trapianti e Terapie cellulari, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Robin Foà
- Department of Translational and Precision Medicine, Sapienza University, Roma, Italy
| | - Giuseppe Saglio
- A.S.O. Ordine Mauriziano, Struttura Complessa a Direzione Universitaria-Ematologia e Terapie Cellulari-, P.O. Umberto I, Torino, Italy
| |
Collapse
|
44
|
Suttorp M, Webster Carrion A, Hijiya N. Chronic Myeloid Leukemia in Children: Immune Function and Vaccinations. J Clin Med 2021; 10:jcm10184056. [PMID: 34575167 PMCID: PMC8470625 DOI: 10.3390/jcm10184056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
Children with CML need TKI treatment for many years, and the lack of knowledge about immune dysfunction with TKI has hindered routine immunizations. This review attempts to provide an overview of the effects of TKIs licensed for children (e.g., imatinib, dasatinib, and nilotinib) on immune function, as well as its implications on immunizations. We discuss surveillance strategies (e.g., immunoglobulin blood serum levels and hepatitis B reactivation) and immunizations. All inactivated vaccines (e.g., influenza, pneumococcal, and streptococcal) can be given during the treatment of CML in the chronic phase, although their efficacy may be lower. As shown in single cases of children and adults with CML, live vaccines (e.g., varicella, measles, mumps, rubella, and yellow fever) may be administered under defined circumstances with great precautions. We also highlight important aspects of COVID-19 in this patient population (e.g., the outcome of COVID-19 infection in adults with CML and in children with varying hemato-oncological diseases) and discuss the highly dynamic field of presently available different vaccination options. In conclusion, TKI treatment for CML causes humoral and cellular immune dysfunction, which is mild in most patients, and thus infectious complications are rare. Routine immunizations are important for health maintenance of children, but vaccinations for children with CML on TKI therapy should be carefully considered.
Collapse
Affiliation(s)
- Meinolf Suttorp
- Hematology and Oncology, Medical Faculty, Technical University, D-01307 Dresden, Germany
- Correspondence:
| | - Andrea Webster Carrion
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Columbia University Medical Center, New York, NY 10032, USA; (A.W.C.); (N.H.)
| | - Nobuko Hijiya
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Columbia University Medical Center, New York, NY 10032, USA; (A.W.C.); (N.H.)
| |
Collapse
|
45
|
Dowarah J, Marak BN, Yadav UCS, Singh VP. Potential drug development and therapeutic approaches for clinical intervention in COVID-19. Bioorg Chem 2021; 114:105016. [PMID: 34144277 PMCID: PMC8143914 DOI: 10.1016/j.bioorg.2021.105016] [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: 03/28/2021] [Accepted: 05/20/2021] [Indexed: 01/25/2023]
Abstract
While the vaccination is now available to many countries and will slowly dissipate to others, effective therapeutics for COVID-19 is still illusive. The SARS-CoV-2 pandemic has posed an unprecedented challenge to researchers, scientists, and clinicians and affected the wellbeing of millions of people worldwide. Since the beginning of the pandemic, a multitude of existing anti-viral, antibiotic, antimalarial, and anticancer drugs have been tested, and some have shown potency in the treatment and management of COVID-19, albeit others failed to leave any positive impact and a few also became controversial as they showed mixed clinical outcomes. In the present article, we have brought together some of the candidate therapeutic drugs being repurposed or used in the clinical trials and discussed their clinical efficacy and safety for COVID-19.
Collapse
Affiliation(s)
- Jayanta Dowarah
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | - Brilliant N Marak
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | | | - Ved Prakash Singh
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India; Department of Industrial Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India.
| |
Collapse
|
46
|
Karnik M, Beeraka NM, Uthaiah CA, Nataraj SM, Bettadapura ADS, Aliev G, Madhunapantula SV. A Review on SARS-CoV-2-Induced Neuroinflammation, Neurodevelopmental Complications, and Recent Updates on the Vaccine Development. Mol Neurobiol 2021; 58:4535-4563. [PMID: 34089508 PMCID: PMC8179092 DOI: 10.1007/s12035-021-02399-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a devastating viral infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The incidence and mortality of COVID-19 patients have been increasing at an alarming rate. The mortality is much higher in older individuals, especially the ones suffering from respiratory distress, cardiac abnormalities, renal diseases, diabetes, and hypertension. Existing evidence demonstrated that SARS-CoV-2 makes its entry into human cells through angiotensin-converting enzyme 2 (ACE-2) followed by the uptake of virions through cathepsin L or transmembrane protease serine 2 (TMPRSS2). SARS-CoV-2-mediated abnormalities in particular cardiovascular and neurological ones and the damaged coagulation systems require extensive research to develop better therapeutic modalities. As SARS-CoV-2 uses its S-protein to enter into the host cells of several organs, the S-protein of the virus is considered as the ideal target to develop a potential vaccine. In this review, we have attempted to highlight the landmark discoveries that lead to the development of various vaccines that are currently under different stages of clinical progression. Besides, a brief account of various drug candidates that are being tested to mitigate the burden of COVID-19 was also covered. Further, in a dedicated section, the impact of SARS-CoV-2 infection on neuronal inflammation and neuronal disorders was discussed. In summary, it is expected that the content covered in this article help to understand the pathophysiology of COVID-19 and the impact on neuronal complications induced by SARS-CoV-2 infection while providing an update on the vaccine development.
Collapse
Affiliation(s)
- Medha Karnik
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Narasimha M Beeraka
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Chinnappa A Uthaiah
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Suma M Nataraj
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Anjali Devi S Bettadapura
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region, 142432, Russia
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418, Russia
- GALLY International Research Institute, 7733 Louis Pasteur Drive, San Antonio, TX, #330, USA
| | - SubbaRao V Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| |
Collapse
|
47
|
Bonifacio M, Tiribelli M, Miggiano MC, Abruzzese E, Binotto G, Scaffidi L, Cordioli M, Damiani D, Di Bona E, Trawinska MM, Tanasi I, Dubbini MV, Velotta V, Ceccarelli G, Pierdomenico E, Lo Schirico M, Semenzato G, Ruggeri M, Fanin R, Tacconelli E, Pizzolo G, Krampera M. The serological prevalence of SARS-CoV-2 infection in patients with chronic myeloid leukemia is similar to that in the general population. Cancer Med 2021; 10:6310-6316. [PMID: 34464516 PMCID: PMC8446554 DOI: 10.1002/cam4.4179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/30/2021] [Accepted: 07/28/2021] [Indexed: 12/18/2022] Open
Abstract
Background Patients with hematological malignancies are at an increased risk of SARS‐CoV‐2 disease (COVID‐19) and adverse outcome. However, a low mortality rate has been reported in patients with chronic myeloid leukemia (CML). Preclinical evidence suggests that tyrosine kinase inhibitors (TKIs) may have a protective role against severe COVID‐19. Methods We conducted a cross‐sectional study of 564 consecutive patients with CML who were tested for anti‐SARS‐CoV‐2 IgG/IgM antibodies at their first outpatient visit between May and early November 2020 in five hematologic centers representative of three Italian regions. Results The estimated serological prevalence of SARS‐CoV‐2 infection in patients with CML after the first pandemic wave was similar to that in the general population (about 2%), both at national and regional levels. CML patients with positive anti‐SARS‐CoV‐2 serology were more frequently male (p = 0.027) and active workers (p = 0.012), while there was no significant association with TKI treatment type. Only 3 out of 11 IgG‐positive patients had previously received a molecular diagnosis of COVID‐19, while the remainders were asymptomatic or with mild symptoms. Conclusions Our data confirm that the course of SARS‐CoV‐2 infection in patients with CML is generally mild and reassure about the safety of continuing TKIs during the COVID‐19 pandemic. Furthermore, we suggest that patients with CML succeed to mount an antibody response after exposure to SARS‐CoV‐2, similar to the general population.
Collapse
Affiliation(s)
| | - Mario Tiribelli
- Division of Hematology and BMT, Department of Medical Area, University of Udine, Udine, Italy
| | - Maria Cristina Miggiano
- Hematology Department, San Bortolo Hospital, Azienda ULSS8 "Berica" of Vicenza, Vicenza, Italy
| | | | - Gianni Binotto
- Padua School of Medicine, Department of Medicine, Hematology and Clinical Immunology, Padua, Italy
| | - Luigi Scaffidi
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Maddalena Cordioli
- Department of Diagnostics and Public Health, Section of Infectious Diseases, University of Verona, Verona, Italy
| | - Daniela Damiani
- Division of Hematology and BMT, Department of Medical Area, University of Udine, Udine, Italy
| | - Eros Di Bona
- Hematology Department, San Bortolo Hospital, Azienda ULSS8 "Berica" of Vicenza, Vicenza, Italy
| | | | - Ilaria Tanasi
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Maria Vittoria Dubbini
- Division of Hematology and BMT, Department of Medical Area, University of Udine, Udine, Italy
| | - Vanessa Velotta
- Hematology, S. Eugenio Hospital, ASL Roma2, Tor Vergata University, Rome, Italy
| | - Giulia Ceccarelli
- Hematology, S. Eugenio Hospital, ASL Roma2, Tor Vergata University, Rome, Italy
| | - Elisabetta Pierdomenico
- Padua School of Medicine, Department of Medicine, Hematology and Clinical Immunology, Padua, Italy
| | - Mariella Lo Schirico
- Padua School of Medicine, Department of Medicine, Hematology and Clinical Immunology, Padua, Italy
| | - Gianpietro Semenzato
- Padua School of Medicine, Department of Medicine, Hematology and Clinical Immunology, Padua, Italy
| | - Marco Ruggeri
- Hematology Department, San Bortolo Hospital, Azienda ULSS8 "Berica" of Vicenza, Vicenza, Italy
| | - Renato Fanin
- Division of Hematology and BMT, Department of Medical Area, University of Udine, Udine, Italy
| | - Evelina Tacconelli
- Department of Diagnostics and Public Health, Section of Infectious Diseases, University of Verona, Verona, Italy
| | - Giovanni Pizzolo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| |
Collapse
|
48
|
Razizadeh M, Nikfar M, Liu Y. Small molecule therapeutics to destabilize the ACE2-RBD complex: A molecular dynamics study. Biophys J 2021; 120:2793-2804. [PMID: 34214539 PMCID: PMC8241573 DOI: 10.1016/j.bpj.2021.06.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/23/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
The ongoing coronavirus disease 19 (COVID-19) pandemic has infected millions of people, claimed hundreds of thousands of lives, and made a worldwide health emergency. Understanding the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mechanism of infection is crucial in the development of potential therapeutics and vaccines. The infection process is triggered by direct binding of the SARS-CoV-2 receptor-binding domain (RBD) to the host-cell receptor angiotensin-converting enzyme 2 (ACE2). Many efforts have been made to design or repurpose therapeutics to deactivate the RBD or ACE2 and prevent the initial binding. In addition to direct inhibition strategies, small chemical compounds might be able to interfere and destabilize the metastable, prefusion complex of ACE2-RBD. This approach can be employed to prevent the further progress of virus infection at its early stages. In this study, molecular docking was employed to analyze the binding of two chemical compounds, SSAA09E2 and Nilotinib, with the druggable pocket of the ACE2-RBD complex. The structural changes as a result of the interference with the ACE2-RBD complex were analyzed by molecular dynamics simulations. Results show that both Nilotinib and SSAA09E2 can induce significant conformational changes in the ACE2-RBD complex, intervene with the hydrogen bonds, and influence the flexibility of proteins. Moreover, essential dynamics analysis suggests that the presence of small molecules can trigger large-scale conformational changes that may destabilize the ACE2-RBD complex.
Collapse
Affiliation(s)
- Meghdad Razizadeh
- Department of Mechanical Engineering and Mechanics, Bethlehem, Pennsylvania
| | - Mehdi Nikfar
- Department of Mechanical Engineering and Mechanics, Bethlehem, Pennsylvania
| | - Yaling Liu
- Department of Mechanical Engineering and Mechanics, Bethlehem, Pennsylvania; Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania.
| |
Collapse
|
49
|
Pagnano KB, Peralta EH, Navarro JR, David Salas LDR, Delgado N, Moiraghi B, Toreli ACM, Perobelli LM, Fechio L, Quixada ATS, Funke V, Bendit I, Seguro FS, Pilleux L, Bortolini J, Lourenço ALG, Sapelli J, Nucci FM, Pavlovsky C, Oliveira LDC, Moura MS, Palma LC, Gonçalves NN, Conchon M, Hokama POM, Almeida LL, Zulli R, de Souza CA, Boquimpani CM. COVID-19 in chronic myeloid leukemia patients in Latin America. Leuk Lymphoma 2021; 62:3212-3218. [PMID: 34254886 DOI: 10.1080/10428194.2021.1950709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
This observational, multicenter study aimed to report the clinical evolution of COVID-19 in patients with chronic myeloid leukemia in Latin America. A total of 92 patients presented with COVID-19 between March and December 2020, 26% of whom were severe or critical. The median age at COVID-19 diagnosis was 48 years (22-79 years), 32% were 60 years or older, and 61% were male. Thirty-nine patients presented with at least one comorbidity (42.3%). Eighty-one patients recovered (88%), and 11 (11.9%) died from COVID-19. There was one case of reinfection. Patients with a major molecular response presented superior overall survival compared to patients with no major molecular response (91 vs. 61%, respectively; p = 0.004). Patients in treatment-free remission and receiving tyrosine kinase inhibitors showed higher survival rates than patients who underwent hematopoietic stem cell transplantation and those who did not receive tyrosine kinase inhibitors (100, 89, 50, and 33%, respectively; p < 0.001).
Collapse
Affiliation(s)
- Katia B Pagnano
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| | | | | | | | - Nancy Delgado
- Centro Médico Nacional Siglo XXI IMSS, Cuauhtémoc, México Ciudad de México
| | | | | | - Leila M Perobelli
- Hospital de Transplantes Euryclides de Jesus Zerbini, São Paulo, Brazil
| | - Leonardo Fechio
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| | - Acy T S Quixada
- Faculdade de Medicina da Universidade Federal do Ceará, Hospital Universitário Walter Cantídio, Fortaleza, Brazil
| | - Vaneuza Funke
- Complexo Hospital de Clínicas - Universidade Federal do Paraná, Curitiba, Brazil
| | - Israel Bendit
- Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda S Seguro
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | - Fabio M Nucci
- Hospital Universitário Antonio Pedro, Universidade Federal Fluminense, Niterói, Brazil
| | | | | | | | - Leonardo C Palma
- Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | - Paula O M Hokama
- São Paulo State University, UNESP, Medical School, Campus de Botucatu, Botucatu, Brazil
| | | | - Roberto Zulli
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| | | | | |
Collapse
|
50
|
Touret F, Driouich JS, Cochin M, Petit PR, Gilles M, Barthélémy K, Moureau G, Mahon FX, Malvy D, Solas C, de Lamballerie X, Nougairède A. Preclinical evaluation of Imatinib does not support its use as an antiviral drug against SARS-CoV-2. Antiviral Res 2021; 193:105137. [PMID: 34265358 PMCID: PMC8274277 DOI: 10.1016/j.antiviral.2021.105137] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/09/2022]
Abstract
Following the emergence of SARS-CoV-2, the search for an effective and rapidly available treatment was initiated worldwide based on repurposing of available drugs. Previous reports described the antiviral activity of certain tyrosine kinase inhibitors (TKIs) targeting the Abelson kinase 2 against pathogenic coronaviruses. Imatinib, one of them, has more than twenty years of safe utilization for the treatment of hematological malignancies. In this context, Imatinib was rapidly evaluated in clinical trials against Covid-19. Here, we present the pre-clinical evaluation of imatinib in multiple models. Our results indicated that imatinib and another TKI, the masitinib, exhibit an antiviral activity in VeroE6 cells. However, imatinib was inactive in a reconstructed bronchial human airway epithelium model. In vivo, imatinib therapy failed to impair SARS-CoV-2 replication in a golden Syrian hamster model despite high concentrations in plasma and in the lung. Overall, these results do not support the use of imatinib and similar TKIs as antivirals in the treatment of Covid-19.
Collapse
Affiliation(s)
- Franck Touret
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France.
| | - Jean-Sélim Driouich
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| | - Maxime Cochin
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| | - Paul Rémi Petit
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| | - Magali Gilles
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| | - Karine Barthélémy
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| | - Grégory Moureau
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| | - Francois-Xavier Mahon
- Cancer Center of Bordeaux, Institut Bergonié, INSERM U1218, University of Bordeaux, Bordeaux, France
| | - Denis Malvy
- Department for Infectious and Tropical Diseases, University Hospital Center of Bordeaux, Bordeaux, France; Inserm 1219, University of Bordeaux, Bordeaux, France
| | - Caroline Solas
- APHM, Unité des Virus Émergents (UVE: Aix Marseille University IRD 190-Inserm 1207-IHU Méditerranée Infection), Laboratoire de Pharmacocinétique et Toxicologie, Hôpital La Timone, Marseille, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| | - Antoine Nougairède
- Unité des Virus Émergents (UVE: Aix-Marseille University -IRD 190-Inserm 1207-IHU Méditerranée Infection), Marseille, France
| |
Collapse
|