1
|
Marcos-Villar L, Perdiguero B, López-Bravo M, Zamora C, Sin L, Álvarez E, Sorzano CÓS, Sánchez-Cordón PJ, Casasnovas JM, Astorgano D, García-Arriaza J, Anthiya S, Borrajo ML, Lou G, Cuesta B, Franceschini L, Gelpí JL, Thielemans K, Sisteré-Oró M, Meyerhans A, García F, Esteban I, López-Bigas N, Plana M, Alonso MJ, Esteban M, Gómez CE. Heterologous mRNA/MVA delivering trimeric-RBD as effective vaccination regimen against SARS-CoV-2: COVARNA Consortium. Emerg Microbes Infect 2024; 13:2387906. [PMID: 39087555 PMCID: PMC11313003 DOI: 10.1080/22221751.2024.2387906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024]
Abstract
Despite the high efficiency of current SARS-CoV-2 mRNA vaccines in reducing COVID-19 morbidity and mortality, waning immunity and the emergence of resistant variants underscore the need for novel vaccination strategies. This study explores a heterologous mRNA/Modified Vaccinia virus Ankara (MVA) prime/boost regimen employing a trimeric form of the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein compared to a homologous MVA/MVA regimen. In C57BL/6 mice, the RBD was delivered during priming via an mRNA vector encapsulated in nanoemulsions (NE) or lipid nanoparticles (LNP), followed by a booster with a replication-deficient MVA-based recombinant virus (MVA-RBD). This heterologous mRNA/MVA regimen elicited strong anti-RBD binding and neutralizing antibodies (BAbs and NAbs) against both the ancestral SARS-CoV-2 strain and different variants of concern (VoCs). Additionally, this protocol induced robust and polyfunctional RBD-specific CD4 and CD8 T cell responses, particularly in animals primed with mLNP-RBD. In K18-hACE2 transgenic mice, the LNP-RBD/MVA combination provided complete protection from morbidity and mortality following a live SARS-CoV-2 challenge compared with the partial protection observed with mNE-RBD/MVA or MVA/MVA regimens. Although the mNE-RBD/MVA regimen only protects half of the animals, it was able to induce antibodies with Fc-mediated effector functions besides NAbs. Moreover, viral replication and viral load in the respiratory tract were markedly reduced and decreased pro-inflammatory cytokine levels were observed. These results support the efficacy of heterologous mRNA/MVA vaccine combinations over homologous MVA/MVA regimen, using alternative nanocarriers that circumvent intellectual property restrictions of current mRNA vaccine formulations.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Mice
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Mice, Inbred C57BL
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Humans
- Female
- Nanoparticles/administration & dosage
- Vaccination
- mRNA Vaccines/administration & dosage
- Mice, Transgenic
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- CD8-Positive T-Lymphocytes/immunology
- Angiotensin-Converting Enzyme 2/immunology
- Angiotensin-Converting Enzyme 2/genetics
- Liposomes
Collapse
Affiliation(s)
- Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Carmen Zamora
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | - Pedro J. Sánchez-Cordón
- Veterinary Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Madrid, Spain
| | | | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Shubaash Anthiya
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mireya L. Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Gustavo Lou
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Belén Cuesta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Lorenzo Franceschini
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Josep L. Gelpí
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona (UB), Barcelona, Spain
| | - Kris Thielemans
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marta Sisteré-Oró
- Infection Biology Laboratory, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Felipe García
- Infectious Diseases Department, Hospital Clínic, UB,Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - Ignasi Esteban
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - Núria López-Bigas
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institute for Research in Biomedicine (IRB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, Madrid, Spain
| | - Montserrat Plana
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - María J. Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigaciones Sanitarias (IDIS), Santiago de Compostela, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
2
|
Xu Y, Kong X, Huang W, Liang Z, Huang J, Li Y, Zhang N, Liu D, Guo W, Mei J. Efficacy of the COVID-19 vaccination in patients with asymptomatic or mild illness during the Omicron epidemic in Guangzhou: a multi-centre retrospective cohort study. Ann Med 2024; 56:2307504. [PMID: 38465636 PMCID: PMC10930140 DOI: 10.1080/07853890.2024.2307504] [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: 08/13/2023] [Accepted: 01/15/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Despite the widespread administration of coronavirus disease 2019 (COVID-19) vaccines, the impact on patients with asymptomatic to mild illness remains unclear. Here, we aimed to assess the efficacy of various vaccine doses and types on the duration of isolation duration and discharge rates, the viral shedding duration, and negative rates in asymptomatic to mild COVID-19 patients. METHODS We included adult patients at the Fangcang isolation centres in Pazhou or Yongning between November and December 2022. We analysed data on basic demographics, admission details, laboratory indicators and vaccination information. RESULTS A total of 6560 infected patients were included (3584 from Pazhou and 2976 from Yongning). Of these, 90.6% received inactivated vaccines, 3.66% received recombinant SARS-CoV-2 spike protein subunit vaccines and 0.91% received adenovirus vaccines. Among the 6173 vaccinated individuals, 71.9% received a booster dose. By day 9, the isolation rate reached 50% among vaccinated patients. On day 7.5, the positive rate among vaccinated individuals reached 50%. CONCLUSIONS Full vaccination was effective, with heterologous vaccines showing greater efficacy than inactivated vaccines alone. However, there was no significant difference in the vaccine protective effect 12 months after vaccination.
Collapse
Affiliation(s)
- Yuanda Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Xuetao Kong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
- Department of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Weiqing Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Zijing Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Jinkun Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Yimin Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Nuofu Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Dan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Wenwei Guo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Jiang Mei
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
3
|
Yu Y, Silva-Ayala D, Zhou Z, Peng Y, Fang RH, Gao W, Griffiths A, Zhang L. Cellular Nanoparticles Treat Coronavirus Infection in Vivo. NANO LETTERS 2024. [PMID: 39535430 DOI: 10.1021/acs.nanolett.4c04653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Cellular nanoparticles (CNPs), which refer to nanoparticles coated with natural cell membranes, are promising for neutralizing pathological agents. Here, we use CNPs as a medical countermeasure against the infection of SARS-CoV-2 variants in an animal model. CNPs comprise polymeric cores coated with the plasma membranes of human macrophages. The resulting nanoparticles (MΦ-NPs) act as host cell decoys to intercept SARS-CoV-2 and block its cellular entry, thus inhibiting subsequent viral infection. Our findings indicate that MΦ-NPs bind to the spike proteins of SARS-CoV-2 variants in a dose-dependent manner and inhibit the infectivity of live viruses. In hamsters infected with SARS-CoV-2 variants, MΦ-NPs significantly reduce the viral burden in the lungs, demonstrating their effectiveness in inhibiting viral infectivity in vivo. Furthermore, MΦ-NPs are primarily taken up by alveolar macrophages without inducing noticeable adverse effects. Given the crucial role of macrophages in viral infections, MΦ-NPs present a promising approach to combating emerging viral threats.
Collapse
Affiliation(s)
- Yiyan Yu
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Daniela Silva-Ayala
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, School of Medicine, Boston University, Boston, Massachusetts 02115, United States
| | - Zhidong Zhou
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Yifei Peng
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Ronnie H Fang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Weiwei Gao
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Anthony Griffiths
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, School of Medicine, Boston University, Boston, Massachusetts 02115, United States
- Laboratory for Infectious Disease Research and Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri 65211, United States
| | - Liangfang Zhang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
4
|
Einhauser S, Asam C, Weps M, Senninger A, Peterhoff D, Bauernfeind S, Asbach B, Carnell GW, Heeney JL, Wytopil M, Fuchs A, Messmann H, Prelog M, Liese J, Jeske SD, Protzer U, Hoelscher M, Geldmacher C, Überla K, Steininger P, Wagner R. Longitudinal effects of SARS-CoV-2 breakthrough infection on imprinting of neutralizing antibody responses. EBioMedicine 2024; 110:105438. [PMID: 39522353 DOI: 10.1016/j.ebiom.2024.105438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The impact of the infecting SARS-CoV-2 variant of concern (VOC) and the vaccination status was determined on the magnitude, breadth, and durability of the neutralizing antibody (nAb) profile in a longitudinal multicentre cohort study. METHODS 173 vaccinated and 56 non-vaccinated individuals were enrolled after SARS-CoV-2 Alpha, Delta, or Omicron infection and visited four times within 6 months and nAbs were measured for D614G, Alpha, Delta, BA.1, BA.2, BA.5, BQ.1.1, XBB.1.5 and JN.1. FINDINGS Magnitude-breadth-analysis showed enhanced neutralization capacity in vaccinated individuals against multiple VOCs. Longitudinal analysis revealed sustained neutralization magnitude-breadth after antigenically distant Delta or Omicron breakthrough infection (BTI), with triple-vaccinated individuals showing significantly elevated titres and improved breadth. Antigenic mapping and antibody landscaping revealed initial boosting of vaccine-induced WT-specific responses after BTI, a shift in neutralization towards infecting VOCs at peak responses and an immune imprinted bias towards dominating WT immunity in the long-term. Despite that bias, machine-learning models confirmed a sustained shift of the immune-profiles following BTI. INTERPRETATION In summary, our longitudinal analysis revealed delayed and short lived nAb shifts towards the infecting VOC, but an immune imprinted bias towards long-term vaccine induced immunity after BTI. FUNDING This work was funded by the Bavarian State Ministry of Science and the Arts for the CoVaKo study and the ForCovid project. The funders had no influence on the study design, data analysis or data interpretation.
Collapse
Affiliation(s)
- Sebastian Einhauser
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Claudia Asam
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Manuela Weps
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Antonia Senninger
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Stilla Bauernfeind
- Department of Infection Prevention and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
| | - Benedikt Asbach
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - George William Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan Luke Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom; DIOSynVax, Ltd., Cambridge, United Kingdom
| | - Monika Wytopil
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - André Fuchs
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Helmut Messmann
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Martina Prelog
- Pediatric Rheumatology / Special Immunology, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Liese
- Pediatric Infectious Diseases, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Samuel D Jeske
- Institute of Virology, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany; Institute of Virology, Helmholtz Munich, Munich, Germany; German Centre for Infection Research, Munich Partner Site, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
5
|
Peña S, Zhou Z, Kestilä L, Galanti MR, Shaaban AN, Caspersen IH, Magnus P, Geraldo P, Rojas-Saunero P, Parikka S, Nohynek H, Karvonen S. Tobacco Use and Uptake of COVID-19 Vaccinations in Finland: A Population-Based Study. Nicotine Tob Res 2024; 26:1553-1562. [PMID: 38196092 DOI: 10.1093/ntr/ntad234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/18/2023] [Accepted: 11/21/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION People who smoke are at higher risk of Coronavirus Disease-2019 (COVID-19) hospitalizations and deaths and might benefit greatly from high COVID-19 vaccination coverage. Studies on tobacco use and COVID-19 vaccine uptake in the general population are lacking. AIMS AND METHODS We conducted a cohort study utilizing linked data from 42 935 participants from two national surveys in Finland (FinSote 2018 and 2020). Exposures were smoking and smokeless tobacco (snus) use. The primary outcome was the uptake of two COVID-19 vaccine doses. Secondary outcomes were the uptake of one COVID-19 vaccine dose; three COVID-19 vaccine doses; time between the first and second dose; and time between the second and third dose. We examined the association between tobacco use and COVID-19 vaccine uptake and between-dose spacing in Finland. RESULTS People who smoke had a 7% lower risk of receiving two COVID-19 vaccine doses (95% confidence interval [CI] = 0.91; 0.96) and a 14% lower risk of receiving three doses (95% CI = 0.78; 0.94) compared to never smokers. People who smoked occasionally had a lower risk of receiving three vaccine doses. People who currently used snus had a 28% lower uptake of three doses (95% CI = 0.56; 0.93) compared to never users but we did not find evidence of an association for one or two doses. We did not find evidence of an association between tobacco use and spacing between COVID-19 vaccine doses. CONCLUSIONS People who smoke tobacco products daily, occasionally, and use snus had a lower uptake of COVID-19 vaccines. Our findings support a growing body of literature on lower vaccination uptake among people who use tobacco products. IMPLICATIONS People who smoke or use snus might be a crucial target group of public health efforts to increase COVID-19 vaccinations and plan future vaccination campaigns. CLINICAL TRIALS REGISTRATION NUMBER NCT05479383.
Collapse
Affiliation(s)
- Sebastián Peña
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Zhi Zhou
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Laura Kestilä
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Maria Rosaria Galanti
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Centre for Epidemiology and Community Medicine, Stockholm, Sweden
| | - Ahmed Nabil Shaaban
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | | | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Pablo Geraldo
- Department of Sociology and Department of Statistics, University of California Los Angeles, Los Angeles, CA, USA
| | - Paloma Rojas-Saunero
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Suvi Parikka
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Hanna Nohynek
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Sakari Karvonen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| |
Collapse
|
6
|
Muraoka D, Moi ML, Muto O, Nakatsukasa T, Deng S, Takashima C, Yamaguchi R, Sawada SI, Hayakawa H, Nguyen TTN, Haseda Y, Soga T, Matsushita H, Ikeda H, Akiyoshi K, Harada N. Low-frequency CD8 + T cells induced by SIGN-R1 + macrophage-targeted vaccine confer SARS-CoV-2 clearance in mice. NPJ Vaccines 2024; 9:173. [PMID: 39294173 PMCID: PMC11411095 DOI: 10.1038/s41541-024-00961-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/01/2024] [Indexed: 09/20/2024] Open
Abstract
Vaccine-induced T cells and neutralizing antibodies are essential for protection against SARS-CoV-2. Previously, we demonstrated that an antigen delivery system, pullulan nanogel (PNG), delivers vaccine antigen to lymph node medullary macrophages and thereby enhances the induction of specific CD8+ T cells. In this study, we revealed that medullary macrophage-selective delivery by PNG depends on its binding to a C-type lectin SIGN-R1. In a K18-hACE2 mouse model of SARS-CoV-2 infection, vaccination with a PNG-encapsulated receptor-binding domain of spike protein decreased the viral load and prolonged the survival in the CD8+ T cell- and B cell-dependent manners. T cell receptor repertoire analysis revealed that although the vaccine induced T cells at various frequencies, low-frequency specific T cells mainly promoted virus clearance. Thus, the induction of specific CD8+ T cells that respond quickly to viral infection, even at low frequencies, is important for vaccine efficacy and can be achieved by SIGN-R1+ medullary macrophage-targeted antigen delivery.
Collapse
Affiliation(s)
- Daisuke Muraoka
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan.
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan.
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.
| | - Osamu Muto
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takaaki Nakatsukasa
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Situo Deng
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Chieko Takashima
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shin-Ichi Sawada
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Haruka Hayakawa
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | | | | | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazunari Akiyoshi
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
7
|
Ceja-Gálvez HR, Hernández-Ramírez CO, Vega-Magaña AN, Hernández-Bello J, Arellano-Arteaga KJ, Turrubiates-Hernández FJ, Padilla-Borquez DL, Muñoz-Valle JF. Clinical characteristics and severity markers in hospitalized COVID-19 patients from western Mexico: a comparative analysis of Delta and Omicron variants. Front Public Health 2024; 12:1425372. [PMID: 39281077 PMCID: PMC11392767 DOI: 10.3389/fpubh.2024.1425372] [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: 04/29/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction COVID-19 is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a virus notable for its rapid mutation rate, which has led to the emergence of various variants such as Delta and Omicron, each with potentially different levels of transmissibility and virulence. Therefore, this study aims to compare clinical charactheristics and markers associated with the severity of COVID-19 in hospitalized patients from western Mexico who were infected with the Delta and Omicron variants of SARS-CoV-2. Methods This cross-sectional study involved 66 patients hospitalized for COVID-19, diagnosed by RT-qPCR. SARS-CoV-2 variants were identified through whole genome sequencing using the COVIDseq platform from Illumina. Upon admission, patients underwent a clinical history assessment, blood gas analysis, and blood biometry. Additionally, several tests and markers were measured, including the percentage of neutralizing antibodies, erythrocyte sedimentation rate (ESR), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNFα), D-dimer, lactate dehydrogenase (LDH), C-reactive protein (CRP), and ferritin. Results and discussion Patients hospitalized with the Omicron were found to be older, compared to those infected with the Delta (64 vs. 54 years, p = 0.006). Additionally, a higher proportion of male patients were observed in the Omicron compared to the Delta (p = 0.029). Both Omicron and Delta variants were associated with lymphopenia, although the lymphocyte count was lower in Omicron (0.9 vs. 0.56 10x3/L; p = 0.007). The COVID-GRAM scale indicated a high risk for severe disease in both groups, but the score was higher in Omicron compared to Delta (157 vs. 128 points; p = 0.0004). Patients infected with Omicron exhibited a lower percentage of neutralizing antibodies than those with Delta (35.99 vs. 81%; p < 0.05), regardless of their vaccination status. Among the markers assessed, globular ESR was found to be lower in Omicron compared to Delta (30.5 vs. 41.5 mm/h; p = 0.001), while ferritin levels were higher in patients infected with the Omicron (1,359 vs. 960.6 μg/L; p = 0.007). In patients with severe COVID-19, markers such as lymphopenia, neutralizing antibody levels, ferritin, and COVID-GRAM scores are elevated in the Omicron variant, while only the leukocyte count and ESR for the Delta variant.
Collapse
Affiliation(s)
- Hazael Ramiro Ceja-Gálvez
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Cristian Oswaldo Hernández-Ramírez
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Alejandra Natali Vega-Magaña
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jorge Hernández-Bello
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Francisco Javier Turrubiates-Hernández
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Diana Lourdes Padilla-Borquez
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
8
|
Frasson I, Diamante L, Zangrossi M, Carbognin E, Pietà AD, Penna A, Rosato A, Verin R, Torrigiani F, Salata C, Dizanzo MP, Vaccaro L, Cacchiarelli D, Richter SN, Montagner M, Martello G. Identification of druggable host dependency factors shared by multiple SARS-CoV-2 variants of concern. J Mol Cell Biol 2024; 16:mjae004. [PMID: 38305139 PMCID: PMC11411213 DOI: 10.1093/jmcb/mjae004] [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: 02/03/2023] [Revised: 06/23/2023] [Accepted: 01/31/2024] [Indexed: 02/03/2024] Open
Abstract
The high mutation rate of SARS-CoV-2 leads to the emergence of multiple variants, some of which are resistant to vaccines and drugs targeting viral elements. Targeting host dependency factors, e.g. cellular proteins required for viral replication, would help prevent the development of resistance. However, it remains unclear whether different SARS-CoV-2 variants induce conserved cellular responses and exploit the same core host factors. To this end, we compared three variants of concern and found that the host transcriptional response was conserved, differing only in kinetics and magnitude. Clustered regularly interspaced short palindromic repeats screening identified host genes required for each variant during infection. Most of the genes were shared by multiple variants. We validated our hits with small molecules and repurposed the US Food and Drug Administration-approved drugs. All the drugs were highly active against all the tested variants, including new variants that emerged during the study (Delta and Omicron). Mechanistically, we identified reactive oxygen species production as a key step in early viral replication. Antioxidants such as N-acetyl cysteine (NAC) were effective against all the variants in both human lung cells and a humanized mouse model. Our study supports the use of available antioxidant drugs, such as NAC, as a general and effective anti-COVID-19 approach.
Collapse
Affiliation(s)
- Ilaria Frasson
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Linda Diamante
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Manuela Zangrossi
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Elena Carbognin
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Alessandro Penna
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua 35128, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | | | - Lorenzo Vaccaro
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
- School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples Federico II, Naples 80138, Italy
| | - Sara N Richter
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Microbiology and Virology Unit, Padua University Hospital, Padua 35128, Italy
| | - Marco Montagner
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Graziano Martello
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| |
Collapse
|
9
|
Kim W. Germinal Center Response to mRNA Vaccination and Impact of Immunological Imprinting on Subsequent Vaccination. Immune Netw 2024; 24:e28. [PMID: 39246619 PMCID: PMC11377948 DOI: 10.4110/in.2024.24.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/29/2024] [Indexed: 09/10/2024] Open
Abstract
Vaccines are the most effective intervention currently available, offering protective immunity against targeted pathogens. The emergence of the coronavirus disease 2019 pandemic has prompted rapid development and deployment of lipid nanoparticle encapsulated, mRNA-based vaccines. While these vaccines have demonstrated remarkable immunogenicity, concerns persist regarding their ability to confer durable protective immunity to continuously evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. This review focuses on human B cell responses induced by SARS-CoV-2 mRNA vaccination, with particular emphasis on the crucial role of germinal center reactions in shaping enduring protective immunity. Additionally, we explored observations of immunological imprinting and dynamics of recalled pre-existing immunity following variants of concern-based booster vaccination. Insights from this review contribute to comprehensive understanding B cell responses to mRNA vaccination in humans, thereby refining vaccination strategies for optimal and sustained protection against evolving coronavirus variants.
Collapse
Affiliation(s)
- Wooseob Kim
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
10
|
Rajaram RB, Jayaraman T, Khoo X, Saravanaa N, Kukreja A, Johari BM, Fareeda Muhammad Gowdh N, Lee W, Sooi C, Basri S, Ng R, Ong H, Wong P, Syed Omar SF, Mahadeva S. Liver dysfunction in adults with COVID-19 infection: A longitudinal study with transient elastography evaluation. JGH Open 2024; 8:e13118. [PMID: 39114430 PMCID: PMC11304265 DOI: 10.1002/jgh3.13118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/13/2024] [Accepted: 06/21/2024] [Indexed: 08/10/2024]
Abstract
Background and Aim Abnormal liver biochemistry (ALB) is common among patients with COVID-19 infection due to various factors. It is uncertain if it persists after the acute infection. We aimed to investigate this. Methods A multicenter study of adult patients hospitalized for COVID-19 infection, with at least a single abnormal liver function test, was conducted. Detailed laboratory and imaging tests, including transabdominal ultrasound and FibroScan, were performed at assessment and at 6-month follow-up after hospital discharge. Results From an initial cohort of 1246 patients who were hospitalized, 731 (58.7%) had ALB. A total of 174/731 patients fulfilled the inclusion criteria with the following characteristics: 48.9% patients had severe COVID-19; 62.1% had chronic liver disease (CLD); and 56.9% had metabolic-associated fatty liver disease (MAFLD). ALB was predominantly of a mixed pattern (67.8%). Among those (55.2%) who had liver injury (aspartate aminotransferase/alanine aminotransferase >3 times the upper limit of normal, or alkaline phosphatase/γ-glutamyl transferase/bilirubin >2 times the upper limit of normal), a mixed pattern was similarly predominant. Approximately 52.3% had normalization of the liver lunction test in the 6-month period post discharge. Patients with persistent ALB had significantly higher mean body mass index (BMI) and serum low-density lipoprotein (LDL), higher rates of MAFLD and CLD, higher mean liver stiffness measurement and continuous attenuated parameter score on FibroScan, and higher rates of liver injury on univariate analysis. Multivariate analysis was not statistically significant. Conclusions Approximately 47.7% of COVID-19 patients were found to have persistent ALB up to 6 months following the acute infection, and it was associated with raised BMI, elevated serum LDL, increased rates of MAFLD and CLD, and higher rates of liver injury on univariate analysis, but not on multivariate analysis.
Collapse
Affiliation(s)
- Ruveena Bhavani Rajaram
- Gastroenterology Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | - Thevaraajan Jayaraman
- Gastroenterology Unit, Department of Medicine, Faculty of MedicineUniversiti Teknologi MARASungai BulohMalaysia
| | - Xin‐Hui Khoo
- Gastroenterology Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | - Nalliah Saravanaa
- Gastroenterology Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | - Anjanna Kukreja
- Infectious Disease Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | - Bushra Megat Johari
- Infectious Disease Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | | | - Wai‐Kin Lee
- Medical DepartmentHospital Seberang JayaSeberang JayaMalaysia
| | | | - Sazali Basri
- Infectious Disease Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | - Rong‐Xiang Ng
- Infectious Disease Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | - Hang‐Cheng Ong
- Infectious Disease Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | - Pui‐Li Wong
- Infectious Disease Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| | | | - Sanjiv Mahadeva
- Gastroenterology Unit, Medical DepartmentUniversiti Malaya Medical CentreKuala LumpurMalaysia
| |
Collapse
|
11
|
Torres-Fernandez D, Dalsuco J, Bramugy J, Bassat Q, Varo R. Innovative strategies for the surveillance, prevention, and management of pediatric infections applied to low-income settings. Expert Rev Anti Infect Ther 2024; 22:413-422. [PMID: 38739471 DOI: 10.1080/14787210.2024.2354839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION Infectious diseases still cause a significant burden of morbidity and mortality among children in low- and middle-income countries (LMICs). There are ample opportunities for innovation in surveillance, prevention, and management, with the ultimate goal of improving survival. AREAS COVERED This review discusses the current status in the use and development of innovative strategies for pediatric infectious diseases in LMICs by focusing on surveillance, diagnosis, prevention, and management. Topics covered are: Minimally Invasive Tissue Sampling as a technique to accurately ascertain the cause of death; Genetic Surveillance to trace the pathogen genomic diversity and emergence of resistance; Artificial Intelligence as a multidisciplinary tool; Portable noninvasive imaging methods; and Prognostic Biomarkers to triage and risk stratify pediatric patients. EXPERT OPINION To overcome the specific hurdles in child health for LMICs, some innovative strategies appear at the forefront of research. If the development of these next-generation tools remains focused on accessibility, sustainability and capacity building, reshaping epidemiological surveillance, diagnosis, and treatment in LMICs, can become a reality and result in a significant public health impact. Their integration with existing healthcare infrastructures may revolutionize disease detection and surveillance, and improve child health and survival.
Collapse
Affiliation(s)
- David Torres-Fernandez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Jessica Dalsuco
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Justina Bramugy
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- ICREA, Pg. Lluís Companys, Barcelona, Spain
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
- CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosauro Varo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| |
Collapse
|
12
|
Kumar V, Shefrin S, Sundar D. Molecular insights into the binding interactions and energetics of the omicron spike variant with hACE2 and a neutralizing antibody. J Struct Biol 2024; 216:108087. [PMID: 38494148 DOI: 10.1016/j.jsb.2024.108087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/19/2024]
Abstract
The global spread of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) since 2019 has led to a continuous evolution of viral variants, with the latest concern being the Omicron (B.1.1.529) variant. In this study, classical molecular dynamics simulations were conducted to elucidate the biophysical aspects of the Omicron spike protein's receptor-binding domain (RBD) in its interaction with human angiotensin-converting enzyme 2 (hACE2) and a neutralizing antibody, comparing it to the wildtype (WT). To model the Omicron variant, 15 in silico mutations were introduced in the RBD region of WT (retrieved from PDB). The simulations of WT spike-hACE2 and Omicron spike-hACE2 complexes revealed comparable binding stability and dynamics. Notably, the Q493R mutation in the Omicron spike increased interactions with hACE2, particularly with ASP38 and ASP355. Additionally, mutations such as N417K, T478K, and Y505H contributed to enhanced structural stability in the Omicron variant. Conversely, when comparing WT with Omicron in complex with a neutralizing antibody, simulation results demonstrated poorer binding dynamics and stability for the Omicron variant. The E484K mutation significantly decreased binding interactions, resulting in an overall decrease in binding energy (∼-57 kcal/mol) compared to WT (∼-84 kcal/mol). This study provides valuable molecular insights into the heightened infectivity of the Omicron variant, shedding light on the specific mutations influencing its interactions with hACE2 and neutralizing antibodies.
Collapse
Affiliation(s)
- Vipul Kumar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India.
| | - Seyad Shefrin
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India.
| | - Durai Sundar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India.
| |
Collapse
|
13
|
Jin G, Wang R, Jin Y, Song Y, Wang T. From intramuscular to nasal: unleashing the potential of nasal spray vaccines against coronavirus disease 2019. Clin Transl Immunology 2024; 13:e1514. [PMID: 38770238 PMCID: PMC11103645 DOI: 10.1002/cti2.1514] [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: 11/10/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Coronavirus disease 2019, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has affected 700 million people worldwide since its outbreak in 2019. The current pandemic strains, including Omicron and its large subvariant series, exhibit strong transmission and stealth. After entering the human body, the virus first infects nasal epithelial cells and invades host cells through the angiotensin-converting enzyme 2 receptor and transmembrane serine protease 2 on the host cell surface. The nasal cavity is an important body part that protects against the virus. Immunisation of the nasal mucosa produces immunoglobulin A antibodies that effectively neutralise viruses. Saline nasal irrigation, a type of physical therapy, can reduce the viral load in the nasal cavity and prevent viral infections to some extent. As a commonly used means to fight SARS-CoV-2, the intramuscular (IM) vaccine can induce the human body to produce a systemic immune response and immunoglobulin G antibody; however, the antibody is difficult to distribute to the nasal mucosa in time and cannot achieve a good preventive effect. Intranasal (IN) vaccines compensate for the shortcomings of IM vaccines, induce mucosal immune responses, and have a better effect in preventing infection. In this review, we discuss the nasal defence barrier, the harm caused by SARS-CoV-2, the mechanism of its invasion into host cells, nasal cleaning, IM vaccines and IN vaccines, and suggest increasing the development of IN vaccines, and use of IN vaccines as a supplement to IM vaccines.
Collapse
Affiliation(s)
- Ge Jin
- Faculty of MedicineDalian University of TechnologyDalianLiaoningChina
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Runze Wang
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Yi Jin
- Department of Breast SurgeryLiaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Yingqiu Song
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Tianlu Wang
- Faculty of MedicineDalian University of TechnologyDalianLiaoningChina
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
- Department of RadiotherapyCancer Hospital of Dalian University of TechnologyDalianLiaoningChina
| |
Collapse
|
14
|
Majumder V, Choudhury C, Goswami B, Sengupta S, Bhattacharjee B. Severity of respiratory illness among Covid-19-vaccinated and non-vaccinated admitted patients-An observational study from a teaching hospital of Tripura. J Family Med Prim Care 2024; 13:2111-2115. [PMID: 38948609 PMCID: PMC11213422 DOI: 10.4103/jfmpc.jfmpc_1643_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 07/02/2024] Open
Abstract
Objective To determine the association between vaccination status and mortality among critically ill patients admitted in a dedicated Covid hospital of Tripura who required invasive mechanical ventilation. Material and Methods This study was conducted at a dedicated Covid hospital of Tripura for a period of six months, i.e., from June 2021 to November 2021. A total of 304 patients were enrolled for this study. Baseline epidemiological, radiological data along with other information like heart rate, pulse rate, oxygen saturation (SpO2), etc., were collected through patient record sheet in all cases during hospitalization. Statistical analysis was done by using SPSS 25 version. Results Admission and mortality rates in hospital and advanced oxygen support like bi-level positive airway pressure (BiPAP), high-flow nasal cannula (HFNOC), and ventilator use incidences were higher in non-vaccinated patients (17.1%) in comparison to double-dose-vaccinated (0.98%) and single-dose (2.3%)-vaccinated patients. Conclusion This retrospective data analysis of Covid-19 positive patients admitted in the dedicated Covid Hospital of Tripura suggests that severe infection, need for invasive and non-invasive ventilation, and death were significantly less in the vaccinated patients as compared to the vaccine-naive one.
Collapse
Affiliation(s)
- Vaskar Majumder
- Department of Anaesthesiology, AGMC and GBP Hospital, P. O. Kunjavan, Agartala, Tripura, India
| | - Chirasree Choudhury
- Department of Anaesthesiology, AGMC and GBP Hospital, P. O. Kunjavan, Agartala, Tripura, India
| | - Bidhan Goswami
- Department of Microbiology, Agartala Government Medical College, P. O. Kunjavan, Agartala, Tripura, India
| | - Shauli Sengupta
- Multidisciplinary Research Unit, Agartala Government Medical College, P. O. Kunjavan, Agartala, Tripura, India
| | - Bhaskar Bhattacharjee
- Multidisciplinary Research Unit, Agartala Government Medical College, P. O. Kunjavan, Agartala, Tripura, India
| |
Collapse
|
15
|
Trabelsi K, Ben Khalaf N, Ramadan AR, Elsharkawy A, Ashoor D, Chlif S, Boussoffara T, Ben-Ahmed M, Kumar M, Fathallah MD. A novel approach to designing viral precision vaccines applied to SARS-CoV-2. Front Cell Infect Microbiol 2024; 14:1346349. [PMID: 38628551 PMCID: PMC11018900 DOI: 10.3389/fcimb.2024.1346349] [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: 11/29/2023] [Accepted: 03/08/2024] [Indexed: 04/19/2024] Open
Abstract
Efficient precision vaccines against several highly pathogenic zoonotic viruses are currently lacking. Proteolytic activation is instrumental for a number of these viruses to gain host-cell entry and develop infectivity. For SARS-CoV-2, this process is enhanced by the insertion of a furin cleavage site at the junction of the spike protein S1/S2 subunits upstream of the metalloprotease TMPRSS2 common proteolytic site. Here, we describe a new approach based on specific epitopes selection from the region involved in proteolytic activation and infectivity for the engineering of precision candidate vaccinating antigens. This approach was developed through its application to the design of SARS-CoV-2 cross-variant candidates vaccinating antigens. It includes an in silico structural analysis of the viral region involved in infectivity, the identification of conserved immunogenic epitopes and the selection of those eliciting specific immune responses in infected people. The following step consists of engineering vaccinating antigens that carry the selected epitopes and mimic their 3D native structure. Using this approach, we demonstrated through a Covid-19 patient-centered study of a 500 patients' cohort, that the epitopes selected from SARS-CoV-2 protein S1/S2 junction elicited a neutralizing antibody response significantly associated with mild and asymptomatic COVID-19 (p<0.001), which strongly suggests protective immunity. Engineered antigens containing the SARS-CoV-2 selected epitopes and mimicking the native epitopes 3D structure generated neutralizing antibody response in mice. Our data show the potential of this combined computational and experimental approach for designing precision vaccines against viruses whose pathogenicity is contingent upon proteolytic activation.
Collapse
Affiliation(s)
- Khaled Trabelsi
- Health Biotechnology Program, King Fahad Chair for Health Biotechnology, Department of Life Sciences College of Graduate Studies, Arabian Gulf University, Manama, Bahrain
| | - Noureddin Ben Khalaf
- Health Biotechnology Program, King Fahad Chair for Health Biotechnology, Department of Life Sciences College of Graduate Studies, Arabian Gulf University, Manama, Bahrain
| | - Ahmed R. Ramadan
- Health Biotechnology Program, King Fahad Chair for Health Biotechnology, Department of Life Sciences College of Graduate Studies, Arabian Gulf University, Manama, Bahrain
| | - Amany Elsharkawy
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| | - Dana Ashoor
- Health Biotechnology Program, King Fahad Chair for Health Biotechnology, Department of Life Sciences College of Graduate Studies, Arabian Gulf University, Manama, Bahrain
| | - Sadok Chlif
- Department of Family and Community Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Thouraya Boussoffara
- Transmission, Control and Immunobiology of Infections Laboratory, Institute Pasteur of Tunis, Tunis, Tunisia
| | - Melika Ben-Ahmed
- Transmission, Control and Immunobiology of Infections Laboratory, Institute Pasteur of Tunis, Tunis, Tunisia
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| | - M-Dahmani Fathallah
- Health Biotechnology Program, King Fahad Chair for Health Biotechnology, Department of Life Sciences College of Graduate Studies, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
16
|
Silva MDO, Castro-Amarante MF, Venceslau-Carvalho AA, Almeida BDS, Daher IP, de Souza-Silva GA, Yamamoto MM, Koike G, de Souza EE, Wrenger C, Ferreira LCDS, Boscardin SB. Enhanced Immunogenicity and Protective Effects against SARS-CoV-2 Following Immunization with a Recombinant RBD-IgG Chimeric Protein. Vaccines (Basel) 2024; 12:356. [PMID: 38675739 PMCID: PMC11054318 DOI: 10.3390/vaccines12040356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 04/28/2024] Open
Abstract
The unprecedented global impact caused by SARS-CoV-2 imposed huge health and economic challenges, highlighting the urgent need for safe and effective vaccines. The receptor-binding domain (RBD) of SARS-CoV-2 is the major target for neutralizing antibodies and for vaccine formulations. Nonetheless, the low immunogenicity of the RBD requires the use of alternative strategies to enhance its immunological properties. Here, we evaluated the use of a subunit vaccine antigen generated after the genetic fusing of the RBD with a mouse IgG antibody. Subcutaneous administration of RBD-IgG led to the extended presence of the protein in the blood of immunized animals and enhanced RBD-specific IgG titers. Furthermore, RBD-IgG immunized mice elicited increased virus neutralizing antibody titers, measured both with pseudoviruses and with live original (Wuhan) SARS-CoV-2. Immunized K18-hACE2 mice were fully resistant to the lethal challenge of the Wuhan SARS-CoV-2, demonstrated by the control of body-weight loss and virus loads in their lungs and brains. Thus, we conclude that the genetic fusion of the RBD with an IgG molecule enhanced the immunogenicity of the antigen and the generation of virus-neutralizing antibodies, supporting the use of IgG chimeric antigens as an approach to improve the performance of SARS-CoV-2 subunit vaccines.
Collapse
Affiliation(s)
- Mariângela de Oliveira Silva
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Maria Fernanda Castro-Amarante
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Alexia Adrianne Venceslau-Carvalho
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Bianca da Silva Almeida
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Isabela Pazotti Daher
- Laboratory of Immunology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo 05403-000, Brazil
| | - Guilherme Antonio de Souza-Silva
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Marcio Massao Yamamoto
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Gabriela Koike
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Edmarcia Elisa de Souza
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Luís Carlos de Souza Ferreira
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| |
Collapse
|
17
|
Muneer A, Xie L, Xie X, Zhang F, Wrobel JA, Xiong Y, Yu X, Wang C, Gheorghe C, Wu P, Song J, Ming GL, Jin J, Song H, Shi PY, Chen X. Targeting G9a translational mechanism of SARS-CoV-2 pathogenesis for multifaceted therapeutics of COVID-19 and its sequalae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583415. [PMID: 38496599 PMCID: PMC10942352 DOI: 10.1101/2024.03.04.583415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
By largely unknown mechanism(s), SARS-CoV-2 hijacks the host translation apparatus to promote COVID-19 pathogenesis. We report that the histone methyltransferase G9a noncanonically regulates viral hijacking of the translation machinery to bring about COVID-19 symptoms of hyperinflammation, lymphopenia, and blood coagulation. Chemoproteomic analysis of COVID-19 patient peripheral mononuclear blood cells (PBMC) identified enhanced interactions between SARS-CoV-2-upregulated G9a and distinct translation regulators, particularly the N 6 -methyladenosine (m 6 A) RNA methylase METTL3. These interactions with translation regulators implicated G9a in translational regulation of COVID-19. Inhibition of G9a activity suppressed SARS-CoV-2 replication in human alveolar epithelial cells. Accordingly, multi-omics analysis of the same alveolar cells identified SARS-CoV-2-induced changes at the transcriptional, m 6 A-epitranscriptional, translational, and post-translational (phosphorylation or secretion) levels that were reversed by inhibitor treatment. As suggested by the aforesaid chemoproteomic analysis, these multi-omics-correlated changes revealed a G9a-regulated translational mechanism of COVID-19 pathogenesis in which G9a directs translation of viral and host proteins associated with SARS-CoV-2 replication and with dysregulation of host response. Comparison of proteomic analyses of G9a inhibitor-treated, SARS-CoV-2 infected cells, or ex vivo culture of patient PBMCs, with COVID-19 patient data revealed that G9a inhibition reversed the patient proteomic landscape that correlated with COVID-19 pathology/symptoms. These data also indicated that the G9a-regulated, inhibitor-reversed, translational mechanism outperformed G9a-transcriptional suppression to ultimately determine COVID-19 pathogenesis and to define the inhibitor action, from which biomarkers of serve symptom vulnerability were mechanistically derived. This cell line-to-patient conservation of G9a-translated, COVID-19 proteome suggests that G9a inhibitors can be used to treat patients with COVID-19, particularly patients with long-lasting COVID-19 sequelae.
Collapse
|
18
|
Zheng Y, Li Y, Li M, Wang R, Jiang Y, Zhao M, Lu J, Li R, Li X, Shi S. COVID-19 cooling: Nanostrategies targeting cytokine storm for controlling severe and critical symptoms. Med Res Rev 2024; 44:738-811. [PMID: 37990647 DOI: 10.1002/med.21997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/16/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants continue to wreak havoc worldwide, the "Cytokine Storm" (CS, also known as the inflammatory storm) or Cytokine Release Syndrome has reemerged in the public consciousness. CS is a significant contributor to the deterioration of infected individuals. Therefore, CS control is of great significance for the treatment of critically ill patients and the reduction of mortality rates. With the occurrence of variants, concerns regarding the efficacy of vaccines and antiviral drugs with a broad spectrum have grown. We should make an effort to modernize treatment strategies to address the challenges posed by mutations. Thus, in addition to the requirement for additional clinical data to monitor the long-term effects of vaccines and broad-spectrum antiviral drugs, we can use CS as an entry point and therapeutic target to alleviate the severity of the disease in patients. To effectively combat the mutation, new technologies for neutralizing or controlling CS must be developed. In recent years, nanotechnology has been widely applied in the biomedical field, opening up a plethora of opportunities for CS. Here, we put forward the view of cytokine storm as a therapeutic target can be used to treat critically ill patients by expounding the relationship between coronavirus disease 2019 (COVID-19) and CS and the mechanisms associated with CS. We pay special attention to the representative strategies of nanomaterials in current neutral and CS research, as well as their potential chemical design and principles. We hope that the nanostrategies described in this review provide attractive treatment options for severe and critical COVID-19 caused by CS.
Collapse
Affiliation(s)
- Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mao Li
- Health Management Centre, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
19
|
Yeh LT, Chan CH, Wang YH, Lee CY, Yang SF, Yeh CB. Exploring the incidence of peripheral arterial occlusive disease following COVID-19 infection: A retrospective cohort study. J Med Virol 2024; 96:e29519. [PMID: 38465773 DOI: 10.1002/jmv.29519] [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: 01/03/2024] [Revised: 02/15/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
Peripheral arterial occlusive disease (PAOD) is a clinical manifestation of systemic atherosclerosis and is always associated with cerebrovascular disease and various complications. The aim of our study is to evaluate the relationship between the coronavirus disease 2019 (COVID-19) infection and the subsequent PAOD development. A retrospective cohort study was conducted and individuals with COVID-19 infection were identified from the TriNetX analytics platform. A total of 2 206 065 patients with COVID-19 infection and 2 206 065 patients without COVID-19 infection were recruited after exclusion and matching. The primary outcome was the development of PAOD after the COVID-19 infection. The Cox proportional hazard regression was adopted to yield the hazard ratio (HR) and 95% confidence interval (CI) of PAOD between groups. After the whole follow-up period, the incidence of PAOD was significantly higher in the COVID-19 group at both the 3-month follow-up (HR: 1.27, 95% CI: 1.24-1.30) and the 12-month follow-up (HR: 1.33, 95% CI: 1.31-1.35) The Kaplan-Meier analysis with the log-rank test demonstrated a higher cumulative probability of PAOD in the COVID-19 group compared to the non-COVID-19 group (p < 0.001). In stratified analysis using 65 years as the threshold, both age groups in the COVID-19 group exhibited a higher risk of PAOD. Similarly, in the sex and race stratified analysis, the COVID-19 group performed a higher risk of PAOD in both subgroups. In conclusion, the COVID-19 infections are strongly associated with an increment of PAOD incidence.
Collapse
Affiliation(s)
- Liang-Tsai Yeh
- Department of Anesthesiology, Changhua Christian Hospital, Changhua, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chi-Ho Chan
- Department of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Yi Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Ophthalmology, Nobel Eye Institute, Taipei, Taiwan
| | - Shun-Fa Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chao-Bin Yeh
- Department of Emergency Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
20
|
Bekdash R, Yoshida K, Nair MS, Qiu L, Ahdout J, Tsai HY, Uryu K, Soni RK, Huang Y, Ho DD, Yazawa M. Developing inhibitory peptides against SARS-CoV-2 envelope protein. PLoS Biol 2024; 22:e3002522. [PMID: 38483887 PMCID: PMC10939250 DOI: 10.1371/journal.pbio.3002522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/25/2024] [Indexed: 03/17/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has affected approximately 800 million people since the start of the Coronavirus Disease 2019 (COVID-19) pandemic. Because of the high rate of mutagenesis in SARS-CoV-2, it is difficult to develop a sustainable approach for prevention and treatment. The Envelope (E) protein is highly conserved among human coronaviruses. Previous studies reported that SARS-CoV-1 E deficiency reduced viral propagation, suggesting that E inhibition might be an effective therapeutic strategy for SARS-CoV-2. Here, we report inhibitory peptides against SARS-CoV-2 E protein named iPep-SARS2-E. Leveraging E-induced alterations in proton homeostasis and NFAT/AP-1 pathway in mammalian cells, we developed screening platforms to design and optimize the peptides that bind and inhibit E protein. Using Vero-E6 cells, human-induced pluripotent stem cell-derived branching lung organoid and mouse models with SARS-CoV-2, we found that iPep-SARS2-E significantly inhibits virus egress and reduces viral cytotoxicity and propagation in vitro and in vivo. Furthermore, the peptide can be customizable for E protein of other human coronaviruses such as Middle East Respiratory Syndrome Coronavirus (MERS-CoV). The results indicate that E protein can be a potential therapeutic target for human coronaviruses.
Collapse
Affiliation(s)
- Ramsey Bekdash
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York, United States of America
- Columbia Stem Cell Initiative, Columbia University, New York, New York, United States of America
- Department of Pharmacology, Columbia University, New York, New York, United States of America
| | - Kazushige Yoshida
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York, United States of America
- Columbia Stem Cell Initiative, Columbia University, New York, New York, United States of America
| | - Manoj S. Nair
- Aaron Diamond AIDS Research Center, Columbia University, New York, New York, United States of America
| | - Lauren Qiu
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York, United States of America
- Columbia Stem Cell Initiative, Columbia University, New York, New York, United States of America
- Department of Biological Science, Columbia University, New York, New York, United States of America
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Johnathan Ahdout
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Hsiang-Yi Tsai
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kunihiro Uryu
- EMSCOPIC, New York, New York, United States of America
| | - Rajesh K. Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Columbia University, New York, New York, United States of America
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University, New York, New York, United States of America
| | - David D. Ho
- Aaron Diamond AIDS Research Center, Columbia University, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University, New York, New York, United States of America
- Division of Infectious Diseases, Department of Medicine, Columbia University, New York, New York, United States of America
| | - Masayuki Yazawa
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York, United States of America
- Columbia Stem Cell Initiative, Columbia University, New York, New York, United States of America
- Department of Pharmacology, Columbia University, New York, New York, United States of America
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
21
|
Armario X, Carron J, Simpkin AJ, Elhadi M, Kennedy C, Abdel-Wahab M, Bleiziffer S, Lefèvre T, Wolf A, Pilgrim T, Villablanca PA, Blackman DJ, Van Mieghem NM, Hengstenberg C, Swaans MJ, Prendergast BD, Patterson T, Barbanti M, Webb JG, Behan M, Resar J, Chen M, Hildick-Smith D, Spence MS, Zweiker D, Bagur R, Teles R, Ribichini FL, Jagielak D, Park DW, Kornowski R, Wykrzykowska JJ, Bunc M, Estévez-Loureiro R, Poon K, Götberg M, Jeger RV, Ince H, Packer EJS, Angelillis M, Nombela-Franco L, Guo Y, Savontaus M, Al-Moghairi AM, Parasca CA, Kliger C, Roy D, Molnár L, Silva M, White J, Yamamoto M, Carrilho-Ferreira P, Toggweiler S, Voudris V, Ohno Y, Rodrigues I, Parma R, Ojeda S, Toutouzas K, Regueiro A, Grygier M, AlMerri K, Cruz-González I, Fridrich V, de la Torre Hernández JM, Noble S, Kala P, Asmarats L, Kurt IH, Bosmans J, Erglis M, Casserly I, Iskandarani D, Bhindi R, Kefer J, Yin WH, Rosseel L, Kim HS, O'Connor S, Hellig F, Sztejfman M, Mendiz O, Pineda AM, Seth A, Pllaha E, de Brito FS, Bajoras V, Balghith MA, Lee M, Eid-Lidt G, Vandeloo B, Vaz VD, Alasnag M, Ussia GP, Tay E, Mayol J, Gunasekaran S, Sardella G, Buddhari W, Kao HL, Dager A, Tzikas A, Gudmundsdottir IJ, Edris A, Gutiérrez Jaikel LA, Arias EA, Al-Hijji M, Ertürk M, Conde-Vela C, Boljević D, Ferrero Guadagnoli A, Hermlin T, ElGuindy AM, Lima-Filho MDO, de Moura Santos L, Perez L, Maluenda G, Akyüz AR, Alhaddad IA, Amin H, So CY, Al Nooryani AA, Vaca C, Albistur J, Nguyen QN, Arzamendi D, Grube E, Modine T, Tchétché D, Hayashida K, Latib A, Makkar RR, Piazza N, Søndergaard L, McEvoy JW, Mylotte D. Impact of the COVID-19 Pandemic on Global TAVR Activity: The COVID-TAVI Study. JACC Cardiovasc Interv 2024; 17:374-387. [PMID: 38180419 DOI: 10.1016/j.jcin.2023.10.041] [Citation(s) in RCA: 1] [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] [Received: 08/09/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND The COVID-19 pandemic adversely affected health care systems. Patients in need of transcatheter aortic valve replacement (TAVR) are especially susceptible to treatment delays. OBJECTIVES This study sought to evaluate the impact of the COVID-19 pandemic on global TAVR activity. METHODS This international registry reported monthly TAVR case volume in participating institutions prior to and during the COVID-19 pandemic (January 2018 to December 2021). Hospital-level information on public vs private, urban vs rural, and TAVR volume was collected, as was country-level information on socioeconomic status, COVID-19 incidence, and governmental public health responses. RESULTS We included 130 centers from 61 countries, including 65,980 TAVR procedures. The first and second pandemic waves were associated with a significant reduction of 15% (P < 0.001) and 7% (P < 0.001) in monthly TAVR case volume, respectively, compared with the prepandemic period. The third pandemic wave was not associated with reduced TAVR activity. A greater reduction in TAVR activity was observed in Africa (-52%; P = 0.001), Central-South America (-33%; P < 0.001), and Asia (-29%; P < 0.001). Private hospitals (P = 0.005), urban areas (P = 0.011), low-volume centers (P = 0.002), countries with lower development (P < 0.001) and economic status (P < 0.001), higher COVID-19 incidence (P < 0.001), and more stringent public health restrictions (P < 0.001) experienced a greater reduction in TAVR activity. CONCLUSIONS TAVR procedural volume declined substantially during the first and second waves of the COVID-19 pandemic, especially in Africa, Central-South America, and Asia. National socioeconomic status, COVID-19 incidence, and public health responses were associated with treatment delays. This information should inform public health policy in case of future global health crises.
Collapse
Affiliation(s)
- Xavier Armario
- Department of Cardiology, Galway University Hospital, Galway, Ireland; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jennifer Carron
- Department of Cardiology, Galway University Hospital, Galway, Ireland
| | - Andrew J Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
| | - Mohamed Elhadi
- Department of Cardiology, Galway University Hospital, Galway, Ireland
| | - Ciara Kennedy
- Department of Cardiology, Galway University Hospital, Galway, Ireland
| | | | - Sabine Bleiziffer
- Heart and Diabetes Center Northrhine-Westfalia, Clinic for Thoracic and Cardiovascular Surgery, Bad Oeynhausen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | - John G Webb
- St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Miles Behan
- Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Jon Resar
- John Hopkins Hospital, Baltimore, Maryland, USA
| | - Mao Chen
- West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | - Rodrigo Bagur
- University Hospital, London Health Sciences Center, London, Ontario, Canada
| | - Rui Teles
- Hospital de Santa Cruz, CHLO, Nova Medical School, CEDOC, Lisbon, Portugal
| | | | | | | | | | | | - Matjaz Bunc
- Ljubljana University Medical Center, Ljubljana, Slovenia
| | | | - Karl Poon
- The Prince Charles Hospital, Brisbane, Australia
| | - Matthias Götberg
- Clinical Sciences, Lund University, Skane University Hospital, Lund, Sweden
| | | | | | | | | | | | - Yingqiang Guo
- West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | - Chad Kliger
- Lenox Hill/Northwell Health, New York, New York, USA
| | - David Roy
- St. Vincent's Hospital, Sydney, Australia
| | - Levente Molnár
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Mariana Silva
- Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | | | - Masanori Yamamoto
- Toyohashi Heart Center, Toyohashi, Japan; Nagoya Heart Center, Nagoya, Japan; Gifu Heart Center, Gifu, Japan
| | | | | | | | - Yohei Ohno
- Tokai University School of Medicine, Isehara, Japan
| | | | | | - Soledad Ojeda
- Hospital Universitario Reina Sofía, Universidad de Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain
| | | | | | - Marek Grygier
- Poznan University School of Medical Sciences, Poznan, Poland
| | | | | | - Viliam Fridrich
- National Institute of Cardiovascular Diseases, Bratislava, Slovakia
| | | | | | - Petr Kala
- Centrum Kardiovaskulární a Transplantační Chirurgie, Brno, Czechia
| | | | | | | | | | - Ivan Casserly
- Mater Misericordiae University Hospital, Dublin, Ireland; Mater Private Hospital, Dublin, Ireland
| | | | | | - Joelle Kefer
- Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | | | | | - Hyo-Soo Kim
- Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | - Andres M Pineda
- University of Florida College of Medicine Jacksonville, Jacksonville, Florida, USA
| | - Ashok Seth
- Fortis Escorts Heart Institute, New Delhi, India
| | | | | | - Vilhelmas Bajoras
- Division of Cardiology and Vascular Diseases, Vilnius University Hospital Santaros Clinics, Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Michael Lee
- Queen Elizabeth Hospital, Hong Kong, Hong Kong
| | - Guering Eid-Lidt
- Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Bert Vandeloo
- Department of Cardiology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Belgium
| | | | | | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Edgar Tay
- National University Heart Center, Singapore, Singapore
| | | | | | | | | | - Hsien-Li Kao
- National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | - Ahmad Edris
- Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | | | | | | | - Mehmet Ertürk
- Istanbul Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, University of Health Science, Istanbul, Turkey
| | | | | | | | | | | | - Moysés de Oliveira Lima-Filho
- Hospital das Clínicas de Ribeirão Preto, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Luis Perez
- Hospital Clínico Regional Guillermo Grant Benavente, Concepción, Chile
| | - Gabriel Maluenda
- Hospital San Borja Arriaran, Santiago, Chile; Clínica Alemana, Santiago, Chile
| | - Ali Rıza Akyüz
- Ahi Evren Thoracic and Cardiovascular Surgery Training and Research Hospital, Health Sciences University, Trabzon, Turkey
| | | | - Haitham Amin
- Mohammed Bin Khalifa Cardiac Center, Royal Medical Services, Awali, Bahrain
| | - Chak-Yu So
- Prince of Wales Hospital, Hong Kong, Hong Kong
| | | | - Carlos Vaca
- Instituto de Cardiología Intervencionista Cardiovida SRL, Santa Cruz, Bolivia
| | - Juan Albistur
- Hospital de Clínicas Dr Manuel Quintela, Montevideo, Uruguay
| | | | | | | | | | | | | | - Azeem Latib
- Montefiore Medical Center, Bronx, New York, USA
| | - Raj R Makkar
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nicolo Piazza
- McGill University Health Center, Montréal, Quebec, Canada
| | | | - John William McEvoy
- Department of Cardiology, Galway University Hospital, Galway, Ireland; School of Medicine, University of Galway, Galway, Ireland
| | - Darren Mylotte
- Department of Cardiology, Galway University Hospital, Galway, Ireland; School of Medicine, University of Galway, Galway, Ireland.
| |
Collapse
|
22
|
Tabarsi P, Mamishi S, Anjidani N, Shahpari R, Kafi H, Fallah N, Yazdani B, Ebrahimi A, Roshanzamir K, Ebrahimi H, Oveisi S, Soltani A, Petrovsky N, Barati S. Comparative immunogenicity and safety of SpikoGen®, a recombinant SARS-CoV-2 spike protein vaccine in children and young adults: An immuno-bridging clinical trial. Int Immunopharmacol 2024; 127:111436. [PMID: 38147778 DOI: 10.1016/j.intimp.2023.111436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND SpikoGen® is a recombinant subunit spike protein ectodomain vaccine manufactured in insect cells and formulated with the novel polysaccharide-based Advax-CpG55.2 adjuvant. This study aimed to compare the immunogenicity and safety of SpikoGen® vaccine in children, adolescents and young adults. METHODS This was a non-randomized, three-arm, open-label, parallel-group, immuno-bridging, non-inferiority trial to compare the immunogenicity and safety of a primary course of two intramuscular doses of SpikoGen® vaccine in children aged 5 to < 12 years, adolescents aged 12 to < 18 years and young adults aged 18 to 40 years. Children 5-12 years received a half dose of 12.5 μg spike protein, whereas the other groups received the full vaccine dose. Vaccine immunogenicity was evaluated via assessment of serum anti-spike and neutralizing antibodies 14 days after the second dose. Solicited adverse events were recorded for 7 days after each vaccination. Safety assessments including serious adverse events were continued through six months after the second dose in children and adolescents. RESULTS Two weeks after the second dose, seroconversion rates for neutralizing antibody levels were not significantly different for children (59.50 %), adolescents (52.06 %) and adults (56.01 %). The 95 % confidence interval of the difference in seroconversion rates between children and adults was within the prespecified non-inferiority margin of 10 % (-12 % to 5 %). SpikoGen® vaccine was well tolerated in all age groups with the most common solicited adverse events being injection site pain and fatigue which were generally transient and mild. CONCLUSION SpikoGen® vaccine was shown to be safe, well tolerated and immunogenic in children as young as 5 years of age, with non-inferior responses to those seen in adults. The Iranian FDA authorisation of SpikoGen® vaccine is now extended down to 5 years of age.
Collapse
Affiliation(s)
- Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute for Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Setareh Mamishi
- Department of Infectious Diseases, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Pediatric Infectious Diseases Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ramin Shahpari
- Medical Department, Orchid Pharmed Company, Tehran, Iran
| | - Hamidreza Kafi
- Medical Department, Orchid Pharmed Company, Tehran, Iran
| | - Newsha Fallah
- Medical Department, Orchid Pharmed Company, Tehran, Iran
| | - Babak Yazdani
- Medical Department, Orchid Pharmed Company, Tehran, Iran
| | - Ali Ebrahimi
- Medical Department, Orchid Pharmed Company, Tehran, Iran
| | - Khashayar Roshanzamir
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Hamidreza Ebrahimi
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Soudabeh Oveisi
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Adele Soltani
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Saghar Barati
- Medical Department, Orchid Pharmed Company, Tehran, Iran.
| |
Collapse
|
23
|
Kim YJ, Min J. Advances in nanobiosensors during the COVID-19 pandemic and future perspectives for the post-COVID era. NANO CONVERGENCE 2024; 11:3. [PMID: 38206526 PMCID: PMC10784265 DOI: 10.1186/s40580-023-00410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2024]
Abstract
The unprecedented threat of the highly contagious virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes exponentially increased infections of coronavirus disease 2019 (COVID-19), highlights the weak spots of the current diagnostic toolbox. In the midst of catastrophe, nanobiosensors offer a new opportunity as an alternative tool to fill a gap among molecular tests, rapid antigen tests, and serological tests. Nanobiosensors surpass the potential of antigen tests because of their enhanced sensitivity, thus enabling us to see antigens as stable and easy-to-access targets. During the first three years of the COVID-19 pandemic, a substantial number of studies have reported nanobiosensors for the detection of SARS-CoV-2 antigens. The number of articles on nanobiosensors and SARS-CoV-2 exceeds the amount of nanobiosensor research on detecting previous infectious diseases, from influenza to SARS-CoV and MERS-CoV. This unprecedented publishing pace also implies the significance of SARS-CoV-2 and the present pandemic. In this review, 158 studies reporting nanobiosensors for detecting SARS-CoV-2 antigens are collected to discuss the current challenges of nanobiosensors using the criteria of point-of-care (POC) diagnostics along with COVID-specific issues. These advances and lessons during the pandemic pave the way for preparing for the post-COVID era and potential upcoming infectious diseases.
Collapse
Affiliation(s)
- Young Jun Kim
- School of Integrative Engineering, Chung-Ang University, Heukseok-Dong, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Junhong Min
- School of Integrative Engineering, Chung-Ang University, Heukseok-Dong, Dongjak-Gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
24
|
Berenguer J, Calvo-Alcántara MJ, Alvaro-Meca A, Estévez JC, Basanta M, Ruiz S, Matáix ÁL, Bienzóbas C, Cosano L, Silva AP, Salas P, Gullón P, Franco M, Arribas JR, Molero JM, Hernán MA. Three Years of the Coronavirus Disease 2019 Pandemic in a European Region: A Population-Based Longitudinal Assessment in Madrid Between 2020 and 2022. Open Forum Infect Dis 2024; 11:ofad635. [PMID: 38173846 PMCID: PMC10763997 DOI: 10.1093/ofid/ofad635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Background Our objective was to assess the health impact of coronavirus disease 2019 (COVID-19) during 2020-2022 in the Madrid region. Methods We included all individuals registered in the Madrid Health System Registry as of 31 December 2019, and followed them until 31 December 2022. Using a unique personal identifier, we linked the databases of primary care, hospitals, pharmacies, certified laboratories performing diagnostic tests, vaccines, and mortality. Results Of 6 833 423 individuals, 21.4% had a confirmed COVID-19 diagnosis, and 1.5% had a COVID-19 hospitalization (primary diagnosis). Thirty-day mortality was 1.6% for confirmed COVID-19 (from 11.4% in first semester 2020 to 0.4% in first semester 2022). Thirty-day mortality was 10.8% for COVID-19 hospitalizations (from 14.0% in first semester 2020 to 6.0% in second semester 2022). There were 24 073 deaths within 30 days of a confirmed COVID-19 diagnosis. Advanced age, male sex, higher socioeconomic deprivation, and comorbidities were associated with higher mortality. Conclusions By linking administrative and clinical databases, we characterized the burden of the COVID-19 pandemic in Madrid over 3 years. Our analysis proposes a high-level framework for comparisons of the burden of COVID-19 across areas worldwide.
Collapse
Affiliation(s)
- Juan Berenguer
- Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Madrid, Spain
| | | | - Alejandro Alvaro-Meca
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Madrid, Spain
- Medicina Preventiva y Salud Pública, Universidad Rey Juan Carlos, Madrid, Spain
| | - José C Estévez
- Gerencia Asistencial de Atención Primaria, Madrid, Spain
| | - Miguel Basanta
- Dirección General de Sistemas de Información y Equipamientos Sanitarios, Madrid, Spain
| | - Sergio Ruiz
- Gerencia Asistencial de Atención Primaria, Madrid, Spain
| | - Ángel L Matáix
- Subdirección General de Farmacia y Productos Sanitarios, Madrid, Spain
| | - César Bienzóbas
- Dirección General de Inspección y Ordenación Sanitaria, Madrid, Spain
| | - Lourdes Cosano
- Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Aura P Silva
- Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pilar Salas
- Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pedro Gullón
- Departamento de Cirugía, Ciencias Médicas y Sociales, Grupo de Investigación en Epidemiología y Salud Pública, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Manuel Franco
- Departamento de Cirugía, Ciencias Médicas y Sociales, Grupo de Investigación en Epidemiología y Salud Pública, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - José R Arribas
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Madrid, Spain
- Infectious Diseases, Internal Medicine, Hospital Universitario La Paz, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Miguel A Hernán
- CAUSALab, Departments of Epidemiology and Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Chavda V, Yadav D, Parmar H, Brahmbhatt R, Patel B, Madhwani K, Jain M, Song M, Patel S. A Narrative Overview of Coronavirus Infection: Clinical Signs and Symptoms, Viral Entry and Replication, Treatment Modalities, and Management. Curr Top Med Chem 2024; 24:1883-1916. [PMID: 38859776 DOI: 10.2174/0115680266296095240529114058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 06/12/2024]
Abstract
The global pandemic known as coronavirus disease (COVID-19) is causing morbidity and mortality on a daily basis. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV- -2) virus has been around since December 2019 and has infected a high number of patients due to its idiopathic pathophysiology and rapid transmission. COVID-19 is now deemed a newly identified "syndrome" condition since it causes a variety of unpleasant symptoms and systemic side effects following the pandemic. Simultaneously, it always becomes potentially hazardous when new variants develop during evolution. Its random viral etiology prevents accurate and suitable therapy. Despite the fact that multiple preclinical and research studies have been conducted to combat this lethal virus, and various therapeutic targets have been identified, the precise course of therapy remains uncertain. However, just a few drugs have shown efficacy in treating this viral infection in its early stages. Currently, several medicines and vaccinations have been licensed following clinical trial research, and many countries are competing to find the most potent and effective immunizations against this highly transmissible illness. For this narrative review, we used PubMed, Google Scholar, and Scopus to obtain epidemiological data, pre-clinical and clinical trial outcomes, and recent therapeutic alternatives for treating COVID-19 viral infection. In this study, we discussed the disease's origin, etiology, transmission, current advances in clinical diagnostic technologies, different new therapeutic targets, pathophysiology, and future therapy options for this devastating virus. Finally, this review delves further into the hype surrounding the SARS-CoV-2 illness, as well as present and potential COVID-19 therapies.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pathology, Stanford School of Medicine, Stanford University Medical Center, Palo Alto94305, CA, USA
- Department of Medicine, Multispeciality, Trauma and ICCU Center, Sardar Hospital, Ahmedabad, 382352, Gujarat, India
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, South Korea
| | - Harisinh Parmar
- Department of Neurosurgery, Krishna institute of medical sciences, Karad, Maharashtra, India
| | - Raxit Brahmbhatt
- Department of Medicine, Multispeciality, Trauma and ICCU Center, Sardar Hospital, Ahmedabad, 382352, Gujarat, India
| | - Bipin Patel
- Department of Medicine, Multispeciality, Trauma and ICCU Center, Sardar Hospital, Ahmedabad, 382352, Gujarat, India
| | - Kajal Madhwani
- Department of Life Science, University of Westminster, London, W1B 2HW, United Kingdom
| | - Meenu Jain
- Gajra Raja Medical College, Gwalior, 474009, Madhya Pradesh, India
| | - Minseok Song
- Department of Life Science, Yeungnam University, South Korea
| | - Snehal Patel
- Department of Pharmacology, Nirma University, Ahmedabad, 382481, Gujarat, India
| |
Collapse
|
26
|
Wu Y, Wu N, Jia X, Wu Y, Zhang X, Liu Y, Hou Y, Shen Y, Li E, Wang W, Wang Y, Chiu S. Long-term immune response to Omicron-specific mRNA vaccination in mice, hamsters, and nonhuman primates. MedComm (Beijing) 2023; 4:e460. [PMID: 38107058 PMCID: PMC10724501 DOI: 10.1002/mco2.460] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron and its subvariants (such as BQ.1, XBB and the latest variants, including XBB.1.16, EG.5, and BA.2.86), as the dominant variants, currently account for almost all new infections in the world due to their high transmissibility and immune escape ability. Omicron-specific mRNA vaccines showed great potential to protect against Omicron infections. However, whether the vaccine could provide long-term protection is unknown. Toward this goal, we evaluated the immunogenicity of a preclinical Omicron (BA.1)-specific mRNA vaccine (SOmicron-6P) in different animal models. SOmicron-6P induced the highest levels of antibody titers at 1-2 weeks in different animals after the second dose. Even 9 months after the immunization, we observed modest neutralizing activity against Omicron subvariants in macaques. In addition, immunological memory cells can be rapidly reactivated upon stimulation. SOmicron-6P at concentrations higher than 10 μg effectively protected hamsters from BA.1 challenge 253 days after the first immunization, which could be attributed to the reactivation of immune systems. In addition, the toxicity tests conducted in rats revealed a highly favorable biosafety profile for SOmicron-6P, even at high dosages. Our data suggest that the Omicron-specific mRNA vaccine is highly effective and safe in animal models and provides long-term immunologic protection against SARS-CoV-2 Omicron infections.
Collapse
Affiliation(s)
- Yi Wu
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Namei Wu
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Xiaoying Jia
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanP. R. China
| | - Yan Wu
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanP. R. China
| | - Xinghai Zhang
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanP. R. China
| | - Yang Liu
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanP. R. China
| | - Yuxia Hou
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanP. R. China
- University of Chinese Academy of SciencesBeijingP. R. China
| | | | - Entao Li
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
- DepartmentKey Laboratory of Anhui Province for Emerging and Reemerging Infectious DiseasesHefeiAnhuiP. R. China
| | - Wei Wang
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanP. R. China
- University of Chinese Academy of SciencesBeijingP. R. China
| | - Yucai Wang
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
- RNAlfa BiotechHefeiAnhuiP. R. China
| | - Sandra Chiu
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiP. R. China
- DepartmentKey Laboratory of Anhui Province for Emerging and Reemerging Infectious DiseasesHefeiAnhuiP. R. China
- Core Unit of National Clinical Research Center for Laboratory MedicineHefeiAnhuiP. R. China
| |
Collapse
|
27
|
Rudramurthy GR, Naveenkumar CN, Bharathkumar K, Shandil RK, Narayanan S. Genomic Mutations in SARS-CoV-2 Genome following Infection in Syrian Golden Hamster and Associated Lung Pathologies. Pathogens 2023; 12:1328. [PMID: 38003792 PMCID: PMC10674674 DOI: 10.3390/pathogens12111328] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The continuous evolution of the SARS-CoV-2 virus led to constant developments and efforts in understanding the significance and impacts of SARS-CoV-2 variants on human health. Our study aimed to determine the accumulation of genetic mutations and associated lung pathologies in male and female hamsters infected with the ancestral Wuhan strain of SARS-CoV-2. The present study showed no significant difference in the viral load between male and female hamsters and peak infection was found to be on day four post infection in both sexes of the animals. Live virus particles were detected up to 5 days post infection (dpi) through the TCID-50 assay, while qRT-PCR could detect viral RNA up to 14 dpi from all the infected animals. Further, the determination of the neutralizing antibody titer showed the onset of the humoral immune response as early as 4 dpi in both sexes against SARS-CoV-2, and a significant cross-protection against the delta variant of SARS-CoV-2 was observed. Histopathology showed edema, inflammation, inflammatory cell infiltration, necrosis, and degeneration of alveolar and bronchial epithelium cells from 3 dpi to 14 dpi in both sexes. Furthermore, next-generation sequencing (NGS) showed up to 10 single-nucleotide polymorphisms (SNPs) in the SARS-CoV-2 (ancestral Wuhan strain) genome isolated from both male and female hamsters. The mutation observed at the 23014 position (Glu484Asp) in the SARS-CoV-2 genome isolated from both sexes of the hamsters plays a significant role in the antiviral efficacy of small molecules, vaccines, and the Mabs-targeting S protein. The present study shows that either of the genders can be used in the pre-clinical efficacy of antiviral agents against SARS-CoV-2 in hamsters. However, considering the major mutation in the S protein, the understanding of the genetic mutation in SARS-CoV-2 after passing through hamsters is crucial in deciding the efficacy of the antiviral agents targeting the S protein. Importance: Our study findings indicate the accumulation of genomic mutations in SARS-CoV-2 after passing through the Syrian golden hamsters. Understanding the genomic mutations showed that either of the hamster genders can be used in the pre-clinical efficacy of antiviral agents and vaccines.
Collapse
Affiliation(s)
- Gudepalya Renukaiah Rudramurthy
- Foundation for Neglected Disease Research (FNDR), Plot No. 20A, KIADB Industrial Area, Bengaluru 561203, Karnataka, India; (C.N.N.); (K.B.); (R.K.S.); (S.N.)
| | | | | | | | | |
Collapse
|
28
|
Gonçalves M, Guimarães A, Carvalho T, Antunes P, Mendes S, Soares J, Gonçalves R, Arroja B, Rebelo A. Safety of Endoscopy Units during the COVID-19 Pandemic. GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2023; 30:4-10. [PMID: 38020825 PMCID: PMC10661713 DOI: 10.1159/000526125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/18/2022] [Indexed: 12/01/2023]
Abstract
Introduction The COVID-19 pandemic drastically changed the daily routine of all healthcare systems worldwide, and endoscopy units were no exception. Endoscopic exams were considered to have a high risk of transmission, and therefore, the safety of endoscopy units and the consequent need for pre-endoscopy SARS-CoV-2 screening were questioned early on. The aim of our study was to assess the safety of endoscopy units during the COVID-19 pandemic, as well as the effectiveness/necessity for SARS-CoV-2 screening prior to endoscopies. Material and Methods This is a retrospective and single-center study carried out in a Portuguese tertiary hospital. All patients who underwent endoscopic procedures between September 1, 2020 and February 28, 2021 were included. The pre-endoscopy screening consisted of a specific questionnaire or a RT-PCR test for SARS-CoV-2 (nasal and oropharyngeal swab). Data were obtained through patient's clinical records and the Trace COVID platform. Results A total of 2,166 patients were included. Patients had a mean age of 61.8 years and were predominantly male (56.2%, n = 1,218). Eighty-one (3.7%) patients had previous SARS-CoV-2 infection, with a median difference of 74 days (IQ 40.5:160.5) between infection and endoscopy. Most patients (70.2%, n = 1,521) underwent PCR screening for SARS-CoV-2 up to 72 h before the procedure, with the remaining patients (29.8%, n = 645) answering a questionnaire of symptoms and risk contacts up to 3 days before endoscopy. Of the patients who underwent RT-PCR screening for SARS-CoV-2, 21 (1.4%) tested positive, and all were asymptomatic at the time of the screening. The evaluation for SARS-CoV-2 infection up to 14 days after the endoscopic exams identified 9 positive patients (0.42%) for SARS-CoV-2. The median difference in days between endoscopy and the diagnosis of infection was 10 days. Discussion/Conclusion Pre-endoscopy screening with RT-PCR test for SARS-CoV-2 identified a very small number of patients with COVID-19 infection as well as patients with COVID-19 infection in the following 14 days. Therefore, the risk of infection in endoscopy units is negligible if screening of symptoms and risk contacts is applied and individual protective equipment is used.
Collapse
Affiliation(s)
| | | | - Tânia Carvalho
- Gastroenterology Department, Hospital de Braga, Braga, Portugal
| | - Pedro Antunes
- Gastroenterology Department, Hospital de Braga, Braga, Portugal
| | - Sofia Mendes
- Gastroenterology Department, Hospital de Braga, Braga, Portugal
| | - João Soares
- Gastroenterology Department, Hospital de Braga, Braga, Portugal
| | | | - Bruno Arroja
- Gastroenterology Department, Hospital de Braga, Braga, Portugal
| | - Ana Rebelo
- Gastroenterology Department, Hospital de Braga, Braga, Portugal
| |
Collapse
|
29
|
Khan MS, Kim E, Le Hingrat Q, Kleinman A, Ferrari A, Sammartino JC, Percivalle E, Xu C, Huang S, Kenniston TW, Cassaniti I, Baldanti F, Pandrea I, Gambotto A, Apetrei C. Tetravalent SARS-CoV-2 S1 subunit protein vaccination elicits robust humoral and cellular immune responses in SIV-infected rhesus macaque controllers. mBio 2023; 14:e0207023. [PMID: 37830800 PMCID: PMC10653869 DOI: 10.1128/mbio.02070-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/30/2023] [Indexed: 10/14/2023] Open
Abstract
IMPORTANCE The study provides important insights into the immunogenicity and efficacy of a tetravalent protein subunit vaccine candidate against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The vaccine induced both humoral and cellular immune responses in nonhuman primates with controlled SIVagm infection and was able to generate Omicron variant-specific antibodies without specifically vaccinating with Omicron. These findings suggest that the tetravalent composition of the vaccine candidate could provide broad protection against multiple SARS-CoV-2 variants while minimizing the risk of immune escape and the emergence of new variants. Additionally, the use of rhesus macaques with controlled SIVsab infection may better represent vaccine immunogenicity in humans with chronic viral diseases, highlighting the importance of preclinical animal models in vaccine development. Overall, the study provides valuable information for the development and implementation of coronavirus disease 2019 vaccines, particularly for achieving global vaccine equity and addressing emerging variants.
Collapse
Affiliation(s)
- Muhammad S. Khan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Eun Kim
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Quentin Le Hingrat
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Adam Kleinman
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Alessandro Ferrari
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jose C. Sammartino
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elena Percivalle
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Cuiling Xu
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Shaohua Huang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas W. Kenniston
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Irene Cassaniti
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Ivona Pandrea
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
30
|
Shaver N, Katz M, Darko Asamoah G, Linkins LA, Abdelkader W, Beck A, Bennett A, Hughes SE, Smith M, Begin M, Coyle D, Piggott T, Kagina BM, Welch V, Colijn C, Earn DJD, El Emam K, Heffernan J, O'Brien SF, Wilson K, Collins E, Navarro T, Beyene J, Boutron I, Bowdish D, Cooper C, Costa A, Curran J, Griffith L, Hsu A, Grimshaw J, Langlois MA, Li X, Pham-Huy A, Raina P, Rubini M, Thabane L, Wang H, Xu L, Brouwers M, Horsley T, Lavis J, Iorio A, Little J. Protocol for a living evidence synthesis on variants of concern and COVID-19 vaccine effectiveness. Vaccine 2023; 41:6411-6418. [PMID: 37718186 DOI: 10.1016/j.vaccine.2023.09.012] [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: 02/08/2023] [Revised: 07/17/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND It is evident that COVID-19 will remain a public health concern in the coming years, largely driven by variants of concern (VOC). It is critical to continuously monitor vaccine effectiveness as new variants emerge and new vaccines and/or boosters are developed. Systematic surveillance of the scientific evidence base is necessary to inform public health action and identify key uncertainties. Evidence syntheses may also be used to populate models to fill in research gaps and help to prepare for future public health crises. This protocol outlines the rationale and methods for a living evidence synthesis of the effectiveness of COVID-19 vaccines in reducing the morbidity and mortality associated with, and transmission of, VOC of SARS-CoV-2. METHODS Living evidence syntheses of vaccine effectiveness will be carried out over one year for (1) a range of potential outcomes in the index individual associated with VOC (pathogenesis); and (2) transmission of VOC. The literature search will be conducted up to May 2023. Observational and database-linkage primary studies will be included, as well as RCTs. Information sources include electronic databases (MEDLINE; Embase; Cochrane, L*OVE; the CNKI and Wangfang platforms), pre-print servers (medRxiv, BiorXiv), and online repositories of grey literature. Title and abstract and full-text screening will be performed by two reviewers using a liberal accelerated method. Data extraction and risk of bias assessment will be completed by one reviewer with verification of the assessment by a second reviewer. Results from included studies will be pooled via random effects meta-analysis when appropriate, or otherwise summarized narratively. DISCUSSION Evidence generated from our living evidence synthesis will be used to inform policy making, modelling, and prioritization of future research on the effectiveness of COVID-19 vaccines against VOC.
Collapse
Affiliation(s)
- Nicole Shaver
- Synthesis and Application Unit, School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Melanie Katz
- Public Health Agency of Canada, Ottawa, Ontario, Canada.
| | - Gideon Darko Asamoah
- Synthesis and Application Unit, School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Lori-Ann Linkins
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | | | - Andrew Beck
- Synthesis and Application Unit, School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Alexandria Bennett
- Synthesis and Application Unit, School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Sarah E Hughes
- Centre for Patient Reported Outcome Research, Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom.
| | - Maureen Smith
- The Cochrane Consumer Network, Ottawa, Ontario, Canada.
| | - Mpho Begin
- Cassidy e-Care Solutions Inc, Winnipeg, Manitoba, Canada.
| | - Doug Coyle
- Faculty of Medicine, University of Ottawa, Ontario, Canada.
| | - Thomas Piggott
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada; Peterborough Public Health, Peterborough, Ontario, Canada; Department of Family Medicine, Queens University, Kingston, Ontario, Canada.
| | - Benjamin M Kagina
- Vaccines for Africa Initiative; NITAGs Support Hub (NISH), Faculty of Health Sciences, School of Public Health, University of Cape Town, South Africa.
| | - Vivian Welch
- Bruyère Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ontario, Canada.
| | - Caroline Colijn
- Department of mathematics, Faculty of Science, Simon Fraser University, Burnaby, British Columbia, Canada.
| | - David J D Earn
- Department of Mathematics and Statistics, McMaster University, Hamilton, Ontario, Canada.
| | | | - Jane Heffernan
- Centre for Disease Modelling, Department of Mathematics & Statistics, Faculty of Science, York University, Toronto, Ontario, Canada.
| | - Sheila F O'Brien
- Epidemiology and Surveillance, Canadian Blood Services, Ottawa, Ontario, Canada; School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ontario, Canada.
| | - Kumanan Wilson
- Department of Medicine, University of Ottawa, Ontario, Canada; Bruyère Research Institute, Ottawa, Ontario, Canada; Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; O'Neill Institute for National and Global Health Law, Georgetown University, Washington, DC, USA.
| | - Erin Collins
- Public Health Agency of Canada, Ottawa, Ontario, Canada.
| | | | - Joseph Beyene
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Isabelle Boutron
- Université Paris Cité, Inserm, INRAE, Centre for Research in Epidemiology and Statistics (CRESS), F75004 Paris, France.
| | - Dawn Bowdish
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Curtis Cooper
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Andrew Costa
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Janet Curran
- School of Nursing, Faculty of Health, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Lauren Griffith
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Amy Hsu
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Jeremy Grimshaw
- Ottawa Hospital Research Institute and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | | | - Xiaoguang Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Anne Pham-Huy
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Parminder Raina
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Michele Rubini
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy.
| | - Lehana Thabane
- Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada; St Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada; Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa.
| | - Hui Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Lan Xu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Melissa Brouwers
- Synthesis and Application Unit, School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Tanya Horsley
- Royal College of Physicians and Surgeons of Canada, Ottawa, Ontario, Canada.
| | - John Lavis
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada; McMaster Health Forum, McMaster University, Hamilton, Ontario, Canada.
| | - Alfonso Iorio
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Julian Little
- Knowledge Synthesis and Application Unit, School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
31
|
Yıldırım S, Erkoyun E, Alpdoğan Ö, Yılmaz HO, Yılmaz B, Erdal Dönmez G, Sarıtaş A, Gökmen N, Ergan B, Bayrak V, Yakar MN, Kılıç Ö, Kılınç A, Saygılı S, Gaygısız Ü, Aydın K, Özel Yeşilyurt A, Cankar Dal H, Bayındır Dicle Ç, Turan S, Binay S, Yarıcı M, Yıldırım F, Hancı P, İnal MT, Akbaş T, Eyüpoğlu S, Albayrak T, Koçak G, Çakır T, Yüksel RC, Sarı A, Güneş M, Menteş O, Yamanel HL, Kirakli C. Vaccination status of COVID-19 patients followed up in the ICU in a country with heterologous vaccination policy: A multicenter national study in Turkey. J Infect Chemother 2023; 29:959-964. [PMID: 37343924 PMCID: PMC10278896 DOI: 10.1016/j.jiac.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/07/2023] [Accepted: 06/18/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVE Vaccination against severe acute respiratory syndrome coronavirus-2 (SARS-2) prevents the development of serious diseases has been shown in many studies. However, the effect of vaccination on outcomes in COVID-19 patients requiring intensive care is not clear. METHODS This is a retrospective multicenter study conducted in 17 intensive care unit (ICU) in Turkey between January 1, 2021, and December 31, 2021. Patients aged 18 years and older who were diagnosed with COVID-19 and followed in ICU were included in the study. Patients who have never been vaccinated and patients who have been vaccinated with a single dose were considered unvaccinated. Logistic regression models were fit for the two outcomes (28-day mortality and in-hospital mortality). RESULTS A total of 2968 patients were included final analysis. The most of patients followed in the ICU during the study period were unvaccinated (58.5%). Vaccinated patients were older, had higher Charlson comorbidity index (CCI), and had higher APACHE-2 scores than unvaccinated patients. Risk for 28-day mortality and in-hospital mortality was similar in across the year both vaccinated and unvaccinated patients. However, risk for in-hospital mortality and 28-day mortality was higher in the unvaccinated patients in quarter 4 adjusted for gender and CCI (OR: 1.45, 95% CI: 1.06-1.99 and OR: 1.42, 95% CI: 1.03-1.96, respectively) compared to the vaccinated group. CONCLUSION Despite effective vaccination, fully vaccinated patients may be admitted to ICU because of disease severity. Unvaccinated patients were younger and had fewer comorbid conditions. Unvaccinated patients have an increased risk of 28-day mortality when adjusted for gender and CCI.
Collapse
Affiliation(s)
- Süleyman Yıldırım
- University of Health Sciences Turkey, Dr. Suat Seren Chest Disease and Surgery Training and Research Hospital, Intensive Care Unit, İzmir, Turkey.
| | | | - Özcan Alpdoğan
- University of Health Sciences, İzmir Tepecik Training and Research Hospital, Intensive Care Unit, İzmir, Turkey
| | | | - Barış Yılmaz
- University of Health Sciences, Istanbul Sureyyapasa Chest Diseases and Thoracic Surgery Training and Research Hospital, İstanbul, Turkey
| | - Gül Erdal Dönmez
- University of Health Sciences, Istanbul Sureyyapasa Chest Diseases and Thoracic Surgery Training and Research Hospital, İstanbul, Turkey
| | - Aykut Sarıtaş
- University of Health Sciences, İzmir Tepecik Training and Research Hospital, Intensive Care Unit, İzmir, Turkey
| | - Necati Gökmen
- Dokuz Eylül University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Division of Intensive Care, İzmir, Turkey
| | - Begüm Ergan
- Dokuz Eylül University, Faculty of Medicine, Department of Chest Disease, Division of Intensive Care, İzmir, Turkey
| | - Vecihe Bayrak
- Dokuz Eylül University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Division of Intensive Care, İzmir, Turkey
| | - Mehmet Nuri Yakar
- Dokuz Eylül University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Division of Intensive Care, İzmir, Turkey
| | - Özgür Kılıç
- On Dokuz Mayıs University, Faculty of Medicine, Department of Internal Medicine, Division of Intensive Care, Samsun, Turkey
| | - Ahmet Kılınç
- On Dokuz Mayıs University, Faculty of Medicine, Department of Internal Medicine, Division of Intensive Care, Samsun, Turkey
| | - Saba Saygılı
- University of Health Sciences Turkey, Dr. Suat Seren Chest Disease and Surgery Training and Research Hospital, Intensive Care Unit, İzmir, Turkey
| | - Ümmügülsüm Gaygısız
- Gazi University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Division of Intensive Care, Ankara, Turkey
| | - Kaniye Aydın
- Çukurova University, Faculty of Medicine, Department of Internal Medicine, Division of Intensive Care Medicine, Adana, Turkey
| | - Aysun Özel Yeşilyurt
- Çukurova University, Faculty of Medicine, Department of Internal Medicine, Division of Intensive Care Medicine, Adana, Turkey
| | - Hayriye Cankar Dal
- University of Health Sciences, Ankara City Hospital, Intensive Care Unit, Ankara, Turkey
| | - Çilem Bayındır Dicle
- University of Health Sciences, Ankara City Hospital, Intensive Care Unit, Ankara, Turkey
| | - Sema Turan
- University of Health Sciences, Ankara City Hospital, Intensive Care Unit, Ankara, Turkey
| | - Songül Binay
- University of Health Sciences, Ankara Dr Abdurrahman Yurtaslan Oncology Training and Research Hospital, Intensive Care Unit, Ankara, Turkey
| | - Metin Yarıcı
- Dışkapı Yıldırım Beyazıt Training and Research Hospital, Department of General Surgery, Intensive Care Unit, Ankara, Turkey
| | - Fatma Yıldırım
- University of Health Sciences, Dışkapı Yıldırım Beyazıt Research and Education Hospital, Department of Chest Diseases, Pulmonary Intensive Care Unit, Ankara, Turkey
| | - Pervin Hancı
- Trakya University Faculty of Medicine, Department of Pulmonology, Division of Intensive Care, Edirne, Turkey
| | - Mehmet Turan İnal
- Trakya University Faculty of Medicine, Department of Pulmonology, Division of Intensive Care, Edirne, Turkey
| | - Türkay Akbaş
- Düzce University, Faculty of Medicine, Department of Internal Medicine, Division of Intensive Care, Düzce, Turkey
| | - Selin Eyüpoğlu
- Giresun Training and Research Hospital, Intensive Care Unit, Giresun, Turkey
| | - Tuna Albayrak
- Giresun University, Giresun Training and Research Hospital, Department of Anesthesiology and Reanimation, Giresun, Turkey
| | - Gamze Koçak
- Mersin City Hospital, Intensive Care Unit, Mersin, Turkey
| | - Tümay Çakır
- Muğla Training and Research Hospital, Intensive Care Unit, Muğla, Turkey
| | - Recep Civan Yüksel
- Ministry of Health, Kayseri City Hospital, Intensive Care Unit, Kayseri, Turkey
| | - Ali Sarı
- Gaziantep Abdulkadir Yüksel State Hospital, Intensive Care Unit, Gaziantep, Turkey
| | - Murat Güneş
- Gümüşhane State Hospital, Intensive Care Unit, Gümüşhane, Turkey
| | - Oral Menteş
- Gülhane Training and Research Hospital, Intensive Care Unit, Ankara, Turkey
| | - H Levent Yamanel
- Gülhane Training and Research Hospital, Intensive Care Unit, Ankara, Turkey
| | - Cenk Kirakli
- University of Health Sciences Turkey, Dr. Suat Seren Chest Disease and Surgery Training and Research Hospital, Intensive Care Unit, İzmir, Turkey
| |
Collapse
|
32
|
Guseman AJ, Rennick LJ, Nambulli S, Roy CN, Martinez DR, Yang DT, Bhinderwala F, Vergara S, Schaefer A, Baric RS, Ambrose Z, Duprex WP, Gronenborn AM. Targeting spike glycans to inhibit SARS-CoV2 viral entry. Proc Natl Acad Sci U S A 2023; 120:e2301518120. [PMID: 37695910 PMCID: PMC10515186 DOI: 10.1073/pnas.2301518120] [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: 01/27/2023] [Accepted: 07/08/2023] [Indexed: 09/13/2023] Open
Abstract
SARS-CoV-2 spike harbors glycans which function as ligands for lectins. Therefore, it should be possible to exploit lectins to target SARS-CoV-2 and inhibit cellular entry by binding glycans on the spike protein. Burkholderia oklahomensis agglutinin (BOA) is an antiviral lectin that interacts with viral glycoproteins via N-linked high mannose glycans. Here, we show that BOA binds to the spike protein and is a potent inhibitor of SARS-CoV-2 viral entry at nanomolar concentrations. Using a variety of biophysical approaches, we demonstrate that the interaction is avidity driven and that BOA cross-links the spike protein into soluble aggregates. Furthermore, using virus neutralization assays, we demonstrate that BOA effectively inhibits all tested variants of concern as well as SARS-CoV 2003, establishing that multivalent glycan-targeting molecules have the potential to act as pan-coronavirus inhibitors.
Collapse
Affiliation(s)
- Alex J. Guseman
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15261
| | - Linda J. Rennick
- Center for Vaccine Research and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
| | - Sham Nambulli
- Center for Vaccine Research and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
| | - Chandra N. Roy
- Center for Vaccine Research and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
| | - David R. Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Darian T. Yang
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15261
| | - Fatema Bhinderwala
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15261
| | - Sandra Vergara
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15261
| | - Alexandra Schaefer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Zandrea Ambrose
- Center for Vaccine Research and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
| | - W. Paul Duprex
- Center for Vaccine Research and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
| | - Angela M. Gronenborn
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15261
| |
Collapse
|
33
|
Sayedahmed EE, Araújo MV, Silva-Pereira TT, Chothe SK, Elkashif A, Alhashimi M, Wang WC, Santos AP, Nair MS, Gontu A, Nissly R, Francisco de Souza Filho A, Tavares MS, Ayupe MC, Salgado CL, Donizetti de Oliveira Candido É, Leal Oliveira DB, Durigon EL, Heinemann MB, Morais da Fonseca D, Jagannath C, Sá Guimarães AM, Kuchipudi SV, Mittal SK. Impact of an autophagy-inducing peptide on immunogenicity and protection efficacy of an adenovirus-vectored SARS-CoV-2 vaccine. Mol Ther Methods Clin Dev 2023; 30:194-207. [PMID: 37502665 PMCID: PMC10299838 DOI: 10.1016/j.omtm.2023.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023]
Abstract
Because of continual generation of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is critical to design the next generation of vaccines to combat the threat posed by SARS-CoV-2 variants. We developed human adenovirus (HAd) vector-based vaccines (HAd-Spike/C5 and HAd-Spike) that express the whole Spike (S) protein of SARS-CoV-2 with or without autophagy-inducing peptide C5 (AIP-C5), respectively. Mice or golden Syrian hamsters immunized intranasally (i.n.) with HAd-Spike/C5 induced similar levels of S-specific humoral immune responses and significantly higher levels of S-specific cell-mediated immune (CMI) responses compared with HAd-Spike vaccinated groups. These results indicated that inclusion of AIP-C5 induced enhanced S-specific CMI responses and similar levels of virus-neutralizing titers against SARS-CoV-2 variants. To investigate the protection efficacy, golden Syrian hamsters immunized i.n. either with HAd-Spike/C5 or HAd-Spike were challenged with SARS-CoV-2. The lungs and nasal turbinates were collected 3, 5, 7, and 14 days post challenge. Significant reductions in morbidity, virus titers, and lung histopathological scores were observed in immunized groups compared with the mock- or empty vector-inoculated groups. Overall, slightly better protection was seen in the HAd-Spike/C5 group compared with the HAd-Spike group.
Collapse
Affiliation(s)
- Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Marcelo Valdemir Araújo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Butantan Institute, São Paulo, Brazil
| | - Taiana Tainá Silva-Pereira
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Shubhada K. Chothe
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Ahmed Elkashif
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Marwa Alhashimi
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Wen-Chien Wang
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Andrea P. Santos
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Meera Surendran Nair
- Department of Veterinary and Biomedical Sciences, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Abhinay Gontu
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Ruth Nissly
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | | | - Mariana Silva Tavares
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marina Caçador Ayupe
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Caio Loureiro Salgado
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Edison Luiz Durigon
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcos Bryan Heinemann
- Department of Preventive Veterinary Medicine and Animal Health, College of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - Denise Morais da Fonseca
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Center for Infectious Diseases and Translational Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ana Marcia Sá Guimarães
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Suresh V. Kuchipudi
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
34
|
Ren P, Li H, Nie T, Jian X, Yu C, Li J, Su H, Zhang X, Li S, Yang X, Peng C, Yin Y, Zhang L, Xu Y, Liu H, Bai F. Discovery and Mechanism Study of SARS-CoV-2 3C-like Protease Inhibitors with a New Reactive Group. J Med Chem 2023; 66:12266-12283. [PMID: 37594952 DOI: 10.1021/acs.jmedchem.3c00818] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2023]
Abstract
3CLpro is an attractive target for the treatment of COVID-19. Using the scaffold hopping strategy, we identified a potent inhibitor of 3CLpro (3a) that contains a thiocyanate moiety as a novel warhead that can form a covalent bond with Cys145 of the protein. Tandem mass spectrometry (MS/MS) and X-ray crystallography confirmed the mechanism of covalent formation between 3a and the protein in its catalytic pocket. Moreover, several analogues of compound 3a were designed and synthesized. Among them, compound 3h shows the best inhibition of 3CLpro with an IC50 of 0.322 μM and a kinact/Ki value of 1669.34 M-1 s-1, and it exhibits good target selectivity for 3CLpro against host proteases. Compound 3c inhibits SARS-CoV-2 in Vero E6 cells (EC50 = 2.499 μM) with low cytotoxicity (CC50 > 200 μM). These studies provide ideas and insights to explore and develop new 3CLpro inhibitors in the future.
Collapse
Affiliation(s)
- Pengxuan Ren
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Hui Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tianqing Nie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoqin Jian
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Changyue Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haixia Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xianglei Zhang
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Shiwei Li
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Xin Yang
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai 201210, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai 201210, China
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yechun Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Hong Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Fang Bai
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| |
Collapse
|
35
|
Monteiro HS, Lima Neto AS, Kahn R, Sousa GS, Carmona HA, Andrade JS, Castro MC. Impact of CoronaVac on Covid-19 outcomes of elderly adults in a large and socially unequal Brazilian city: A target trial emulation study. Vaccine 2023; 41:5742-5751. [PMID: 37573202 DOI: 10.1016/j.vaccine.2023.07.065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Although CoronaVac was the only Covid-19 vaccine adopted in the first months of the Brazilian vaccination campaign, randomized clinical trials to evaluate its efficacy in elderly adults were limited. In this study, we use routinely collected surveillance and SARS-CoV-2 vaccination and testing data comprising the population of the fifth largest city of Brazil to evaluate the effectiveness of CoronaVac in adults 60+ years old against severe outcomes. METHODS Using large observational databases on vaccination and surveillance data from the city of Fortaleza, Brazil, we defined a retrospective cohort including 324,302 eligible adults aged ≥60 years to evaluate the effectiveness of the CoronaVac vaccine. The cohort included individuals vaccinated between January 21, 2021, and August 31, 2021, who were matched with unvaccinated persons at the time of rollout following a 1:1 ratio according to baseline covariates of age, sex, and Human Development Index of the neighborhood of residence. Only Covid-19-related severe outcomes were included in the analysis: hospitalization, ICU admission, and death. Vaccine effectiveness for each outcome was calculated by using the risk ratio between the two groups, with the risk obtained by the Kaplan-Meier estimator. RESULTS We obtained 62,643 matched pairs for assessing the effectiveness of the two-dose regimen of CoronaVac. The demographic profile of the matched population was statistically representative of the population of Fortaleza. Using the cumulative incidence as the risk associated with each group, starting at day 14 since the receipt of the second dose, we found an 82.3 % (95 % CI 66.3-93.9) effectiveness against Covid-19-related death, 68.4 % (95 % CI 42.3-86.4) against ICU admission, and 55.8 % (95 % CI 42.7-68.3) against hospital admission. CONCLUSIONS Our results show that, despite critical delays in vaccine delivery and limited evidence in efficacy trial estimates, CoronaVac contributed to preventing deaths and severe morbidity due to Covid-19 in elderly adults.
Collapse
Affiliation(s)
- Higor S Monteiro
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, Ceará, Brazil; Secretaria Municipal de Saúde de Fortaleza (SMS-Fortaleza), Fortaleza, Ceará, Brazil.
| | - Antonio S Lima Neto
- Secretaria Municipal de Saúde de Fortaleza (SMS-Fortaleza), Fortaleza, Ceará, Brazil; Centro de Ciências da Saúde, Universidade de Fortaleza (UNIFOR), Fortaleza, Ceará, Brazil
| | - Rebecca Kahn
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | - Geziel S Sousa
- Secretaria Municipal de Saúde de Fortaleza (SMS-Fortaleza), Fortaleza, Ceará, Brazil; Centro de Ciências da Saúde, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Humberto A Carmona
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, Ceará, Brazil.
| | - José S Andrade
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, Ceará, Brazil.
| | - Marcia C Castro
- Department of Global Health and Population, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
36
|
Zhang Y, Li Q, Liu N, Hu J, Lin X, Huang M, Wei Y, Qi X, Chen X. Secure reversal of immune evasion from refractory NSCLC and highly contagious CoV-2 mutants by using 3D-engineered multifunctional biologics. Bioeng Transl Med 2023; 8:e10554. [PMID: 37693048 PMCID: PMC10487317 DOI: 10.1002/btm2.10554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 04/22/2023] [Accepted: 05/16/2023] [Indexed: 09/12/2023] Open
Abstract
There is an imperative choice to develop a secure feasible strategy to address evasion dynamics of refractory tumors and SARS-CoV-2-variants, while stem cell-based protocol may be more reliable as its unique ability for resetting multifunctional immunity to address progressive tumor and the constantly-evolving virus. In this study, spheroid-embryonoid stem cells from mature somatic cells were engineered as multifunctional biologics (3D-E/BSC) and inoculated in senile rhesus to identify secure potential against immune-evasion from viral-variants. Meanwhile, a cohort of eligible patients with stage IV NSCLC were approved for phase I clinical trials. Subsequently, long-lasting security and efficacy were validated by primate and clinical trials (p < 0.01) in that it could not only stimulate serological immunity, but also reset core immunity for hosts to address variant evasion after 3D-E/BSC withdrawal. Particularly, illustrated by single-cell evolving trajectory, 3D-E/BSC had securely reset senile thymus of aging hosts to remodel core immunity by rearranging naive rhythm to evolve TRGC2+/JCHAIN+NKT clusters to abolish tumoral and viral evasion dynamics with path-feedbacks of NSCLC and COVID-19 simultaneously activated, leading to continuous blockade of breakthrough infection of viral-mutants and long-term survival in one-third of terminal patients without adjuvant required. Our study may pioneer a practical multifunctional strategy to eliminate evasion of SARS-CoV-2 variants and refractory NSCLC so as for victims to restart a new life-equation.
Collapse
Affiliation(s)
- Yanna Zhang
- Department of Blood Transfusion, Sichuan Provincial People’s HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Qian Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Nanxi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Jianchuan Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xiaojuan Lin
- Department of Gynecology & Obstetrics, West China Second HospitalSichuan UniversityChengduChina
| | - Meijuan Huang
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Yuquan Wei
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Xiaorong Qi
- Department of Gynecology & Obstetrics, West China Second HospitalSichuan UniversityChengduChina
| | - Xiancheng Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
37
|
Kambe R, Sato M, Uehara D, Iizuka Y, Kakizaki S. Prolonged SARS-CoV-2 infection during obinutuzumab and bendamustine treatment for follicular lymphoma: A case report. Clin Case Rep 2023; 11:e7861. [PMID: 37649899 PMCID: PMC10462774 DOI: 10.1002/ccr3.7861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023] Open
Abstract
Key Clinical Message SARS-CoV-2 infection has been associated with a prolonged course and a poor prognosis in patients who receive anti-CD20 antibodies. However, there are no established treatments for such patients. Serial changes in the SARS-CoV-2 antigen titer during the clinical course and treatment strategies for immunosuppressed patients are discussed. Abstract We report a case of prolonged SARS-CoV-2 infection during obinutuzumab and bendamustine treatment for follicular lymphoma. Four years previously, the patient had been diagnosed with follicular lymphoma (Stage IIIA, Grade 2). She received several chemotherapy regimens, including rituximab and radiation therapy. Although these therapies achieved complete response temporally, they did not continue and recurred at 8 months before. Obinutuzumab and bendamustine therapy was selected, and she received five courses of obinutuzumab and bendamustine. She also received a SARS-CoV-2 mRNA vaccine two times. Although she did not have any symptoms, a routine check-up just before the 6th course of obinutuzumab and bendamustine revealed SARS-CoV-2 infection. Because she was immunosuppressed and was considered to be at high risk for the exacerbation of her disease, molnupiravir was immediately administered, and her SARS-CoV-2 antigen decreased. However, it was not completely cleared and flared-up at 6 weeks, with symptoms of COVID-19 appearing. Despite intensive treatment for SARS-CoV-2 infection, including remdesivir, baricitinib, tocilizumab and intravenous immunoglobulin, her SARS-CoV-2 antigen titer never became negative, and she finally died of respiratory failure caused by prolonged SARS-CoV-2 infection. Serial changes in the SARS-CoV-2 antigen titer during the clinical course and treatment strategies for immunosuppressed patients are discussed.
Collapse
Affiliation(s)
- Ryosuke Kambe
- Department of General Internal MedicineNational Hospital Organization Takasaki General Medical CenterTakasakiGunmaJapan
| | - Masamichi Sato
- Department of General Internal MedicineNational Hospital Organization Takasaki General Medical CenterTakasakiGunmaJapan
| | - Daisuke Uehara
- Department of General Internal MedicineNational Hospital Organization Takasaki General Medical CenterTakasakiGunmaJapan
| | - Yutaka Iizuka
- Department of General Internal MedicineNational Hospital Organization Takasaki General Medical CenterTakasakiGunmaJapan
| | - Satoru Kakizaki
- Department of Clinical ResearchNational Hospital Organization Takasaki General Medical CenterTakasakiGunmaJapan
| |
Collapse
|
38
|
Montes-Cobos E, Bastos VC, Monteiro C, de Freitas JC, Fernandes HD, Constancio CS, Rodrigues DA, Gama AM, Vidal VM, Alves LS, Zalcberg-Renault L, de Lira GS, Ota VA, Caloba C, Conde L, Leitão IC, Tanuri A, Ferreira OD, Pereira RM, Vale AM, Castiñeiras TM, Kaiserlian D, Echevarria-Lima J, Bozza MT. Oligosymptomatic long-term carriers of SARS-CoV-2 display impaired innate resistance but increased high-affinity anti-spike antibodies. iScience 2023; 26:107219. [PMID: 37529320 PMCID: PMC10300054 DOI: 10.1016/j.isci.2023.107219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/05/2023] [Accepted: 06/22/2023] [Indexed: 08/03/2023] Open
Abstract
The vast spectrum of clinical features of COVID-19 keeps challenging scientists and clinicians. Low resistance to infection might result in long-term viral persistence, but the underlying mechanisms remain unclear. Here, we studied the immune response of immunocompetent COVID-19 patients with prolonged SARS-CoV-2 infection by immunophenotyping, cytokine and serological analysis. Despite viral loads and symptoms comparable to regular mildly symptomatic patients, long-term carriers displayed weaker systemic IFN-I responses and fewer circulating pDCs and NK cells at disease onset. Type 1 cytokines remained low, while type-3 cytokines were in turn enhanced. Of interest, we observed no defects in antigen-specific cytotoxic T cell responses, and circulating antibodies displayed higher affinity against different variants of SARS-CoV-2 Spike protein in these patients. The identification of distinct immune responses in long-term carriers adds up to our understanding of essential host protective mechanisms to ensure tissue damage control despite prolonged viral infection.
Collapse
Affiliation(s)
- Elena Montes-Cobos
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victoria C. Bastos
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice Monteiro
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João C.R. de Freitas
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heiny D.P. Fernandes
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice S. Constancio
- Laboratório de Imunologia Básica e Aplicada, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle A.S. Rodrigues
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andreza M.D.S. Gama
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinicius M. Vidal
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia S. Alves
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laura Zalcberg-Renault
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme S. de Lira
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor A. Ota
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina Caloba
- Laboratório de Imunologia Molecular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isabela C. Leitão
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amilcar Tanuri
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Orlando D.C. Ferreira
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata M. Pereira
- Laboratório de Imunologia Molecular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André M. Vale
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Terezinha M. Castiñeiras
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dominique Kaiserlian
- INSERM U1060, Université Claude Bernard Lyon 1, Centre hospitalier Lyon-Sud, Pierre-Benite, France
| | - Juliana Echevarria-Lima
- Laboratório de Imunologia Básica e Aplicada, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
39
|
Wei J, Alfajaro MM, Cai WL, Graziano VR, Strine MS, Filler RB, Biering SB, Sarnik SA, Patel S, Menasche BL, Compton SR, Konermann S, Hsu PD, Orchard RC, Yan Q, Wilen CB. The KDM6A-KMT2D-p300 axis regulates susceptibility to diverse coronaviruses by mediating viral receptor expression. PLoS Pathog 2023; 19:e1011351. [PMID: 37410700 PMCID: PMC10325096 DOI: 10.1371/journal.ppat.1011351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/10/2023] [Indexed: 07/08/2023] Open
Abstract
Identification of host determinants of coronavirus infection informs mechanisms of pathogenesis and may provide novel therapeutic targets. Here, we demonstrate that the histone demethylase KDM6A promotes infection of diverse coronaviruses, including SARS-CoV, SARS-CoV-2, MERS-CoV and mouse hepatitis virus (MHV) in a demethylase activity-independent manner. Mechanistic studies reveal that KDM6A promotes viral entry by regulating expression of multiple coronavirus receptors, including ACE2, DPP4 and Ceacam1. Importantly, the TPR domain of KDM6A is required for recruitment of the histone methyltransferase KMT2D and histone deacetylase p300. Together this KDM6A-KMT2D-p300 complex localizes to the proximal and distal enhancers of ACE2 and regulates receptor expression. Notably, small molecule inhibition of p300 catalytic activity abrogates ACE2 and DPP4 expression and confers resistance to all major SARS-CoV-2 variants and MERS-CoV in primary human airway and intestinal epithelial cells. These data highlight the role for KDM6A-KMT2D-p300 complex activities in conferring diverse coronaviruses susceptibility and reveal a potential pan-coronavirus therapeutic target to combat current and emerging coronaviruses. One Sentence Summary: The KDM6A/KMT2D/EP300 axis promotes expression of multiple viral receptors and represents a potential drug target for diverse coronaviruses.
Collapse
Affiliation(s)
- Jin Wei
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Mia Madel Alfajaro
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Wesley L. Cai
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Vincent R. Graziano
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Madison S. Strine
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Renata B. Filler
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Sylvia A. Sarnik
- University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Sonam Patel
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Bridget L. Menasche
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Susan R. Compton
- Department of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Silvana Konermann
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, USA
- Arc Institute, Palo Alto, California, United States of America
| | - Patrick D. Hsu
- Arc Institute, Palo Alto, California, United States of America
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, United States of America
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, California, United States of America
- Center for Computational Biology, University of California, Berkeley, California, United States of America
| | - Robert C. Orchard
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Craig B. Wilen
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
40
|
Cheng FJ, Ho CY, Li TS, Chen Y, Yeh YL, Wei YL, Huynh TK, Chen BR, Ko HY, Hsueh CS, Tan M, Wu YC, Huang HC, Tang CH, Chen CH, Tu CY, Huang WC. Umbelliferone and eriodictyol suppress the cellular entry of SARS-CoV-2. Cell Biosci 2023; 13:118. [PMID: 37381062 DOI: 10.1186/s13578-023-01070-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 06/13/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Artemisia argyi (A. argyi), also called Chinese mugwort, has been widely used to control pandemic diseases for thousands of years since ancient China due to its anti-microbial infection, anti-allergy, and anti-inflammation activities. Therefore, the potential of A. argyi and its constituents in reducing the infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was investigated in this study. RESULTS Among the phytochemicals in A. argyi, eriodictyol and umbelliferone were identified to target transmembrane serine protease 2 (TMPRSS2) and angiotensin-converting enzyme 2 (ACE2) proteins, the essential factors for the cellular entry of SARS-CoV-2, in both FRET-based enzymatic assays and molecular docking analyses. These two ingredients of A. argyi suppressed the infection of ACE2-expressed HEK-293 T cells with lentiviral-based pseudo-particles (Vpp) expressing wild-type and variants of SARS-CoV-2 spike (S) protein (SARS-CoV-2 S-Vpp) via interrupting the interaction between S protein and cellular receptor ACE2 and reducing the expressions of ACE2 and TMPRSS2. Oral administration with umbelliferone efficiently prevented the SARS-CoV-2 S-Vpp-induced inflammation in the lung tissues of BALB/c mice. CONCLUSIONS Eriodictyol and umbelliferone, the phytochemicals of Artemisia argyi, potentially suppress the cellular entry of SARS-CoV-2 by preventing the protein binding activity of the S protein to ACE2.
Collapse
Affiliation(s)
- Fang-Ju Cheng
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
- School of Medicine, China Medical University, Taichung, 404, Taiwan
| | - Chien-Yi Ho
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 404, Taiwan
- Division of Family Medicine, Physical Examination Center, China Medical University Hsinchu Hospital, Hsinchu, 302, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, 302, Taiwan
| | - Tzong-Shiun Li
- Department of Plastic Surgery, and Innovation Research Center, Show Chwan Memorial Hospital, Changhua, 500, Taiwan
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 402, Taiwan
| | - Yeh Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, 402, Taiwan
| | - Yi-Lun Yeh
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Ya-Ling Wei
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Thanh Kieu Huynh
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Bo-Rong Chen
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
- School of Medicine, China Medical University, Taichung, 404, Taiwan
| | - Hung-Yu Ko
- Cognitive Science, University of California San Diego, San Diego, CA, 92093, USA
| | - Chen-Si Hsueh
- Taichung Girls' Senior High School, Taichung, 403, Taiwan
| | - Ming Tan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 404, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, 404, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, 413, Taiwan
| | - Hui-Chi Huang
- Department of Chinese Pharmaceutical Science and Chinese Medicine Resources, China Medical University, Taichung, 404, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, 404, Taiwan
| | - Chia-Hung Chen
- School of Medicine, China Medical University, Taichung, 404, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Department of Respiratory Therapy, China Medical University, Taichung, 404, Taiwan
| | - Chih-Yen Tu
- School of Medicine, China Medical University, Taichung, 404, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Department of Respiratory Therapy, China Medical University, Taichung, 404, Taiwan
| | - Wei-Chien Huang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, 302, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, 413, Taiwan.
- Drug Development Center, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
41
|
Zhang X, Zhou Z. The Mechanism of bnAb Production and Its Application in Mutable Virus Broad-Spectrum Vaccines: Inspiration from HIV-1 Broad Neutralization Research. Vaccines (Basel) 2023; 11:1143. [PMID: 37514959 PMCID: PMC10384589 DOI: 10.3390/vaccines11071143] [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: 05/18/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Elite controllers among HIV-1-infected individuals have demonstrated a stronger ability to control the viral load in their bodies. Scientists have isolated antibodies with strong neutralizing ability from these individuals, which can neutralize HIV-1 variations; these are known as broadly neutralizing antibodies. The nucleic acid of some viruses will constantly mutate during replication (such as SARS-CoV-2), which will reduce the protective ability of the corresponding vaccines. The immune escape caused by this mutation is the most severe challenge faced by humans in the battle against the virus. Therefore, developing broad-spectrum vaccines that can induce broadly neutralizing antibodies against various viruses and their mutated strains is the best way to combat virus mutations. Exploring the mechanism by which the human immune system produces broadly neutralizing antibodies and its induction strategies is crucial in the design process of broad-spectrum vaccines.
Collapse
Affiliation(s)
- Xinyu Zhang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, No. 31 Huatuo Street, Daxing District, Beijing 102629, China
- College of Life Science, Jilin University, Changchun 130012, China
| | - Zehua Zhou
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
42
|
Gozzi N, Chinazzi M, Dean NE, Longini IM, Halloran ME, Perra N, Vespignani A. Estimating the impact of COVID-19 vaccine inequities: a modeling study. Nat Commun 2023; 14:3272. [PMID: 37277329 DOI: 10.1038/s41467-023-39098-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/25/2023] [Indexed: 06/07/2023] Open
Abstract
Access to COVID-19 vaccines on the global scale has been drastically hindered by structural socio-economic disparities. Here, we develop a data-driven, age-stratified epidemic model to evaluate the effects of COVID-19 vaccine inequities in twenty lower middle and low income countries (LMIC) selected from all WHO regions. We investigate and quantify the potential effects of higher or earlier doses availability. In doing so, we focus on the crucial initial months of vaccine distribution and administration, exploring counterfactual scenarios where we assume the same per capita daily vaccination rate reported in selected high income countries. We estimate that more than 50% of deaths (min-max range: [54-94%]) that occurred in the analyzed countries could have been averted. We further consider scenarios where LMIC had similarly early access to vaccine doses as high income countries. Even without increasing the number of doses, we estimate an important fraction of deaths (min-max range: [6-50%]) could have been averted. In the absence of the availability of high-income countries, the model suggests that additional non-pharmaceutical interventions inducing a considerable relative decrease of transmissibility (min-max range: [15-70%]) would have been required to offset the lack of vaccines. Overall, our results quantify the negative impacts of vaccine inequities and underscore the need for intensified global efforts devoted to provide faster access to vaccine programs in low and lower-middle-income countries.
Collapse
Affiliation(s)
- Nicolò Gozzi
- Networks and Urban Systems Centre, University of Greenwich, London, UK
- ISI Foundation, Turin, Italy
| | - Matteo Chinazzi
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, MA, USA
| | - Natalie E Dean
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Ira M Longini
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
| | - M Elizabeth Halloran
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Nicola Perra
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, MA, USA.
- School of Mathematical Sciences, Queen Mary University, London, UK.
| | - Alessandro Vespignani
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, MA, USA
| |
Collapse
|
43
|
Cho A, Mendenhall E, Griffith DM. Power, place, and access: Why history is at the center of black D.C. residents of wards 7 and 8 decisions to receive the COVID-19 vaccine. SSM. QUALITATIVE RESEARCH IN HEALTH 2023; 3:100270. [PMID: 37073370 PMCID: PMC10101486 DOI: 10.1016/j.ssmqr.2023.100270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/26/2023] [Accepted: 04/08/2023] [Indexed: 04/20/2023]
Abstract
Vaccines have played an essential role in curbing case and mortality rates due to SARS-CoV-2 in the United Sates. Still, many communities display high rates of unwillingness or inability to get a COVID-19 vaccine, limiting overall vaccination efforts and contributing to viral spread. Black Americans have expressed skepticism towards vaccines because of limited access to the technology, mistrust in its safety and efficacy, and a lack of confidence in the healthcare authorities that distribute it. This article investigates how Black residents of Wards 7 and 8 in Washington, D.C. thought about COVID-19 vaccination and why or why not they decided to vaccinate. These Wards' vaccination rates were markedly lower than those from Wards 1-6, which have substantially higher populations of White residents, affluence, access, and resources. This study involved 31 interviews with Ward 7 and 8 residents recruited through snowball sampling. We found that residents navigated the dual perceived risks of coronavirus infection and vaccination through three key frames: their relationship to their place or location, their desires to maintain autonomy over their health, and their abilities to access COVID-19 vaccines. This case study advances knowledge of vaccine utilization among marginalized communities, and how this phenomenon varies depending on local social, cultural, and political dynamics. Moreover, this research has implications for vaccine rollout efforts and the D.C. health system, as it reveals gaps in confidence and care that undermine health outcomes for Black residents.
Collapse
Affiliation(s)
- Allison Cho
- Edmund A Walsh School of Foreign Service, Georgetown University, 3700 O St. NW, Washington, DC, 20057, USA
| | - Emily Mendenhall
- Edmund A Walsh School of Foreign Service, Georgetown University, 3700 O St. NW, Washington, DC, 20057, USA
| | - Derek M Griffith
- Racial Justice Institute and School of Health, Georgetown University, 3700 O St. NW, Washington, DC, 20057, USA
| |
Collapse
|
44
|
de Bock E, Filipe MD, Herman ES, Pronk A, Boerma D, Heikens JT, Verheijen PM, Vriens MR, Richir MC. Risk factors of postoperative intensive care unit admission during the COVID-19 pandemic: A multicentre retrospective cohort study. INTERNATIONAL JOURNAL OF SURGERY OPEN 2023; 55:100620. [PMID: 37163195 PMCID: PMC10159662 DOI: 10.1016/j.ijso.2023.100620] [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: 09/19/2022] [Revised: 04/12/2023] [Accepted: 04/30/2023] [Indexed: 05/11/2023]
Abstract
Background During the Coronavirus disease 2019 (COVID-19) pandemic, intensive care unit (ICU) capacity was scarce. Since surgical patients also require ICU admission, determining which factors lead to an increased risk of postoperative ICU admission is essential. This study aims to determine which factors led to an increased risk of unplanned postoperative ICU admission during the COVID-19 pandemic. Methods This multicentre retrospective cohort study investigated all patients who underwent surgery between 9 March 2020 and 30 June 2020. The primary endpoint was the number of surgical patients requiring postoperative ICU admission. The secondary endpoint was to determine factors leading to an increased risk of unplanned postoperative ICU admission, calculated by multivariate analysis with odds ratios (OR's) and 95% confidence (CI) intervals. Results One hundred eighty-five (4.6%) of the 4051 included patients required unplanned postoperative ICU admission. COVID-19 positive patients were at an increased risk of being admitted to the ICU compared to COVID-19 negative (OR 3.14; 95% CI 1.06-9.33; p = 0.040) and untested patients (OR 0.48; 95% CI 0.32-0.70; p = 0.001). Other predictors were male gender (OR 1.36; 95% CI 1.02-1.82; p = 0.046), body mass index (BMI) (OR 1.05; 95% CI 1.02-1.08; p = 0.001), surgical urgency and surgical discipline. Conclusion A confirmed COVID-19 infection, male gender, elevated BMI, surgical urgency, and surgical discipline were independent factors for an increased risk of unplanned postoperative ICU admission. In the event of similar pandemics, postponing surgery in patients with an increased risk of postoperative ICU admission may be considered.
Collapse
Affiliation(s)
- Ellen de Bock
- Department of Surgery, Cancer Centre, University Medical Centre Utrecht, the Netherlands
| | - Mando D Filipe
- Department of Surgery, Cancer Centre, University Medical Centre Utrecht, the Netherlands
| | - Eline S Herman
- Department of Surgery, Diakonessenhuis, Utrecht, the Netherlands
| | - Apollo Pronk
- Department of Surgery, Diakonessenhuis, Utrecht, the Netherlands
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Joost T Heikens
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
- Department of Surgery, Rivierenland Hospital, Tiel, the Netherlands
| | - Paul M Verheijen
- Department of Surgery, Meander Medical Centre, Amersfoort, the Netherlands
| | - Menno R Vriens
- Department of Surgery, Cancer Centre, University Medical Centre Utrecht, the Netherlands
| | - Milan C Richir
- Department of Surgery, Cancer Centre, University Medical Centre Utrecht, the Netherlands
| |
Collapse
|
45
|
Radcliffe C, Malinis M, Azar MM. Antiviral Treatment of Coronavirus Disease-2019 Pneumonia. Clin Chest Med 2023; 44:279-297. [PMID: 37085220 PMCID: PMC9701636 DOI: 10.1016/j.ccm.2022.11.008] [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] [Indexed: 11/29/2022]
Abstract
Direct acting antivirals and monoclonal antibodies reduce morbidity and mortality associated with severe acute respiratory syndrome coronavirus 2 infection. Persons at higher risk for disease progression and hospitalized patients with coronavirus disease-2019 (COVID-19) benefit most from available therapies. Following an emphasis on inpatient treatment of COVID-19 during the early pandemic, several therapeutic options were developed for outpatients with COVID-19. Additional clinical trials and real-world studies are needed to keep pace with the evolving pandemic.
Collapse
Affiliation(s)
- Christopher Radcliffe
- Section of Infectious Diseases, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Maricar Malinis
- Section of Infectious Diseases, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Marwan M Azar
- Section of Infectious Diseases, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
46
|
Abduljalil JM, Elghareib AM, Samir A, Ezat AA, Elfiky AA. How helpful were molecular dynamics simulations in shaping our understanding of SARS-CoV-2 spike protein dynamics? Int J Biol Macromol 2023:125153. [PMID: 37268078 DOI: 10.1016/j.ijbiomac.2023.125153] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/22/2023] [Accepted: 05/27/2023] [Indexed: 06/04/2023]
Abstract
The SARS-CoV-2 spike protein (S) represents an important viral component that is required for successful viral infection in humans owing to its essential role in recognition of and entry to host cells. The spike is also an appealing target for drug designers who develop vaccines and antivirals. This article is important as it summarizes how molecular simulations successfully shaped our understanding of spike conformational behavior and its role in viral infection. MD simulations found that the higher affinity of SARS-CoV-2-S to ACE2 is linked to its unique residues that add extra electrostatic and van der Waal interactions in comparison to the SARS-CoV S. This illustrates the spread potential of the pandemic SARS-CoV-2 relative to the epidemic SARS-CoV. Different mutations at the S-ACE2 interface, which is believed to increase the transmission of the new variants, affected the behavior and binding interactions in different simulations. The contributions of glycans to the opening of S were revealed via simulations. The immune evasion of S was linked to the spatial distribution of glycans. This help the virus to escape the immune system recognition. This article is important as it summarizes how molecular simulations successfully shaped our understanding of spike conformational behavior and its role in viral infection. This will pave the way to us preparing for the next pandemic as the computational tools are tailored to help fight new challenges.
Collapse
Affiliation(s)
- Jameel M Abduljalil
- Department of Biological Sciences, Faculty of Applied Sciences, Thamar University, Dhamar, Yemen; Department of Botany and Microbiology, College of Science, Cairo University, Giza, Egypt
| | - Ahmed M Elghareib
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmed Samir
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmed A Ezat
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Abdo A Elfiky
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt.
| |
Collapse
|
47
|
Alsoussi WB, Malladi SK, Zhou JQ, Liu Z, Ying B, Kim W, Schmitz AJ, Lei T, Horvath SC, Sturtz AJ, McIntire KM, Evavold B, Han F, Scheaffer SM, Fox IF, Mirza SF, Parra-Rodriguez L, Nachbagauer R, Nestorova B, Chalkias S, Farnsworth CW, Klebert MK, Pusic I, Strnad BS, Middleton WD, Teefey SA, Whelan SPJ, Diamond MS, Paris R, O'Halloran JA, Presti RM, Turner JS, Ellebedy AH. SARS-CoV-2 Omicron boosting induces de novo B cell response in humans. Nature 2023; 617:592-598. [PMID: 37011668 DOI: 10.1038/s41586-023-06025-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
The primary two-dose SARS-CoV-2 mRNA vaccine series are strongly immunogenic in humans, but the emergence of highly infectious variants necessitated additional doses and the development of vaccines aimed at the new variants1-4. SARS-CoV-2 booster immunizations in humans primarily recruit pre-existing memory B cells5-9. However, it remains unclear whether the additional doses induce germinal centre reactions whereby re-engaged B cells can further mature, and whether variant-derived vaccines can elicit responses to variant-specific epitopes. Here we show that boosting with an mRNA vaccine against the original monovalent SARS-CoV-2 mRNA vaccine or the bivalent B.1.351 and B.1.617.2 (Beta/Delta) mRNA vaccine induced robust spike-specific germinal centre B cell responses in humans. The germinal centre response persisted for at least eight weeks, leading to significantly more mutated antigen-specific bone marrow plasma cell and memory B cell compartments. Spike-binding monoclonal antibodies derived from memory B cells isolated from individuals boosted with either the original SARS-CoV-2 spike protein, bivalent Beta/Delta vaccine or a monovalent Omicron BA.1-based vaccine predominantly recognized the original SARS-CoV-2 spike protein. Nonetheless, using a more targeted sorting approach, we isolated monoclonal antibodies that recognized the BA.1 spike protein but not the original SARS-CoV-2 spike protein from individuals who received the mRNA-1273.529 booster; these antibodies were less mutated and recognized novel epitopes within the spike protein, suggesting that they originated from naive B cells. Thus, SARS-CoV-2 booster immunizations in humans induce robust germinal centre B cell responses and can generate de novo B cell responses targeting variant-specific epitopes.
Collapse
Affiliation(s)
- Wafaa B Alsoussi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Sameer Kumar Malladi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Julian Q Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Baoling Ying
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Aaron J Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Tingting Lei
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Stephen C Horvath
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Alexandria J Sturtz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Katherine M McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Birk Evavold
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Fangjie Han
- Department of Emergency Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Suzanne M Scheaffer
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Isabella F Fox
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Senaa F Mirza
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Luis Parra-Rodriguez
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | - Christopher W Farnsworth
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Michael K Klebert
- Infectious Disease Clinical Research Unit, Washington University School of Medicine, St Louis, MO, USA
| | - Iskra Pusic
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Benjamin S Strnad
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - William D Middleton
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Sharlene A Teefey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | | | - Jane A O'Halloran
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
- Infectious Disease Clinical Research Unit, Washington University School of Medicine, St Louis, MO, USA
| | - Rachel M Presti
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
- Infectious Disease Clinical Research Unit, Washington University School of Medicine, St Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
48
|
Chiara M, Horner DS, Ferrandi E, Gissi C, Pesole G. HaploCoV: unsupervised classification and rapid detection of novel emerging variants of SARS-CoV-2. Commun Biol 2023; 6:443. [PMID: 37087497 PMCID: PMC10122080 DOI: 10.1038/s42003-023-04784-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 03/30/2023] [Indexed: 04/24/2023] Open
Abstract
Accurate and timely monitoring of the evolution of SARS-CoV-2 is crucial for identifying and tracking potentially more transmissible/virulent viral variants, and implement mitigation strategies to limit their spread. Here we introduce HaploCoV, a novel software framework that enables the exploration of SARS-CoV-2 genomic diversity through space and time, to identify novel emerging viral variants and prioritize variants of potential epidemiological interest in a rapid and unsupervised manner. HaploCoV can integrate with any classification/nomenclature and incorporates an effective scoring system for the prioritization of SARS-CoV-2 variants. By performing retrospective analyses of more than 11.5 M genome sequences we show that HaploCoV demonstrates high levels of accuracy and reproducibility and identifies the large majority of epidemiologically relevant viral variants - as flagged by international health authorities - automatically and with rapid turn-around times.Our results highlight the importance of the application of strategies based on the systematic analysis and integration of regional data for rapid identification of novel, emerging variants of SARS-CoV-2. We believe that the approach outlined in this study will contribute to relevant advances to current and future genomic surveillance methods.
Collapse
Affiliation(s)
- Matteo Chiara
- Department of Biosciences, University of Milan, Milan, Italy.
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy.
| | - David S Horner
- Department of Biosciences, University of Milan, Milan, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Erika Ferrandi
- Department of Biosciences, University of Milan, Milan, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Carmela Gissi
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
- Department of Biosciences, Biotechnology and Environment, University of Bari "A. Moro", Bari, Italy
| | - Graziano Pesole
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy.
- Department of Biosciences, Biotechnology and Environment, University of Bari "A. Moro", Bari, Italy.
| |
Collapse
|
49
|
Liu Y, Wang Z, Zhuang X, Zhang S, Chen Z, Zou Y, Sheng J, Li T, Tai W, Yu J, Wang Y, Zhang Z, Chen Y, Tong L, Yu X, Wu L, Chen D, Zhang R, Jin N, Shen W, Zhao J, Tian M, Wang X, Cheng G. Inactivated vaccine-elicited potent antibodies can broadly neutralize SARS-CoV-2 circulating variants. Nat Commun 2023; 14:2179. [PMID: 37069158 PMCID: PMC10107573 DOI: 10.1038/s41467-023-37926-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/30/2023] [Indexed: 04/19/2023] Open
Abstract
A full understanding of the inactivated COVID-19 vaccine-mediated antibody responses to SARS-CoV-2 circulating variants will inform vaccine effectiveness and vaccination development strategies. Here, we offer insights into the inactivated vaccine-induced antibody responses after prime-boost vaccination at both the polyclonal and monoclonal levels. We characterized the VDJ sequence of 118 monoclonal antibodies (mAbs) and found that 20 neutralizing mAbs showed varied potency and breadth against a range of variants including XBB.1.5, BQ.1.1, and BN.1. Bispecific antibodies (bsAbs) based on nonoverlapping mAbs exhibited enhanced neutralizing potency and breadth against the most antibody-evasive strains, such as XBB.1.5, BQ.1.1, and BN.1. The passive transfer of mAbs or their bsAb effectively protected female hACE2 transgenic mice from challenge with an infectious Delta or Omicron BA.2 variant. The neutralization mechanisms of these antibodies were determined by structural characterization. Overall, a broad spectrum of potent and distinct neutralizing antibodies can be induced in individuals immunized with the SARS-CoV-2 inactivated vaccine BBIBP-CorV, suggesting the application potential of inactivated vaccines and these antibodies for preventing infection by SARS-CoV-2 circulating variants.
Collapse
Grants
- National Key R&D Program of China (2021YFC2300200, 2020YFC1200104, 2018YFA0507202, 2021YFC2302405, 2022YFC2302204), the National Natural Science Foundation of China (32188101, 31825001, 81730063, and 81961160737), the Yunnan Cheng gong expert workstation (202005AF150034), Innovation Team Project of Yunnan Science and Technology Department (202105AE160020), and Tsinghua-Foshan Innovation Special Fund (2022THFS6124).
- National Key R&D Program of China (2022YFC2303403)
- National Key R&D Program of China (2021YFC2300104, 2022YFF1203103), the National Natural Science Foundation of China (32171202), and Vanke Special Fund for Public Health and Health Discipline Development, Tsinghua University (20221080056).
Collapse
Affiliation(s)
- Yubin Liu
- Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Ziyi Wang
- The Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Collaborative Innovation Center for Biotherapy, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xinyu Zhuang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Shengnan Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China
| | - Zhicheng Chen
- Center for Translational Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Yan Zou
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Jie Sheng
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Tianpeng Li
- Center for Translational Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Jinfang Yu
- The Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Collaborative Innovation Center for Biotherapy, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China
| | - Yunfeng Chen
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Liangqin Tong
- Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Xi Yu
- Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Linjuan Wu
- Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Dong Chen
- Wenzhou Central Hospital, Wenzhou, 325000, China
| | - Renli Zhang
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China
| | - Ningyi Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Weijun Shen
- Center for Translational Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China.
| | - Mingyao Tian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China.
| | - Xinquan Wang
- The Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Collaborative Innovation Center for Biotherapy, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Gong Cheng
- Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China.
| |
Collapse
|
50
|
Ding Y, Fan F, Xu X, Zhao G, Zhang X, Zhao H, Wang L, Wang B, Gao XM. A COVID-19 DNA Vaccine Candidate Elicits Broadly Neutralizing Antibodies against Multiple SARS-CoV-2 Variants including the Currently Circulating Omicron BA.5, BF.7, BQ.1 and XBB. Vaccines (Basel) 2023; 11:vaccines11040778. [PMID: 37112691 PMCID: PMC10144402 DOI: 10.3390/vaccines11040778] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Waves of breakthrough infections by SARS-CoV-2 Omicron subvariants currently pose a global challenge to the control of the COVID-19 pandemic. We previously reported a pVAX1-based DNA vaccine candidate, pAD1002, that encodes a receptor-binding domain (RBD) chimera of SARS-CoV-1 and Omicron BA.1. In mouse and rabbit models, pAD1002 plasmid induced cross-neutralizing Abs against heterologous sarbecoviruses, including SARS-CoV-1 and SARS-CoV-2 wildtype, Delta and Omicron variants. However, these antisera failed to block the recent emerging Omicron subvariants BF.7 and BQ.1. To solve this problem, we replaced the BA.1 RBD-encoding DNA sequence in pAD1002 with that of BA.4/5. The resulting construct, namely pAD1016, elicited SARS-CoV-1 and SARS-CoV-2 RBD-specific IFN-γ+ cellular responses in BALB/c and C57BL/6 mice. More importantly, pAD1016 vaccination in mice, rabbits and pigs generated serum Abs capable of neutralizing pseudoviruses representing multiple SARS-CoV-2 Omicron subvariants including BA.2, BA.4/5, BF.7, BQ.1 and XBB. As a booster vaccine for inactivated SARS-CoV-2 virus preimmunization in mice, pAD1016 broadened the serum Ab neutralization spectrum to cover the Omicron BA.4/5, BF7 and BQ.1 subvariants. These preliminary data highlight the potential benefit of pAD1016 in eliciting neutralizing Abs against broad-spectrum Omicron subvariants in individuals previously vaccinated with inactivated prototype SARS-CoV-2 virus and suggests that pAD1016 is worthy of further translational study as a COVID-19 vaccine candidate.
Collapse
|