1
|
Oruçoğlu B, Çetin İ, Şimşek H, Topçul M, Çalışkan M, Aydın C, Kavaklı IH, Okyar A, Gül Ş. Identification of potential SARS-CoV-2 inhibitors among well-tolerated drugs using drug repurposing and in vitro approaches. Sci Rep 2025; 15:13975. [PMID: 40263343 PMCID: PMC12015351 DOI: 10.1038/s41598-025-88388-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/28/2025] [Indexed: 04/24/2025] Open
Abstract
The 3C-like protease (3CLpro) is essential in the SARS-CoV-2 life cycle and a promising target for antiviral drug discovery, as no similar proteases exist in humans. This study aimed to identify effective SARS-CoV-2 inhibitors among FDA-approved drugs. Previous computational analysis revealed several drugs with high binding affinity to the 3CLpro active site. In vitro enzymatic assays confirmed that ten of these drugs effectively inhibited the enzyme. To evaluate their impact on viral replication, we used non-infectious SARS-CoV-2 sub-genomic replicons in lung and intestinal cells. Amcinonide, eltrombopag, lumacaftor, candesartan, and nelfinavir inhibited replication at low micromolar concentrations. Lumacaftor showed IC50 values of 964 nM in Caco-2 cells and 458 nM in Calu-3 cells, while candesartan had IC50 values of 714 nM and 1.05 µM, respectively. Furthermore, dual combination experiments revealed that amcinonide, pimozide, lumacaftor, and eltrombopag acted as potent inhibitors at nanomolar concentrations when combined with candesartan. This study highlights lumacaftor, candesartan, and nelfinavir as effective inhibitors of SARS-CoV-2 replication in vitro and emphasizes their potential for repurposing as antiviral treatments. These findings support future clinical trials and may lead to breakthroughs in COVID-19 treatment strategies.
Collapse
Affiliation(s)
- Betül Oruçoğlu
- Biotechnology Division, Department of Biology, Istanbul University, Istanbul, Türkiye
- Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Türkiye
| | - İdil Çetin
- Biotechnology Division, Department of Biology, Istanbul University, Istanbul, Türkiye
| | - Handan Şimşek
- Biotechnology Division, Department of Biology, Istanbul University, Istanbul, Türkiye
| | - Mehmet Topçul
- Biotechnology Division, Department of Biology, Istanbul University, Istanbul, Türkiye
| | - Mahmut Çalışkan
- Biotechnology Division, Department of Biology, Istanbul University, Istanbul, Türkiye
| | - Cihan Aydın
- Department of Molecular Biology, Faculty of Engineering and Natural Sciences, Istanbul Medeniyet University, Istanbul, Türkiye
- Istanbul Medeniyet University Science and Advanced Technology Research Center (IMU-BILTAM), Istanbul, Türkiye
| | - I Halil Kavaklı
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Türkiye
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Türkiye
| | - Alper Okyar
- Faculty of Pharmacy, Department of Pharmacology, İstanbul University, Istanbul, Türkiye
| | - Şeref Gül
- Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Türkiye.
| |
Collapse
|
2
|
Goto K, Amano R, Ichinose A, Michishita A, Hamada M, Nakamura Y, Takahashi M. Generation of RNA aptamers against chikungunya virus E2 envelope protein. J Virol 2025; 99:e0209524. [PMID: 39927773 PMCID: PMC11915788 DOI: 10.1128/jvi.02095-24] [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: 12/02/2024] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Nucleic acid aptamers are a promising drug modality, whereas the generation of virus-neutralizing aptamers has remained difficult due to the lack of a robust system for targeting the viral particles of interest. Here, we took advantage of our latest platform technology of Systematic Evolution of Ligands by EXponential enrichment (SELEX) with virus-like particles (VLPs) and targeted chikungunya virus (CHIKV) as a model, the pathogenic reemerging virus with an unmet need for control. The identified aptamer against CHIKV-VLPs, Apt#1, and its truncated derivatives showed neutralizing activity with nanomolar IC50 values in a cell-based assay system using a pseudoviral particle of CHIKV (CHIKVpp). An antiviral-based chemical genetics approach revealed significant competition of Apt#1 with suramin, a reported interactant with domain A of the E2 envelope protein (E2DA), in both CHIKVpp and surface plasmon resonance (SPR) analyses, predicting E2DA to be the Apt#1 interface. In addition, Apt#1 interfered with the attachment of CHIKVpp, collectively suggesting its property as an attachment inhibitor via E2DA of CHIKV. Thus, the generation of the VLP-targeted aptamers proved to contribute to anti-CHIKV strategies and confirmed the utility of the platform as a novel and viable option for the development of neutralizing agents against viral particles of interest.IMPORTANCEOur latest SELEX technology using VLPs has generated aptamers that bind the native conformation of the incorporated envelope protein and achieve the virus binding and neutralizing effects. Indeed, the aptamer-probed target E2DA is a representative neutralization site on the surface of the viral particle, validating the utility of the VLP-driven procedure. Simultaneously, the enhanced antiviral effects of the aptamer in combination with approved drugs using the CHIKVpp assay with human cells indicated potential therapeutic strategies that are expected to help address unmet needs in CHIKV control. The robust affinity of the aptamer to viral particles demonstrated by SPR analysis can also lead to conjugates with antivirals as guiding molecules and aptasensors for diagnostic tools. Overall, our VLP-based method provided anti-CHIKV as well as a versatile platform applicable to other emerging and reemerging viruses, in preparation for outbreaks with the need for rapid development of antiviral strategies as next-generation theranostics.
Collapse
Affiliation(s)
- Kaku Goto
- Project Division of RNA Medical Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryo Amano
- Project Division of RNA Medical Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akiko Ichinose
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Akiya Michishita
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Michiaki Hamada
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yoshikazu Nakamura
- Project Division of RNA Medical Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- RIBOMIC Inc., Tokyo, Japan
| | - Masaki Takahashi
- Project Division of RNA Medical Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
3
|
Jittamala P, Boyd S, Schilling WH, Watson JA, Ngamprasertchai T, Siripoon T, Luvira V, Batty EM, Wongnak P, Esper LM, Almeida PJ, Cruz C, Ascencao FR, Aguiar RS, Ghanchi NK, Callery JJ, Singh S, Kruabkontho V, Ngernseng T, Tubprasert J, Madmanee W, Suwannasin K, Promsongsil A, Hanboonkunupakarn B, Poovorawan K, Potaporn M, Srisubat A, Loharjun B, Taylor WR, Qamar F, Kazi AM, Beg MA, Chommanam D, Vidhamaly S, Chotivanich K, Imwong M, Pukrittayakamee S, Dondorp AM, Day NP, Teixeira MM, Piyaphanee W, Phumratanaprapin W, White NJ, PLATCOV Collaborative Group. Antiviral efficacy of fluoxetine in early symptomatic COVID-19: an open-label, randomised, controlled, adaptive platform trial (PLATCOV). EClinicalMedicine 2025; 80:103036. [PMID: 39896880 PMCID: PMC11787712 DOI: 10.1016/j.eclinm.2024.103036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/24/2024] [Accepted: 12/16/2024] [Indexed: 02/04/2025] Open
Abstract
Background The selective serotonin reuptake inhibitors (SSRIs) fluoxetine and fluvoxamine were repurposed for the treatment of early COVID-19 based on their antiviral activity in vitro, and observational and clinical trial evidence suggesting they prevented progression to severe disease. However, these SSRIs have not been recommended in therapeutic guidelines and their antiviral activity in vivo has not been characterised. Methods PLATCOV is an open-label, multicentre, phase 2, randomised, controlled, adaptive pharmacometric platform trial running in Thailand, Brazil, Pakistan, and Laos. We recruited low-risk adult outpatients aged 18-50 with early symptomatic COVID-19 (symptoms <4 days) between 5 April 2022 and 8 May 2023. Patients were assigned using block randomisation to one of eleven treatment arms including oral fluoxetine (40 mg/day for 7 days), or no study drug. Uniform randomisation ratios were applied across the active treatment groups while the no study drug group comprised ≥20% of patients at all times. The primary endpoint was the rate of oropharyngeal viral clearance assessed until day 7. Measurements were taken daily between days 0 and 7 and analysed in a modified intention-to-treat population (>2 days follow-up).The viral clearance rate was estimated under a Bayesian hierarchical linear model fitted to the log10 viral densities measured in standardised duplicate oropharyngeal swab eluates taken daily over one week (18 measurements per patient). Secondary endpoints were all-cause hospital admission at 28 days, and time to resolution of fever and symptoms. This ongoing trial is registered at ClinicalTrials.gov (NCT05041907). Findings 271 patients were concurrently randomised to either fluoxetine (n = 120) or no study drug (n = 151). All patients had received at least one COVID-19 vaccine dose and 67% were female (182/271). In the primary analysis, viral clearance rates following fluoxetine were compatible with a small or no increase relative to the no study drug arm (15% increase; 95% credible interval (CrI): -2 to 34%). There were no deaths or hospitalisations in either arm. There were no significant differences in times to symptom resolution or fever clearance between the fluoxetine and the no study drug arms (although only a quarter of patients were febrile at baseline). Fluoxetine was well tolerated, there were no serious adverse events and only one grade 3 adverse event in the intervention arm. Interpretation Overall, the evidence from this study is compatible with fluoxetine having a weak in vivo antiviral activity against SARS-CoV-2, although the primary endpoint is also compatible with no effect. This level of antiviral efficacy is substantially less than with other currently available antiviral drugs. Funding Wellcome Trust Grant ref: 223195/Z/21/Z through the COVID-19 Therapeutics Accelerator.
Collapse
Affiliation(s)
- Podjanee Jittamala
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Simon Boyd
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - William H.K. Schilling
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - James A. Watson
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory, Big Data Institute, Oxford, United Kingdom
| | - Thundon Ngamprasertchai
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tanaya Siripoon
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Viravarn Luvira
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Elizabeth M. Batty
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Phrutsamon Wongnak
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Lisia M. Esper
- Clinical Research Unit, Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pedro J. Almeida
- Clinical Research Unit, Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cintia Cruz
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Fernando R. Ascencao
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Renato S. Aguiar
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - James J. Callery
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Shivani Singh
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Varaporn Kruabkontho
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thatsanun Ngernseng
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jaruwan Tubprasert
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanassanan Madmanee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kanokon Suwannasin
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Amornrat Promsongsil
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Borimas Hanboonkunupakarn
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kittiyod Poovorawan
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Manus Potaporn
- Department of Medical Services, Ministry of Public Health, Bangkok, Thailand
| | - Attasit Srisubat
- Department of Medical Services, Ministry of Public Health, Bangkok, Thailand
| | - Bootsakorn Loharjun
- Department of Medical Services, Ministry of Public Health, Bangkok, Thailand
| | - Walter R.J. Taylor
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | - Danoy Chommanam
- Laos-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Laos
| | | | - Kesinee Chotivanich
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mallika Imwong
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sasithon Pukrittayakamee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Arjen M. Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nicholas P.J. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mauro M. Teixeira
- Clinical Research Unit, Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Watcharapong Piyaphanee
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Weerapong Phumratanaprapin
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - PLATCOV Collaborative Group
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory, Big Data Institute, Oxford, United Kingdom
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Clinical Research Unit, Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Aga Khan University, Karachi, Pakistan
- Department of Medical Services, Ministry of Public Health, Bangkok, Thailand
- Laos-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Laos
- Pulmonology Department, Mahosot Hospital, Vientiane, Laos
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
4
|
Monistrol-Mula A, Giné-Vázquez I, Caggiu G, Conflitti C, Gemes K, Hecker I, Mediavilla R, Monzio Compagnoni M, Pinucci I, Stoffers-Winterling J, Witteveen AB, Smith P, Walter H, Ayuso-Mateos JL, Melchior M, Mittendorfer-Rutz E, Sijbrandij M, Haro JM, Felez-Nobrega M. SARS-CoV-2 infection and COVID-19 outcomes across mental disorders and the role of sex: A register-based study from Catalonia. Psychiatry Res 2025; 344:116325. [PMID: 39708616 DOI: 10.1016/j.psychres.2024.116325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
INTRODUCTION This study investigated the risk of SARS-CoV-2 infection and severe COVID-19 outcomes among different mental health diagnoses and the role of sex in these associations. METHODS Using electronic records from Catalonia, we identified adults receiving mental health care from 2017-2019 with diagnoses of non-affective psychosis (NAP), bipolar disorder (BD), depressive disorder (DEP), stress-related disorders, neurotic/somatoform disorders (NSD), and substance misuse (SUB) (exposed). The outcomes assessed were SARS-CoV-2 infection, COVID-19 hospitalization, and COVID-19-related death, compared to matched individuals without these mental disorders (unexposed). Adjusted logistic regression analyses were conducted. RESULTS 785,378 adults were included (70.3% < 65 years old; 57.1% women). Compared to unexposed, those with NAP, BD, DEP, and SUB had a lower risk of SARS-CoV-2 infection, while those with NSD had an increased risk. Infected individuals with DEP, NSD, and SUB had a lower risk of hospitalization but a higher risk of COVID-19-related death. Higher COVID-19-related death was also observed in individuals with NAP and BD. Sex-stratified analysis revealed that women with NSD were especially vulnerable to infection, and women with DEP and NSD had a higher risk of COVID-19-related death. CONCLUSIONS These findings emphasize the need for tailored public health strategies to reduce excess mortality risk among individuals with certain mental disorders, while accounting for sex differences.
Collapse
Affiliation(s)
- Anna Monistrol-Mula
- Group of Epidemiology of Mental Disorders and Ageing, Sant Joan de Déu Research Institute, Esplugues de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Iago Giné-Vázquez
- Group of Epidemiology of Mental Disorders and Ageing, Sant Joan de Déu Research Institute, Esplugues de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Giulia Caggiu
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy; National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy; Department of Mental Health and Addiction Services, ASST Lecco, 23900 Lecco, Italy
| | - Claudia Conflitti
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy; Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - Katalin Gemes
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Irwin Hecker
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie Et de Santé Publique, IPLESP, Equipe de Recherche en Epidémiologie Sociale, ERES, Paris, France
| | - Roberto Mediavilla
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, Madrid, Spain
| | - Matteo Monzio Compagnoni
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy; National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy
| | - Irene Pinucci
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Jutta Stoffers-Winterling
- Leibniz Institute for Resilience Research, Mainz, Germany; University Medical Center of the Johannes Gutenberg Universität Mainz, Mainz, Germany
| | - Anke B Witteveen
- Department of Clinical, Neuro- and Developmental Psychology, WHO Collaborating Centre of Research and Dissemination of Psychological Interventions, Amsterdam Public Health Research Institute, VU University, Amsterdam, the Netherlands
| | - Pierre Smith
- Sciensano (Belgian Institute for Health), Department of Epidemiology and Public Health, Health Information Service, Brussels, Belgium; Université catholique de Louvain, Institute of Health and Society (IRSS), Brussels, Belgium
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jose Luis Ayuso-Mateos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, Madrid, Spain
| | - Maria Melchior
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie Et de Santé Publique, IPLESP, Equipe de Recherche en Epidémiologie Sociale, ERES, Paris, France
| | - Ellenor Mittendorfer-Rutz
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Marit Sijbrandij
- Department of Clinical, Neuro- and Developmental Psychology, WHO Collaborating Centre of Research and Dissemination of Psychological Interventions, Amsterdam Public Health Research Institute, VU University, Amsterdam, the Netherlands
| | - Josep Maria Haro
- Group of Epidemiology of Mental Disorders and Ageing, Sant Joan de Déu Research Institute, Esplugues de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, Universitat de Barcelona, Barcelona, Spain.
| | - Mireia Felez-Nobrega
- Group of Epidemiology of Mental Disorders and Ageing, Sant Joan de Déu Research Institute, Esplugues de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Jung J, Kim TH, Park JY, Kwon S, Sung JS, Kang MJ, Jose J, Lee M, Shin HJ, Pyun JC. SARS-CoV-2 vaccine based on ferritin complexes with screened immunogenic sequences from the Fv-antibody library. J Mater Chem B 2025; 13:1383-1394. [PMID: 39668674 DOI: 10.1039/d4tb01595a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
In this study, the vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was developed using ferritin complexes with the immunogenic sequences screened against the SARS-CoV-2 spike protein (SP) from the Fv-antibody library. The Fv-antibody library was prepared on the outer membrane of E. coli by the expression of the VH region of immunoglobulin G (IgG) with a randomized complementarity-determining region 3 (CDR3). Four Fv-antibodies to the receptor-binding domain (RBD) were screened from the Fv-antibody library, which had a comparable binding constant (KD) between SARS-CoV-2 SP and the angiotensin-converting enzyme 2 (ACE2) receptor. The binding sites of screened Fv-antibodies on the RBD were analyzed using a docking analysis, and these binding sites were used as immunogenic sequences for the vaccine. The four immunogenic sequences were modified and co-expressed as a part of ferritin which was assembled into a ferritin complex. After the vaccination of ferritin complexes to mice, the anti-sera were analyzed to have a high enough titer. Additionally, the immune responses were found to be activated by vaccination, such as the expression of IgG subclasses and the increased level of cytokines. The neutralizing activity of the anti-sera was estimated using a cell-based infection assay based on pseudo-virus expressing the SP of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Tae-Hun Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Jae-Yeon Park
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul 02456, Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, University of Munster, Münster (48149), Germany
| | - Misu Lee
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Hyun-Jin Shin
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| |
Collapse
|
6
|
Monistrol-Mula A, Felez-Nobrega M, Byrne EM, Lind PA, Hickie IB, Martin NG, Medland SE, Colodro-Conde L, Mitchell BL. The effect of polygenic liability to mental disorders on COVID-19 outcomes in people with depression: the mediating role of anxiety. Psychol Med 2024:1-10. [PMID: 39552393 DOI: 10.1017/s0033291724001983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
BACKGROUND Genetic vulnerability to mental disorders has been associated with coronavirus disease-19 (COVID-19) outcomes. We explored whether polygenic risk scores (PRSs) for several mental disorders predicted poorer clinical and psychological COVID-19 outcomes in people with pre-existing depression. METHODS Data from three assessments of the Australian Genetics of Depression Study (N = 4405; 52.2 years ± 14.9; 76.2% females) were analyzed. Outcomes included COVID-19 clinical outcomes (severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2] infection and long COVID, noting the low incidence of COVID-19 cases in Australia at that time) and COVID-19 psychological outcomes (COVID-related stress and COVID-19 burnout). Predictors included PRS for depression, bipolar disorder, schizophrenia, and anxiety. The associations between these PRSs and the outcomes were assessed with adjusted linear/logistic/multinomial regressions. Mediation (N = 4338) and moderation (N = 3326) analyses were performed to explore the potential influence of anxiety symptoms and resilience on the identified associations between the PRSs and COVID-19 psychological outcomes. RESULTS None of the selected PRS predicted SARS-CoV-2 infection or long COVID. In contrast, the depression PRS predicted higher levels of COVID-19 burnout. Anxiety symptoms fully mediated the association between the depression PRS and COVID-19 burnout. Resilience did not moderate this association. CONCLUSIONS A higher genetic risk for depression predicted higher COVID-19 burnout and this association was fully mediated by anxiety symptoms. Interventions targeting anxiety symptoms may be effective in mitigating the psychological effects of a pandemic among people with depression.
Collapse
Affiliation(s)
- Anna Monistrol-Mula
- Group of Epidemiology of Psychiatric disorders and Ageing, Sant Joan de Déu Research Institute, Sant Boi de Llobregat, Barcelona, Spain
- Centre for Biomedical Research on Mental Health (CIBERSAM), Madrid, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Mireia Felez-Nobrega
- Group of Epidemiology of Psychiatric disorders and Ageing, Sant Joan de Déu Research Institute, Sant Boi de Llobregat, Barcelona, Spain
- Centre for Biomedical Research on Mental Health (CIBERSAM), Madrid, Spain
| | - Enda M Byrne
- Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Penelope A Lind
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Ian B Hickie
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas G Martin
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sarah E Medland
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Psychology, The University of Queensland, Brisbane, QLD, Australia
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Lucía Colodro-Conde
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Psychology, The University of Queensland, Brisbane, QLD, Australia
| | - Brittany L Mitchell
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
7
|
Lopez Quezada L, Mba Medie F, Luu RJ, Gaibler RB, Gabriel EP, Rubio LD, Mulhern TJ, Marr EE, Borenstein JT, Fisher CR, Gard AL. Predicting Clinical Outcomes of SARS-CoV-2 Drug Efficacy with a High-Throughput Human Airway Microphysiological System. Adv Biol (Weinh) 2024; 8:e2300511. [PMID: 39123296 DOI: 10.1002/adbi.202300511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/23/2023] [Indexed: 08/12/2024]
Abstract
The average cost to bring a new drug from its initial discovery to a patient's bedside is estimated to surpass $2 billion and requires over a decade of research and development. There is a need for new drug screening technologies that can parse drug candidates with increased likelihood of clinical utility early in development in order to increase the cost-effectiveness of this pipeline. For example, during the COVID-19 pandemic, resources were rapidly mobilized to identify effective therapeutic treatments but many lead antiviral compounds failed to demonstrate efficacy when progressed to human trials. To address the lack of predictive preclinical drug screening tools, PREDICT96-ALI, a high-throughput (n = 96) microphysiological system (MPS) that recapitulates primary human tracheobronchial tissue,is adapted for the evaluation of differential antiviral efficacy of native SARS-CoV-2 variants of concern. Here, PREDICT96-ALI resolves both the differential viral kinetics between variants and the efficacy of antiviral compounds over a range of drug doses. PREDICT96-ALI is able to distinguish clinically efficacious antiviral therapies like remdesivir and nirmatrelvir from promising lead compounds that do not show clinical efficacy. Importantly, results from this proof-of-concept study track with known clinical outcomes, demonstrate the feasibility of this technology as a prognostic drug discovery tool.
Collapse
Affiliation(s)
| | | | - Rebeccah J Luu
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | - Logan D Rubio
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | | | | | - Ashley L Gard
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
8
|
Chen W, Jiang S, Li C, Li S, Wang J, Xu R. Potential association between COVID-19 and neurological disorders: analysis of common genes and therapeutics. Front Neurol 2024; 15:1417183. [PMID: 39469068 PMCID: PMC11513677 DOI: 10.3389/fneur.2024.1417183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/29/2024] [Indexed: 10/30/2024] Open
Abstract
As the COVID-19 pandemic persists, the increasing evidences suggest that the patients with COVID-19 may face the risks of the neurological complications and sequelae. To address this issue, we conducted a comprehensive study aimed at exploring the relationship between COVID-19 and various neurological disorders, with a particular focus on the shared dysregulated genes and the potential therapeutic targets. We selected six neurological disorders for investigation, including Alzheimer's disease, epilepsy, stroke, Parkinson's disease, and the sleep disorders. Through the bioinformatics analysis of the association between these disorders and COVID-19, we aimed to uncover the common molecular mechanisms and the potential treatment pathways. In this study, we utilized the publicly available RNA-Seq and microarray datasets, and employed tools such as Limma and DESeq2 for the differential gene analysis. Through the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, we explored the common biological features and pathways. Additionally, we focused on analyzing the regulatory roles of miRNA and transcription factors on the shared differentially expressed genes, and predicted the potential drugs interacting with these genes. These analyses contribute to a better understanding of the relationship between COVID-19 and the neurological disorders, and provide a theoretical basis for the future treatment strategies. Through this research, we aim to offer the deeper insights to the scientific community and present the new perspectives for the clinical practice in addressing the challenges of the neurological complications and sequelae faced by the COVID-19 patients.
Collapse
Affiliation(s)
- Wenzhi Chen
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Shishi Jiang
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Cheng Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Shu Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| |
Collapse
|
9
|
Lorkiewicz P, Waszkiewicz N. Viral infections in etiology of mental disorders: a broad analysis of cytokine profile similarities - a narrative review. Front Cell Infect Microbiol 2024; 14:1423739. [PMID: 39206043 PMCID: PMC11349683 DOI: 10.3389/fcimb.2024.1423739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
The recent pandemic caused by the SARS-CoV-2 virus and the associated mental health complications have renewed scholarly interest in the relationship between viral infections and the development of mental illnesses, a topic that was extensively discussed in the previous century in the context of other viruses, such as influenza. The most probable and analyzable mechanism through which viruses influence the onset of mental illnesses is the inflammation they provoke. Both infections and mental illnesses share a common characteristic: an imbalance in inflammatory factors. In this study, we sought to analyze and compare cytokine profiles in individuals infected with viruses and those suffering from mental illnesses. The objective was to determine whether specific viral diseases can increase the risk of specific mental disorders and whether this risk can be predicted based on the cytokine profile of the viral disease. To this end, we reviewed existing literature, constructed cytokine profiles for various mental and viral diseases, and conducted comparative analyses. The collected data indicate that the risk of developing a specific mental illness cannot be determined solely based on cytokine profiles. However, it was observed that the combination of IL-8 and IL-10 is frequently associated with psychotic symptoms. Therefore, to assess the risk of mental disorders in infected patients, it is imperative to consider the type of virus, the mental complications commonly associated with it, the predominant cytokines to evaluate the risk of psychotic symptoms, and additional patient-specific risk factors.
Collapse
Affiliation(s)
- Piotr Lorkiewicz
- Department of Psychiatry, Medical University of Bialystok, Białystok, Poland
| | | |
Collapse
|
10
|
Khawaja T, Kajova M, Levonen I, Pietilä JP, Välimaa H, Paajanen J, Pakkanen SH, Patjas A, Montonen R, Miettinen S, Virtanen J, Smura T, Sironen T, Fagerlund R, Ugurlu H, Iheozor-Ejiofor R, Saksela K, Vahlberg T, Ranki A, Vierikko A, Ihalainen J, Vapalahti O, Kantele A. Double-blinded, randomised, placebo-controlled trial of convalescent plasma for COVID-19: analyses by neutralising antibodies homologous to recipients' variants. Infect Dis (Lond) 2024; 56:423-433. [PMID: 38513074 DOI: 10.1080/23744235.2024.2329957] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Convalescent plasma (CP) emerged as potential treatment for COVID-19 early in the pandemic. While efficacy in hospitalised patients has been lacklustre, CP may be beneficial at the first stages of disease. Despite multiple new variants emerging, no trials have involved analyses on variant-specific antibody titres of CP. METHODS We recruited hospitalised COVID-19 patients within 10 days of symptom onset and, employing a double-blinded approach, randomised them to receive 200 ml convalescent plasma with high (HCP) or low (LCP) neutralising antibody (NAb) titre against the ancestral strain (Wuhan-like variant) or placebo in 1:1:1 ratio. Primary endpoints comprised intubation, corticosteroids for symptom aggravation, and safety assessed as serious adverse events. For a preplanned ad hoc analysis, the patients were regrouped by infused CP's NAb titers to variants infecting the recipients i.e. by titres of homologous HCP (hHCP) or LCP (hLCP). RESULTS Of the 57 patients, 18 received HCP, 19 LCP and 20 placebo, all groups smaller than planned. No significant differences were found for primary endpoints. In ad hoc analysis, hHCPrecipients needed significantly less respiratory support, and appeared to be given corticosteroids less frequently (1/14; 7.1%) than those receiving hLCP (9/23; 39.1%) or placebo (8/20; 40%), (p = 0.077). DISCUSSION Our double-blinded, placebo-controlled CP therapy trial remained underpowered and does not allow any firm conclusions for early-stage hospitalised COVID-19 patients. Interestingly, however, regrouping by homologous - recipients' variant-specific - CP titres suggested benefits for hHCP. We encourage similar re-analysis of ongoing/previous larger CP studies. TRIAL REGISTRATION ClinTrials.gov identifier: NCT0473040.
Collapse
Affiliation(s)
- T Khawaja
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - M Kajova
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - I Levonen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J P Pietilä
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - H Välimaa
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - J Paajanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Pulmonary Medicine, Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - S H Pakkanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - A Patjas
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - R Montonen
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - S Miettinen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - J Virtanen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - T Smura
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - T Sironen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - R Fagerlund
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - H Ugurlu
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - R Iheozor-Ejiofor
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - K Saksela
- Department of Virology, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Centre, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - T Vahlberg
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - A Ranki
- Department of Dermatology, Allergology and Venereology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - A Vierikko
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - J Ihalainen
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - O Vapalahti
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Centre, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - A Kantele
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Thümmler L, Beckmann N, Sehl C, Soddemann M, Braß P, Bormann M, Brochhagen L, Elsner C, Hoertel N, Cougoule C, Ciesek S, Widera M, Dittmer U, Lindemann M, Horn PA, Witzke O, Kadow S, Kamler M, Gulbins E, Becker KA, Krawczyk A. Fluoxetine and Sertraline Potently Neutralize the Replication of Distinct SARS-CoV-2 Variants. Viruses 2024; 16:545. [PMID: 38675888 PMCID: PMC11053511 DOI: 10.3390/v16040545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
The pandemic caused by SARS-CoV-2 is still a major health problem. Newly emerging variants and long-COVID-19 represent a challenge for the global health system. In particular, individuals in developing countries with insufficient health care need easily accessible, affordable and effective treatments of COVID-19. Previous studies have demonstrated the efficacy of functional inhibitors of acid sphingomyelinase against infections with various viruses, including early variants of SARS-CoV-2. This work investigated whether the acid sphingomyelinase inhibitors fluoxetine and sertraline, usually used as antidepressant molecules in clinical practice, can inhibit the replication of the former and recently emerged SARS-CoV-2 variants in vitro. Fluoxetine and sertraline potently inhibited the infection with pseudotyped virus-like particles and SARS-CoV-2 variants D614G, alpha, delta, omicron BA.1 and omicron BA.5. These results highlight fluoxetine and sertraline as priority candidates for large-scale phase 3 clinical trials at different stages of SARS-CoV-2 infections, either alone or in combination with other medications.
Collapse
Affiliation(s)
- Laura Thümmler
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (P.B.); (M.B.); (L.B.); (O.W.)
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (M.L.); (P.A.H.)
| | - Nadine Beckmann
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany (C.S.); (M.S.); (S.K.); (E.G.); (K.A.B.)
| | - Carolin Sehl
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany (C.S.); (M.S.); (S.K.); (E.G.); (K.A.B.)
| | - Matthias Soddemann
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany (C.S.); (M.S.); (S.K.); (E.G.); (K.A.B.)
| | - Peer Braß
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (P.B.); (M.B.); (L.B.); (O.W.)
| | - Maren Bormann
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (P.B.); (M.B.); (L.B.); (O.W.)
| | - Leonie Brochhagen
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (P.B.); (M.B.); (L.B.); (O.W.)
| | - Carina Elsner
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (C.E.); (U.D.)
| | - Nicolas Hoertel
- Institute Psychiatry and Neuroscience de Paris, INSERM U1266, Paris Cité University, 75014 Paris, France;
- Psychiatry and Addiction Department Corentin-Celton Hospital (AP-HP), 92130 Paris, France
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), CNRS, University of Toulouse, UPS, 31000 Toulouse, France;
| | - Sandra Ciesek
- Institute of Medical Virology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany; (S.C.); (M.W.)
- Institute of Pharmaceutical Biology, Goethe-University, 60323 Frankfurt am Main, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Branch Translational Medicine and Pharmacology, 60311 Frankfurt am Main, Germany
| | - Marek Widera
- Institute of Medical Virology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany; (S.C.); (M.W.)
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (C.E.); (U.D.)
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (M.L.); (P.A.H.)
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (M.L.); (P.A.H.)
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (P.B.); (M.B.); (L.B.); (O.W.)
| | - Stephanie Kadow
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany (C.S.); (M.S.); (S.K.); (E.G.); (K.A.B.)
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, West German Heart Center, University Hospital Essen, 45147 Essen, Germany;
| | - Erich Gulbins
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany (C.S.); (M.S.); (S.K.); (E.G.); (K.A.B.)
| | - Katrin Anne Becker
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany (C.S.); (M.S.); (S.K.); (E.G.); (K.A.B.)
| | - Adalbert Krawczyk
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (P.B.); (M.B.); (L.B.); (O.W.)
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (C.E.); (U.D.)
| |
Collapse
|
12
|
Alkafaas SS, Abdallah AM, Hassan MH, Hussien AM, Elkafas SS, Loutfy SA, Mikhail A, Murad OG, Elsalahaty MI, Hessien M, Elshazli RM, Alsaeed FA, Ahmed AE, Kamal HK, Hafez W, El-Saadony MT, El-Tarabily KA, Ghosh S. Molecular docking as a tool for the discovery of novel insight about the role of acid sphingomyelinase inhibitors in SARS- CoV-2 infectivity. BMC Public Health 2024; 24:395. [PMID: 38321448 PMCID: PMC10848368 DOI: 10.1186/s12889-024-17747-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Recently, COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants, caused > 6 million deaths. Symptoms included respiratory strain and complications, leading to severe pneumonia. SARS-CoV-2 attaches to the ACE-2 receptor of the host cell membrane to enter. Targeting the SARS-CoV-2 entry may effectively inhibit infection. Acid sphingomyelinase (ASMase) is a lysosomal protein that catalyzes the conversion of sphingolipid (sphingomyelin) to ceramide. Ceramide molecules aggregate/assemble on the plasma membrane to form "platforms" that facilitate the viral intake into the cell. Impairing the ASMase activity will eventually disrupt viral entry into the cell. In this review, we identified the metabolism of sphingolipids, sphingolipids' role in cell signal transduction cascades, and viral infection mechanisms. Also, we outlined ASMase structure and underlying mechanisms inhibiting viral entry 40 with the aid of inhibitors of acid sphingomyelinase (FIASMAs). In silico molecular docking analyses of FIASMAs with inhibitors revealed that dilazep (S = - 12.58 kcal/mol), emetine (S = - 11.65 kcal/mol), pimozide (S = - 11.29 kcal/mol), carvedilol (S = - 11.28 kcal/mol), mebeverine (S = - 11.14 kcal/mol), cepharanthine (S = - 11.06 kcal/mol), hydroxyzin (S = - 10.96 kcal/mol), astemizole (S = - 10.81 kcal/mol), sertindole (S = - 10.55 kcal/mol), and bepridil (S = - 10.47 kcal/mol) have higher inhibition activity than the candidate drug amiodarone (S = - 10.43 kcal/mol), making them better options for inhibition.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Abanoub Mosaad Abdallah
- Narcotic Research Department, National Center for Social and Criminological Research (NCSCR), Giza, 11561, Egypt
| | - Mai H Hassan
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Aya Misbah Hussien
- Biotechnology department at Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Abanoub Mikhail
- Department of Physics, Faculty of Science, Minia University, Minia, Egypt
- Faculty of Physics, ITMO University, Saint Petersburg, Russia
| | - Omnia G Murad
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed I Elsalahaty
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University - Egypt, New Damietta, 34517, Egypt
| | - Fatimah A Alsaeed
- Department of Biology, College of Science, King Khalid University, Muhayl, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16Th Street, 35233, Khalifa City, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, 12622, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate, Egypt
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, 15551, United Arab Emirates
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, 9301, South Africa
- Natural & Medical Science Research Center, University of Nizwa, Nizwa, Oman
| |
Collapse
|
13
|
Virtanen J, Korhonen EM, Salonen S, Vapalahti O, Sironen T, Jääskeläinen AJ. SARS-CoV-2 infections among pregnant women, 2020, Finland-Cross-testing of neutralization assays. J Med Virol 2024; 96:e29415. [PMID: 38293724 DOI: 10.1002/jmv.29415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
We studied the development of the severe acute respiratory syndrome-related coronavirus (SARS-CoV-2) pandemic in southern Finland in 2020 and evaluated the performance of two surrogate immunoassays for the detection of neutralizing antibodies (NAbs). The data set consisted of 12 000 retrospectively collected samples from pregnant women in their first trimester throughout 2020. All the samples were initially screened for immunoglobulin G (IgG) with SARS-CoV-2 spike antibody assay (EIM-S1, Euroimmun) followed by confirmation with nucleocapsid antibody assay (Architect SARS-CoV-2, Abbott). Samples that were reactive (positive or borderline) with both assays were subjected to testing with commercial surrogate immunoassays of NeutraLISA (EIM) and cPassTM (GenScript Biotech Corporation) by using pseudoneutralization assay (PNAbA) as a golden standard. No seropositive cases were detected between January and March. Between April and December, IgG (EIM-S1 and Abbott positive) and NAb (PNAbA positive) seroprevalences were between 0.4% and 1.4%. NeutraLISA showed 90% and cPass 55% concordant results with PNAbA among PNAbA negative samples and 49% and 92% among PNAbA positive samples giving NeutraLISA better specificity but lower sensitivity than cPass. To conclude, seroprevalence in pregnant women reflected that of the general population but the variability of the performance of serological protocols needs to be taken into account in inter-study comparison.
Collapse
Affiliation(s)
- Jenni Virtanen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Essi M Korhonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Sami Salonen
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Olli Vapalahti
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tarja Sironen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Anne J Jääskeläinen
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
14
|
Fei L, Bozza B, Melani G, Righi L, Santarelli G, Boy OB, Benedetti D, Falone A, Flaccomio D, Giuranno G, Martelli M, Merola P, Moretti S, Ndoci E, Pecoraro V, Siviglia S, Berni A, Fanelli A, Giovagnini E, Morettini A, Nozzoli C, Para O, Rostagno C, Tozzetti C. SSRIs in the course of COVID-19 pneumonia: Evidence of effectiveness of antidepressants on acute inflammation. A retrospective study. Hum Psychopharmacol 2024; 39:e2887. [PMID: 38059650 DOI: 10.1002/hup.2887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION Relationships between inflammation and mood have been observed in terms of pro-inflammatory effects induced by depressive conditions and, in parallel, by an antidepressant-induced favorable effect on the recovery of inflammatory states. Selective serotonin reuptake inhibitor (SSRI) drugs were hypothesized to improve the prognosis of COVID-19 pneumonia, a typical acute inflammation, in terms of decreased mortality rate and pro-inflammatory cytokine serum levels. METHODS The medical records of COVID-19 pneumonia inpatients at Careggi University Hospital (Florence) were analyzed for prognosis and Interleukin 6 (IL-6) after admission for over a period of 22 months. Medical records of patients treated at admission and not discontinued until discharge with an SSRI or with vortioxetine were identified. Two groups, one treated with antidepressants, the other not treated, were evaluated according to the mentioned parameters. Multiple linear regression and logistic regression were performed. RESULTS The entire sample composed of 1236 records (recovered patients 77.1%, deceased patients 22.9%). The treated group (n = 107) had a better prognosis than the untreated group in spite of age and comorbidity both being greater than in the untreated group. Correspondingly, IL-6 levels in the treated group were significantly lower (p < 0.01) than the levels in the untreated group, in every comparison. CONCLUSIONS Outcomes of this study support the hypothesis of the favorable influence of some antidepressants on the prognosis of COVID-19, possibly mediated by IL-6 modulation. Reduction in acute inflammation induced by the action of antidepressants was confirmed.
Collapse
Affiliation(s)
- Leonardo Fei
- Careggi University Hospital (IT), Chair of Psycho-Oncology, Florence, Italy
| | - Bernardo Bozza
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | - Giulia Melani
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | | | | | | | - Davide Benedetti
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | - Andrea Falone
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | - Dario Flaccomio
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | | | - Michela Martelli
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | - Pierpaolo Merola
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | | | - Endrit Ndoci
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | | | - Serena Siviglia
- School of Psychiatry, University of Florence (IT), Florence, Italy
| | - Andrea Berni
- Careggi University Hospital (IT), Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Kirenga BJ, Mugenyi L, Sánchez-Rico M, Kyobe H, Muttamba W, Mugume R, Mwesigwa E, Kalimo E, Nyombi V, Segawa I, Namakula LO, Sekibira R, Kabweru W, Byanyima R, Aanyu H, Byakika-Kibwika P, Mwebesa HG, Hoertel N, Bazeyo W. Association of fluvoxamine with mortality and symptom resolution among inpatients with COVID-19 in Uganda: a prospective interventional open-label cohort study. Mol Psychiatry 2023; 28:5411-5418. [PMID: 36869228 PMCID: PMC9982784 DOI: 10.1038/s41380-023-02004-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 03/05/2023]
Abstract
Prior research suggests that fluvoxamine, a selective serotonin reuptake inhibitor (SSRI) used for the treatment of obsessive-compulsive disorder and major depressive disorder, could be repurposed against COVID-19. We undertook a prospective interventional open-label cohort study to evaluate the efficacy and tolerability of fluvoxamine among inpatients with laboratory-confirmed COVID-19 in Uganda. The main outcome was all-cause mortality. Secondary outcomes were hospital discharge and complete symptom resolution. We included 316 patients, of whom 94 received fluvoxamine in addition to standard care [median age, 60 years (IQR = 37.0); women, 52.2%]. Fluvoxamine use was significantly associated with reduced mortality [AHR = 0.32; 95% CI = 0.19-0.53; p < 0.001, NNT = 4.46] and with increased complete symptom resolution [AOR = 2.56; 95% CI = 1.53-5.51; p < 0.001, NNT = 4.44]. Sensitivity analyses yielded similar results. These effects did not significantly differ by clinical characteristic, including vaccination status. Among the 161 survivors, fluvoxamine was not significantly associated with time to hospital discharge [AHR 0.81, 95% CI (0.54-1.23), p = 0.32]. There was a trend toward greater side effects with fluvoxamine (7.45% versus 3.15%; SMD = 0.21; χ2 = 3.46, p = 0.06), most of which were light or mild in severity and none of which were serious. One hundred mg of fluvoxamine prescribed twice daily for 10 days was well tolerated and significantly associated with reduced mortality and with increased complete symptom resolution, without a significant increase in time to hospital discharge, among inpatients with COVID-19. Large-scale randomized trials are urgently needed to confirm these findings, especially for low- and middle-income countries, where access to vaccines and approved treatments against COVID-19 is limited.
Collapse
Affiliation(s)
- Bruce J Kirenga
- Department of Internal Medicine, Makerere University, Kampala, Uganda.
- Makerere University Lung Institute, Kampala, Uganda.
| | - Levicatus Mugenyi
- Makerere University Lung Institute, Kampala, Uganda
- Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Entebbe Unit, Entebbe, Uganda
| | - Marina Sánchez-Rico
- Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, F-92130, Issy-les-Moulineaux, France
| | | | - Winters Muttamba
- Makerere University Lung Institute, Kampala, Uganda
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, UK
| | | | - Eliya Mwesigwa
- Makerere University Lung Institute, Kampala, Uganda
- Mulago National Referral Hospital, Kampala, Uganda
| | - Ezra Kalimo
- Mulago National Referral Hospital, Kampala, Uganda
| | - Vicky Nyombi
- Mulago National Referral Hospital, Kampala, Uganda
| | - Ivan Segawa
- Makerere University Lung Institute, Kampala, Uganda
| | - Loryndah Olive Namakula
- Makerere University Lung Institute, Kampala, Uganda
- Mulago National Referral Hospital, Kampala, Uganda
| | | | | | | | - Hellen Aanyu
- Mulago National Referral Hospital, Kampala, Uganda
| | | | | | - Nicolas Hoertel
- Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, F-92130, Issy-les-Moulineaux, France
- Université Paris Cité, Paris, France
- INSERM U1266, Institut de Psychiatrie et Neuroscience de Paris, Paris, France
| | | |
Collapse
|
16
|
Pauletto P, Bortoli M, Bright FO, Delgado CP, Nogara PA, Orian L, da Rocha JBT. In silico analysis of the antidepressant fluoxetine and similar drugs as inhibitors of the human protein acid sphingomyelinase: a related SARS-CoV-2 inhibition pathway. J Biomol Struct Dyn 2023; 41:9562-9575. [PMID: 36447407 DOI: 10.1080/07391102.2022.2148124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/29/2022] [Indexed: 12/05/2022]
Abstract
Acid Sphingomyelinase (ASM) is a human phosphodiesterase that catalyzes the metabolism of sphingomyelin (SM) to ceramide and phosphocholine. ASM is involved in the plasma membrane cell repair and is associated with the lysosomal inner lipid membrane by nonbonding interactions. The disruption of those interaction would result in ASM release into the lysosomal lumen and consequent degradation of its structure. Furthermore, SARS-CoV-2 infection has been linked with ASM activation and with a ceramide domain formation in the outer leaflet of the plasma membrane that is thought to be crucial for the viral particles recognition by the host cells. In this study, we have explored in silico the behavior of fluoxetine and related drugs as potential inhibitors of ASM. Theoretically, these drugs would be able to overpass lysosomal membrane and reach the interactions that sustain ASM structure, breaking them and inhibiting the ASM. The analyses of docking data indicated that fluoxetine allocated mainly in the N-terminal saposin domain via nonbonding interactions, mostly of hydrophobic nature. Similar results were obtained for venlafaxine, citalopram, atomoxetine, nisoxetine and fluoxetine's main metabolite norfluoxetine. In conclusion, it was observed that the saposin allocation may be a good indicative of the drugs inhibition mechanism, once this domain is responsible for the binding of ASM to lysosomal membrane and some of those drugs have previously been reported to inhibit the phosphodiesterase by releasing its structure in the lysosomal lumen. Our MD data also provides some insight about natural ligand C18 sphingomyelin conformations on saposin.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pedro Pauletto
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Marco Bortoli
- Institut de Química Computacional i Catàlisi (IQCC) i Departament de Química, Facultat de Ciències, Universitat de Girona, Girona, Spain
| | - Folorunsho Omage Bright
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Cássia Pereira Delgado
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Pablo Andrei Nogara
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Laura Orian
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Padova, Italy
| | | |
Collapse
|
17
|
Sánchez-Rico M, Edán-Sánchez A, Olfson M, Alvarado JM, Airagnes G, Rezaei K, Delcuze A, Peyre H, Limosin F, Hoertel N. Antipsychotic use and 28-day mortality in patients hospitalized with COVID-19: A multicenter observational retrospective study. Eur Neuropsychopharmacol 2023; 75:93-104. [PMID: 37713738 PMCID: PMC10272945 DOI: 10.1016/j.euroneuro.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 09/17/2023]
Abstract
Prior research has yielded conflicting results about the potential influence of antipsychotics in patients with COVID-19. In this multicenter retrospective study, we examined the association of antipsychotic use at admission with 28-day all-cause mortality in a sample of 59,021 adult patients hospitalized with COVID-19 from January 2020 to November 2021. In a 1:1 ratio matched analytic sample (N=1,454) accounting for age, sex, hospital, hospitalization period, the Elixhauser Comorbidity Index, other psychotropic medications, medications prescribed according to compassionate use or as part of a clinical trial, current diagnoses of psychiatric disorders, and clinical and biological markers of COVID-19 severity, antipsychotic use was not associated with 28-day mortality [23.5% (N=727) versus 18.6% (N=727); OR=1.16; 95%CI=0.89-1.51; p=0.280]. This association remained non-significant in exploratory analyses across all classes of antipsychotics and individual molecules, except for typical antipsychotics and loxapine, which were significantly linked to increased 28-day mortality, associations likely due to residual indication bias. Contrariwise, antipsychotics prescribed at daily doses higher than 200 mg of chlorpromazine-equivalents might be associated with reduced 28-day mortality when compared to patients not taking antipsychotics in the matched analytic sample [10.4% (N=154) versus 18.6% (N=727); AOR=0.56; 95%CI=0.31-0.96; p=0.040]. These results suggest that antipsychotic use, when prescribed at usual doses, are not be associated with 28-day mortality in patients hospitalized with COVID-19.
Collapse
Affiliation(s)
- Marina Sánchez-Rico
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain.
| | - Alejandro Edán-Sánchez
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Mark Olfson
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Jesús M Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Guillaume Airagnes
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Université Paris Cité, Paris, France
| | - Katayoun Rezaei
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France
| | - Aude Delcuze
- CLINEA, Clinique Les Orchidées, Service de Psychiatrie, Andilly, France
| | | | - Frédéric Limosin
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Université Paris Cité, Paris, France
| | - Nicolas Hoertel
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; INSERM UMR_1266, Institut de Psychiatrie et Neuroscience de Paris, F-75014 Paris, France
| |
Collapse
|
18
|
Diesendorf V, Roll V, Geiger N, Fähr S, Obernolte H, Sewald K, Bodem J. Drug-induced phospholipidosis is not correlated with the inhibition of SARS-CoV-2 - inhibition of SARS-CoV-2 is cell line-specific. Front Cell Infect Microbiol 2023; 13:1100028. [PMID: 37637460 PMCID: PMC10450944 DOI: 10.3389/fcimb.2023.1100028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Recently, Tummino et al. reported that 34 compounds, including Chloroquine and Fluoxetine, inhibit SARS-CoV-2 replication by inducing phospholipidosis, although Chloroquine failed to suppress viral replication in Calu-3 cells and patients. In contrast, Fluoxetine represses viral replication in human precision-cut lung slices (PCLS) and Calu-3 cells. Thus, it is unlikely that these compounds have similar mechanisms of action. Here, we analysed a subset of these compounds in the viral replication and phospholipidosis assays using the Calu-3 cells and PCLS as the patient-near system. Trimipramine and Chloroquine induced phospholipidosis but failed to inhibit SARS-CoV-2 replication in Calu-3 cells, which contradicts the reported findings and the proposed mechanism. Fluoxetine, only slightly induced phospholipidosis in Calu-3 cells but reduced viral replication by 2.7 orders of magnitude. Tilorone suppressed viral replication by 1.9 orders of magnitude in Calu-3 cells without causing phospholipidosis. Thus, induction of phospholipidosis is not correlated with the inhibition of SARS-CoV-2, and the compounds act via other mechanisms. However, we show that compounds, such as Amiodarone, Tamoxifen and Tilorone, with antiviral activity on Calu-3 cells, also inhibited viral replication in human PCLS. Our results indicate that antiviral assays against SARS-CoV-2 are cell-line specific. Data from Vero E6 can lead to non-transferable results, underlining the importance of an appropriate cell system for analysing antiviral compounds against SARS-CoV-2. We observed a correlation between the active compounds in Calu-3 cells and PCLS.
Collapse
Affiliation(s)
- Viktoria Diesendorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Valeria Roll
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Nina Geiger
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Sofie Fähr
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Helena Obernolte
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
Hoertel N, Rezaei K, Sánchez-Rico M, Delgado-Álvarez A, Kornhuber J, Gulbins E, Olfson M, Ouazana-Vedrines C, Carpinteiro A, Cougoule C, Becker KA, Alvarado JM, Limosin F, on behalf of the AP-HP/Université Paris Cité/INSERM COVID-19 Research Collaboration, AP-HP COVID CDR Initiative and “Entrepôt de Données de Santé” AP-HP Consortium. Medications Modulating the Acid Sphingomyelinase/Ceramide System and 28-Day Mortality among Patients with SARS-CoV-2: An Observational Study. Pharmaceuticals (Basel) 2023; 16:1107. [PMID: 37631022 PMCID: PMC10458150 DOI: 10.3390/ph16081107] [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: 05/26/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 08/27/2023] Open
Abstract
Prior evidence indicates the potential central role of the acid sphingomyelinase (ASM)/ceramide system in the infection of cells with SARS-CoV-2. We conducted a multicenter retrospective observational study including 72,105 adult patients with laboratory-confirmed SARS-CoV-2 infection who were admitted to 36 AP-HP (Assistance Publique-Hôpitaux de Paris) hospitals from 2 May 2020 to 31 August 2022. We examined the association between the ongoing use of medications functionally inhibiting acid sphingomyelinase (FIASMA), which reduces the infection of cells with SARS-CoV-2 in vitro, upon hospital admission with 28-day all-cause mortality in a 1:1 ratio matched analytic sample based on clinical characteristics, disease severity and other medications (N = 9714). The univariate Cox regression model of the matched analytic sample showed that FIASMA medication use at admission was associated with significantly lower risks of 28-day mortality (HR = 0.80; 95% CI = 0.72-0.88; p < 0.001). In this multicenter observational study, the use of FIASMA medications was significantly and substantially associated with reduced 28-day mortality among adult patients hospitalized with COVID-19. These findings support the continuation of these medications during the treatment of SARS-CoV-2 infections. Randomized clinical trials (RCTs) are needed to confirm these results, starting with the molecules with the greatest effect size in the study, e.g., fluoxetine, escitalopram, and amlodipine.
Collapse
Affiliation(s)
- Nicolas Hoertel
- INSERM U1266, Université Paris Cité, F-75014 Paris, France
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
| | - Katayoun Rezaei
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
| | - Marina Sánchez-Rico
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, 28223 Madrid, Spain
| | - Alfonso Delgado-Álvarez
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, 28223 Madrid, Spain
- Department of Biological and Health Psychology, Faculty of Psychology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany (K.A.B.)
| | - Mark Olfson
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University, New York, NY 10032, USA
| | - Charles Ouazana-Vedrines
- Service de Psychiatrie de l’Adulte, DMU Psychiatrie et Addictologie, Hôpital Hôtel-Dieu, AP-HP, Université Paris Cité, F-75004 Paris, France
| | - Alexander Carpinteiro
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany (K.A.B.)
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Katrin Anne Becker
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany (K.A.B.)
| | - Jesús M. Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, 28223 Madrid, Spain
| | - Frédéric Limosin
- INSERM U1266, Université Paris Cité, F-75014 Paris, France
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
| | | |
Collapse
|
20
|
Tamouza R, Meyer U, Lucas A, Richard JR, Nkam I, Pinot A, Djonouma N, Boukouaci W, Charvet B, Pierquin J, Brunel J, Fourati S, Rodriguez C, Barau C, Le Corvoisier P, El Abdellati K, De Picker L, Perron H, Leboyer M. Patients with psychosis spectrum disorders hospitalized during the COVID-19 pandemic unravel overlooked SARS-CoV-2 past infection clustering with HERV-W ENV expression and chronic inflammation. Transl Psychiatry 2023; 13:272. [PMID: 37524719 PMCID: PMC10390536 DOI: 10.1038/s41398-023-02575-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Epidemiology has repeatedly associated certain infections with a risk of further developing psychiatric diseases. Such infections can activate retro-transposable genetic elements (HERV) known to trigger immune receptors and impair synaptic plasticity of neuroreceptors. Since the HERV-W ENV protein was recently shown to co-cluster with pro-inflammatory cytokines in a subgroup of patients with schizophrenia or bipolar disorder, we questioned the influence of the COVID-19 pandemic on patients with psychosis spectrum disorders (PSD). Present results revealed that (i) SARS-CoV-2 serology shows high prevalence and titers of antibodies in PSD, (ii) HERV-W ENV is detected in seropositive individuals only and (iii) SARS-CoV-2 and HERV-W ENV positivity co-clustered with high serum levels of pro-inflammatory cytokines in psychotic patients. These results thus suggest that SARS-CoV-2 infection in many patients with psychotic disorders now admitted in the psychiatry department did not cause severe COVID-19. They also confirm the previously reported association of elevated serum pro-inflammatory cytokines and HERV-W ENV in a subgroup of psychotic patients. In the context of the COVID-19 pandemic, this cluster is only found in SARS-CoV-2 seropositive PSD cases, suggesting a dominant influence of this virus on HERV-W ENV and cytokine expression, and/or patients' greater susceptibility to SARS-CoV-2 infection. Further investigation on an interplay between this viral infection and the clinical evolution of such PSD patients is needed. However, this repeatedly defined subgroup of psychotic patients with a pro-inflammatory phenotype and HERV expression calls for a differential therapeutic approach in psychoses, therefore for further precision medicine development.
Collapse
Affiliation(s)
- Ryad Tamouza
- AP-HP, Hôpital Henri Mondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie (DMU IMPACT), Fédération Hospitalo-Universitaire de Médecine de Précision (FHU ADAPT), Créteil, F-94010, France.
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuro-Psychiatrie translationnelle, F-94010, Créteil, France.
- Fondation FondaMental, Créteil, France.
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Alexandre Lucas
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), We-Met Platform, Inserm UMR1297 and Université Paul Sabatier, Toulouse, France
| | - Jean Romain Richard
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuro-Psychiatrie translationnelle, F-94010, Créteil, France
| | - Irène Nkam
- AP-HP, Hôpital Henri Mondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie (DMU IMPACT), Fédération Hospitalo-Universitaire de Médecine de Précision (FHU ADAPT), Créteil, F-94010, France
| | - Armand Pinot
- AP-HP, Hôpital Henri Mondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie (DMU IMPACT), Fédération Hospitalo-Universitaire de Médecine de Précision (FHU ADAPT), Créteil, F-94010, France
| | - Ndilyam Djonouma
- AP-HP, Hôpital Henri Mondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie (DMU IMPACT), Fédération Hospitalo-Universitaire de Médecine de Précision (FHU ADAPT), Créteil, F-94010, France
| | - Wahid Boukouaci
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuro-Psychiatrie translationnelle, F-94010, Créteil, France
| | - Benjamin Charvet
- GeNeuro, 18, chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
- Université de Lyon-UCBL, Lyon, France
| | - Justine Pierquin
- GeNeuro, 18, chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
- Université de Lyon-UCBL, Lyon, France
| | - Joanna Brunel
- GeNeuro, 18, chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
- Université de Lyon-UCBL, Lyon, France
| | - Slim Fourati
- Virology Unit, Department of Prevention, Diagnosis and Treatment of Infections, Hôpital Henri Mondor (AP-HP) and Institut Mondor de Recherche Biomédicale, INSERM U955, Université Paris-Est, Créteil, France
| | - Christophe Rodriguez
- Virology Unit, Department of Prevention, Diagnosis and Treatment of Infections, Hôpital Henri Mondor (AP-HP) and Institut Mondor de Recherche Biomédicale, INSERM U955, Université Paris-Est, Créteil, France
| | - Caroline Barau
- APHP, Hôpital Henri Mondor, Plateforme de Ressources Biologiques, F94010, Créteil, France
| | - Philippe Le Corvoisier
- Université Paris Est Créteil, Centre Investigation Clinique, CIC Henri Mondor, Créteil, F94010, France
| | - Kawtar El Abdellati
- CAPRI, University of Antwerp, Antwerp, Belgium
- University Psychiatric Centre, Duffel, Belgium
- ECNP Immuno-NeuroPsychiatry Network, Utrecht, The Netherlands
| | - Livia De Picker
- CAPRI, University of Antwerp, Antwerp, Belgium
- University Psychiatric Centre, Duffel, Belgium
- ECNP Immuno-NeuroPsychiatry Network, Utrecht, The Netherlands
| | - Hervé Perron
- GeNeuro, 18, chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
- Université de Lyon-UCBL, Lyon, France
| | - Marion Leboyer
- AP-HP, Hôpital Henri Mondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie (DMU IMPACT), Fédération Hospitalo-Universitaire de Médecine de Précision (FHU ADAPT), Créteil, F-94010, France
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuro-Psychiatrie translationnelle, F-94010, Créteil, France
- Fondation FondaMental, Créteil, France
| |
Collapse
|
21
|
Glebov OO, Mueller C, Stewart R, Aarsland D, Perera G. Antidepressant drug prescription and incidence of COVID-19 in mental health outpatients: a retrospective cohort study. BMC Med 2023; 21:209. [PMID: 37340474 PMCID: PMC10283271 DOI: 10.1186/s12916-023-02877-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/20/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Currently, the main pharmaceutical intervention for COVID-19 is vaccination. While antidepressant (AD) drugs have shown some efficacy in treatment of symptomatic COVID-19, their preventative potential remains largely unexplored. Analysis of association between prescription of ADs and COVID-19 incidence in the population would be beneficial for assessing the utility of ADs in COVID-19 prevention. METHODS Retrospective study of association between AD prescription and COVID-19 diagnosis was performed in a cohort of community-dwelling adult mental health outpatients during the 1st wave of COVID-19 pandemic in the UK. Clinical record interactive search (CRIS) was performed for mentions of ADs within 3 months preceding admission to inpatient care of the South London and Maudsley (SLaM) NHS Foundation Trust. Incidence of positive COVID-19 tests upon admission and during inpatient treatment was the primary outcome measure. RESULTS AD mention was associated with approximately 40% lower incidence of positive COVID-19 test results when adjusted for socioeconomic parameters and physical health. This association was also observed for prescription of ADs of the selective serotonin reuptake inhibitor (SSRI) class. CONCLUSIONS This preliminary study suggests that ADs, and SSRIs in particular, may be of benefit for preventing COVID-19 infection spread in the community. The key limitations of the study are its retrospective nature and the focus on a mental health patient cohort. A more definitive assessment of AD and SSRI preventative potential warrants prospective studies in the wider demographic.
Collapse
Affiliation(s)
- Oleg O Glebov
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Shandong, China.
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Christoph Mueller
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- South London and Maudsley National Health Service Foundation Trust, London, UK
| | - Robert Stewart
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- South London and Maudsley National Health Service Foundation Trust, London, UK
| | - Dag Aarsland
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for Age-Related Research, Stavanger University Hospital, Stavanger, Norway
| | - Gayan Perera
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
22
|
Sánchez-Rico M, Rezaei K, Delgado-Álvarez A, Limosin F, Hoertel N, Alvarado JM. Comorbidity Patterns and Mortality Among Hospitalized Patients with Psychiatric Disorders and COVID-19. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2023; 45. [PMID: 37290011 PMCID: PMC10668315 DOI: 10.47626/1516-4446-2023-3076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/10/2023] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To examine the association between psychiatric and non-psychiatric comorbidity and 28-day mortality among patients with psychiatric disorders and COVID-19. METHODS We performed a multicenter observational retrospective cohort study of adult patients with psychiatric disorders hospitalized with laboratory-confirmed COVID-19 at 36 Greater Paris University hospitals (January 2020-May 2021) (N=3,768). First, we searched for different subgroups of patients according to their psychiatric and non-psychiatric comorbidities through cluster analysis. Next, we compared 28-day all-cause mortality rates across the identified clusters, while taking into account sex, age, and the number of medical conditions. RESULTS We found 5 clusters of patients with distinct psychiatric and non-psychiatric comorbidity patterns. Twenty-eight-day mortality in the cluster of patients with mood disorders was significantly lower than in other clusters. There were no significant differences in mortality across other clusters. CONCLUSIONS All psychiatric and non-psychiatric conditions may be associated with increased mortality in patients with psychiatric disorders and COVID-19. The lower risk of death among patients with mood disorders might be in line with the potential beneficial effect of certain antidepressants in COVID-19, but requires further research. These findings help identify at-risk patients with psychiatric disorders who should benefit from vaccine booster prioritization and other prevention measures.
Collapse
Affiliation(s)
- Marina Sánchez-Rico
- Département Médico-Universitaire de Psychiatrie et Addictologie, Assistance Publique-Hôpitaux de Paris, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
- Departamento de Psicobiología y Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| | - Katayoun Rezaei
- Département Médico-Universitaire de Psychiatrie et Addictologie, Assistance Publique-Hôpitaux de Paris, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
| | - Alfonso Delgado-Álvarez
- Departamento de Psicobiología y Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| | - Frédéric Limosin
- Département Médico-Universitaire de Psychiatrie et Addictologie, Assistance Publique-Hôpitaux de Paris, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
- Institut National de la Santé et de la Recherche Médicale 1266, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
- Faculté de Santé, Unité de Formation et de Recherche de Médecine, Université Paris Cité, Paris, France
| | - Nicolas Hoertel
- Département Médico-Universitaire de Psychiatrie et Addictologie, Assistance Publique-Hôpitaux de Paris, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
- Institut National de la Santé et de la Recherche Médicale 1266, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
- Faculté de Santé, Unité de Formation et de Recherche de Médecine, Université Paris Cité, Paris, France
| | - Jesús M. Alvarado
- Departamento de Psicobiología y Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| |
Collapse
|
23
|
Gupta Y, Savytskyi OV, Coban M, Venugopal A, Pleqi V, Weber CA, Chitale R, Durvasula R, Hopkins C, Kempaiah P, Caulfield TR. Protein structure-based in-silico approaches to drug discovery: Guide to COVID-19 therapeutics. Mol Aspects Med 2023; 91:101151. [PMID: 36371228 PMCID: PMC9613808 DOI: 10.1016/j.mam.2022.101151] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
With more than 5 million fatalities and close to 300 million reported cases, COVID-19 is the first documented pandemic due to a coronavirus that continues to be a major health challenge. Despite being rapid, uncontrollable, and highly infectious in its spread, it also created incentives for technology development and redefined public health needs and research agendas to fast-track innovations to be translated. Breakthroughs in computational biology peaked during the pandemic with renewed attention to making all cutting-edge technology deliver agents to combat the disease. The demand to develop effective treatments yielded surprising collaborations from previously segregated fields of science and technology. The long-standing pharmaceutical industry's aversion to repurposing existing drugs due to a lack of exponential financial gain was overrun by the health crisis and pressures created by front-line researchers and providers. Effective vaccine development even at an unprecedented pace took more than a year to develop and commence trials. Now the emergence of variants and waning protections during the booster shots is resulting in breakthrough infections that continue to strain health care systems. As of now, every protein of SARS-CoV-2 has been structurally characterized and related host pathways have been extensively mapped out. The research community has addressed the druggability of a multitude of possible targets. This has been made possible due to existing technology for virtual computer-assisted drug development as well as new tools and technologies such as artificial intelligence to deliver new leads. Here in this article, we are discussing advances in the drug discovery field related to target-based drug discovery and exploring the implications of known target-specific agents on COVID-19 therapeutic management. The current scenario calls for more personalized medicine efforts and stratifying patient populations early on for their need for different combinations of prognosis-specific therapeutics. We intend to highlight target hotspots and their potential agents, with the ultimate goal of using rational design of new therapeutics to not only end this pandemic but also uncover a generalizable platform for use in future pandemics.
Collapse
Affiliation(s)
- Yash Gupta
- Department of Medicine, Infectious Diseases, Mayo Clinic, Jacksonville, FL, USA
| | - Oleksandr V Savytskyi
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; In Vivo Biosystems, Eugene, OR, USA
| | - Matt Coban
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Vasili Pleqi
- Department of Medicine, Infectious Diseases, Mayo Clinic, Jacksonville, FL, USA
| | - Caleb A Weber
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Rohit Chitale
- Department of Medicine, Infectious Diseases, Mayo Clinic, Jacksonville, FL, USA; The Council on Strategic Risks, 1025 Connecticut Ave NW, Washington, DC, USA
| | - Ravi Durvasula
- Department of Medicine, Infectious Diseases, Mayo Clinic, Jacksonville, FL, USA
| | | | - Prakasha Kempaiah
- Department of Medicine, Infectious Diseases, Mayo Clinic, Jacksonville, FL, USA
| | - Thomas R Caulfield
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of QHS Computational Biology, Mayo Clinic, Jacksonville, FL, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
24
|
Repurposing selective serotonin reuptake inhibitors for severity of COVID-19: a population-based study. Eur Neuropsychopharmacol 2023; 71:96-108. [PMID: 37094487 PMCID: PMC10070770 DOI: 10.1016/j.euroneuro.2023.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
The World Health Organization has proposed that a search be made for alternatives to vaccines for the prevention and treatment of COVID-19, with one such alternative being selective serotonin reuptake inhibitors (SSRIs). This study thus sought to assess: the impact of previous treatment with SSRI antidepressants on the severity of COVID-19 (risk of hospitalisation, admission to an intensive care unit [ICU], and mortality), its influence on susceptibility to SARS-CoV-2 and progression to severe COVID-19. We conducted a population-based multiple case-control study in a region in the north-west of Spain. Data were sourced from electronic health records. Adjusted odds ratios (aORs) and 95%CIs were calculated using multilevel logistic regression. We collected data from a total of 86 602 subjects: 3060 cases PCR+, 26 757 non-hospitalised cases PCR+ and 56 785 controls (without PCR+). Citalopram displayed a statistically significant decrease in the risk of hospitalisation (aOR=0.70; 95% CI 0.49-0.99, p=0.049) and progression to severe COVID-19 (aOR=0.64; 95% CI 0.43-0.96, p=0.032). Paroxetine was associated with a statistically significant decrease in risk of mortality (aOR=0.34; 95% CI 0.12 – 0.94, p=0.039). No class effect was observed for SSRIs overall, nor was any other effect found for the remaining SSRIs. The results of this large-scale, real-world data study indicate that, citalopram, could be a candidate drug for being repurposed as preventive treatment aimed at reducing COVID-19 patients’ risk of progressing to severe stages of the disease.
Collapse
|
25
|
Knip M, Parviainen A, Turtinen M, But A, Härkönen T, Hepojoki J, Sironen T, Iheozor-Ejiofor R, Uğurlu H, Saksela K, Lempainen J, Ilonen J, Vapalahti O. SARS-CoV-2 and type 1 diabetes in children in Finland: an observational study. Lancet Diabetes Endocrinol 2023; 11:251-260. [PMID: 36958868 DOI: 10.1016/s2213-8587(23)00041-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Some epidemiological studies have suggested an increase in incidence of type 1 diabetes during the COVID-19 pandemic, however the mechanism(s) behind such an increase have yet to be identified. In this study we aimed to evaluate the possible role of the SARS-CoV-2 virus in the reported increase in the rate of type 1 diabetes. METHODS In this observational cohort study using data from the Finnish Pediatric Diabetes Register (FPDR), we assessed the incidence of type 1 diabetes (number of children with newly diagnosed type 1 diabetes per 100 000 person-years during the pandemic and the reference period) during the first 18 months of the COVID-19 pandemic in children in Finland younger than 15 years old compared with a reference period which included three corresponding pre-pandemic periods also obtained from the FPDR. Children with confirmed monogenic diabetes were excluded. We also compared the phenotype and HLA genotype of the disease between these two cohorts, and analysed the proportion of newly diagnosed people with type 1 diabetes testing positive for SARS-CoV-2 antibodies. FINDINGS 785 children and adolescents in Finland were diagnosed with type 1 diabetes from March 1, 2020, to Aug 31, 2021. In the reference period, which comprised three similar 18-month terms (from March 1, 2014, to Aug 31, 2015; March 1, 2016, to Aug 31, 2017; and March 1, 2018, to Aug 31, 2019) 2096 children and adolescents were diagnosed. The incidence of type 1 diabetes was 61·0 per 100 000 person-years (95% CI 56·8-65·4) among children younger than 15 years old during the pandemic, which was significantly higher than during the reference period (52·3 per 100 000 person-years; 50·1-54·6). The incidence rate ratio adjusted for age and sex for the COVID-19 pandemic was 1·16 (1·06-1·25; p=0·0006) when compared with the reference period. The children diagnosed during the COVID-19 pandemic had more often diabetic ketoacidosis (p<0·001), had a higher HbA1c (p<0·001), and tested more frequently positive for glutamic acid debarboxylase antibodies at diagnosis (p<0·001) than those diagnosed before the pandemic. There were no significant differences in the distribution of HLA genotypes between the two periods. Only five of those diagnosed during the pandemic (0·9%) of 583 tested positive for infection-induced SARS-CoV-2 antibodies. INTERPRETATION Children and adolescents diagnosed with type 1 diabetes during the pandemic had a more severe disease at diagnosis. The observed increase in type 1 diabetes incidence during the first 18 months could be a consequence of lockdown and physical distancing rather than a direct effect of SARS-CoV-2 infection. FUNDING Helsinki University Hospital Research Funds, EU Horizon 2020 (Versatile emerging infectious disease observatory project), Academy of Finland, Sigrid Jusélius Foundation, Jane & Aatos Erkko Foundation, and Medicinska understödsföreningen Liv och Hälsa. TRANSLATIONS For the Finnish and Swedish translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Mikael Knip
- Pediatric Research Center, New Children's Hospital, Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Anna Parviainen
- Pediatric Research Center, New Children's Hospital, Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maaret Turtinen
- Pediatric Research Center, New Children's Hospital, Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anna But
- Biostatistics consulting, Department of Public Health, University Hospital, University of Helsinki, Helsinki, Finland
| | - Taina Härkönen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Tarja Sironen
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | | | - Hasan Uğurlu
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Kalle Saksela
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Virology and Immunology, Diagnostic Center, Helsinki University Hospital (HUSLAB), Helsinki, Finland
| | - Johanna Lempainen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Olli Vapalahti
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Virology and Immunology, Diagnostic Center, Helsinki University Hospital (HUSLAB), Helsinki, Finland; Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
26
|
Pauletto PJT, Delgado CP, da Rocha JBT. Acid sphingomyelinase (ASM) and COVID-19: A review of the potential use of ASM inhibitors against SARS-CoV-2. Cell Biochem Funct 2023; 41:284-295. [PMID: 36929117 DOI: 10.1002/cbf.3789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/18/2023]
Abstract
In the last 2 years, different pharmacological agents have been indicated as potential inhibitors of SARS-CoV-2 in vitro. Specifically, drugs termed as functional inhibitors of acid sphingomyelinase (FIASMAs) have proved to inhibit the SARS-CoV-2 replication using different types of cells. Those therapeutic agents share several chemical structure characteristics and some well-known representatives are fluoxetine, escitalopram, fluvoxamine, and others. Most of the FIASMAs are primarily used as effective therapeutic agents to treat different pathologies, therefore, they are natural drug candidates for repositioning strategy. In this review, we summarize the two main proposed mechanisms mediating acid sphingomyelinase (ASM) inhibition and how they can explain the inhibition of SARS-CoV-2 replication by FIASMAs. The first mechanism implies a disruption in the lysosomal pH fall as the endosome-lysosome moves toward the interior of the cell. In fact, changes in cholesterol levels in endosome-lysosome membranes, which are associated with ASM inhibition is thought to be mediated by lysosomal proton pump (ATP-ase) inactivation. The second mechanism involves the formation of an extracellular ceramide-rich domain, which is blocked by FIASMAs. The ceramide-rich domains are believed to facilitate the SARS-CoV-2 entrance into the host cells.
Collapse
Affiliation(s)
- Pedro José Tronco Pauletto
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - Cassia Pereira Delgado
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - João Batista Teixeira da Rocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
27
|
Roy SS, Sharma S, Rizvi ZA, Sinha D, Gupta D, Rophina M, Sehgal P, Sadhu S, Tripathy MR, Samal S, Maiti S, Scaria V, Sivasubbu S, Awasthi A, Harshan KH, Jain S, Chowdhury S. G4-binding drugs, chlorpromazine and prochlorperazine, repurposed against COVID-19 infection in hamsters. Front Mol Biosci 2023; 10:1133123. [PMID: 37006620 PMCID: PMC10061221 DOI: 10.3389/fmolb.2023.1133123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has caused millions of infections and deaths worldwide. Limited treatment options and the threat from emerging variants underline the need for novel and widely accessible therapeutics. G-quadruplexes (G4s) are nucleic acid secondary structures known to affect many cellular processes including viral replication and transcription. We identified heretofore not reported G4s with remarkably low mutation frequency across >5 million SARS-CoV-2 genomes. The G4 structure was targeted using FDA-approved drugs that can bind G4s - Chlorpromazine (CPZ) and Prochlorperazine (PCZ). We found significant inhibition in lung pathology and lung viral load of SARS-CoV-2 challenged hamsters when treated with CPZ or PCZ that was comparable to the widely used antiviral drug Remdesivir. In support, in vitro G4 binding, inhibition of reverse transcription from RNA isolated from COVID-infected humans, and attenuated viral replication and infectivity in Vero cell cultures were clear in case of both CPZ and PCZ. Apart from the wide accessibility of CPZ/PCZ, targeting relatively invariant nucleic acid structures poses an attractive strategy against viruses like SARS-CoV-2, which spread fast and accumulate mutations quickly.
Collapse
Affiliation(s)
- Shuvra Shekhar Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shalu Sharma
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Zaigham Abbas Rizvi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Dipanjali Sinha
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Divya Gupta
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Mercy Rophina
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Paras Sehgal
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srikanth Sadhu
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Manas Ranjan Tripathy
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Sweety Samal
- Translational Health Science and Technology Institute, Faridabad, 411008, India
| | - Souvik Maiti
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- CSIR-National Chemical Laboratory, Pune, 121001, India
| | - Vinod Scaria
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amit Awasthi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Krishnan H. Harshan
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Sanjeev Jain
- Molecular Genetics Laboratory, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Shantanu Chowdhury
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- *Correspondence: Shantanu Chowdhury,
| |
Collapse
|
28
|
Hoertel N, Sánchez-Rico M, de la Muela P, Abellán M, Blanco C, Leboyer M, Cougoule C, Gulbins E, Kornhuber J, Carpinteiro A, Becker KA, Vernet R, Beeker N, Neuraz A, Alvarado JM, Herrera-Morueco JJ, Airagnes G, Lemogne C, Limosin F. Risk of Death in Individuals Hospitalized for COVID-19 With and Without Psychiatric Disorders: An Observational Multicenter Study in France. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:56-67. [PMID: 35013734 PMCID: PMC8730644 DOI: 10.1016/j.bpsgos.2021.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/03/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023] Open
Abstract
Background Prior research suggests that psychiatric disorders could be linked to increased mortality among patients with COVID-19. However, whether all or specific psychiatric disorders are intrinsic risk factors of death in COVID-19 or whether these associations reflect the greater prevalence of medical risk factors in people with psychiatric disorders has yet to be evaluated. Methods We performed an observational, multicenter, retrospective cohort study to examine the association between psychiatric disorders and mortality among patients hospitalized for laboratory-confirmed COVID-19 at 36 Greater Paris University hospitals. Results Of 15,168 adult patients, 857 (5.7%) had an ICD-10 diagnosis of psychiatric disorder. Over a mean follow-up period of 14.6 days (SD = 17.9), 326 of 857 (38.0%) patients with a diagnosis of psychiatric disorder died compared with 1276 of 14,311 (8.9%) patients without such a diagnosis (odds ratio 6.27, 95% CI 5.40-7.28, p < .01). When adjusting for age, sex, hospital, current smoking status, and medications according to compassionate use or as part of a clinical trial, this association remained significant (adjusted odds ratio 3.27, 95% CI 2.78-3.85, p < .01). However, additional adjustments for obesity and number of medical conditions resulted in a nonsignificant association (adjusted odds ratio 1.02, 95% CI 0.84-1.23, p = .86). Exploratory analyses after the same adjustments suggested that a diagnosis of mood disorders was significantly associated with reduced mortality, which might be explained by the use of antidepressants. Conclusions These findings suggest that the increased risk of COVID-19-related mortality in individuals with psychiatric disorders hospitalized for COVID-19 might be explained by the greater number of medical conditions and the higher prevalence of obesity in this population and not by the underlying psychiatric disease.
Collapse
Affiliation(s)
- Nicolas Hoertel
- Département Médico-Universitaire Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, Assistance Publique–Hôpitaux de Paris Centre, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1266, Paris, France
- Université de Paris, Paris, France
| | - Marina Sánchez-Rico
- Département Médico-Universitaire Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, Assistance Publique–Hôpitaux de Paris Centre, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
- Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcón, Spain
| | - Pedro de la Muela
- Département Médico-Universitaire Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, Assistance Publique–Hôpitaux de Paris Centre, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
- Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcón, Spain
| | - Miriam Abellán
- Département Médico-Universitaire Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, Assistance Publique–Hôpitaux de Paris Centre, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
| | - Carlos Blanco
- Division of Epidemiology, Services and Prevention Research, National Institute on Drug Abuse, Bethesda, Maryland
| | - Marion Leboyer
- INSERM U955, Neuro-Psychiatrie Translationnelle, Université Paris-Est, Paris, France
- Département Médico-Universitaire IMPACT, Département Médical Universitaire de Psychiatrie, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Toulouse, France
| | - Erich Gulbins
- Department of Molecular Biology, University Medicine Essen, Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich Alexander University of Erlangen Nuremberg, Erlangen, Germany
| | - Alexander Carpinteiro
- Department of Molecular Biology, University Medicine Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University Medicine Essen, Essen, Germany
| | - Raphaël Vernet
- Department of Medical Informatics, Biostatistics and Public Health Department, L'Assistance Publique–Hôpitaux de Paris Centre-Université de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Nathanaël Beeker
- Unité de Recherche clinique, L'Assistance Publique–Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Antoine Neuraz
- INSERM UMR_S 1138, Cordeliers Research Center, Université de Paris, Paris, France
- Department of Medical Informatics, L'Assistance Publique–Hôpitaux de Paris, Necker-Enfants Malades Hospital, Paris, France
| | - Jesús M. Alvarado
- Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcón, Spain
| | - Juan José Herrera-Morueco
- Département Médico-Universitaire Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, Assistance Publique–Hôpitaux de Paris Centre, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
- Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcón, Spain
| | - Guillaume Airagnes
- Département Médico-Universitaire Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, Assistance Publique–Hôpitaux de Paris Centre, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
| | - Cédric Lemogne
- Département Médico-Universitaire Psychiatrie et Addictologie, L'Assistance Publique–Hôpitaux de Paris, Hôpital Hôtel-Dieu, Université de Paris, Service de Psychiatrie de l’adulte, INSERM, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Frédéric Limosin
- Département Médico-Universitaire Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, Assistance Publique–Hôpitaux de Paris Centre, Hôpital Corentin-Celton, Issy-les-Moulineaux, France
| |
Collapse
|
29
|
Asadi Anar M, Foroughi E, Sohrabi E, Peiravi S, Tavakoli Y, Kameli Khouzani M, Behshood P, Shamshiri M, Faridzadeh A, Keylani K, Langari SF, Ansari A, Khalaji A, Garousi S, Mottahedi M, Honari S, Deravi N. Selective serotonin reuptake inhibitors: New hope in the fight against COVID-19. Front Pharmacol 2022; 13:1036093. [PMID: 36532776 PMCID: PMC9748354 DOI: 10.3389/fphar.2022.1036093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
The emerging COVID-19 pandemic led to a dramatic increase in global mortality and morbidity rates. As in most infections, fatal complications of coronavirus affliction are triggered by an untrammeled host inflammatory response. Cytokine storms created by high levels of interleukin and other cytokines elucidate the pathology of severe COVID-19. In this respect, repurposing drugs that are already available and might exhibit anti-inflammatory effects have received significant attention. With the in vitro and clinical investigation of several studies on the effect of antidepressants on COVID-19 prognosis, previous data suggest that selective serotonin reuptake inhibitors (SSRIs) might be the new hope for the early treatment of severely afflicted patients. SSRIs' low cost and availability make them potentially eligible for COVID-19 repurposing. This review summarizes current achievements and literature about the connection between SSRIs administration and COVID-19 prognosis.
Collapse
Affiliation(s)
- Mahsa Asadi Anar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elaheh Foroughi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elika Sohrabi
- Department of Medicine, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Samira Peiravi
- Department of Emergency Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yasaman Tavakoli
- Department of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | | | - Parisa Behshood
- Department of Microbiology, Young Researchers and Elite Club, Islamic Azad University, Shahrekord, Iran
| | - Melika Shamshiri
- School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kimia Keylani
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Faride Langari
- Department of Ophthalmology, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Ansari
- Shantou University Medical College, Shantou, Guangdong, China
| | | | - Setareh Garousi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Mottahedi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Honari
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Zhu J, Li Y, Liang J, Mubareka S, Slutsky AS, Zhang H. The Potential Protective Role of GS-441524, a Metabolite of the Prodrug Remdesivir, in Vaccine Breakthrough SARS-CoV-2 Infections. INTENSIVE CARE RESEARCH 2022; 2:49-60. [PMID: 36407474 PMCID: PMC9645326 DOI: 10.1007/s44231-022-00021-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
Cases of vaccine breakthrough, especially in variants of concern (VOCs) infections, are emerging in coronavirus disease (COVID-19). Due to mutations of structural proteins (SPs) (e.g., Spike proteins), increased transmissibility and risk of escaping from vaccine-induced immunity have been reported amongst the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Remdesivir was the first to be granted emergency use authorization but showed little impact on survival in patients with severe COVID-19. Remdesivir is a prodrug of the nucleoside analogue GS-441524 which is converted into the active nucleotide triphosphate to disrupt viral genome of the conserved non-structural proteins (NSPs) and thus block viral replication. GS-441524 exerts a number of pharmacological advantages over Remdesivir: (1) it needs fewer conversions for bioactivation to nucleotide triphosphate; (2) it requires only nucleoside kinase, while Remdesivir requires several hepato-renal enzymes, for bioactivation; (3) it is a smaller molecule and has a potency for aerosol and oral administration; (4) it is less toxic allowing higher pulmonary concentrations; (5) it is easier to be synthesized. The current article will focus on the discussion of interactions between GS-441524 and NSPs of VOCs to suggest potential application of GS-441524 in breakthrough SARS-CoV-2 infections. Supplementary Information The online version contains supplementary material available at 10.1007/s44231-022-00021-4.
Collapse
Affiliation(s)
- JiaYi Zhu
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON Canada
- Department of Physiology, University of Toronto, Toronto, ON Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Yuchong Li
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON Canada
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jady Liang
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON Canada
- Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Samira Mubareka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
- Department of Medical Microbiology and Infectious Disease, Sunnybrook Health Science Centre, Toronto, ON Canada
| | - Arthur S. Slutsky
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON Canada
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON Canada
| | - Haibo Zhang
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON Canada
- Department of Physiology, University of Toronto, Toronto, ON Canada
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON Canada
- Department of Anaesthesiology and Pain Medicine, University of Toronto, Toronto, ON Canada
| |
Collapse
|
31
|
Péricat D, Leon-Icaza SA, Sanchez Rico M, Mühle C, Zoicas I, Schumacher F, Planès R, Mazars R, Gros G, Carpinteiro A, Becker KA, Izopet J, Strub-Wourgaft N, Sjö P, Neyrolles O, Kleuser B, Limosin F, Gulbins E, Kornhuber J, Meunier E, Hoertel N, Cougoule C. Antiviral and Anti-Inflammatory Activities of Fluoxetine in a SARS-CoV-2 Infection Mouse Model. Int J Mol Sci 2022; 23:13623. [PMID: 36362409 PMCID: PMC9657171 DOI: 10.3390/ijms232113623] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/11/2022] [Accepted: 10/26/2022] [Indexed: 08/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic continues to cause significant morbidity and mortality worldwide. Since a large portion of the world's population is currently unvaccinated or incompletely vaccinated and has limited access to approved treatments against COVID-19, there is an urgent need to continue research on treatment options, especially those at low cost and which are immediately available to patients, particularly in low- and middle-income countries. Prior in vitro and observational studies have shown that fluoxetine, possibly through its inhibitory effect on the acid sphingomyelinase/ceramide system, could be a promising antiviral and anti-inflammatory treatment against COVID-19. In this report, we evaluated the potential antiviral and anti-inflammatory activities of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and against variants of concern in vitro, i.e., SARS-CoV-2 ancestral strain, Alpha B.1.1.7, Gamma P1, Delta B1.617 and Omicron BA.5. Fluoxetine, administrated after SARS-CoV-2 infection, significantly reduced lung tissue viral titres and expression of several inflammatory markers (i.e., IL-6, TNFα, CCL2 and CXCL10). It also inhibited the replication of all variants of concern in vitro. A modulation of the ceramide system in the lung tissues, as reflected by the increase in the ratio HexCer 16:0/Cer 16:0 in fluoxetine-treated mice, may contribute to explain these effects. Our findings demonstrate the antiviral and anti-inflammatory properties of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and its in vitro antiviral activity against variants of concern, establishing fluoxetine as a very promising candidate for the prevention and treatment of SARS-CoV-2 infection and disease pathogenesis.
Collapse
Affiliation(s)
- David Péricat
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Stephen Adonai Leon-Icaza
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Marina Sanchez Rico
- Faculté de Santé, Université Paris Cité, 75006 Paris, France
- Département de Psychiatrie et d’Addictologie de l’Adulte et du Sujet Agé, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, 92130 Issy-les-Moulineaux, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, 75014 Paris, France
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Iulia Zoicas
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - Rémi Planès
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Raoul Mazars
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Germain Gros
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Alexander Carpinteiro
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Katrin Anne Becker
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Jacques Izopet
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Université Toulouse, CNRS, INSERM, UPS, 31300 Toulouse, France
- Laboratoire de Virologie, CHU Toulouse, Hôpital Purpan, 31300 Toulouse, France
| | | | - Peter Sjö
- Drugs for Neglected Diseases Initiative, 1202 Geneva, Switzerland
| | - Olivier Neyrolles
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - Frédéric Limosin
- Faculté de Santé, Université Paris Cité, 75006 Paris, France
- Département de Psychiatrie et d’Addictologie de l’Adulte et du Sujet Agé, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, 92130 Issy-les-Moulineaux, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, 75014 Paris, France
| | - Erich Gulbins
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Nicolas Hoertel
- Faculté de Santé, Université Paris Cité, 75006 Paris, France
- Département de Psychiatrie et d’Addictologie de l’Adulte et du Sujet Agé, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, 92130 Issy-les-Moulineaux, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, 75014 Paris, France
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| |
Collapse
|
32
|
Use of Human Lung Tissue Models for Screening of Drugs against SARS-CoV-2 Infection. Viruses 2022; 14:v14112417. [PMID: 36366514 PMCID: PMC9693925 DOI: 10.3390/v14112417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
The repurposing of licenced drugs for use against COVID-19 is one of the most rapid ways to develop new and alternative therapeutic options to manage the ongoing pandemic. Given circa 7817 licenced compounds available from Compounds Australia that can be screened, this paper demonstrates the utility of commercially available ex vivo/3D airway and alveolar tissue models. These models are a closer representation of in vivo studies than in vitro models, but retain the benefits of rapid in vitro screening for drug efficacy. We demonstrate that several existing drugs appear to show anti-SARS-CoV-2 activity against both SARS-CoV-2 Delta and Omicron Variants of Concern in the airway model. In particular, fluvoxamine, as well as aprepitant, everolimus, and sirolimus, has virus reduction efficacy comparable to the current standard of care (remdesivir, molnupiravir, nirmatrelvir). Whilst these results are encouraging, further testing and efficacy studies are required before clinical use can be considered.
Collapse
|
33
|
Hoertel N, Sánchez-Rico M, Kornhuber J, Gulbins E, Reiersen AM, Lenze EJ, Fritz BA, Jalali F, Mills EJ, Cougoule C, Carpinteiro A, Mühle C, Becker KA, Boulware DR, Blanco C, Alvarado JM, Strub-Wourgaft N, Lemogne C, Limosin F, on behalf of AP-HP/Université Paris Cité/INSERM COVID-19 Research Collaboration, AP-HP COVID CDR Initiative and “Entrepôt de Données de Santé” AP-HP Consortium. Antidepressant Use and Its Association with 28-Day Mortality in Inpatients with SARS-CoV-2: Support for the FIASMA Model against COVID-19. J Clin Med 2022; 11:5882. [PMID: 36233753 PMCID: PMC9572995 DOI: 10.3390/jcm11195882] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 01/29/2023] Open
Abstract
To reduce Coronavirus Disease 2019 (COVID-19)-related mortality and morbidity, widely available oral COVID-19 treatments are urgently needed. Certain antidepressants, such as fluvoxamine or fluoxetine, may be beneficial against COVID-19. We included 388,945 adult inpatients who tested positive for SARS-CoV-2 at 36 AP−HP (Assistance Publique−Hôpitaux de Paris) hospitals from 2 May 2020 to 2 November 2021. We compared the prevalence of antidepressant use at admission in a 1:1 ratio matched analytic sample with and without COVID-19 (N = 82,586), and assessed its association with 28-day all-cause mortality in a 1:1 ratio matched analytic sample of COVID-19 inpatients with and without antidepressant use at admission (N = 1482). Antidepressant use was significantly less prevalent in inpatients with COVID-19 than in a matched control group of inpatients without COVID-19 (1.9% versus 4.8%; Odds Ratio (OR) = 0.38; 95%CI = 0.35−0.41, p < 0.001). Antidepressant use was significantly associated with reduced 28-day mortality among COVID-19 inpatients (12.8% versus 21.2%; OR = 0.55; 95%CI = 0.41−0.72, p < 0.001), particularly at daily doses of at least 40 mg fluoxetine equivalents. Antidepressants with high FIASMA (Functional Inhibitors of Acid Sphingomyelinase) activity seem to drive both associations. These treatments may reduce SARS-CoV-2 infections and COVID-19-related mortality in inpatients, and may be appropriate for prophylaxis and/or COVID-19 therapy for outpatients or inpatients.
Collapse
Affiliation(s)
- Nicolas Hoertel
- Institut de Psychiatrie et Neuroscience de Paris, Université Paris Cité, INSERM U1266, F-75014 Paris, France
- AP-HP, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Issy-les-Moulineaux, F-92130 Paris, France
| | - Marina Sánchez-Rico
- AP-HP, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Issy-les-Moulineaux, F-92130 Paris, France
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, 28223 Pozuelo de Alarcón (Madrid), Spain
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Erich Gulbins
- Institute for Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Angela M. Reiersen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric J. Lenze
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bradley A. Fritz
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Farid Jalali
- Department of Gastroenterology, Saddleback Medical Group, Laguna Hills, CA 92653, USA
| | - Edward J. Mills
- Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, F-31400 Toulouse, France
| | - Alexander Carpinteiro
- Institute for Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Katrin Anne Becker
- Institute for Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - David R. Boulware
- Department of Medicine, Division of Infectious Diseases and International Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carlos Blanco
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Bethesda, MD 20852, USA
| | - Jesús M. Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, 28223 Pozuelo de Alarcón (Madrid), Spain
| | - Nathalie Strub-Wourgaft
- COVID-19 Response & Pandemic Preparedness, Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Cédric Lemogne
- Institut de Psychiatrie et Neuroscience de Paris, Université Paris Cité, INSERM U1266, F-75014 Paris, France
- Service de Psychiatrie de l’adulte, AP-HP, Hôpital Hôtel-Dieu, DMU Psychiatrie et Addictologie, F-75004 Paris, France
| | - Frédéric Limosin
- Institut de Psychiatrie et Neuroscience de Paris, Université Paris Cité, INSERM U1266, F-75014 Paris, France
- AP-HP, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Issy-les-Moulineaux, F-92130 Paris, France
| | | |
Collapse
|
34
|
Foletto VS, da Rosa TF, Serafin MB, Hörner R. Selective serotonin reuptake inhibitor (SSRI) antidepressants reduce COVID-19 infection: prospects for use. Eur J Clin Pharmacol 2022; 78:1601-1611. [PMID: 35943535 PMCID: PMC9360648 DOI: 10.1007/s00228-022-03372-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/30/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE The absence of specific treatments for COVID-19 leads to an intense global effort in the search for new therapeutic interventions and better clinical outcomes for patients. This review aimed to present a selection of accepted studies that reported the activity of antidepressant drugs belonging to the selective serotonin receptor inhibitor (SSRI) class for treating the novel coronavirus. METHODS A search was performed in PubMed and SciELO databases using the following search strategies: [(coronavirus) OR (COVID) OR (SARS-CoV-2) AND (antidepressant) OR (serotonin) OR (selective serotonin receptor inhibitors)]. In the end, eleven articles were included. We also covered information obtained from ClinicalTrials.gov in our research. RESULTS Although several clinical trials are ongoing, only a few drugs have been officially approved to treat the infection. Remdesivir, an antiviral drug, despite favorable preliminary results, has restricted the use due to the risk of toxicity and methodological flaws. Antidepressant drugs were able to reduce the risk of intubation or death related to COVID-19, decrease the need for intensive medical care, and severely inhibit viral titers by up to 99%. Among the SSRIs studied so far, fluoxetine and fluvoxamine have shown to be the most promising against SARS-CoV-2. CONCLUSION If successful, these drugs can substantially reduce hospitalization and mortality rates, as well as allow for fully outpatient treatment for mild-to-moderate infections. Thus, repositioning SSRIs can provide benefits when faced with a rapidly evolving pandemic such as COVID-19.
Collapse
Affiliation(s)
| | - Taciéli Fagundes da Rosa
- Federal University of Santa Maria, Postgraduate Program in Pharmaceutical Sciences, Santa Maria, RS, Brazil
| | - Marissa Bolson Serafin
- Federal University of Santa Maria, Postgraduate Program in Pharmaceutical Sciences, Santa Maria, RS, Brazil
| | - Rosmari Hörner
- Federal University of Santa Maria, Postgraduate Program in Pharmaceutical Sciences, Santa Maria, RS, Brazil.
- Department of Clinical and Toxicological Analysis, Federal University of Santa Maria, UFSM, Building 26, Room 1201, Santa Maria, RS, 97015-900, Brazil.
| |
Collapse
|
35
|
Schloer S, Treuherz D, Faist A, Witt MD, Wunderlich K, Wiewrodt R, Wiebe K, Barth P, Wälzlein JH, Kummer S, Balkema-Buschmann A, Ludwig S, Brunotte L, Rescher U. 3D ex vivo tissue platforms to investigate the early phases of influenza A virus- and SARS-CoV-2-induced respiratory diseases. Emerg Microbes Infect 2022; 11:2160-2175. [PMID: 36000328 PMCID: PMC9518268 DOI: 10.1080/22221751.2022.2117101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pandemic outbreaks of viruses such as influenza virus or SARS-CoV-2 are associated with high morbidity and mortality and thus pose a massive threat to global health and economics. Physiologically relevant models are needed to study the viral life cycle, describe the pathophysiological consequences of viral infection, and explore possible drug targets and treatment options. While simple cell culture-based models do not reflect the tissue environment and systemic responses, animal models are linked with huge direct and indirect costs and ethical questions. Ex vivo platforms based on tissue explants have been introduced as suitable platforms to bridge the gap between cell culture and animal models. We established a murine lung tissue explant platform for two respiratory viruses, influenza A virus (IAV) and SARS-CoV-2. We observed efficient viral replication, associated with the release of inflammatory cytokines and the induction of an antiviral interferon response, comparable to ex vivo infection in human lung explants. Endolysosomal entry could be confirmed as a potential host target for pharmacological intervention, and the potential repurposing potentials of fluoxetine and interferons for host-directed therapy previously seen in vitro could be recapitulated in the ex vivo model.
Collapse
Affiliation(s)
- Sebastian Schloer
- Institute-Associated Research Group "Regulatory Mechanisms of Inflammation", Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany.,Leibniz Institute of Virology, Martinistraße 52, 20251 Hamburg, Germany
| | - Daniel Treuherz
- Institute-Associated Research Group "Regulatory Mechanisms of Inflammation", Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Aileen Faist
- Institute of Virology, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
| | - Marlous de Witt
- Institute of Virology, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
| | - Katharina Wunderlich
- Institute of Virology, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
| | - Rainer Wiewrodt
- Department of Medicine A, Hematology, Oncology and Respiratory Medicine, University Hospital Münster, Münster, Germany
| | - Karsten Wiebe
- Department of Thoracic Surgery, University Hospital Münster, Münster, Germany
| | - Peter Barth
- Gerhard-Domagk-Institute of Pathology, Westfälische Wilhelms-University, Münster, Germany
| | - Joo-Hee Wälzlein
- Center for Biological Threats and Special Pathogens, Robert Koch-Institute, Berlin, Germany
| | - Susann Kummer
- Center for Biological Threats and Special Pathogens, Robert Koch-Institute, Berlin, Germany
| | - Anne Balkema-Buschmann
- Friedrich-Loeffler-Institute, Institute of Novel and Emerging Infectious Diseases, Südufer 10, 17493 Greifswald, Germany
| | - Stephan Ludwig
- Institute of Virology, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
| | - Linda Brunotte
- Institute of Virology, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
| | - Ursula Rescher
- Institute-Associated Research Group "Regulatory Mechanisms of Inflammation", Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, and "Cells in Motion" Interfaculty Center, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| |
Collapse
|
36
|
Assmus F, Driouich JS, Abdelnabi R, Vangeel L, Touret F, Adehin A, Chotsiri P, Cochin M, Foo CS, Jochmans D, Kim S, Luciani L, Moureau G, Park S, Pétit PR, Shum D, Wattanakul T, Weynand B, Fraisse L, Ioset JR, Mowbray CE, Owen A, Hoglund RM, Tarning J, de Lamballerie X, Nougairède A, Neyts J, Sjö P, Escudié F, Scandale I, Chatelain E. Need for a Standardized Translational Drug Development Platform: Lessons Learned from the Repurposing of Drugs for COVID-19. Microorganisms 2022; 10:1639. [PMID: 36014057 PMCID: PMC9460261 DOI: 10.3390/microorganisms10081639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 12/15/2022] Open
Abstract
In the absence of drugs to treat or prevent COVID-19, drug repurposing can be a valuable strategy. Despite a substantial number of clinical trials, drug repurposing did not deliver on its promise. While success was observed with some repurposed drugs (e.g., remdesivir, dexamethasone, tocilizumab, baricitinib), others failed to show clinical efficacy. One reason is the lack of clear translational processes based on adequate preclinical profiling before clinical evaluation. Combined with limitations of existing in vitro and in vivo models, there is a need for a systematic approach to urgent antiviral drug development in the context of a global pandemic. We implemented a methodology to test repurposed and experimental drugs to generate robust preclinical evidence for further clinical development. This translational drug development platform comprises in vitro, ex vivo, and in vivo models of SARS-CoV-2, along with pharmacokinetic modeling and simulation approaches to evaluate exposure levels in plasma and target organs. Here, we provide examples of identified repurposed antiviral drugs tested within our multidisciplinary collaboration to highlight lessons learned in urgent antiviral drug development during the COVID-19 pandemic. Our data confirm the importance of assessing in vitro and in vivo potency in multiple assays to boost the translatability of pre-clinical data. The value of pharmacokinetic modeling and simulations for compound prioritization is also discussed. We advocate the need for a standardized translational drug development platform for mild-to-moderate COVID-19 to generate preclinical evidence in support of clinical trials. We propose clear prerequisites for progression of drug candidates for repurposing into clinical trials. Further research is needed to gain a deeper understanding of the scope and limitations of the presented translational drug development platform.
Collapse
Affiliation(s)
- Frauke Assmus
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Jean-Sélim Driouich
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - Rana Abdelnabi
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Laura Vangeel
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Franck Touret
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - Ayorinde Adehin
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Palang Chotsiri
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Maxime Cochin
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - Caroline S. Foo
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Dirk Jochmans
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Seungtaek Kim
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si 13488, Korea
| | - Léa Luciani
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - Grégory Moureau
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - Soonju Park
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si 13488, Korea
| | - Paul-Rémi Pétit
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - David Shum
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si 13488, Korea
| | - Thanaporn Wattanakul
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Birgit Weynand
- Departmet of Imaging and Pathology, Katholieke Universiteit Leuven, Translational Cell and Tissue Research, 3000 Leuven, Belgium
| | - Laurent Fraisse
- Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Jean-Robert Ioset
- Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Charles E. Mowbray
- Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Andrew Owen
- Centre for Excellence in Long-Acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 7ZX, UK
| | - Richard M. Hoglund
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - Antoine Nougairède
- Unité des Virus Émergents (UVE), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 190-Inserm 1207, 13005 Marseille, France
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
- Global Virus Network (GVN), Baltimore, MD 21201, USA
| | - Peter Sjö
- Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Fanny Escudié
- Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| |
Collapse
|
37
|
Chlorpromazine, a Clinically Approved Drug, Inhibits SARS-CoV-2 Nucleocapsid-Mediated Induction of IL-6 in Human Monocytes. Molecules 2022; 27:molecules27123651. [PMID: 35744777 PMCID: PMC9228867 DOI: 10.3390/molecules27123651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic, caused by the rapidly spreading SARS-CoV-2 virus, led to the unprecedented mobilization of scientists, resulting in the rapid development of vaccines and potential pharmaceuticals. Although COVID-19 symptoms are moderately severe in most people, in some cases the disease can result in pneumonia and acute respiratory failure as well as can be fatal. The severe course of COVID-19 is associated with a hyperinflammatory state called a cytokine storm. One of the key cytokines creating a proinflammatory environment is IL-6, which is secreted mainly by monocytes and macrophages. Therefore, this cytokine has become a target for some therapies that inhibit its biological action; however, these therapies are expensive, and their availability is limited in poorer countries. Thus, new cheaper drugs that can overcome the severe infections of COVID-19 are needed. Here, we show that chlorpromazine inhibits the expression and secretion of IL-6 by monocytes activated by SARS-CoV-2 virus nucleocapsid protein and affects the activity of NF-κB and MEK/ERK signaling. Our results, including others, indicate that chlorpromazine, which has been used for several decades as a neuroleptic, exerts antiviral and immunomodulatory activity, is safe and inexpensive, and might be a desirable drug to support the therapy of patients with COVID-19.
Collapse
|
38
|
Sfera A, Thomas KG, Andronescu CV, Jafri N, Sfera DO, Sasannia S, Zapata-Martín del Campo CM, Maldonado JC. Bromodomains in Human-Immunodeficiency Virus-Associated Neurocognitive Disorders: A Model of Ferroptosis-Induced Neurodegeneration. Front Neurosci 2022; 16:904816. [PMID: 35645713 PMCID: PMC9134113 DOI: 10.3389/fnins.2022.904816] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) comprise a group of illnesses marked by memory and behavioral dysfunction that can occur in up to 50% of HIV patients despite adequate treatment with combination antiretroviral drugs. Iron dyshomeostasis exacerbates HIV-1 infection and plays a major role in Alzheimer's disease pathogenesis. In addition, persons living with HIV demonstrate a high prevalence of neurodegenerative disorders, indicating that HAND provides a unique opportunity to study ferroptosis in these conditions. Both HIV and combination antiretroviral drugs increase the risk of ferroptosis by augmenting ferritin autophagy at the lysosomal level. As many viruses and their proteins exit host cells through lysosomal exocytosis, ferroptosis-driving molecules, iron, cathepsin B and calcium may be released from these organelles. Neurons and glial cells are highly susceptible to ferroptosis and neurodegeneration that engenders white and gray matter damage. Moreover, iron-activated microglia can engage in the aberrant elimination of viable neurons and synapses, further contributing to ferroptosis-induced neurodegeneration. In this mini review, we take a closer look at the role of iron in the pathogenesis of HAND and neurodegenerative disorders. In addition, we describe an epigenetic compensatory system, comprised of bromodomain-containing protein 4 (BRD4) and microRNA-29, that may counteract ferroptosis by activating cystine/glutamate antiporter, while lowering ferritin autophagy and iron regulatory protein-2. We also discuss potential interventions for lysosomal fitness, including ferroptosis blockers, lysosomal acidification, and cathepsin B inhibitors to achieve desirable therapeutic effects of ferroptosis-induced neurodegeneration.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
- Department of Psychiatry, University of California, Riverside, Riverside, CA, United States
| | | | | | - Nyla Jafri
- Patton State Hospital, San Bernardino, CA, United States
| | - Dan O. Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | | | | | - Jose C. Maldonado
- Department of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| |
Collapse
|
39
|
Hoertel N, Sánchez-Rico M, Gulbins E, Kornhuber J, Carpinteiro A, Abellán M, de la Muela P, Vernet R, Beeker N, Neuraz A, Delcuze A, Alvarado JM, Cougoule C, Meneton P, Limosin F. Association between FIASMA psychotropic medications and reduced risk of intubation or death in individuals with psychiatric disorders hospitalized for severe COVID-19: an observational multicenter study. Transl Psychiatry 2022; 12:90. [PMID: 35241663 PMCID: PMC8892828 DOI: 10.1038/s41398-022-01804-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 12/17/2022] Open
Abstract
The acid sphingomyelinase (ASM)/ceramide system may provide a useful framework for better understanding SARS-CoV-2 infection and the repurposing of psychotropic medications functionally inhibiting the acid sphingomyelinase/ceramide system (named FIASMA psychotropic medications) against COVID-19. We examined the potential usefulness of FIASMA psychotropic medications in patients with psychiatric disorders hospitalized for severe COVID-19, in an observational multicenter study conducted at Greater Paris University hospitals. Of 545 adult inpatients, 164 (30.1%) received a FIASMA psychotropic medication upon hospital admission for COVID-19. We compared the composite endpoint of intubation or death between patients who received a psychotropic FIASMA medication at baseline and those who did not in time-to-event analyses adjusted for sociodemographic characteristics, psychiatric and other medical comorbidity, and other medications. FIASMA psychotropic medication use at baseline was significantly associated with reduced risk of intubation or death in both crude (HR = 0.42; 95%CI = 0.31-0.57; p < 0.01) and primary inverse probability weighting (IPW) (HR = 0.50; 95%CI = 0.37-0.67; p < 0.01) analyses. This association was not specific to one FIASMA psychotropic class or medication. Patients taking a FIASMA antidepressant at baseline had a significantly reduced risk of intubation or death compared with those taking a non-FIASMA antidepressant at baseline in both crude (HR = 0.57; 95%CI = 0.38-0.86; p < 0.01) and primary IPW (HR = 0.57; 95%CI = 0.37-0.87; p < 0.01) analyses. These associations remained significant in multiple sensitivity analyses. Our results show the potential importance of the ASM/ceramide system framework in COVID-19 and support the continuation of FIASMA psychotropic medications in these patients and the need of large- scale clinical trials evaluating FIASMA medications, and particularly FIASMA antidepressants, against COVID-19.
Collapse
Affiliation(s)
- Nicolas Hoertel
- AP-HP, Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Département de Psychiatrie, Issy-les-Moulineaux, France.
- Université de Paris, Paris, France.
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR S1266, Paris, France.
| | - Marina Sánchez-Rico
- AP-HP, Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Département de Psychiatrie, Issy-les-Moulineaux, France
- Universidad Complutense de Madrid, Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| | - Erich Gulbins
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg- Essen, Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich- Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexander Carpinteiro
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg- Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Miriam Abellán
- AP-HP, Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Département de Psychiatrie, Issy-les-Moulineaux, France
| | - Pedro de la Muela
- AP-HP, Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Département de Psychiatrie, Issy-les-Moulineaux, France
- Universidad Complutense de Madrid, Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| | - Raphaël Vernet
- AP-HP.Centre-Université de Paris, Hôpital Européen Georges Pompidou, Medical Informatics, Biostatistics and Public Health Department, Paris, France
| | - Nathanaël Beeker
- Assistance Publique-Hopitaux de Paris, Unité de Recherche clinique, Hopital Cochin, Paris, France
| | - Antoine Neuraz
- INSERM, UMR S1138, Cordeliers Research Center, Université de Paris, Paris, France
- AP-HP.Centre-Université de Paris, Department of Medical Informatics, Necker-Enfants Malades Hospital, 75015, Paris, France
| | - Aude Delcuze
- ORPEA - CLINEA, Clinique Les Orchidées, Service de Psychiatrie, Andilly, France
| | - Jesús M Alvarado
- Universidad Complutense de Madrid, Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, Toulouse, France
| | - Pierre Meneton
- INSERM U1142 LIMICS, UMR S1142, Sorbonne Universities, UPMC University of Paris 06, University of Paris 13, Paris, France
| | - Frédéric Limosin
- AP-HP, Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Département de Psychiatrie, Issy-les-Moulineaux, France
- Université de Paris, Paris, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR S1266, Paris, France
| |
Collapse
|
40
|
Eugene AR. Fluoxetine pharmacokinetics and tissue distribution quantitatively supports a therapeutic role in COVID-19 at a minimum dose of 20 mg per day. F1000Res 2021; 10:477. [PMID: 36262792 PMCID: PMC9561539 DOI: 10.12688/f1000research.53275.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/22/2023] Open
Abstract
Background. Various in vitro studies have shown fluoxetine inhibits multiple variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogen causing the coronavirus disease 2019 (COVID-19) worldwide pandemic and multiple observational clinical studies have shown that patients receiving fluoxetine experienced clinical benefit by lowering the risk of intubation and death. The aim of this study is to conduct population pharmacokinetic dosing simulations to quantify the percentage of patients achieving a trough level for the effective concentration resulting in 50% (EC50) and 90% (EC90) inhibition of SARS-CoV-2 as reported in Calu-3 human lung cells. Methods. Pharmacometric parameter estimates used in this study were obtained from the U.S. FDA website from a new drug application for fluoxetine hydrochloride. A population of 1,000 individuals were simulated at standard fluoxetine antidepressant doses (20 mg/day, 30 mg/day, 40 mg/day, 50 mg/day, and 60 mg/day) to estimate the percentage of the patients achieving a trough plasma level for the EC50 and EC90 SARS-CoV-2 inhibition. All analyses and graphing were conducted in R. Results. By day-10 at 20 mg/day 93.2% and 47% of the population will achieve the trough target plasma EC50 and EC90 concentrations, respectively, which translates to a lung tissue distribution coefficient of 60-times higher EC50 (283.6 ng/ml [0.82 mM]) and EC90 (1390.1 ng/ml [4.02 mM]). Further, by day-10 at an ideal dose of 40 mg/day, 99% and 93% of patients will reach the trough EC50 and EC90 concentrations, respectfully. Lastly, only a dose of 60 mg/day will reach the SARS-CoV-2 EC90 inhibitory concentration in the brain. Conclusion. Overall, with a minimum treatment period of 10-days and a minimum dose of 20 mg/day, this study corroborates in vitro studies reporting fluoxetine inhibiting SARS-CoV-2 titers and also multiple observational clinical studies showing therapeutic benefit of fluoxetine in COVID-19 patients.
Collapse
Affiliation(s)
- Andy R. Eugene
- Independent Neurophysiology Laboratory, Department of Psychiatry, Medical University of Lublin, Lublin Voivodship, 20-059, Poland
- Institute for the Study of Child Development, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, 08901, USA
| |
Collapse
|
41
|
Eugene AR. Fluoxetine pharmacokinetics and tissue distribution suggest a possible role in reducing SARS-CoV-2 titers. F1000Res 2021; 10:477. [PMID: 36262792 PMCID: PMC9561539 DOI: 10.12688/f1000research.53275.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 08/27/2023] Open
Abstract
Background. Recent in vitro studies have shown fluoxetine inhibits the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogen, including variants B.1.1.7 and B.1.351, SARS-CoV-2 spike mutations (E484K, K417N, N501Y), and one retrospective clinical study reported fluoxetine exposure at a median dose of 20 mg in patients with the SARS-CoV-2 coronavirus disease 2019 (COVID-19) had a significantly lower risk of intubation and death. The aim of this study is to conduct in silico population pharmacokinetic dosing simulations to quantify the percentage of patients achieving a trough level for the effective concentration resulting in 90% inhibition (EC90) of SARS-CoV-2 as reported in Calu-3 human lung cells. Methods. Population pharmacokinetic parameter estimates for a structural one-compartment model with first-order absorption were used to simulate fluoxetine pharmacokinetic data. A population of 1,000 individuals were simulated at standard fluoxetine doses (20 mg/day, 40 mg/day, and 60 mg/day) to estimate the percentage of the patients achieving a trough plasma level for the EC90 SARS-CoV-2 inhibitory concentration for a 10 day treatment period. All analyses were conducted via statistical programming in R. Results. Standard fluoxetine antidepressant doses resulted in a range of 81% to 97% of the patient population achieving a trough target plasma concentration of 23.2 ng/ml at day 10 and translates to a lung-tissue distribution coefficient of 60-times higher (EC90 of 4.02 mM). At a dose of 40 mg per day, at least 87% of patients will reach the trough target EC90 concentration within three days. Conclusion. Overall, the findings of this population pharmacokinetic dosing study corroborates in vitro and observational clinical studies reporting the first selective serotonin reuptake inhibitor fluoxetine inhibits the SARS-CoV-2 pathogen at commonly treated doses in the practice of psychiatry.
Collapse
Affiliation(s)
- Andy R. Eugene
- Independent Neurophysiology Laboratory, Department of Psychiatry, Medical University of Lublin, Lublin Voivodship, 20-059, Poland
- Institute for the Study of Child Development, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, 08901, USA
| |
Collapse
|