301
|
Wigner-Jeziorska P, Janik-Karpińska E, Niwald M, Saluk J, Miller E. Effect of SARS-CoV-2 Infection and BNT162b2 Vaccination on the mRNA Expression of Genes Associated with Angiogenesis. Int J Mol Sci 2023; 24:16094. [PMID: 38003287 PMCID: PMC10671623 DOI: 10.3390/ijms242216094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), discovered in December 2019 in Wuhan, China, caused the coronavirus disease 2019 (COVID-19). Due to the rate of spread of this virus, the World Health Organization, in March 2020, recognised COVID-19 as a worldwide pandemic. The disease is multisystemic with varying degrees of severity. Unfortunately, despite intensive research, the molecular changes caused by SARS-CoV-2 remain unclear. Mechanisms affected by the virus infection include endothelial dysfunction and angiogenesis. Similarly, the vaccines developed so far affect the process of angiogenesis, contributing to the development of undesirable effects on part of the cardiovascular system. The presented research aimed to investigate the impact of the SARS-CoV-2 infection and the Pfizer Comirnaty vaccine (BNT162b2) on the molecular aspect of angiogenesis. We found that convalescents vaccinated with one dose of BNT162b2 were characterised by higher MMP-7 (metalloproteinases 7) expression than non-vaccinated convalescents and healthy volunteers vaccinated with one dose of BNT162b2. Moreover, non-vaccinated convalescents showed increased mRNA expression of ADAMTS1 (ADAM metallopeptidase with thrombospondin type 1 motif 1) compared to healthy volunteers vaccinated with one dose of BNT162b2. In addition, we showed significant sex differences in the expression of MMP-7. In conclusion, the results of our study suggest a significant impact of SARS-CoV-2 infection and vaccination on the course of angiogenesis at the molecular level.
Collapse
Affiliation(s)
- Paulina Wigner-Jeziorska
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.-J.); (J.S.)
| | - Edyta Janik-Karpińska
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland;
| | - Marta Niwald
- Department of Neurological Rehabilitation, Medical University of Lodz, 90-136 Lodz, Poland;
| | - Joanna Saluk
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.-J.); (J.S.)
| | - Elżbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, 90-136 Lodz, Poland;
| |
Collapse
|
302
|
van Bergen J, Camps MG, Pardieck IN, Veerkamp D, Leung WY, Leijs AA, Myeni SK, Kikkert M, Arens R, Zondag GC, Ossendorp F. Multiantigen pan-sarbecovirus DNA vaccines generate protective T cell immune responses. JCI Insight 2023; 8:e172488. [PMID: 37707962 PMCID: PMC10721273 DOI: 10.1172/jci.insight.172488] [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: 05/23/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023] Open
Abstract
SARS-CoV-2 is the third zoonotic coronavirus to cause a major outbreak in humans in recent years, and many more SARS-like coronaviruses with pandemic potential are circulating in several animal species. Vaccines inducing T cell immunity against broadly conserved viral antigens may protect against hospitalization and death caused by outbreaks of such viruses. We report the design and preclinical testing of 2 T cell-based pan-sarbecovirus vaccines, based on conserved regions within viral proteins of sarbecovirus isolates of human and other carrier animals, like bats and pangolins. One vaccine (CoVAX_ORF1ab) encoded antigens derived from nonstructural proteins, and the other (CoVAX_MNS) encoded antigens from structural proteins. Both multiantigen DNA vaccines contained a large set of antigens shared across sarbecoviruses and were rich in predicted and experimentally validated human T cell epitopes. In mice, the multiantigen vaccines generated both CD8+ and CD4+ T cell responses to shared epitopes. Upon encounter of full-length spike antigen, CoVAX_MNS-induced CD4+ T cells were responsible for accelerated CD8+ T cell and IgG Ab responses specific to the incoming spike, irrespective of its sarbecovirus origin. Finally, both vaccines elicited partial protection against a lethal SARS-CoV-2 challenge in human angiotensin-converting enzyme 2-transgenic mice. These results support clinical testing of these universal sarbecovirus vaccines for pandemic preparedness.
Collapse
Affiliation(s)
| | - Marcel G.M. Camps
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Iris N. Pardieck
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Dominique Veerkamp
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Wing Yan Leung
- Immunetune BV, Leiden, Netherlands
- Synvolux BV, Leiden, Netherlands
| | - Anouk A. Leijs
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Sebenzile K. Myeni
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Marjolein Kikkert
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Gerben C. Zondag
- Immunetune BV, Leiden, Netherlands
- Synvolux BV, Leiden, Netherlands
| | - Ferry Ossendorp
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
303
|
Batta I, Kaur T, Agrawal DK. Distinguishing Swine Flu (H1N1) from COVID-19: Clinical, Virological, and Immunological Perspectives. ARCHIVES OF MICROBIOLOGY & IMMUNOLOGY 2023; 7:271-280. [PMID: 37994372 PMCID: PMC10664801 DOI: 10.26502/ami.936500125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
This article provides an in-depth examination on the differences between the influenza A strain, H1N1 (also called Swine Flu) and Covid-19 focusing on the immune response and clinical symptoms. Flu symptoms due to influenza A strain, H1N1, were initially discovered in 2009. This variant of influenza A is believed to have emerged through reassortment, a process where the resulting virus inherits gene segments from each of its parental viruses. This reassortment event has resulted in a variant with altered characteristics, potentially affecting the level of immunity in humans. The symptoms of this strain typically manifest 1-4 days after exposure and include fever, cough, sore throat, runny/stuffy nose, body aches, fatigue, and gastrointestinal symptoms such as diarrhea. The transmission dynamics of this new variant, including human-to-human transmission, are still under investigation by health authorities. Individuals with weakened immune systems are generally more susceptible to severe illness. Risk factors associated with swine flu can include older adults, young children, pregnant women, and individuals with obesity. Historical variants of swine flu, such as the 2015 variant in India, have been associated with significant case numbers and deaths, often due to respiratory failure. Since the epidemic of Covid-19 due to SARS-CoV2 in early 2020, several symptoms of COVID-19 and swine flu overlap. In this article, we critically reviewed the differences and similarities in the immune response and clinical symptoms due to H1N1 virus and SARS-CoV2 in human.
Collapse
Affiliation(s)
- Irene Batta
- Bothell High School, Bothell, Washington, USA
| | - Tejinder Kaur
- Department of Zoology, DAV University, Jallandhar, Punjab, India
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
304
|
Duerr R, Dimartino D, Marier C, Zappile P, Wang G, François F, Ortigoza MB, Iturrate E, Samanovic MI, Mulligan MJ, Heguy A. Selective adaptation of SARS-CoV-2 Omicron under booster vaccine pressure: a multicentre observational study. EBioMedicine 2023; 97:104843. [PMID: 37866115 PMCID: PMC10623172 DOI: 10.1016/j.ebiom.2023.104843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND High rates of vaccination and natural infection drive immunity and redirect selective viral adaptation. Updated boosters are installed to cope with drifted viruses, yet data on adaptive evolution under increasing immune pressure in a real-world situation are lacking. METHODS Cross-sectional study to characterise SARS-CoV-2 mutational dynamics and selective adaptation over >1 year in relation to vaccine status, viral phylogenetics, and associated clinical and demographic variables. FINDINGS The study of >5400 SARS-CoV-2 infections between July 2021 and August 2022 in metropolitan New York portrayed the evolutionary transition from Delta to Omicron BA.1-BA.5 variants. Booster vaccinations were implemented during the Delta wave, yet booster breakthrough infections and SARS-CoV-2 re-infections were almost exclusive to Omicron. In adjusted logistic regression analyses, BA.1, BA.2, and BA.5 had a significant growth advantage over co-occurring lineages in the boosted population, unlike BA.2.12.1 or BA.4. Selection pressure by booster shots translated into diffuse adaptive evolution in Delta spike, contrasting with strong, receptor-binding motif-focused adaptive evolution in BA.2-BA.5 spike (Fisher Exact tests; non-synonymous/synonymous mutation rates per site). Convergent evolution has become common in Omicron, engaging spike positions crucial for immune escape, receptor binding, or cleavage. INTERPRETATION Booster shots are required to cope with gaps in immunity. Their discriminative immune pressure contributes to their effectiveness but also requires monitoring of selective viral adaptation processes. Omicron BA.2 and BA.5 had a selective advantage under booster vaccination pressure, contributing to the evolution of BA.2 and BA.5 sublineages and recombinant forms that predominate in 2023. FUNDING The study was supported by NYU institutional funds and partly by the Cancer Center Support Grant P30CA016087 at the Laura and Isaac Perlmutter Cancer Center.
Collapse
Affiliation(s)
- Ralf Duerr
- Department of Microbiology, NYU Grossman School of Medicine, USA; Department of Medicine, NYU Grossman School of Medicine, USA; Vaccine Center, NYU Grossman School of Medicine, New York, NY, USA.
| | - Dacia Dimartino
- Genome Technology Center, Office of Science and Research, NYU Langone Health, USA
| | - Christian Marier
- Genome Technology Center, Office of Science and Research, NYU Langone Health, USA
| | - Paul Zappile
- Genome Technology Center, Office of Science and Research, NYU Langone Health, USA
| | - Guiqing Wang
- Department of Pathology, NYU Grossman School of Medicine, USA
| | - Fritz François
- Department of Medicine, NYU Grossman School of Medicine, USA
| | - Mila B Ortigoza
- Department of Microbiology, NYU Grossman School of Medicine, USA; Department of Medicine, NYU Grossman School of Medicine, USA
| | | | - Marie I Samanovic
- Department of Medicine, NYU Grossman School of Medicine, USA; Vaccine Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Mark J Mulligan
- Department of Microbiology, NYU Grossman School of Medicine, USA; Department of Medicine, NYU Grossman School of Medicine, USA; Vaccine Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Adriana Heguy
- Genome Technology Center, Office of Science and Research, NYU Langone Health, USA; Department of Pathology, NYU Grossman School of Medicine, USA.
| |
Collapse
|
305
|
Rose AM, Nicolay N, Sandonis Martín V, Mazagatos C, Petrović G, Baruch J, Denayer S, Seyler L, Domegan L, Launay O, Machado A, Burgui C, Vaikutyte R, Niessen FA, Loghin II, Husa P, Aouali N, Panagiotakopoulos G, Tolksdorf K, Horváth JK, Howard J, Pozo F, Gallardo V, Nonković D, Džiugytė A, Bossuyt N, Demuyser T, Duffy R, Luong Nguyen LB, Kislaya I, Martínez-Baz I, Gefenaite G, Knol MJ, Popescu C, Součková L, Simon M, Michelaki S, Reiche J, Ferenczi A, Delgado-Sanz C, Lovrić Makarić Z, Cauchi JP, Barbezange C, Van Nedervelde E, O'Donnell J, Durier C, Guiomar R, Castilla J, Jonikaite I, Bruijning-Verhagen PC, Lazar M, Demlová R, Wirtz G, Amerali M, Dürrwald R, Kunstár MP, Kissling E, Bacci S, Valenciano M. Vaccine effectiveness against COVID-19 hospitalisation in adults (≥ 20 years) during Omicron-dominant circulation: I-MOVE-COVID-19 and VEBIS SARI VE networks, Europe, 2021 to 2022. Euro Surveill 2023; 28:2300187. [PMID: 37997665 PMCID: PMC10668256 DOI: 10.2807/1560-7917.es.2023.28.47.2300187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/24/2023] [Indexed: 11/25/2023] Open
Abstract
IntroductionThe I-MOVE-COVID-19 and VEBIS hospital networks have been measuring COVID-19 vaccine effectiveness (VE) in participating European countries since early 2021.AimWe aimed to measure VE against PCR-confirmed SARS-CoV-2 in patients ≥ 20 years hospitalised with severe acute respiratory infection (SARI) from December 2021 to July 2022 (Omicron-dominant period).MethodsIn both networks, 46 hospitals (13 countries) follow a similar test-negative case-control protocol. We defined complete primary series vaccination (PSV) and first booster dose vaccination as last dose of either vaccine received ≥ 14 days before symptom onset (stratifying first booster into received < 150 and ≥ 150 days after last PSV dose). We measured VE overall, by vaccine category/product, age group and time since first mRNA booster dose, adjusting by site as a fixed effect, and by swab date, age, sex, and presence/absence of at least one commonly collected chronic condition.ResultsWe included 2,779 cases and 2,362 controls. The VE of all vaccine products combined against hospitalisation for laboratory-confirmed SARS-CoV-2 was 43% (95% CI: 29-54) for complete PSV (with last dose received ≥ 150 days before onset), while it was 59% (95% CI: 51-66) after addition of one booster dose. The VE was 85% (95% CI: 78-89), 70% (95% CI: 61-77) and 36% (95% CI: 17-51) for those with onset 14-59 days, 60-119 days and 120-179 days after booster vaccination, respectively.ConclusionsOur results suggest that, during the Omicron period, observed VE against SARI hospitalisation improved with first mRNA booster dose, particularly for those having symptom onset < 120 days after first booster dose.
Collapse
Affiliation(s)
| | - Nathalie Nicolay
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | | | - Clara Mazagatos
- National Centre for Epidemiology, Institute of Health Carlos III, Madrid, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - Joaquin Baruch
- IDCU within Health promotion and disease prevention Directorate, G'mangia, Malta
| | | | - Lucie Seyler
- Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Lisa Domegan
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | - Odile Launay
- Inserm, CIC Cochin-Pasteur, Paris, France
- AP-HP, Hôpital Cochin, Paris, France
- Faculty of Medicine, University of Paris City, Paris, France
| | - Ausenda Machado
- National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | - Cristina Burgui
- Instituto de Salud Pública de Navarra-IdiSNA, Pamplona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - F Annabel Niessen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Isabela I Loghin
- St. Parascheva Clinical Hospital of Infectious Diseases, Iasi, Romania
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Petr Husa
- Faculty of Medicine, Masaryk University, Brno, Czechia
- University Hospital Brno, Brno, Czechia
| | | | | | | | - Judit Krisztina Horváth
- National Laboratory for Health Security, Epidemiology and Surveillance Centre, Semmelweis University, Budapest, Hungary
| | | | - Francisco Pozo
- National Centre for Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Virtudes Gallardo
- Dirección General de Salud Pública y Ordenación Farmacéutica, Junta de Andalucía, Spain
| | - Diana Nonković
- Teaching Public Health Institute of Split-Dalmatia County, Split, Croatia
| | - Aušra Džiugytė
- IDCU within Health promotion and disease prevention Directorate, G'mangia, Malta
| | | | | | - Róisín Duffy
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | | | - Irina Kislaya
- National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | - Iván Martínez-Baz
- Instituto de Salud Pública de Navarra-IdiSNA, Pamplona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Giedre Gefenaite
- Faculty of Medicine, Lund University, Lund, Sweden
- Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mirjam J Knol
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Corneliu Popescu
- Dr Victor Babes Clinical Hospital of Infectious and Tropical Diseases, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Marc Simon
- Centre Hospitalier de Luxembourg, Luxembourg
| | | | | | - Annamária Ferenczi
- National Laboratory for Health Security, Epidemiology and Surveillance Centre, Semmelweis University, Budapest, Hungary
| | - Concepción Delgado-Sanz
- National Centre for Epidemiology, Institute of Health Carlos III, Madrid, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - John Paul Cauchi
- IDCU within Health promotion and disease prevention Directorate, G'mangia, Malta
| | | | | | - Joan O'Donnell
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | | | - Raquel Guiomar
- National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | - Jesús Castilla
- Instituto de Salud Pública de Navarra-IdiSNA, Pamplona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - Patricia Cjl Bruijning-Verhagen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Mihaela Lazar
- "Cantacuzino" National Military Medical Institute for Research-Development, Bucharest, Romania
| | | | - Gil Wirtz
- Centre Hospitalier de Luxembourg, Luxembourg
| | - Marina Amerali
- National Public Health Organisation (EODY), Athens, Greece
| | | | - Mihály Pál Kunstár
- National Laboratory for Health Security, Epidemiology and Surveillance Centre, Semmelweis University, Budapest, Hungary
| | | | - Sabrina Bacci
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | | |
Collapse
|
306
|
Chappell KJ, Mordant FL, Amarilla AA, Modhiran N, Liang B, Li Z, Wijesundara DK, Lackenby JA, Griffin P, Bennet JK, Hensen L, Zhang W, Nguyen THO, Tran MH, Tapley P, Barnes J, Reading PC, Kedzierska K, Ranasinghe C, Subbarao K, Watterson D, Young PR, Munro TP. Long-term safety and immunogenicity of an MF59-adjuvanted spike glycoprotein-clamp vaccine for SARS-CoV-2 in adults aged 18-55 years or ≥56 years: 12-month results from a randomised, double-blind, placebo-controlled, phase 1 trial. EBioMedicine 2023; 97:104842. [PMID: 37865043 PMCID: PMC10597768 DOI: 10.1016/j.ebiom.2023.104842] [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: 05/10/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023] Open
Abstract
BACKGROUND We previously demonstrated the safety and immunogenicity of an MF59-adjuvanted COVID-19 vaccine based on the SARS-CoV-2 spike glycoprotein stabilised in a pre-fusion conformation by a molecular clamp using HIV-1 glycoprotein 41 sequences. Here, we describe 12-month results in adults aged 18-55 years and ≥56 years. METHODS Phase 1, double-blind, placebo-controlled trial conducted in Australia (July 2020-December 2021; ClinicalTrials.govNCT04495933; active, not recruiting). Healthy adults (Part 1: 18-55 years; Part 2: ≥56 years) received two doses of placebo, 5 μg, 15 μg, or 45 μg vaccine, or one 45 μg dose of vaccine followed by placebo (Part 1 only), 28 days apart (n = 216; 24 per group). Safety, humoral immunogenicity (including against virus variants), and cellular immunogenicity were assessed to day 394 (12 months after second dose). Effects of subsequent COVID-19 vaccination on humoral responses were examined. FINDINGS All two-dose vaccine regimens were well tolerated and elicited strong antigen-specific and neutralising humoral responses, and CD4+ T-cell responses, by day 43 in younger and older adults, although cellular responses were lower in older adults. Humoral responses waned by day 209 but were boosted in those receiving authorised vaccines. Neutralising activity against Delta and Omicron variants was present but lower than against the Wuhan strain. Cross-reactivity in HIV diagnostic tests declined over time but remained detectable in most participants. INTERPRETATION The SARS-CoV-2 molecular clamp vaccine is well tolerated and evokes robust immune responses in adults of all ages. Although the HIV glycoprotein 41-based molecular clamp is not being progressed, the clamp concept represents a viable platform for vaccine development. FUNDING This study was funded by the Coalition for Epidemic Preparedness Innovations, the National Health and Medical Research Council of Australia, and the Queensland Government.
Collapse
Affiliation(s)
- Keith J Chappell
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia; The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia.
| | - Francesca L Mordant
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Benjamin Liang
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Zheyi Li
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Danushka K Wijesundara
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia; The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Julia A Lackenby
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia; The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Paul Griffin
- Nucleus Network Brisbane Clinic, Herston, QLD, Australia; Department of Infectious Diseases, Mater Health, QLD, Australia; School of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | | | - Luca Hensen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Mai H Tran
- Agilex Biolabs, Thebarton, SA, Australia
| | | | - James Barnes
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia; WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Charani Ranasinghe
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia; WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia; The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia; The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Trent P Munro
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia; The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
307
|
Chen CH, Wu MJ, Tsai SF. Safety and effectiveness of COVID-19 vaccines in patients with IgA nephropathy: a retrospective cohort study from the TriNetX global collaborative networks. EClinicalMedicine 2023; 65:102306. [PMID: 38021374 PMCID: PMC10661495 DOI: 10.1016/j.eclinm.2023.102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Background This study aimed to evaluate the renal safety and effectiveness of COVID-19 vaccination in patients with immunoglobulin A nephropathy (IgAN). Methods We conducted a global and retrospective collaborative network analysis using TriNetX data from September 11, 2018 to September 11, 2023, to address this question. The study recorded diagnoses of IgAN, COVID-19 vaccinations, and outcomes of effectiveness using International Classification of Diseases, Tenth Revision, Clinical Modification codes and procedure codes. Propensity score matching (PSM) created matched groups (1:1). Hazard ratios (HR) with 95% confidence intervals (95% CI) were calculated for outcomes of effectiveness, and Kaplan-Meier method assessed survival probability. Safety outcomes regarding renal function were compared with estimated glomerular filtration rate (eGFR), proteinuria, and hematuria. Subgroup analyses were based on sex and age group. Sensitivity analysis was done before the outbreak of Omicron (from September 11, 2018 to October 31, 2021). Findings The study involved 1010 vaccinated and 2776 unvaccinated patients with IgAN without COVID-19 infection at baseline. After PSM (1:1) with 25 variables, both groups consisted of well-matched 979 patients who were relatively young (around 55 years old) and in good health (eGFR: 78-80 ml/min/1.732 m2). Compared to the non-vaccinated group, vaccinated patients had significantly lower risks of COVID-19 infection and complications, including COVID-19 infection (HR: 0.050, 95% CI: 0.026, 0.093), COVID-19 pneumonia (HR: 0), severe lung complication (0.647, 95% CI: 0.421, 0.994), acute respiratory failure (0.625, 95% CI: 0.400, 0.978), sepsis (0.545, 95% CI: 0.334, 0.890), emergency department visits (0.716, 95% CI: 0.615, 0.833), all hospitalizations (0.573, 95% CI: 0.459, 0.715), and mortality (0.595, 95% CI: 0.366, 0.969). However, one month after the follow-up, the vaccinated group exhibited a slightly, but statistically significantly, lower eGFR compared to the non-vaccinated group (73.58 vs. 83.05 ml/min/1.732 m2, p = 0.047). Nine months after the follow-up, the difference in eGFR between the two groups disappeared. The lower risk of COVID-19 infection was observed across genders (male and female) and age groups (young and old). For the period before Omicron outbreak, results were also similar. Interpretation In the largest TriNetX matched cohort study of IgAN, COVID-19 vaccination was associated with a reduced risk of COVID-19 infection and associated complications. However, careful monitoring of renal function, especially GFR, is advisable. Funding This study was supported by grant TCVGH-1103602C, TCVGH-1103601D, and TCVGH-1113602D from Taichung Veterans General Hospital.
Collapse
Affiliation(s)
- Cheng-Hsu Chen
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taiwan
- Department of Life Science, Tunghai University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shang-Feng Tsai
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taiwan
- Department of Life Science, Tunghai University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
308
|
Zerbo O, Ray GT, Fireman B, Layefsky E, Goddard K, Ross P, Greenberg M, Klein NP. Effectiveness of COVID-19 vaccination during pregnancy by circulating viral variant. AJOG GLOBAL REPORTS 2023; 3:100264. [PMID: 37719643 PMCID: PMC10502365 DOI: 10.1016/j.xagr.2023.100264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND SARS-CoV-2 infection in pregnancy can result in a spectrum of asymptomatic to critical COVID-19 outcomes, including hospitalization, admission to the intensive care unit, or death. OBJECTIVE This study aimed to investigate the effectiveness of messenger RNA COVID-19 vaccination during pregnancy against both hospitalization and infection, stratified by different variant circulations and by time since the last vaccine dose. STUDY DESIGN This was a retrospective cohort study among pregnant persons who were members of Kaiser Permanente Northern California and delivered between December 15, 2020, and September 30, 2022. Pregnant persons who received any vaccine dose before the pregnancy onset date were excluded. The primary outcome was hospitalization for COVID-19, and the secondary outcome was polymerase chain reaction-confirmed SARS-CoV-2 infection. Exposure was receipt of a messenger RNA vaccine during pregnancy. Poisson regression was used to estimate the risk ratio of hospitalization by comparing vaccinated pregnant persons with unvaccinated pregnant persons adjusted for sociodemographic factors and calendar time. Cox regression was used to estimate the hazard ratio of infection by comparing vaccinated pregnant persons with unvaccinated pregnant persons. Vaccine effectiveness was estimated as 1 minus the rate ratio or the hazard ratio multiplied by 100. Vaccine effectiveness was estimated overall and by variant periods (before Delta, Delta, Omicron, and subvariants). RESULTS Of 57,688 pregnant persons, 16,153 (28%) received at least 1 dose of a messenger RNA COVID-19 vaccine during pregnancy; moreover, 4404 pregnant persons tested positive for SARS-CoV-2 infection, and 108 pregnant persons were hospitalized during pregnancy. Overall, 2-dose vaccine effectiveness against hospitalization was 91% within <150 days of vaccination and 48% >150 days after vaccination. The 2-dose vaccine effectiveness within <150 days after vaccination was 100% during the original virus strain and Delta variant periods of the virus; vaccine effectiveness was 51% during the Omicron period. Of the hospitalization cases, 97% of pregnant persons were unvaccinated. During hospitalization, none of the vaccinated pregnant persons required ventilation or were admitted to the intensive care unit. Moreover, 2-dose vaccine effectiveness against infection was 54% within <150 days after vaccination and 26% ≥150 days after vaccination. CONCLUSION Messenger RNA COVID-19 vaccination during pregnancy was effective against hospitalization for COVID-19 and SARS-CoV-2 infection. COVID-19 was mild among pregnant persons who were vaccinated compared with those who were unvaccinated. Thus, all pregnant persons should be strongly encouraged to receive messenger RNA COVID-19 vaccines to prevent severe disease.
Collapse
Affiliation(s)
- Ousseny Zerbo
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA (Dr Zerbo, Mr. Ray, Fireman and Layefsky, Ms. Goddard, and Ross, and Dr Klein)
| | - G. Thomas Ray
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA (Dr Zerbo, Mr. Ray, Fireman and Layefsky, Ms. Goddard, and Ross, and Dr Klein)
| | - Bruce Fireman
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA (Dr Zerbo, Mr. Ray, Fireman and Layefsky, Ms. Goddard, and Ross, and Dr Klein)
| | - Evan Layefsky
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA (Dr Zerbo, Mr. Ray, Fireman and Layefsky, Ms. Goddard, and Ross, and Dr Klein)
| | - Kristin Goddard
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA (Dr Zerbo, Mr. Ray, Fireman and Layefsky, Ms. Goddard, and Ross, and Dr Klein)
| | - Pat Ross
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA (Dr Zerbo, Mr. Ray, Fireman and Layefsky, Ms. Goddard, and Ross, and Dr Klein)
| | - Mara Greenberg
- Department of Obstetrics and Gynecology, Kaiser Permanente Northern California, Oakland, CA (Dr Greenberg)
- Regional Perinatal Service Center, Kaiser Permanente Northern California, Santa Clara, CA (Dr Greenberg)
| | - Nicola P. Klein
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA (Dr Zerbo, Mr. Ray, Fireman and Layefsky, Ms. Goddard, and Ross, and Dr Klein)
| |
Collapse
|
309
|
Olayiwola MO, Alaje AI, Olarewaju AY, Adedokun KA. A caputo fractional order epidemic model for evaluating the effectiveness of high-risk quarantine and vaccination strategies on the spread of COVID-19. HEALTHCARE ANALYTICS (NEW YORK, N.Y.) 2023; 3:100179. [PMID: 37101804 PMCID: PMC10118058 DOI: 10.1016/j.health.2023.100179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/15/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023]
Abstract
The recent global Coronavirus disease (COVID-19) threat to the human race requires research on preventing its reemergence without affecting socio-economic factors. This study proposes a fractional-order mathematical model to analyze the impact of high-risk quarantine and vaccination on COVID-19 transmission. The proposed model is used to analyze real-life COVID-19 data to develop and analyze the solutions and their feasibilities. Numerical simulations study the high-risk quarantine and vaccination strategies and show that both strategies effectively reduce the virus prevalence, but their combined application is more effective. We also demonstrate that their effectiveness varies with the volatile rate of change in the system's distribution. The results are analyzed using Caputo fractional order and presented graphically and extensively analyzed to highlight potent ways of curbing the virus.
Collapse
|
310
|
Juhel BC, Brunelle CL, Bernstein MC, Smith LH, Jung AW, Ababneh HS, Hausman EK, Bucci LK, Bernstein T, Naoum GE, Taghian AG. Side effects of COVID-19 vaccinations in patients treated for breast cancer. Clin Exp Med 2023; 23:3671-3680. [PMID: 37031282 PMCID: PMC10098240 DOI: 10.1007/s10238-023-01050-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/17/2023] [Indexed: 04/10/2023]
Abstract
Lymph node swelling is a side effect of the mRNA COVID-19 vaccines, a distressing side effect for women treated for breast cancer. The purpose of this study is to present side effects reported by a cohort of patients treated for breast cancer. A survey link was sent to 4945 women who received breast cancer treatment and were prospectively screened for breast cancer-related lymphedema. In total, 621 patients who received an mRNA vaccine and responded to the survey were included in analysis. We assessed the frequency and predictors of side effects. The most frequent side effects reported were injection site soreness, fatigue, generalized muscle soreness, headache, and chills, with median duration ≤ 48 h. Lymph node swelling occurred most often in the axilla ipsilateral to the vaccine. The median duration was 1 week or less after all doses. These data will inform patient education regarding future vaccine doses, including reassurances about which side effects to expect, particularly lymph node swelling which may impact mammograms after vaccination. Type and duration of side effects were similar to that reported by the general population in Phase 3 testing trials of the mRNA vaccines. Clinical Trial Registration NCT04872738 posted May 4, 2021.
Collapse
Affiliation(s)
- Brooke C Juhel
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cheryl L Brunelle
- Department of Physical and Occupational Therapy, Massachusetts General Hospital, Boston, MA, USA
| | - Madison C Bernstein
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Louisa H Smith
- Department of Health Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Roux Institute, Northeastern University, Portland, ME, USA
| | - Amanda W Jung
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hazim S Ababneh
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth K Hausman
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Loryn K Bucci
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tess Bernstein
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - George E Naoum
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Northwestern University Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, Il, USA
| | - Alphonse G Taghian
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
311
|
Wang CC, Young YH. Comparing the recurrence of audio-vestibular disorders following breakthrough infection of COVID-19 vs. those following vaccine administration. Am J Otolaryngol 2023; 44:103970. [PMID: 37467676 DOI: 10.1016/j.amjoto.2023.103970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/13/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023]
Abstract
PURPOSE The term "breakthrough infection" of COVID-19 indicates that subjects who previously received COVID-19 vaccination became infected with COVID-19. This study compared the recurrence of audio-vestibular disorders following breakthrough infection of COVID-19 vs. those following vaccine administration. PATIENTS AND METHODS Fifty patients with previous known audio-vestibular disorders visited our clinic due to recurrence of inner ear symptoms following breakthrough infection of COVID-19 and were assigned to Group A. Another 50 patients who had recurrent inner ear symptoms following COVID-19 vaccination were assigned to Group B for comparison. The post-breakthrough infection interval is defined from date of breakthrough infection to the onset of inner ear symptoms, while the post-vaccination interval means the time from date of vaccination to the onset of inner ear symptoms. These two intervals were calculated and then compared. RESULTS The time from latest vaccination to the breakthrough infection of COVID-19 was 4 m (median), likely due to waning of IgG response. To the onset of inner ear symptoms, the post-breakthrough infection interval was 40d (median) for Group A, which was significantly longer than 10d (median) of the post-vaccination interval for Group B. CONCLUSION The post-breakthrough infection interval (median, 40d) is significantly longer than the post-vaccination interval (median, 10d) to exacerbate pre-existing audio-vestibular disorders. The reason is probably because an interval of 40d is related to IgG peak response following COVID-19 breakthrough infection, while that of 10d is responsible for IgG production after COVID-19 vaccination.
Collapse
Affiliation(s)
- Chih-Ching Wang
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ho Young
- Department of Otolaryngology, Far Eastern Memorial Hospital, New Taipei, Taiwan.
| |
Collapse
|
312
|
Rose AM, Nicolay N, Sandonis Martín V, Mazagatos C, Petrović G, Niessen FA, Machado A, Launay O, Denayer S, Seyler L, Baruch J, Burgui C, Loghin II, Domegan L, Vaikutytė R, Husa P, Panagiotakopoulos G, Aouali N, Dürrwald R, Howard J, Pozo F, Sastre-Palou B, Nonković D, Knol MJ, Kislaya I, Luong Nguyen LB, Bossuyt N, Demuyser T, Džiugytė A, Martínez-Baz I, Popescu C, Duffy R, Kuliešė M, Součková L, Michelaki S, Simon M, Reiche J, Otero-Barrós MT, Lovrić Makarić Z, Bruijning-Verhagen PC, Gomez V, Lesieur Z, Barbezange C, Van Nedervelde E, Borg ML, Castilla J, Lazar M, O'Donnell J, Jonikaitė I, Demlová R, Amerali M, Wirtz G, Tolksdorf K, Valenciano M, Bacci S, Kissling E. Vaccine effectiveness against COVID-19 hospitalisation in adults (≥ 20 years) during Alpha- and Delta-dominant circulation: I-MOVE-COVID-19 and VEBIS SARI VE networks, Europe, 2021. Euro Surveill 2023; 28:2300186. [PMID: 37997666 PMCID: PMC10668259 DOI: 10.2807/1560-7917.es.2023.28.47.2300186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/24/2023] [Indexed: 11/25/2023] Open
Abstract
IntroductionTwo large multicentre European hospital networks have estimated vaccine effectiveness (VE) against COVID-19 since 2021.AimWe aimed to measure VE against PCR-confirmed SARS-CoV-2 in hospitalised severe acute respiratory illness (SARI) patients ≥ 20 years, combining data from these networks during Alpha (March-June)- and Delta (June-December)-dominant periods, 2021.MethodsForty-six participating hospitals across 14 countries follow a similar generic protocol using the test-negative case-control design. We defined complete primary series vaccination (PSV) as two doses of a two-dose or one of a single-dose vaccine ≥ 14 days before onset.ResultsWe included 1,087 cases (538 controls) and 1,669 cases (1,442 controls) in the Alpha- and Delta-dominant periods, respectively. During the Alpha period, VE against hospitalisation with SARS-CoV2 for complete Comirnaty PSV was 85% (95% CI: 69-92) overall and 75% (95% CI: 42-90) in those aged ≥ 80 years. During the Delta period, among SARI patients ≥ 20 years with symptom onset ≥ 150 days from last PSV dose, VE for complete Comirnaty PSV was 54% (95% CI: 18-74). Among those receiving Comirnaty PSV and mRNA booster (any product) ≥ 150 days after last PSV dose, VE was 91% (95% CI: 57-98). In time-since-vaccination analysis, complete all-product PSV VE was > 90% in those with their last dose < 90 days before onset; ≥ 70% in those 90-179 days before onset.ConclusionsOur results from this EU multi-country hospital setting showed that VE for complete PSV alone was higher in the Alpha- than the Delta-dominant period, and addition of a first booster dose during the latter period increased VE to over 90%.
Collapse
Affiliation(s)
| | - Nathalie Nicolay
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | | | - Clara Mazagatos
- National Centre for Epidemiology, Institute of Health Carlos III, Madrid, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - F Annabel Niessen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Ausenda Machado
- National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | - Odile Launay
- Inserm, CIC Cochin-Pasteur, Paris, France
- AP-HP, Hôpital Cochin, Paris, France
- Faculty of Medicine, University of Paris City, Paris, France
| | | | - Lucie Seyler
- Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Joaquin Baruch
- IDCU within Health promotion and disease prevention Directorate, G'mangia, Malta
| | - Cristina Burgui
- Instituto de Salud Pública de Navarra-IdiSNA, Pamplona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Isabela I Loghin
- St. Parascheva Clinical Hospital of Infectious Diseases, Iasi, Romania
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Lisa Domegan
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | | | - Petr Husa
- Faculty of Medicine, Masaryk University, Brno, Czechia
- University Hospital Brno, Brno, Czechia
| | | | | | | | | | - Francisco Pozo
- National Centre for Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Bartolomé Sastre-Palou
- Servicio de Medicina Preventiva Hospital Universitario Son Espases, Servicio de Epidemiología, Consellería de Salut, Palma, Spain
| | - Diana Nonković
- Teaching Public Health Institute of Split-Dalmatia County, Split, Croatia
| | - Mirjam J Knol
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Irina Kislaya
- National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | | | | | | | - Aušra Džiugytė
- IDCU within Health promotion and disease prevention Directorate, G'mangia, Malta
| | - Iván Martínez-Baz
- Instituto de Salud Pública de Navarra-IdiSNA, Pamplona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Corneliu Popescu
- Dr Victor Babes Clinical Hospital of Infectious and Tropical Diseases, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Róisín Duffy
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | - Monika Kuliešė
- Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | - Marc Simon
- Centre Hospitalier de Luxembourg, Luxembourg
| | | | - María Teresa Otero-Barrós
- Servicio de Epidemiología, Dirección General de Salud Pública, Consejería de Sanidad de Galicia, Santiago de Compostela, A Coruna, Spain
| | | | - Patricia Cjl Bruijning-Verhagen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Verónica Gomez
- National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | | | | | | | - Maria-Louise Borg
- IDCU within Health promotion and disease prevention Directorate, G'mangia, Malta
| | - Jesús Castilla
- Instituto de Salud Pública de Navarra-IdiSNA, Pamplona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Mihaela Lazar
- "Cantacuzino" National Military Medical Institute for Research-Development, Bucharest, Romania
| | - Joan O'Donnell
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | | | | | - Marina Amerali
- National Public Health Organisation (EODY), Athens, Greece
| | - Gil Wirtz
- Luxembourg Institute of Health, Luxembourg
| | | | | | - Sabrina Bacci
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | | |
Collapse
|
313
|
Weinberg AR, Caeg CO, DePalma R, Hernandez F, Rogers JH, Ibrahim HN, Bynon SJ, Nigo M. COVID-19 Vaccine Seroresponse Based on The Timing of The Primary Series; Pre- versus Post-Renal Transplantation. Clin Transplant 2023; 37:e15072. [PMID: 37434417 DOI: 10.1111/ctr.15072] [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: 11/29/2022] [Revised: 06/24/2023] [Accepted: 06/29/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) poses a serious risk to patients with chronic kidney disease (CKD) and renal transplant. While COVID-19 vaccination is recommended before transplant, there are limited data comparing vaccine timing. Our aim is to evaluate serological responses to COVID-19 vaccines pre- and post-renal transplant and the durability of antibody levels. METHODS We retrospectively evaluated the antibody response of adult renal transplant recipients who had received at least a primary series of the COVID-19 vaccine. The patients were divided into two groups based on the timing; pre- or post-transplant. Antibody titer levels were evaluated at least 4 weeks after vaccination for each group. Titer durability was assessed by calculating the median titer level of individuals. RESULTS A total of 139 patients were identified between January 2019 and April 2022. Twenty-nine patients were excluded because of previous COVID-19 infection, and 15 patients were excluded each for insufficient vaccine doses and lack of titer data. Forty patients were included for the pre-transplant group and 40 for post-transplant. The number of pre-transplant patients who developed antibodies (39 patients, 97.5%) was significantly greater than the number of post-transplant patients (21 patients, 52.5%) with p < .01. The median post-vaccination titer levels were significantly greater in the pre-transplant group up to 5 months after vaccination (p < .05). The pre-transplant group's titers seemed sustained even after renal transplantation. CONCLUSION Vaccinating renal transplant patients before transplant results in increased achievement of seroresponse, higher levels of antibody titers, and sustained titers following transplant. Larger and prospective studies are warranted to confirm the findings.
Collapse
Affiliation(s)
- Amy R Weinberg
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Calvin O Caeg
- Transplant Center, Memorial Hermann Hospital Texas Medical Center, Houston, Texas, USA
| | - Robyn DePalma
- Transplant Center, Memorial Hermann Hospital Texas Medical Center, Houston, Texas, USA
| | - Frances Hernandez
- Transplant Center, Memorial Hermann Hospital Texas Medical Center, Houston, Texas, USA
| | - Jackson H Rogers
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hassan N Ibrahim
- Division of Immunology and Organ Transplantation, Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Steve J Bynon
- Division of Immunology and Organ Transplantation, Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Masayuki Nigo
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
314
|
Kocatürk E, Salameh P, Sarac E, Vera Ayala CE, Thomsen SF, Zuberbier T, Ensina LF, Popov TA, van Doorn MBA, Giménez-Arnau AM, Asero R, Criado PR, Aarestrup FM, AbdulHameed Ansari Z, Al Abri S, Al-Ahmad M, Al Hinai B, Allenova A, Al-Nesf M, Altrichter S, Arnaout R, Bartosińska J, Bauer A, Bernstein JA, Bizjak M, Bonnekoh H, Bouillet L, Brzoza Z, Calvalcanti Dela Bianca Melo AC, Campinhos FL, Carne E, Purayil SC, Cherrez-Ojeda I, Chong-Neto HJ, Christoff G, Conlon N, Jardim Criado RF, Cvenkel K, Damadoglu E, Danilycheva I, Day C, de Montjoye L, Demir S, Ferucci SM, Fomina D, Fukunaga A, Garcia E, Gelincik A, Göbel JH, Godse K, Gonçalo M, Gotua M, Grattan C, Gugala A, Guillet C, Kalyoncu AF, Karakaya G, Kasperska-Zając A, Katelaris CH, Khoshkhui M, Kleinheinz A, Kolacinska-Flont M, Kolkhir P, Košnik M, Krasowska D, Kumaran MS, Kuprys-Lipinska I, Kurowski M, Kuznetsova EV, Larenas-Linnemann D, Lebedkina MS, Lee Y, Makris M, Gómez RM, Nasr I, Neisinger S, Oda Y, Kara RÖ, Palitot EB, Papapostolou N, Salvador Parisi CA, Pesque D, Peter J, Petkova E, Ridge K, Rudenko M, Rutkowski K, Saini SS, Salman A, Sanchez J, Şekerel B, Serdotetskova SA, Serpa FS, Dikicier BS, Sidiropoulos N, Sikora A, Sørensen JA, Soria A, Kucuk OS, Thalappil SR, et alKocatürk E, Salameh P, Sarac E, Vera Ayala CE, Thomsen SF, Zuberbier T, Ensina LF, Popov TA, van Doorn MBA, Giménez-Arnau AM, Asero R, Criado PR, Aarestrup FM, AbdulHameed Ansari Z, Al Abri S, Al-Ahmad M, Al Hinai B, Allenova A, Al-Nesf M, Altrichter S, Arnaout R, Bartosińska J, Bauer A, Bernstein JA, Bizjak M, Bonnekoh H, Bouillet L, Brzoza Z, Calvalcanti Dela Bianca Melo AC, Campinhos FL, Carne E, Purayil SC, Cherrez-Ojeda I, Chong-Neto HJ, Christoff G, Conlon N, Jardim Criado RF, Cvenkel K, Damadoglu E, Danilycheva I, Day C, de Montjoye L, Demir S, Ferucci SM, Fomina D, Fukunaga A, Garcia E, Gelincik A, Göbel JH, Godse K, Gonçalo M, Gotua M, Grattan C, Gugala A, Guillet C, Kalyoncu AF, Karakaya G, Kasperska-Zając A, Katelaris CH, Khoshkhui M, Kleinheinz A, Kolacinska-Flont M, Kolkhir P, Košnik M, Krasowska D, Kumaran MS, Kuprys-Lipinska I, Kurowski M, Kuznetsova EV, Larenas-Linnemann D, Lebedkina MS, Lee Y, Makris M, Gómez RM, Nasr I, Neisinger S, Oda Y, Kara RÖ, Palitot EB, Papapostolou N, Salvador Parisi CA, Pesque D, Peter J, Petkova E, Ridge K, Rudenko M, Rutkowski K, Saini SS, Salman A, Sanchez J, Şekerel B, Serdotetskova SA, Serpa FS, Dikicier BS, Sidiropoulos N, Sikora A, Sørensen JA, Soria A, Kucuk OS, Thalappil SR, Tomaszewska K, Tuncay G, Unal D, Valle S, van Lindonk E, Vestergaard C, Meshkova RY, Vitchuk A, Xepapadaki P, Ye YM, Zalewska-Janowska A, Zamlynski M, Maurer M. Urticaria exacerbations and adverse reactions in patients with chronic urticaria receiving COVID-19 vaccination: Results of the UCARE COVAC-CU study. J Allergy Clin Immunol 2023; 152:1095-1106. [PMID: 37574079 DOI: 10.1016/j.jaci.2023.07.019] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Concern about disease exacerbations and fear of reactions after coronavirus disease 2019 (COVID-19) vaccinations are common in chronic urticaria (CU) patients and may lead to vaccine hesitancy. OBJECTIVE We assessed the frequency and risk factors of CU exacerbation and adverse reactions in CU patients after COVID-19 vaccination. METHODS COVAC-CU is an international multicenter study of Urticaria Centers of Reference and Excellence (UCAREs) that retrospectively evaluated the effects of COVID-19 vaccination in CU patients aged ≥18 years and vaccinated with ≥1 dose of any COVID-19 vaccine. We evaluated CU exacerbations and severe allergic reactions as well as other adverse events associated with COVID-19 vaccinations and their association with various CU parameters. RESULTS Across 2769 COVID-19-vaccinated CU patients, most (90%) received at least 2 COVID-19 vaccine doses, and most patients received CU treatment and had well-controlled disease. The rate of COVID-19 vaccination-induced CU exacerbation was 9%. Of 223 patients with CU exacerbation after the first dose, 53.4% experienced recurrence of CU exacerbation after the second dose. CU exacerbation most often started <48 hours after vaccination (59.2%), lasted for a few weeks or less (70%), and was treated mainly with antihistamines (70.3%). Factors that increased the risk for COVID-19 vaccination-induced CU exacerbation included female sex, disease duration shorter than 24 months, having chronic spontaneous versus inducible urticaria, receipt of adenovirus viral vector vaccine, having nonsteroidal anti-inflammatory drug/aspirin intolerance, and having concerns about getting vaccinated; receiving omalizumab treatment and Latino/Hispanic ethnicity lowered the risk. First-dose vaccine-related adverse effects, most commonly local reactions, fever, fatigue, and muscle pain, were reported by 43.5% of CU patients. Seven patients reported severe allergic reactions. CONCLUSIONS COVID-19 vaccination leads to disease exacerbation in only a small number of CU patients and is generally well tolerated.
Collapse
Affiliation(s)
- Emek Kocatürk
- Department of Dermatology, Koc University School of Medicine, Istanbul, Turkey; Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany.
| | - Pascale Salameh
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany; Department of Clinical Epidemiology, Institut National de Santé Publique, Epidémiologie Clinique et Toxicologie-Liban (INSPECT-LB), Beirut, Lebanon; Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia, Cyprus; School of Medicine, Lebanese American University, Beirut, Germany; Faculty of Pharmacy, Lebanese University, Hadat, Lebanon
| | - Esra Sarac
- Department of Dermatology, Koc University School of Medicine, Istanbul, Turkey
| | - Carolina E Vera Ayala
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Simon Francis Thomsen
- Department of Dermatology, Department of Biomedical Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Torsten Zuberbier
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Luis Felipe Ensina
- Department of Dermatology, Federal University of São Paulo, São Paulo, Brazil
| | - Todor A Popov
- Department of Allergology, University Hospital Sv Ivan Rilski, Sofia, Bulgaria
| | - Martijn B A van Doorn
- Department of Dermatology, Erasmus MC Rotterdam, Rotterdam, The Netherlands; Centre for Human Drug Research, Leiden, The Netherlands
| | - Ana Maria Giménez-Arnau
- Department of Dermatology, Hospital del Mar Research Institute, Universitat Pompeu Fabra, Barcelona, Spain
| | - Riccardo Asero
- Ambulatorio di Allergologia, Clinica San Carlo, Paderno Dugnano, Italy
| | - Paulo Ricardo Criado
- Alergoskin Alergia e Dermatologia, Centro Universitário Faculdade de Medicina do ABC, Santo André, Brazil
| | - Fernando M Aarestrup
- Department of Allergy and Immunology, Faculdade de Ciências, Médicas e da Saúde de Juiz de Fora (SUPREMA), Hospital Maternidade Therezinha de Jesus, Minas Gerais, Brazil
| | | | - Salma Al Abri
- Clinical Immunology and Allergy Unit, Royal Hospital, Muscat, Oman
| | - Mona Al-Ahmad
- Microbiology Department, College of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Bushra Al Hinai
- Clinical Immunology and Allergy Unit, Royal Hospital, Muscat, Oman
| | - Anastasiia Allenova
- Laboratory of Immune-Mediated Skin Diseases, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Medical Research and Education Center, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - Maryam Al-Nesf
- Department of Medicine, Allergy and Immunology Division, Hamad Medical Corporation, Doha, Qatar
| | - Sabine Altrichter
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany; Department of Dermatology and Venereology, Kepler University Hospital, Linz, Austria
| | - Rand Arnaout
- Department of Dermatology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Joanna Bartosińska
- Department of Dermatology, Venereology, and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Andrea Bauer
- Department of Dermatology, Technical University Dresden, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Jonathan A Bernstein
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Immunology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mojca Bizjak
- Division of Allergy, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
| | - Hanna Bonnekoh
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Laurence Bouillet
- National Reference Center for Angioedema, Grenoble University Hospital, Grenoble, France
| | - Zenon Brzoza
- Department of Internal Diseases with the Division of Allergology, Institute of Medical Sciences, University of Opole, Opole, Poland
| | | | - Fernanda L Campinhos
- Asthma Reference Center, UCARE Hospital Santa Casa de Misericórdia de Vitória, Esperito Santo, Brazil
| | - Emily Carne
- Department of Dermatology, University Hospital of Wales, Cardiff, United Kingdom
| | | | - Ivan Cherrez-Ojeda
- Research Department, Universidad Espiritu Santo, Samborondon, Ecuador; Department Allergy and Pulmonology, Respiralab Research Group, Guayaquil, Ecuador
| | - Herberto Jose Chong-Neto
- Departamento de Pediatria, Serviço de Alergia e Imunologia, Complexo Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, Brazil
| | - George Christoff
- Faculty of Public Health, Medical University Sofia, Sofia, Bulgaria
| | - Niall Conlon
- School of Medicine, Trinity College Dublin, St James's Hospital Dublin, Dublin, Ireland
| | | | - Klara Cvenkel
- Department of Dermatovenereology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Ebru Damadoglu
- Department of Chest Diseases, Division of Allergy and Immunology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Inna Danilycheva
- Department of Allergology, NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| | - Cascia Day
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Laurence de Montjoye
- Department of Dermatology, Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Semra Demir
- Department of Internal Medicine, Division of Immunology and Allergy Diseases, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Silvia Mariel Ferucci
- Department of Dermatology, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico Milan, Milan, Italy
| | - Daria Fomina
- Moscow Healthcare Department, Moscow City Research and Practical Center of Allergoloy and Immunology, Moscow, Russia; Department of Clinical Immunology and Allergy, Sechenow First Moscow State Medical University, Moscow, Russia; Department of Pulmonology, Astana Medical University, Kazakhstan, Russia
| | - Atsushi Fukunaga
- Department of Dermatology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Elizabeth Garcia
- Faculty of Medicine, Universidad de los Andes-UNIMEQ ORL, Bogota, Colombia
| | - Asli Gelincik
- Department of Internal Medicine, Division of Immunology and Allergy Diseases, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Joe Hannah Göbel
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Kiran Godse
- Department of Dermatology, Dr D. Y. Patil Medical College & Hospital, Navi Mumbai, India
| | - Margarida Gonçalo
- Department of Dermatology, University Hospital and Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maia Gotua
- Center of Allergy and Immunology, David Tvildiani Medical University, Tbilisi, Georgia
| | - Clive Grattan
- Department of Dermatology, St Thomas Hospital London, London, United Kingdom
| | - Agata Gugala
- Department of Immunology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Carole Guillet
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Ali Fuat Kalyoncu
- Department of Chest Diseases, Division of Allergy and Immunology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Gul Karakaya
- Department of Chest Diseases, Division of Allergy and Immunology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Alicja Kasperska-Zając
- Department of Clinical Allergology and Urticaria, Medical University of Silesia, Silesia, Poland
| | | | - Maryam Khoshkhui
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Marta Kolacinska-Flont
- Department of Internal Medicine, Asthma, and Allergy, Barlicki Memorial Hospital, Medical University of Lodz, Lodz, Poland
| | - Pavel Kolkhir
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Mitja Košnik
- Division of Allergy, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
| | - Dorota Krasowska
- Department of Dermatology, Venereology, and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Muthu Sendhil Kumaran
- Department of Dermatology, Venereology, and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Izabela Kuprys-Lipinska
- Department of Internal Medicine, Asthma, and Allergy, Barlicki Memorial Hospital, Medical University of Lodz, Lodz, Poland
| | - Marcin Kurowski
- Department of Immunology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Elizaveta V Kuznetsova
- Department of Clinical Immunology and Allergy, Sechenow First Moscow State Medical University, Moscow, Russia
| | | | - Marina S Lebedkina
- Moscow Healthcare Department, Moscow City Research and Practical Center of Allergoloy and Immunology, Moscow, Russia
| | - Youngsoo Lee
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Michael Makris
- Allergy Unit, 2nd Department of Dermatology and Venereology, University Hospital "Attikon", National and Kapodistrian University of Athens, Athens, Greece
| | | | - Iman Nasr
- Clinical Immunology and Allergy Unit, Royal Hospital, Muscat, Oman
| | - Sophia Neisinger
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Yoshiko Oda
- Department of Dermatology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Rabia Öztaş Kara
- Department of Dermatology, University of Sakarya School of Medicine, Sakarya, Turkey
| | - Esther Bastos Palitot
- Federal University of Paraiba, Department of Infectious, Parasitic and Inflammatory Diseases, João Pessoa, Paraiba, Brazil; Lauro Wanderley University Hospital - Ebserh Network, João Pessoa, Brazil
| | - Niki Papapostolou
- Allergy Unit, 2nd Department of Dermatology and Venereology, University Hospital "Attikon", National and Kapodistrian University of Athens, Athens, Greece
| | | | - David Pesque
- Department of Dermatology, Hospital del Mar Research Institute, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jonathan Peter
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Elena Petkova
- University Hospital "Alexandrovska", Clinic of Allergology, Sofia, Bulgaria
| | - Katie Ridge
- School of Medicine, Trinity College Dublin, St James's Hospital Dublin, Dublin, Ireland
| | - Michael Rudenko
- London Allergy & Immunology Centre, Department of Allergy and Immunology, London, United Kingdom
| | - Krzysztof Rutkowski
- Urticaria Clinic, St John's Institute of Dermatology, London, United Kingdom
| | - Sarbjit S Saini
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins Asthma and Allergy Center, Baltimore, Md
| | - Andac Salman
- Department of Dermatology, Marmara University School of Medicine, Istanbul, Turkey; Department of Dermatology, School of Medicine, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Jorge Sanchez
- Group of Clinical and Experimental Allergy, Hospital "Alma Mater de Antioquia", University of Antioquia, Antioquia, Colombia
| | - Bülent Şekerel
- Pediatric Allergy Asthma Division, Hacettepe University, Ankara, Turkey
| | - Sofia A Serdotetskova
- Moscow Healthcare Department, Moscow City Research and Practical Center of Allergoloy and Immunology, Moscow, Russia
| | - Faradiba S Serpa
- Asthma Reference Center, UCARE Hospital Santa Casa de Misericórdia de Vitória, Esperito Santo, Brazil
| | | | - Nikitas Sidiropoulos
- Allergy Unit, 2nd Department of Dermatology and Venereology, University Hospital "Attikon", National and Kapodistrian University of Athens, Athens, Greece
| | - Agnieszka Sikora
- Department of Clinical Allergology and Urticaria, Medical University of Silesia, Silesia, Poland
| | - Jennifer Astrup Sørensen
- Department of Dermatology, Department of Biomedical Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Angele Soria
- Department of Dermatology and Allergy, Tenon Hospital APHP, Sorbonne Université Paris, Paris, France
| | - Ozlem Su Kucuk
- Department of Dermatology, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| | - Sherin Rahim Thalappil
- Department of Medicine, Allergy and Immunology Division, Hamad Medical Corporation, Doha, Qatar
| | | | - Gulseren Tuncay
- Department of Chest Diseases, Division of Allergy and Immunology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Derya Unal
- Department of Internal Medicine, Division of Immunology and Allergy Diseases, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Solange Valle
- Medicine Department, Immunology Service, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Esmee van Lindonk
- Department of Dermatology, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | | | - Raisa Y Meshkova
- Department of Clinical Immunology and Allergology, Smolensk State Medical University, Smolensk
| | - Aleksandr Vitchuk
- Department of Dermatology, Smolensk State Medical University, Smolensk, Russia
| | - Paraskevi Xepapadaki
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Young-Min Ye
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | | | - Mateusz Zamlynski
- Department of Clinical Allergology and Urticaria, Medical University of Silesia, Silesia, Poland
| | - Marcus Maurer
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology and Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany.
| |
Collapse
|
315
|
Kampouri E, Hill JA, Dioverti V. COVID-19 after hematopoietic cell transplantation and chimeric antigen receptor (CAR)-T-cell therapy. Transpl Infect Dis 2023; 25 Suppl 1:e14144. [PMID: 37767643 DOI: 10.1111/tid.14144] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
More than 3 years have passed since Coronavirus disease 2019 (COVID-19) was declared a global pandemic, yet COVID-19 still severely impacts immunocompromised individuals including those treated with hematopoietic cell transplantation (HCT) and chimeric antigen receptor-T-cell therapies who remain at high risk for severe COVID-19 and mortality. Despite vaccination efforts, these patients have inadequate responses due to immunosuppression, which underscores the need for additional preventive approaches. The optimal timing, schedule of vaccination, and immunological correlates for protective immunity remain unknown. Antiviral therapies used early during disease can reduce mortality and severity due to COVID-19. The combination or sequential use of antivirals could be beneficial to control replication and prevent the development of treatment-related mutations in protracted COVID-19. Despite conflicting data, COVID-19 convalescent plasma remains an option in immunocompromised patients with mild-to-moderate disease to prevent progression. Protracted COVID-19 has been increasingly recognized among these patients and has been implicated in intra-host emergence of SARS-CoV-2 variants. Finally, novel SARS-CoV2-specific T-cells and natural killer cell-boosting (or -containing) products may be active against multiple variants and are promising therapies in immunocompromised patients.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Veronica Dioverti
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
316
|
Guthrie CM, Tan X, Meeker AC, Self AE, Liu L, Cheng Y. Engineering a dual vaccine against COVID-19 and tuberculosis. Front Cell Infect Microbiol 2023; 13:1273019. [PMID: 37965265 PMCID: PMC10641007 DOI: 10.3389/fcimb.2023.1273019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2 virus, has been one of the top public health threats across the world over the past three years. Mycobacterium bovis BCG is currently the only licensed vaccine for tuberculosis, one of the deadliest infectious diseases in the world, that is caused by Mycobacterium tuberculosis. In the past decades, recombinant M.bovis BCG has been studied as a novel vaccine vector for other infectious diseases in humans besides tuberculosis, such as viral infections. In the current study, we generated a recombinant M. bovis BCG strain AspikeRBD that expresses a fusion protein consisting of M. tb Ag85A protein and the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein using synthetic biology technique. Our results show that the recombinant M. bovis BCG strain successfully expressed this fusion protein. Interestingly, the recombinant M. bovis BCG strain AspikeRBD significantly induced SARS-CoV-2 spike-specific T cell activation and IgG production in mice when compared to the parental M.bovis BCG strain, and was more potent than the recombinant M.bovis BCG strain expressing SARS-CoV-2 spike RBD alone. As expected, the recombinant M. bovis BCG strain AspikeRBD activated an increased number of M. tb Ag85A-specific IFNγ-releasing T cells and enhanced IgG production in mice when compared to the parental M.bovis BCG strain or the BCG strain expressing SARS-CoV-2 spike RBD alone. Taken together, our results indicate a potential application of the recombinant M. bovis BCG strain AspikeRBD as a novel dual vaccine against SARS-CoV-2 and M. tb in humans.
Collapse
Affiliation(s)
- Carlyn Monèt Guthrie
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Xuejuan Tan
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Amber Cherry Meeker
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Ashton Elisabeth Self
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Lin Liu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Yong Cheng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
317
|
Gennari AG, Rossi A, Sartoretti T, Maurer A, Skawran S, Treyer V, Sartoretti E, Curioni-Fontecedro A, Schwyzer M, Waelti S, Huellner MW, Messerli M. Characterization of hypermetabolic lymph nodes after SARS-CoV-2 vaccination using PET-CT derived node-RADS, in patients with melanoma. Sci Rep 2023; 13:18357. [PMID: 37884535 PMCID: PMC10603100 DOI: 10.1038/s41598-023-44215-2] [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: 01/18/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
This study aimed to evaluate the diagnostic accuracy of Node Reporting and Data System (Node-RADS) in discriminating between normal, reactive, and metastatic axillary LNs in patients with melanoma who underwent SARS-CoV-2 vaccination. Patients with proven melanoma who underwent a 2-[18F]-fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (2-[18F]-FDG PET/CT) between February and April 2021 were included in this retrospective study. Primary melanoma site, vaccination status, injection site, and 2-[18F]-FDG PET/CT were used to classify axillary LNs into normal, inflammatory, and metastatic (combined classification). An adapted Node-RADS classification (A-Node-RADS) was generated based on LN anatomical characteristics on low-dose CT images and compared to the combined classification. 108 patients were included in the study (54 vaccinated). HALNs were detected in 42 patients (32.8%), of whom 97.6% were vaccinated. 172 LNs were classified as normal, 30 as inflammatory, and 14 as metastatic using the combined classification. 152, 22, 29, 12, and 1 LNs were classified A-Node-RADS 1, 2, 3, 4, and 5, respectively. Hence, 174, 29, and 13 LNs were deemed benign, equivocal, and metastatic. The concordance between the classifications was very good (Cohen's k: 0.91, CI 0.86-0.95; p-value < 0.0001). A-Node-RADS can assist the classification of axillary LNs in melanoma patients who underwent 2-[18F]-FDG PET/CT and SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Antonio G Gennari
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Sartoretti
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Alexander Maurer
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Stephan Skawran
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Elisabeth Sartoretti
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Alessandra Curioni-Fontecedro
- University of Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
| | - Moritz Schwyzer
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Stephan Waelti
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
- Department of Radiology and Nuclear Medicine, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Martin W Huellner
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Michael Messerli
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
318
|
Carmola LR, Roebling AD, Khosravi D, Langsjoen RM, Bombin A, Bixler B, Reid A, Chen C, Wang E, Lu Y, Zheng Z, Zhang R, Nguyen PV, Arthur RA, Fitts E, Gulick DA, Higginbotham D, Taz A, Ahmed A, Crumpler JH, Kraft C, Lam WA, Babiker A, Waggoner JJ, Openo KP, Johnson LM, Westbrook A, Piantadosi A. Viral and host factors associated with SARS-CoV-2 disease severity in Georgia, USA. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.25.23297530. [PMID: 37961729 PMCID: PMC10635197 DOI: 10.1101/2023.10.25.23297530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
While SARS-CoV-2 vaccines have shown strong efficacy, their suboptimal uptake combined with the continued emergence of new viral variants raises concerns about the ongoing and future public health impact of COVID-19. We investigated viral and host factors, including vaccination status, that were associated with SARS-CoV-2 disease severity in a setting with low vaccination rates. We analyzed clinical and demographic data from 1,957 individuals in the state of Georgia, USA, coupled with viral genome sequencing from 1,185 samples. We found no difference in disease severity between individuals infected with Delta and Omicron variants among the participants in this study, after controlling for other factors, and we found no specific mutations associated with disease severity. Compared to those who were unvaccinated, vaccinated individuals experienced less severe SARS-CoV-2 disease, and the effect was similar for both variants. Vaccination within 270 days before infection was associated with decreased odds of moderate and severe outcomes, with the strongest association observed at 91-270 days post-vaccination. Older age and underlying health conditions, especially immunosuppression and renal disease, were associated with increased disease severity. Overall, this study provides insights into the impact of vaccination status, variants/mutations, and clinical factors on disease severity in SARS-CoV-2 infection when vaccination rates are low. Understanding these associations will help refine and reinforce messaging around the crucial importance of vaccination in mitigating the severity of SARS-CoV-2 disease.
Collapse
Affiliation(s)
- Ludy R. Carmola
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Allison Dorothy Roebling
- Georgia Emerging Infections Program; Georgia Department of Health; Atlanta, GA, 30303; USA
- Atlanta Veterans Affairs Medical Center; Decatur, GA, 30033; USA
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Dara Khosravi
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Rose M. Langsjoen
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Andrei Bombin
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Bri Bixler
- Graduate Program in Genetics and Molecular Biology, Emory University; Atlanta, GA, 30322; USA
| | - Alex Reid
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Cara Chen
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Ethan Wang
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Yang Lu
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Ziduo Zheng
- Department of Biostatistics and Bioinformatics; Rollins School of Public Health, Emory University; Atlanta, GA, 30322; USA
| | - Rebecca Zhang
- Department of Biostatistics and Bioinformatics; Rollins School of Public Health, Emory University; Atlanta, GA, 30322; USA
| | - Phuong-Vi Nguyen
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Robert A. Arthur
- Emory Integrated Computational Core; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Eric Fitts
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Dalia Arafat Gulick
- Georgia Clinical & Translational Science Alliance; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Dustin Higginbotham
- Georgia Clinical & Translational Science Alliance; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Azmain Taz
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Alaa Ahmed
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
- Emory Integrated Genomics Core; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - John Hunter Crumpler
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Colleen Kraft
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Wilbur A. Lam
- The Atlanta Center for Microsystems-Engineered Point-of-Care Technologies; Atlanta, GA, 30322; USA
- Department of Pediatrics, Emory University School of Medicine; Atlanta, GA, 30322; USA
- Aflac Cancer and Blood Disorders Center at Children’s Healthcare of Atlanta; Atlanta, GA, 30322; USA
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Ahmed Babiker
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Jesse J. Waggoner
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Kyle P. Openo
- Georgia Emerging Infections Program; Georgia Department of Health; Atlanta, GA, 30303; USA
- Atlanta Veterans Affairs Medical Center; Decatur, GA, 30033; USA
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| | - Laura M. Johnson
- Pediatric Biostatistics Core; Department of Pediatrics; School of Medicine; Emory University; Atlanta, GA, 30322; USA
| | - Adrianna Westbrook
- Pediatric Biostatistics Core; Department of Pediatrics; School of Medicine; Emory University; Atlanta, GA, 30322; USA
| | - Anne Piantadosi
- Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA, 30322; USA
- Division of Infectious Diseases; Department of Medicine, Emory University School of Medicine; Atlanta, GA, 30322; USA
| |
Collapse
|
319
|
Mathew DS, Pandya T, Pandya H, Vaghela Y, Subbian S. An Overview of SARS-CoV-2 Etiopathogenesis and Recent Developments in COVID-19 Vaccines. Biomolecules 2023; 13:1565. [PMID: 38002247 PMCID: PMC10669259 DOI: 10.3390/biom13111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 11/26/2023] Open
Abstract
The Coronavirus disease-2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has significantly impacted the health and socioeconomic status of humans worldwide. Pulmonary infection of SARS-CoV-2 results in exorbitant viral replication and associated onset of inflammatory cytokine storm and disease pathology in various internal organs. However, the etiopathogenesis of SARS-CoV-2 infection is not fully understood. Currently, there are no targeted therapies available to cure COVID-19, and most patients are treated empirically with anti-inflammatory and/or anti-viral drugs, based on the disease symptoms. Although several types of vaccines are currently implemented to control COVID-19 and prevent viral dissemination, the emergence of new variants of SARS-CoV-2 that can evade the vaccine-induced protective immunity poses challenges to current vaccination strategies and highlights the necessity to develop better and improved vaccines. In this review, we summarize the etiopathogenesis of SARS-CoV-2 and elaborately discuss various types of vaccines and vaccination strategies, focusing on those vaccines that are currently in use worldwide to combat COVID-19 or in various stages of clinical development to use in humans.
Collapse
Affiliation(s)
- Dona Susan Mathew
- Department of Microbiology, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 608204, India;
| | - Tirtha Pandya
- Public Health Research Institute (PHRI) Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (T.P.); (H.P.); (Y.V.)
| | - Het Pandya
- Public Health Research Institute (PHRI) Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (T.P.); (H.P.); (Y.V.)
| | - Yuzen Vaghela
- Public Health Research Institute (PHRI) Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (T.P.); (H.P.); (Y.V.)
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI) Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (T.P.); (H.P.); (Y.V.)
| |
Collapse
|
320
|
Lee JM, Figueroa A, Sachithanandham J, Li M, Connolly CM, Shapiro JR, Chen Y, Jones M, Dhara VG, Towns M, Lee JS, Peralta SR, Milstone AM, Betenbaugh M, Debes AK, Blankson J, Sitaras I, Yoon S, Thompson EA, Bingham CO, Klein SL, Pekosz A, Bailey JR. Three doses of COVID-19 mRNA vaccine induce class-switched antibody responses in inflammatory arthritis patients on immunomodulatory therapies. Front Immunol 2023; 14:1266370. [PMID: 38022602 PMCID: PMC10646683 DOI: 10.3389/fimmu.2023.1266370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Patients with inflammatory arthritis (IA) are at increased risk of severe COVID-19 due to medication-induced immunosuppression that impairs host defenses. The aim of this study was to assess antibody and B cell responses to COVID-19 mRNA vaccination in IA patients receiving immunomodulatory therapies. Adults with IA were enrolled through the Johns Hopkins Arthritis Center and compared with healthy controls (HC). Paired plasma and peripheral blood mononuclear cell (PBMC) samples were collected prior to and 30 days or 6 months following the first two doses of mRNA vaccines (D2; HC=77 and IA=31 patients), or 30 days following a third dose of mRNA vaccines (D3; HC=11 and IA=96 patients). Neutralizing antibody titers, total binding antibody titers, and B cell responses to vaccine and Omicron variants were analyzed. Anti-Spike (S) IgG and S-specific B cells developed appropriately in most IA patients following D3, with reduced responses to Omicron variants, and negligible effects of medication type or drug withholding. Neutralizing antibody responses were lower compared to healthy controls after both D2 and D3, with a small number of individuals demonstrating persistently undetectable neutralizing antibody levels. Most IA patients respond as well to mRNA COVID-19 vaccines as immunocompetent individuals by the third dose, with no evidence of improved responses following medication withholding. These data suggest that IA-associated immune impairment may not hinder immunity to COVID-19 mRNA vaccines in most individuals.
Collapse
Affiliation(s)
- Jenny M. Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Alexis Figueroa
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jaiprasath Sachithanandham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Maggie Li
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Caoilfhionn M. Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Janna R. Shapiro
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Yiqun Chen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Michelle Jones
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Venkata Gayatri Dhara
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Marilyn Towns
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - John S. Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Stephanie R. Peralta
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Aaron M. Milstone
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Amanda K. Debes
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Joel Blankson
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Steve Yoon
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Elizabeth A. Thompson
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Clifton O. Bingham
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Justin R. Bailey
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
321
|
Aid M, Stephenson KE, Collier ARY, Nkolola JP, Michael JV, McKenzie SE, Barouch DH. Activation of coagulation and proinflammatory pathways in thrombosis with thrombocytopenia syndrome and following COVID-19 vaccination. Nat Commun 2023; 14:6703. [PMID: 37872311 PMCID: PMC10593859 DOI: 10.1038/s41467-023-42559-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
Thrombosis with thrombocytopenia syndrome (TTS) is a rare but potentially severe adverse event following immunization with adenovirus vector-based COVID-19 vaccines such as Ad26.COV2.S (Janssen) and ChAdOx1 (AstraZeneca). However, no case of TTS has been reported in over 1.5 million individuals who received a second immunization with Ad26.COV2.S in the United States. Here we utilize transcriptomic and proteomic profiling to compare individuals who receive two doses of Ad26.COV2.S with those vaccinated with BNT162b2 or mRNA-1273. Initial Ad26.COV2.S vaccination induces transient activation of platelet and coagulation and innate immune pathways that resolve by day 7; by contrast, patients with TTS show robust upregulation of these pathways on days 15-19 following initial Ad26.COV2.S vaccination. Meanwhile, a second immunization or a reduced initial dose of Ad26.COV2.S induces lower activation of these pathways than does the full initial dose. Our data suggest a role of coagulation and proinflammatory pathways in TTS pathogenesis, which may help optimize vaccination regimens to reduce TTS risk.
Collapse
Affiliation(s)
- Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ai-Ris Y Collier
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joseph P Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - James V Michael
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Steven E McKenzie
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
322
|
Wallace M, Rosenblum HG, Moulia DL, Broder KR, Shimabukuro TT, Taylor CA, Havers FP, Meyer SA, Dooling K, Oliver SE, Hadler SC, Gargano JW. A summary of the Advisory Committee for Immunization Practices (ACIP) use of a benefit-risk assessment framework during the first year of COVID-19 vaccine administration in the United States. Vaccine 2023; 41:6456-6467. [PMID: 37527956 PMCID: PMC11068153 DOI: 10.1016/j.vaccine.2023.07.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/01/2023] [Accepted: 07/18/2023] [Indexed: 08/03/2023]
Abstract
To inform Advisory Committee for Immunization Practices (ACIP) COVID-19 vaccine policy decisions, we developed a benefit-risk assessment framework that directly compared the estimated benefits of COVID-19 vaccination to individuals (e.g., prevention of COVID-19-associated hospitalization) with risks associated with COVID-19 vaccines. This assessment framework originated following the identification of thrombosis with thrombocytopenia syndrome (TTS) after Janssen COVID-19 vaccination in April 2021. We adapted the benefit-risk assessment framework for use in subsequent policy decisions, including the adverse events of myocarditis and Guillain-Barre syndrome (GBS) following mRNA and Janssen COVID-19 vaccination respectively, expansion of COVID-19 vaccine approvals or authorizations to new age groups, and use of booster doses. Over the first year of COVID-19 vaccine administration in the United States (December 2020-December 2021), we used the benefit-risk assessment framework to inform seven different ACIP policy decisions. This framework allowed for rapid and direct comparison of the benefits and potential harms of vaccination, which may be helpful in informing other vaccine policy decisions. The assessments were a useful tool for decision-making but required reliable and granular data to stratify analyses and appropriately focus on populations most at risk for a specific adverse event. Additionally, careful decision-making was needed on parameters for data inputs. Sensitivity analyses were used where data were limited or uncertain; adjustments in the methodology were made over time to ensure the assessments remained relevant and applicable to the policy questions under consideration.
Collapse
Affiliation(s)
- Megan Wallace
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States.
| | - Hannah G Rosenblum
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Danielle L Moulia
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Karen R Broder
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Tom T Shimabukuro
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Christopher A Taylor
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Fiona P Havers
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Sarah A Meyer
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Kathleen Dooling
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Sara E Oliver
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Stephen C Hadler
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Julia W Gargano
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| |
Collapse
|
323
|
Karl T, Schuster A, Stangassinger LM, Stiboller T, Cadamuro J, Oostingh GJ. Factors Affecting SARS-CoV-2 IgG Production after Vaccination and/or Disease: A Large-Scale Seroprevalence Study. Vaccines (Basel) 2023; 11:1615. [PMID: 37897017 PMCID: PMC10611123 DOI: 10.3390/vaccines11101615] [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: 09/19/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
This study aimed at identifying factors influencing SARS-CoV-2-specific IgG antibody levels after vaccination and/or infection. Between January 2022 and March 2023, 2000 adults (≥18 years, Salzburg, Austria) participated in this population-based seroprevalence study by providing 3 mL of blood to detect SARS-CoV-2-specific IgG antibodies using an anti-SARS-CoV-2 IgG quantitative assay and by completing a self-designed questionnaire including anthropometric factors, vaccination information, and medical history. For 77 of the participants, a time-course study up to 24 weeks post vaccination or quarantine end was performed. Convalescent-only subjects had the lowest median antibody titer (65.6 BAU/mL) compared to vaccinated and hybrid immunized subjects (p-value < 0.0001) The type of vaccine as well as vaccine combinations significantly influenced the levels of SARS-CoV-2 spike-protein-specific IgG, ranging from a median antibody level of 770.5 BAU/mL in subjects who were vaccinated only to 3020.0 BAU/mL in hybrid immunized subjects (p-value < 0.0001). Over time, a significant decline in the levels of neutralizing antibodies was found. Depending on the subpopulation analyzed, further significant influencing factors included sex assigned at birth, disease severity, chronic diseases, and medication. A hybrid immunization resulted in more robust immune responses. Nevertheless, there were multiple other factors impacting these responses. This knowledge should be included in future vaccination strategies and serve as a guide in the development of personalized medicine.
Collapse
Affiliation(s)
- Tanja Karl
- Department of Health Sciences, Biomedical Sciences, Salzburg University of Applied Sciences, 5412 Puch/Salzburg, Austria; (A.S.); (L.M.S.); (T.S.); (G.J.O.)
- Research Program of Medical Sciences, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Anja Schuster
- Department of Health Sciences, Biomedical Sciences, Salzburg University of Applied Sciences, 5412 Puch/Salzburg, Austria; (A.S.); (L.M.S.); (T.S.); (G.J.O.)
| | - Lea Maria Stangassinger
- Department of Health Sciences, Biomedical Sciences, Salzburg University of Applied Sciences, 5412 Puch/Salzburg, Austria; (A.S.); (L.M.S.); (T.S.); (G.J.O.)
| | - Tanja Stiboller
- Department of Health Sciences, Biomedical Sciences, Salzburg University of Applied Sciences, 5412 Puch/Salzburg, Austria; (A.S.); (L.M.S.); (T.S.); (G.J.O.)
| | - Janne Cadamuro
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Gertie Janneke Oostingh
- Department of Health Sciences, Biomedical Sciences, Salzburg University of Applied Sciences, 5412 Puch/Salzburg, Austria; (A.S.); (L.M.S.); (T.S.); (G.J.O.)
| |
Collapse
|
324
|
Lécuyer D, Nardacci R, Tannous D, Gutierrez-Mateyron E, Deva Nathan A, Subra F, Di Primio C, Quaranta P, Petit V, Richetta C, Mostefa-Kara A, Del Nonno F, Falasca L, Marlin R, Maisonnasse P, Delahousse J, Pascaud J, Deprez E, Naigeon M, Chaput N, Paci A, Saada V, Ghez D, Mariette X, Costa M, Pistello M, Allouch A, Delelis O, Piacentini M, Le Grand R, Perfettini JL. The purinergic receptor P2X7 and the NLRP3 inflammasome are druggable host factors required for SARS-CoV-2 infection. Front Immunol 2023; 14:1270081. [PMID: 37920468 PMCID: PMC10619763 DOI: 10.3389/fimmu.2023.1270081] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
Purinergic receptors and NOD-like receptor protein 3 (NLRP3) inflammasome regulate inflammation and viral infection, but their effects on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection remain poorly understood. Here, we report that the purinergic receptor P2X7 and NLRP3 inflammasome are cellular host factors required for SARS-CoV-2 infection. Lung autopsies from patients with severe coronavirus disease 2019 (COVID-19) reveal that NLRP3 expression is increased in host cellular targets of SARS-CoV-2 including alveolar macrophages, type II pneumocytes and syncytia arising from the fusion of infected macrophages, thus suggesting a potential role of NLRP3 and associated signaling pathways to both inflammation and viral replication. In vitro studies demonstrate that NLRP3-dependent inflammasome activation is detected upon macrophage abortive infection. More importantly, a weak activation of NLRP3 inflammasome is also detected during the early steps of SARS-CoV-2 infection of epithelial cells and promotes the viral replication in these cells. Interestingly, the purinergic receptor P2X7, which is known to control NLRP3 inflammasome activation, also favors the replication of D614G and alpha SARS-CoV-2 variants. Altogether, our results reveal an unexpected relationship between the purinergic receptor P2X7, the NLRP3 inflammasome and the permissiveness to SARS-CoV-2 infection that offers novel opportunities for COVID-19 treatment.
Collapse
Affiliation(s)
- Déborah Lécuyer
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| | - Roberta Nardacci
- National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
- UniCamillus - Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Désirée Tannous
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
- NH TherAguix SAS, Meylan, France
| | - Emie Gutierrez-Mateyron
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| | - Aurélia Deva Nathan
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| | - Frédéric Subra
- Université Paris-Saclay, ENS Paris-Saclay, CNRS UMR 8113, IDA FR3242, Laboratory of Biology and Applied Pharmacology (LBPA), Gif-sur-Yvette, France
| | - Cristina Di Primio
- Institute of Neuroscience, Italian National Research Council, Pisa, Italy
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Paola Quaranta
- Institute of Neuroscience, Italian National Research Council, Pisa, Italy
- Retrovirus Center, Department of Translational Research, Universita of Pisa, Pisa, Italy
| | - Vanessa Petit
- Université Paris-Saclay, Inserm U1274, CEA, Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
| | - Clémence Richetta
- Université Paris-Saclay, ENS Paris-Saclay, CNRS UMR 8113, IDA FR3242, Laboratory of Biology and Applied Pharmacology (LBPA), Gif-sur-Yvette, France
| | - Ali Mostefa-Kara
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| | - Franca Del Nonno
- National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Laura Falasca
- National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Romain Marlin
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA- HB/IDMIT), Fontenay-aux-Roses, France
| | - Pauline Maisonnasse
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA- HB/IDMIT), Fontenay-aux-Roses, France
| | - Julia Delahousse
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| | - Juliette Pascaud
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA- HB/IDMIT), Fontenay-aux-Roses, France
- Assistance Publique, Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Eric Deprez
- Université Paris-Saclay, ENS Paris-Saclay, CNRS UMR 8113, IDA FR3242, Laboratory of Biology and Applied Pharmacology (LBPA), Gif-sur-Yvette, France
| | - Marie Naigeon
- Gustave Roussy Cancer Center, Villejuif, France
- Université Paris-Saclay, Inserm, CNRS, Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse, Laboratoire d'Immunomonitoring en Oncologie, Villejuif, France
- Université Paris-Saclay, Faculté de Pharmacie, Chatenay-Malabry, France
| | - Nathalie Chaput
- Université Paris-Saclay, Inserm, CNRS, Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse, Laboratoire d'Immunomonitoring en Oncologie, Villejuif, France
- Université Paris-Saclay, Faculté de Pharmacie, Chatenay-Malabry, France
- Université Paris-Saclay, Gustave Roussy Cancer Center, CNRS, Stabilité Génétique et Oncogenèse, Villejuif, France
| | - Angelo Paci
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
- Université Paris-Saclay, Faculté de Pharmacie, Chatenay-Malabry, France
- Department of Biology and Pathology, Gustave Roussy Cancer Center, Villejuif, France
| | - Véronique Saada
- Department of Biology and Pathology, Gustave Roussy Cancer Center, Villejuif, France
| | - David Ghez
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Xavier Mariette
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA- HB/IDMIT), Fontenay-aux-Roses, France
- Assistance Publique, Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, Le Kremlin Bicêtre, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France
| | - Mario Costa
- Institute of Neuroscience, Italian National Research Council, Pisa, Italy
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- Centro Pisano Ricerca e Implementazione Clinical Flash Radiotherapy "CPFR@CISUP", "S. Chiara" Hospital, Pisa, Italy
| | - Mauro Pistello
- Retrovirus Center, Department of Translational Research, Universita of Pisa, Pisa, Italy
- Virology Operative Unit, Pisa University Hospital, Pisa, Italy
| | - Awatef Allouch
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
- NH TherAguix SAS, Meylan, France
| | - Olivier Delelis
- Université Paris-Saclay, ENS Paris-Saclay, CNRS UMR 8113, IDA FR3242, Laboratory of Biology and Applied Pharmacology (LBPA), Gif-sur-Yvette, France
| | - Mauro Piacentini
- National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA- HB/IDMIT), Fontenay-aux-Roses, France
| | - Jean-Luc Perfettini
- Université Paris-Saclay, Inserm UMR1030, Laboratory of Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| |
Collapse
|
325
|
Wang X, Hetzel M, Zhang W, Ehrhardt A, Bayer W. Comparative analysis of the impact of 40 adenovirus types on dendritic cell activation and CD8 + T cell proliferation capacity for the identification of favorable immunization vector candidates. Front Immunol 2023; 14:1286622. [PMID: 37915567 PMCID: PMC10616870 DOI: 10.3389/fimmu.2023.1286622] [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: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
For the development of new adenovirus (AdV)-based vectors, it is important to understand differences in immunogenicity. In a side-by-side in vitro analysis, we evaluated the effect of 40 AdV types covering human AdV (HAdV) species A through G on the expression of 11 activation markers and the secretion of 12 cytokines by AdV-transduced dendritic cells, and the effect on CD8+ T cell proliferation capacity. We found that the expression of activation markers and cytokines differed widely between the different HAdV types, and many types were able to significantly impair the proliferation capacity of CD8+ T cells. Univariate and multivariate regression analyses suggested an important role of type I interferons in mediating this suppression of CD8+ T cells, which we confirmed experimentally in a proliferation assay using a type I interferon receptor blocking antibody. Using Bayesian statistics, we calculated a prediction model that suggests HAdV types HAdV-C1, -D8, -B7, -F41, -D33, -C2, -A31, -B3 and -D65 as the most favorable candidates for vaccine vector development.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mario Hetzel
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
326
|
Sekulovski M, Mileva N, Vasilev GV, Miteva D, Gulinac M, Peshevska-Sekulovska M, Chervenkov L, Batselova H, Vasilev GH, Tomov L, Lazova S, Vassilev D, Velikova T. Blood Coagulation and Thrombotic Disorders following SARS-CoV-2 Infection and COVID-19 Vaccination. Biomedicines 2023; 11:2813. [PMID: 37893186 PMCID: PMC10604891 DOI: 10.3390/biomedicines11102813] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Although abundant data confirm the efficacy and safety profile of the developed vaccines against COVID-19, there are still some concerns regarding vaccination in high-risk populations. This is especially valid for patients susceptible to thrombotic or bleeding events and hesitant people due to the fear of thrombotic incidents following vaccination. This narrative review focuses on various inherited and acquired thrombotic and coagulation disorders and the possible pathophysiologic mechanisms interacting with the coagulation system during immunization in view of the currently available safety data regarding COVID-19 vaccines. Inherited blood coagulation disorders and inherited thrombotic disorders in the light of COVID-19, as well as blood coagulation and thrombotic disorders and bleeding complications following COVID-19 vaccines, along with the possible pathogenesis hypotheses, therapeutic interventions, and imaging for diagnosing are discussed in detail. Lastly, the lack of causality between the bleeding and thrombotic events and COVID-19 vaccines is debated, but still emphasizes the importance of vaccination against COVID-19, outweighing the minimal risk of potential rare adverse events associated with coagulation.
Collapse
Affiliation(s)
- Metodija Sekulovski
- Department of Anesthesiology and Intensive Care, University Hospital Lozenetz, Kozyak Str., 1407 Sofia, Bulgaria
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
| | - Niya Mileva
- Medical Faculty, Medical University of Sofia, 1 Georgi Sofiiski Str., 1431 Sofia, Bulgaria;
| | - Georgi Vasilev Vasilev
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
- Clinic of Endocrinology and Metabolic Disorders, University Multiprofil Hospital Active Treatement “Sv. Georgi”, 4000 Plovdiv, Bulgaria
| | - Dimitrina Miteva
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
- Department of General and Clinical Pathology, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Monika Peshevska-Sekulovska
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
- Department of Gastroenterology, University Hospital Lozenetz, 1407 Sofia, Bulgaria
| | - Lyubomir Chervenkov
- Department of Diagnostic Imaging, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria;
| | - Hristiana Batselova
- Department of Epidemiology and Disaster Medicine, Medical University of Plovdiv, University Hospital “St George”, 4000 Plovdiv, Bulgaria;
| | - Georgi Hristov Vasilev
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
- Laboratory of Hematopathology and Immunology, National Specialized Hospital for Active Treatment of Hematological Diseases, 1756 Sofia, Bulgaria
| | - Latchezar Tomov
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
- Department of Informatics, New Bulgarian University, Montevideo 21 Str., 1618 Sofia, Bulgaria
| | - Snezhina Lazova
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
- Pediatric Clinic, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben” Blvd, 1606 Sofia, Bulgaria
- Department of Healthcare, Faculty of Public Health “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Bialo More 8 Str., 1527 Sofia, Bulgaria
| | - Dobrin Vassilev
- Faculty of Public Health and Healthcare, Ruse University Angel Kanchev, 7017 Ruse, Bulgaria;
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University, St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (G.V.V.); (D.M.); (M.G.); (M.P.-S.); (G.H.V.); (L.T.); (S.L.); (T.V.)
| |
Collapse
|
327
|
Gouda MA, AboShabaan HS, Abdelgawad AS, Abdel Wahed AS, A Abd El-Razik K, Elsaadawy Y, Abdel-Wahab AA, Hawash Y. Association between breakthrough infection with COVID-19 and Toxoplasma gondii: a cross-sectional study. Sci Rep 2023; 13:17636. [PMID: 37848511 PMCID: PMC10582182 DOI: 10.1038/s41598-023-44616-3] [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: 06/14/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023] Open
Abstract
The breakthrough infection following COVID-19 vaccination has been a subject of concern recently. Evidence suggests that COVID-19 vaccine efficacy diminishes over time due to multiple factors related to the host, and vaccine. Coinfection with other pathogens was claimed earlier as a contributing cause for this phenomenon. Hence, we aimed to stratify the association of post-COVID-19 vaccination breakthrough coinfection with Toxoplasma gondii (T. gondii) and its impact on disease severity. This cross-sectional study included 330 COVID-19-vaccinated patients confirmed by RT-PCR. They were also screened for anti- T. gondii antibodies using ELISA. Toxoplasma seropositive cases' whole blood was screened for DNA using PCR to correlate results with COVID-19 severity. Out of 330 COVID-19 vaccinated patients with breakthrough infection, 34.5% (114 patients) showed positivity for Toxoplasma IgG by ELISA, and none of the cases was IgM positive. Eleven patients (9.6%) of the IgG-positive cases were positive by PCR. Positive PCR cases correlated positively with the Toxoplasma IgG titer (P < 0.001), and the Cutoff point was 191.5. Molecular analysis of Toxoplasma and COVID-19 severity showed that 8 (72.7%), 1 (9.1%), and 2 cases (18.2%) had mild, moderate, and severe courses of the disease, respectively, with no significant correlation. Our study reported a heightened prevalence of latent toxoplasmosis among mild cases of COVID-19 breakthrough infection. Nevertheless, a discernible correlation between latent toxoplasmosis and COVID-19 severity is lacking. Hence, implementing studies on a larger scale could provide a more comprehensive comprehension of this association.
Collapse
Affiliation(s)
- Marwa A Gouda
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shibin El Kom, Menoufia, Egypt.
| | - Hind S AboShabaan
- Department of Clinical Pathology, National Liver Institute, Menoufia University, Shibin El Kom, Menoufia, Egypt
| | - Ahmed S Abdelgawad
- Department of Clinical Pathology, National Liver Institute, Menoufia University, Shibin El Kom, Menoufia, Egypt
| | - Aliaa Sabry Abdel Wahed
- Department of Hepatology and Gastroenterology, National Liver Institute, Menoufia University, Shibin El Kom, Menoufia, Egypt
| | - Khaled A Abd El-Razik
- Department of Animal Reproduction, National Research Centre (NRC), Dokki, Giza, Egypt
| | - Yara Elsaadawy
- Department of Medical Microbiology, Immunology, and Infection Control, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ayman A Abdel-Wahab
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shibin El Kom, Menoufia, Egypt
| | - Yousry Hawash
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shibin El Kom, Menoufia, Egypt
| |
Collapse
|
328
|
Harel R, Itchaki G. COVID-19 in Patients with Chronic Lymphocytic Leukemia: What Have We Learned? Acta Haematol 2023; 147:60-72. [PMID: 37820599 PMCID: PMC11251671 DOI: 10.1159/000534540] [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: 05/10/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) is a prevalent hematological malignancy (HM) characterized by inherent immunodeficiency, which is further pronounced by disease-directed therapy. The COVID-19 pandemic has had devastating outcomes, and although its impact has diminished over time, it continues to be a cause of significant morbidity and mortality, particularly among immunodeficient patients. SUMMARY In this review, we describe mechanisms of immune dysfunction in CLL in relation to COVID-19, provide an overview of the clinical outcomes of the disease in this patient population, and identify risk factors associated with severe morbidity and mortality. Additionally, we acknowledge the influence of the rapidly evolving landscape of new disease variants. The review further delineates the humoral and cellular responses to vaccination and their clinical efficacy in preventing COVID-19 in CLL patients. Moreover, we explore potential approaches to enhance these immune responses. Pre- and post-exposure prophylaxis strategies are discussed, along with description of common agents in the treatment of the disease in both outpatient and inpatient setting. Throughout the review, we emphasize the interplay between novel therapies for CLL and COVID-19 outcomes, prevention, and treatment and describe the impact of COVID-19 on the utilization of these novel agents. This information has the potential to guide clinical decision making in the management CLL patients. KEY MESSAGES CLL patients are at risk for severe COVID-19 infection. Vaccinations and COVID-19 directed therapy have improved outcomes in patients with CLL, yet clinical challenges persist.
Collapse
Affiliation(s)
- Reut Harel
- Department of Hematology, Emek Medical Center, Afula, Israel
| | - Gilad Itchaki
- Hematology, Meir Medical Center, Kefar Sava, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
329
|
Sedighikamal H, Sattarzadeh A, Karimi Mostofi R, Dinarvand B, Nazarpour M. High-Titer Recombinant Adenovirus 26 Vector GMP Manufacturing in HEK 293 Cells with a Stirred Single-Use Bioreactor for COVID-19 Vaccination Purposes. ACS OMEGA 2023; 8:36720-36728. [PMID: 37841195 PMCID: PMC10568722 DOI: 10.1021/acsomega.3c03007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 virus) pandemic has shown the importance of pursuing various vaccine manufacturing strategies. In the present study, the HEK 293 cells were infected with recombinant adenovirus serotype 26 (rAd26), and the effects of critical process parameters (CPPs) including viable cell density (VCD) at infection time (0.5 × 106, 0.8 × 106, 1.4 × 106, 1.8 × 106, and 2.5 × 106 cells/mL), the multiplicity of infection (MOI) = 3, 6, 9, 12, and 15, and two aeration strategies (high-speed agitation with a sparging system and low-speed agitation with an overlay system) were investigated experimentally. The results of small-scale experiments in 2 L shake flasks (SF 2L) demonstrated that the initial VCD and MOI could affect the cell proliferation and viability. The results at these experiments showed that VCD = 1.4 × 106 cells/mL and MOI = 9 yielded TCID50 /mL = 108.9, at 72 h post-infection (hpi), while the virus titer at VCD = 0.5 × 106 and 0.8 × 106 cells/mL was lower compared to that of VCD = 1.4 × 106 cells/mL. Moreover, our findings showed that VCDs > 1.8 × 106 cells/m with MOI = 9 did not have a positive effect on TCID50 /mL and MOI = 3 and 6 were less efficient, whereas MOI > 12 decreased the viability drastically. In the next step, the optimized CPPs in a small scale were exploited in a 200 L single-use bioreactor (SUB), with good manufacturing practice (GMP) conditions, at RPM = 25 with an overlay system, yielding high-titer rAd26 manufacturing, i.e., TCID50/mL = 108.9, at 72 hpi.
Collapse
Affiliation(s)
- Hossein Sedighikamal
- API
Production Plant, Actoverco Biotech Company, Alborz 331325489, Iran
- Division
of Industrial Biotechnology, Department of Chemical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | | | - Reza Karimi Mostofi
- API
Production Plant, Actoverco Biotech Company, Alborz 331325489, Iran
- Department
of Pharmaceutics, Faculty of Pharmacy, Tehran
University of Medical Sciences, Tehran 8741253641, Iran
| | | | - Madineh Nazarpour
- API
Production Plant, Actoverco Biotech Company, Alborz 331325489, Iran
| |
Collapse
|
330
|
Chien KS, Peterson CB, Young E, Chihara D, Manasanch EE, Ramdial JL, Thompson PA. Outcomes of breakthrough COVID-19 infections in patients with hematologic malignancies. Blood Adv 2023; 7:5691-5697. [PMID: 36696472 PMCID: PMC9896882 DOI: 10.1182/bloodadvances.2022008827] [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: 08/31/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Patients with hematologic malignancies have both an increased risk for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and higher morbidity/mortality. They have lower seroconversion rates after vaccination, potentially leading to inferior coronavirus disease 2019 (COVID-19) outcomes, despite vaccination. We consequently evaluated the clinical outcomes of COVID-19 infections in 243 vaccinated and 175 unvaccinated patients with hematologic malignancies. Hospitalization rates were lower in the vaccinated group when compared with the unvaccinated group (31.3% vs 52.6%). However, the rates of COVID-19-associated death were similar at 7.0% and 8.6% in vaccinated and unvaccinated patients, respectively. By univariate logistic regression, females, older patients, and individuals with higher modified Charlson Comorbidity Index scores were at a higher risk of death from COVID-19 infections. To account for the nonrandomized nature of COVID-19 vaccination status, a propensity score weighting approach was used. In the final propensity-weighted model, vaccination status was not significantly associated with the risk of death from COVID-19 infections but was associated with the risk of hospitalization. The predicted benefit of vaccination was an absolute decrease in the probability of death and hospitalization from COVID-19 infections by 2.3% and 22.9%, respectively. In conclusion, COVID-19 vaccination status in patients with hematologic malignancies was associated with a decreased risk of hospitalization but not associated with a decreased risk of death from COVID-19 infections in the pre-Omicron era. Protective strategies, in addition to immunization, are warranted in this vulnerable patient population.
Collapse
Affiliation(s)
- Kelly S. Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christine B. Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elliana Young
- Department of Enterprise Data Engineering and Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dai Chihara
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elizabet E. Manasanch
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeremy L. Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Philip A. Thompson
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
331
|
Xu J, Zhang Y, Qu P, Shamseldin MM, Yoo SJ, Misny J, Thongpan I, KC M, Hall JM, Evans JP, Eltobgy M, Lu M, Ye C, Chamblee M, Liang X, Martinez-Sobrido L, Amer AO, Yount JS, Boyaka PN, Peeples ME, Liu SL, Dubey P, Li J. A next-generation intranasal trivalent MMS vaccine induces durable and broad protection against SARS-CoV-2 variants of concern. Proc Natl Acad Sci U S A 2023; 120:e2220403120. [PMID: 37796985 PMCID: PMC10576135 DOI: 10.1073/pnas.2220403120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/24/2023] [Indexed: 10/07/2023] Open
Abstract
As SARS-CoV-2 variants of concern (VoCs) that evade immunity continue to emerge, next-generation adaptable COVID-19 vaccines which protect the respiratory tract and provide broader, more effective, and durable protection are urgently needed. Here, we have developed one such approach, a highly efficacious, intranasally delivered, trivalent measles-mumps-SARS-CoV-2 spike (S) protein (MMS) vaccine candidate that induces robust systemic and mucosal immunity with broad protection. This vaccine candidate is based on three components of the MMR vaccine, a measles virus Edmonston and the two mumps virus strains [Jeryl Lynn 1 (JL1) and JL2] that are known to provide safe, effective, and long-lasting protective immunity. The six proline-stabilized prefusion S protein (preS-6P) genes for ancestral SARS-CoV-2 WA1 and two important SARS-CoV-2 VoCs (Delta and Omicron BA.1) were each inserted into one of these three viruses which were then combined into a trivalent "MMS" candidate vaccine. Intranasal immunization of MMS in IFNAR1-/- mice induced a strong SARS-CoV-2-specific serum IgG response, cross-variant neutralizing antibodies, mucosal IgA, and systemic and tissue-resident T cells. Immunization of golden Syrian hamsters with MMS vaccine induced similarly high levels of antibodies that efficiently neutralized SARS-CoV-2 VoCs and provided broad and complete protection against challenge with any of these VoCs. This MMS vaccine is an efficacious, broadly protective next-generation COVID-19 vaccine candidate, which is readily adaptable to new variants, built on a platform with a 50-y safety record that also protects against measles and mumps.
Collapse
Affiliation(s)
- Jiayu Xu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Yuexiu Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Panke Qu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Mohamed M. Shamseldin
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Helwan11795, Egypt
| | - Sung J. Yoo
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Jack Misny
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205
| | - Ilada Thongpan
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205
| | - Mahesh KC
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205
| | - Jesse M. Hall
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - John P. Evans
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Chengjin Ye
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227
| | - Michelle Chamblee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Luis Martinez-Sobrido
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227
| | - Amal O. Amer
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| | - Prosper N. Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| | - Mark E. Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Shan-Lu Liu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
- Center for Retrovirus Research, The Ohio State University, Columbus, OH43210
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
332
|
Patel B, Chapman SA, Neumann JT, Visaria A, Ogungbe O, Wen S, Khodaverdi M, Makwana P, Singh JA, Sokos G. Outcomes of patients with active cancers and pre-existing cardiovascular diseases infected with SARS-CoV-2. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:36. [PMID: 37803479 PMCID: PMC10557272 DOI: 10.1186/s40959-023-00187-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVE To determine the impact of acute SARS-CoV-2 infection on patient with concomitant active cancer and CVD. METHODS The researchers extracted and analyzed data from the National COVID Cohort Collaborative (N3C) database between January 1, 2020, and July 22, 2022. They included only patients with acute SARS-CoV-2 infection, defined as a positive test by PCR 21 days before and 5 days after the day of index hospitalization. Active cancers were defined as last cancer drug administered within 30 days of index admission. The "Cardioonc" group consisted of patients with CVD and active cancers. The cohort was divided into four groups: (1) CVD (-), (2) CVD ( +), (3) Cardioonc (-), and (4) Cardioonc ( +), where (-) or ( +) denotes acute SARS-CoV-2 infection status. The primary outcome of the study was major adverse cardiovascular events (MACE), including acute stroke, acute heart failure, myocardial infarction, or all-cause mortality. The researchers analyzed the outcomes by different phases of the pandemic and performed competing-risk analysis for other MACE components and death as a competing event. RESULTS The study analyzed 418,306 patients, of which 74%, 10%, 15.7%, and 0.3% had CVD (-), CVD ( +), Cardioonc (-), and Cardioonc ( +), respectively. The Cardioonc ( +) group had the highest MACE events in all four phases of the pandemic. Compared to CVD (-), the Cardioonc ( +) group had an odds ratio of 1.66 for MACE. However, during the Omicron era, there was a statistically significant increased risk for MACE in the Cardioonc ( +) group compared to CVD (-). Competing risk analysis showed that all-cause mortality was significantly higher in the Cardioonc ( +) group and limited other MACE events from occurring. When the researchers identified specific cancer types, patients with colon cancer had higher MACE. CONCLUSION In conclusion, the study found that patients with both CVD and active cancer suffered relatively worse outcomes when they had acute SARS-CoV-2 infection during early and alpha surges in the United States. These findings highlight the need for improved management strategies and further research to better understand the impact of the virus on vulnerable populations during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Brijesh Patel
- Heart and Vascular Institute, West Virginia University, Morgantown, WV, USA.
- WVU School of Medicine, Non-Invasive Cardiologist and Cardio-Oncology, WVU Heart & Vascular Institute, 1 Medical Center Drive, Box 8500, Morgantown, WV, 26505, USA.
| | - Scott A Chapman
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Jake T Neumann
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Aayush Visaria
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | | | - Sijin Wen
- Department of Biostatistics, School of Public Health, West Virginia University, Morgantown, WV, USA
| | - Maryam Khodaverdi
- West Virginia Clinical and Transitional Science Institute, Morgantown, WV, USA
| | - Priyal Makwana
- West Virginia Clinical and Transitional Science Institute, Morgantown, WV, USA
| | - Jasvinder A Singh
- West Virginia Clinical and Transitional Science Institute, Morgantown, WV, USA
- Medicine Service, VA Medical Center, 700 19Th St S, Birmingham, AL, 35233, USA
- Department of Medicine at the School of Medicine, University of Alabama at Birmingham (UAB), 510 20th Street S, Birmingham, AL, 35294-0022, USA
- Department of Epidemiology at the UAB School of Public Health, Ryals Public Health Building, 1665 University Blvd, Birmingham, AL, 35294-0022, USA
| | - George Sokos
- Heart and Vascular Institute, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
333
|
Fallahi P, Elia G, Ragusa F, Paparo SR, Patrizio A, Balestri E, Mazzi V, Benvenga S, Varricchi G, Gragnani L, Botrini C, Baldini E, Centanni M, Ferri C, Antonelli A, Ferrari SM. Thyroid Autoimmunity and SARS-CoV-2 Infection. J Clin Med 2023; 12:6365. [PMID: 37835009 PMCID: PMC10573843 DOI: 10.3390/jcm12196365] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological culprit of COronaVIrus Disease 19 (COVID-19), can enter the cells via the angiotensin-converting enzyme 2 (ACE2) receptor, which has been found in several tissues including in endocrine organs, such as the ovaries, testes, pancreas, and thyroid. Several thyroid disorders have been associated with SARS-CoV-2 infection [subacute thyroiditis (SAT), thyrotoxicosis, and non-thyroidal illness syndrome (NTIS)] and, in part, they are believed to be secondary to the local virus replication within the gland cells. However, as documented for other viruses, SARS-CoV-2 seems to interfere with several aspects of the immune system, inducing the synthesis of autoantibodies and triggering latent or new onset autoimmune disease (AID), including autoimmune thyroid disease (AITD), such as Hashimoto Thyroiditis (HT) and Graves' disease (GD). Several mechanisms have been hypothesized to explain this induction of autoimmunity by SARS-CoV-2 infection: the immune system hyper-stimulation, the molecular mimicry between the self-antigens of the host and the virus, neutrophils extracellular traps, and finally, the virus induced transcriptional changes in the immune genes; nonetheless, more evidence is needed especially from large, long-term cohort studies involving COVID-19 patients, to establish or reject this pathogenetic relationship.
Collapse
Affiliation(s)
- Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.F.); (S.R.P.); (L.G.)
| | - Giusy Elia
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Francesca Ragusa
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Sabrina Rosaria Paparo
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.F.); (S.R.P.); (L.G.)
| | - Armando Patrizio
- Department of Emergency Medicine, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy;
| | - Eugenia Balestri
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Valeria Mazzi
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine—Endocrinology, University of Messina, 98122 Messina, Italy;
- Master Program on Childhood, Adolescent and Women’s Endocrine Health, University of Messina, 98122 Messina, Italy
- Interdepartmental Program of Molecular & Clinical Endocrinology and Women’s Endocrine Health, University Hospital Policlinico “G. Martino”, 98124 Messina, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy;
- Center for Basic and Clinical Immunology Research, University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization Center of Excellence, University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council, 80131 Naples, Italy
| | - Laura Gragnani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.F.); (S.R.P.); (L.G.)
| | - Chiara Botrini
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Enke Baldini
- Department of Experimental Medicine, “Sapienza” University of Rome, 00185 Rome, Italy;
| | - Marco Centanni
- Department of Medico-Surgical Sciences and Biotechnologies, Endocrinology Section, ‘‘Sapienza’’ University of Rome, 00185 Rome, Italy;
- Endocrine Unit, Azienda Unità Sanitaria Locale (AUSL) Latina, 04100 Latina, Italy
| | - Clodoveo Ferri
- Rheumatology Unit, School of Medicine, University of Modena and Reggio Emilia, 41100 Modena, Italy;
- Rheumatology Clinic ‘Madonna Dello Scoglio’ Cotronei, 88836 Crotone, Italy
| | - Alessandro Antonelli
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| |
Collapse
|
334
|
Desai A, Deepak P, Cross RK, Murone J, Farraye FA, Ungaro RC, Kochhar GS. Effect of 2 vs 3 Doses of COVID-19 Vaccine in Patients With Inflammatory Bowel Disease: A Population-based Propensity Matched Analysis. Inflamm Bowel Dis 2023; 29:1563-1571. [PMID: 36576102 DOI: 10.1093/ibd/izac252] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Indexed: 12/29/2022]
Abstract
AIM There are limited data on the impact of 2 vs 3 doses of COVID-19 vaccine in patients with inflammatory bowel disease (IBD). The primary aim of the study was to assess the efficacy of COVID-19 vaccine based on number of administered doses in patients with IBD. METHODS We conducted a retrospective cohort study using TriNetX, a multi-institutional database to compare patients with IBD who received 1, 2, or 3 doses of BNT162b2 or mRNA-1273 to unvaccinated IBD patients (1.1.2020-7.26.2022) to assess the risk of COVID-19 after 1:1 propensity score matching. We also evaluated the impact of vaccine on a composite of severe COVID-19 outcomes including hospitalization, intubation, intensive care unit care, acute kidney injury, or mortality. RESULTS After propensity score matching, vaccinated patients with 2 (adjusted OR [aOR], 0.8; 95% confidence interval [CI], 0.6-0.9) and 3 doses (aOR, 0.7; 95% CI, 0.5-0.9) were found to have a lower risk of COVID-19 compared with unvaccinated patients. Vaccinated patients with IBD had a lower risk of severe COVID-19 outcomes (aOR, 0.7; 95% CI, 0.6-0.9) compared with unvaccinated patients. There was no difference in the risk of COVID-19 in IBD patients with 2 compared with 3 doses (aOR, 0.97; 95% CI, 0.7-1.3). However, IBD patients with 2 doses were at an increased risk for hospitalization due to COVID-19 (aOR, 1.78; 95% CI, 1.02-3.11) compared with those that received 3 doses. CONCLUSION Vaccinated patients with IBD had a lower risk of severe COVID-19 outcomes compared with unvaccinated patients. A third dose of COVID-19 vaccine compared with 2 doses decreases the risk of hospitalization but not breakthrough infection in patients with IBD.
Collapse
Affiliation(s)
- Aakash Desai
- Division of Gastroenterology & Hepatology, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Parakkal Deepak
- Division of Gastroenterology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Raymond K Cross
- Division of Gastroenterology, Hepatology & Nutrition, University of Maryland, Baltimore, MD, USA
| | - Julie Murone
- Division of Gastroenterology, Hepatology & Nutrition, Allegheny Health Network, Pittsburgh, PA, USA
| | - Francis A Farraye
- Division of Gastroenterology, Hepatology & Nutrition, Mayo Clinic, Jacksonville, FL, USA
| | - Ryan C Ungaro
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Gursimran S Kochhar
- Division of Gastroenterology, Hepatology & Nutrition, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
335
|
Buzas D, Bunzel AH, Staufer O, Milodowski EJ, Edmunds GL, Bufton JC, Vidana Mateo BV, Yadav SKN, Gupta K, Fletcher C, Williamson MK, Harrison A, Borucu U, Capin J, Francis O, Balchin G, Hall S, Vega MV, Durbesson F, Lingappa S, Vincentelli R, Roe J, Wooldridge L, Burt R, Anderson RJL, Mulholland AJ, Bristol UNCOVER Group, Hare J, Bailey M, Davidson AD, Finn A, Morgan D, Mann J, Spatz J, Garzoni F, Schaffitzel C, Berger I. In vitro generated antibodies guide thermostable ADDomer nanoparticle design for nasal vaccination and passive immunization against SARS-CoV-2. Antib Ther 2023; 6:277-297. [PMID: 38075238 PMCID: PMC10702856 DOI: 10.1093/abt/tbad024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 01/10/2024] Open
Abstract
Background Due to COVID-19, pandemic preparedness emerges as a key imperative, necessitating new approaches to accelerate development of reagents against infectious pathogens. Methods Here, we developed an integrated approach combining synthetic, computational and structural methods with in vitro antibody selection and in vivo immunization to design, produce and validate nature-inspired nanoparticle-based reagents against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Results Our approach resulted in two innovations: (i) a thermostable nasal vaccine called ADDoCoV, displaying multiple copies of a SARS-CoV-2 receptor binding motif derived epitope and (ii) a multivalent nanoparticle superbinder, called Gigabody, against SARS-CoV-2 including immune-evasive variants of concern (VOCs). In vitro generated neutralizing nanobodies and electron cryo-microscopy established authenticity and accessibility of epitopes displayed by ADDoCoV. Gigabody comprising multimerized nanobodies prevented SARS-CoV-2 virion attachment with picomolar EC50. Vaccinating mice resulted in antibodies cross-reacting with VOCs including Delta and Omicron. Conclusion Our study elucidates Adenovirus-derived dodecamer (ADDomer)-based nanoparticles for use in active and passive immunization and provides a blueprint for crafting reagents to combat respiratory viral infections.
Collapse
Affiliation(s)
- Dora Buzas
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Adrian H Bunzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Oskar Staufer
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Leibniz Institute for New Materials, Helmholtz Institute for Pharmaceutical Research and Center for Biophysics, Saarland University, Saarbrücken 66123, Germany
| | | | - Grace L Edmunds
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joshua C Bufton
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | - Kapil Gupta
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | | | - Maia K Williamson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Ufuk Borucu
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Julien Capin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Ore Francis
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Georgia Balchin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Sophie Hall
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Mirella V Vega
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Fabien Durbesson
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | | | - Renaud Vincentelli
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | - Joe Roe
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Linda Wooldridge
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Rachel Burt
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | | | | | | | - Jonathan Hare
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Mick Bailey
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Andrew D Davidson
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Adam Finn
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
- Children's Vaccine Centre, Bristol Medical School, Bristol BS2 8EF UK
| | - David Morgan
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Jamie Mann
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joachim Spatz
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Frederic Garzoni
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Christiane Schaffitzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| | - Imre Berger
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| |
Collapse
|
336
|
Wang D, Zhou C, Wang C, Guo S, Zhang Y, Lv H, Zhou FH. COVID-19 Vaccine in Renal Transplant Recipients: A Bibliometric-Based Analysis of Trends. Transplant Proc 2023; 55:1771-1783. [PMID: 37481393 DOI: 10.1016/j.transproceed.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND The global community has been affected by COVID-19, which emerged in December 2019. Since then, many studies have been conducted on kidney transplant recipients (KTRs) and COVID-19. This study aimed to perform a bibliometric and visual analysis of the published relationship between KTRs and COVID-19. OBJECTIVE To discuss the current status, hot spots, and development trend of research on KTRs vaccination with the COVID-19 vaccine and to provide a reference for researchers in related fields. METHODS Visual analysis of countries/regions, institutions, authors, references cited, and keywords for 2020 to 2023 via Microsoft Office Excel 2019 and CiteSpace (6.1.R6) based on the Web of Science core database. RESULTS A total of 366 publications were included after screening, with a rapid increase in the global literature studying the COVID-19 vaccine of KTRs. The US has the highest number of publications, indicating that it is the leading country in this field of research. Charite University of Medicine Berlin and Schrezenmeier E are the most published institutions and authors, respectively. "Antibody Response After a Third Dose of the messenger RNA-1273 SARS-CoV-2 Vaccine in Kidney Transplant Recipients With Minimal Serologic Response to 2 Doses" is the most central co-cited reference; The keywords "kidney transplant recipient," "covid 19 vaccine," and "mortality" have become hot topics of research. The keywords "humoral response" and "bnt162b2" are the latest research frontiers for detecting bursts. CONCLUSIONS This paper analyzed the current status and trends of vaccination studies in KTRs through bibliometric analysis. Several studies support the vaccination of KTRs with the COVID-19 vaccine. However, the evidence for improving vaccine efficacy by adjustment of immunosuppression is still limited, and future studies on vaccination will remain a hot topic in this field.
Collapse
Affiliation(s)
- Dong Wang
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Chuan Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Chao Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Sheng Guo
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Yunfeng Zhang
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Haoxuan Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Feng-Hai Zhou
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China.
| |
Collapse
|
337
|
Naik R, Avula S, Palleti SK, Gummadi J, Ramachandran R, Chandramohan D, Dhillon G, Gill AS, Paiwal K, Shaik B, Balachandran M, Patel B, Gurugubelli S, Mariswamy Arun Kumar AK, Nanjundappa A, Bellamkonda M, Rathi K, Sakhamuri PL, Nassar M, Bali A. From Emergence to Endemicity: A Comprehensive Review of COVID-19. Cureus 2023; 15:e48046. [PMID: 37916248 PMCID: PMC10617653 DOI: 10.7759/cureus.48046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2023] [Indexed: 11/03/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), later renamed coronavirus disease 2019 (COVID-19), was first identified in Wuhan, China, in early December 2019. Initially, the China office of the World Health Organization was informed of numerous cases of pneumonia of unidentified etiology in Wuhan, Hubei Province at the end of 2019. This would subsequently result in a global pandemic with millions of confirmed cases of COVID-19 and millions of deaths reported to the WHO. We have analyzed most of the data published since the beginning of the pandemic to compile this comprehensive review of SARS-CoV-2. We looked at the core ideas, such as the etiology, epidemiology, pathogenesis, clinical symptoms, diagnostics, histopathologic findings, consequences, therapies, and vaccines. We have also included the long-term effects and myths associated with some therapeutics of COVID-19. This study presents a comprehensive assessment of the SARS-CoV-2 virology, vaccines, medicines, and significant variants identified during the course of the pandemic. Our review article is intended to provide medical practitioners with a better understanding of the fundamental sciences, clinical treatment, and prevention of COVID-19. As of May 2023, this paper contains the most recent data made accessible.
Collapse
Affiliation(s)
- Roopa Naik
- Medicine, Geisinger Commonwealth School of Medicine, Scranton, USA
- Internal Medicine/Hospital Medicine, Geisinger Health System, Wilkes Barre, USA
| | - Sreekant Avula
- Diabetes, Endocrinology, and Metabolism, University of Minnesota, Minneapolis, USA
| | - Sujith K Palleti
- Nephrology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Jyotsna Gummadi
- Internal Medicine, MedStar Franklin Square Medical Center, Baltimore, USA
| | | | | | - Gagandeep Dhillon
- Physician Executive MBA, University of Tennessee, Knoxville, USA
- Internal Medicine, University of Maryland Baltimore Washington Medical Center, Glen Burnie, USA
| | | | - Kapil Paiwal
- Oral & Maxillofacial Pathology, Daswani Dental College & Research Center, Kota, IND
| | - Bushra Shaik
- Internal Medicine, Onslow Memorial Hospital, Jacksonville, USA
| | | | - Bhumika Patel
- Oral Medicine and Radiology, Howard University, Washington, D.C., USA
| | | | | | | | - Mahita Bellamkonda
- Hospital Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Kanika Rathi
- Internal Medicine, University of Florida, Gainesville, USA
| | | | - Mahmoud Nassar
- Endocrinology, Diabetes, and Metabolism, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Atul Bali
- Internal Medicine/Nephrology, Geisinger Medical Center, Danville, USA
- Internal Medicine/Nephrology, Geisinger Health System, Wilkes-Barre, USA
- Medicine, Geisinger Commonwealth School of Medicine, Scranton, USA
| |
Collapse
|
338
|
Castel AD, Barth S, Wilbourn BC, Horberg M, Monroe AK, Greenberg AE. Trends in COVID-19 Vaccine Hesitancy and Uptake Among Persons Living With HIV in Washington, DC. J Acquir Immune Defic Syndr 2023; 94:124-134. [PMID: 37368934 PMCID: PMC10529778 DOI: 10.1097/qai.0000000000003243] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/30/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE The COVID-19 pandemic has disproportionately affected older people, people with underlying health conditions, racial and ethnic minorities, socioeconomically disadvantaged, and people living with HIV (PWH). We sought to describe vaccine hesitancy and associated factors, reasons for vaccine hesitancy, and vaccine uptake over time in PWH in Washington, DC. METHODS We conducted a cross-sectional survey between October 2020 and December 2021 among PWH enrolled in a prospective longitudinal cohort in DC. Survey data were linked to electronic health record data and descriptively analyzed. Multivariable logistic regression was performed to identify factors associated with vaccine hesitancy. The most common reasons for vaccine hesitancy and uptake were assessed. RESULTS Among 1029 participants (66% men, 74% Black, median age 54 years), 13% were vaccine hesitant and 9% refused. Women were 2.6-3.5 times, non-Hispanic Blacks were 2.2 times, Hispanics and those of other race/ethnicities were 3.5-8.8 times, and younger PWH were significantly more likely to express hesitancy or refusal than men, non-Hispanic Whites, and older PWH, respectively. The most reported reasons for vaccine hesitancy were side effect concerns (76%), plans to use other precautions/masks (73%), and speed of vaccine development (70%). Vaccine hesitancy and refusal declined over time (33% in October 2020 vs. 4% in December 2021, P < 0.0001). CONCLUSIONS This study is one of the largest analyses of vaccine hesitancy among PWH in a US urban area highly affected by HIV and COVID-19. Multilevel culturally appropriate approaches are needed to effectively address COVID-19 vaccine concerns raised among PWH.
Collapse
Affiliation(s)
- Amanda D Castel
- Department of Epidemiology, The George Washington University School of Public Health, Washington, DC; and
| | - Shannon Barth
- Department of Epidemiology, The George Washington University School of Public Health, Washington, DC; and
| | - Brittany C Wilbourn
- Department of Epidemiology, The George Washington University School of Public Health, Washington, DC; and
| | | | - Anne K Monroe
- Department of Epidemiology, The George Washington University School of Public Health, Washington, DC; and
| | - Alan E Greenberg
- Department of Epidemiology, The George Washington University School of Public Health, Washington, DC; and
| |
Collapse
|
339
|
Dari A, Solforosi L, Roozendaal R, Hoetelmans RMW, Pérez-Ruixo JJ, Boulton M. Mechanistic Model Describing the Time Course of Humoral Immunity Following Ad26.COV2.S Vaccination in Non-Human Primates. J Pharmacol Exp Ther 2023; 387:121-130. [PMID: 37536955 DOI: 10.1124/jpet.123.001591] [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: 01/23/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023] Open
Abstract
Mechanistic modeling can be used to describe the time course of vaccine-induced humoral immunity and to identify key biologic drivers in antibody production. We used a six-compartment mechanistic model to describe a 20-week time course of humoral immune responses in 56 non-human primates (NHPs) elicited by vaccination with Ad26.COV2.S according to either a single-dose regimen (1 × 1011 or 5 × 1010 viral particles [vp]) or a two-dose homologous regimen (5 × 1010 vp) given in an interval of 4 or 8 weeks. Humoral immune responses were quantified by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike-specific binding antibody concentrations as determined by spike protein-enzyme-linked immunosorbent assay. The mechanistic model adequately described the central tendency and variability of binding antibody concentrations through 20 weeks in all vaccination arms. The estimation of mechanistic modeling parameters revealed greater contribution of the antibody production mediated by short-lived cells as compared with long-lived cells in driving the peak response, especially post second dose when a more rapid peak response was observed. The antibody production mediated by long-lived cells was identified as relevant for generating the first peak and for contributing to the long-term time course of sustained antibody concentrations in all vaccination arms. The findings contribute evidence on the key biologic components responsible for the observed time course of vaccine-induced humoral immunity in NHPs and constitute a step toward defining immune biomarkers of protection against SARS-CoV-2 that might translate across species. SIGNIFICANCE STATEMENT: We demonstrate the adequacy of a mechanistic modeling approach describing the time course of binding antibody concentrations in non-human primates (NHPs) elicited by different dose levels and regimens of Ad26.COV2.S. The findings are relevant for informing the mechanism-based accounts of vaccine-induced humoral immunity in NHPs and translational research efforts aimed at identifying immune biomarkers of protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Laura Solforosi
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Ramon Roozendaal
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Richard M W Hoetelmans
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Juan-José Pérez-Ruixo
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Muriel Boulton
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| |
Collapse
|
340
|
Fundora MP, Kamidani S, Oster ME. COVID Vaccination as a Strategy for Cardiovascular Disease Prevention. Curr Cardiol Rep 2023; 25:1327-1335. [PMID: 37688764 DOI: 10.1007/s11886-023-01950-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/21/2023] [Indexed: 09/11/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular (CV) disease is a known complication of SARS-CoV-2 infection. A clear benefit of COVID-19 vaccination is a reduction mortality; however, COVID-19 vaccination may also prevent cardiovascular disease (CVD). We aim to describe CV pathology associated with SARS-CoV-2 infection and describe how COVID-19 vaccination is a strategy for CVD prevention. RECENT FINDINGS The risks and benefits of COVID-19 vaccination have been widely studied. Analysis of individuals with and without pre-existing CVD has shown that COVID-19 vaccination can prevent morbidity associated with SARS-CoV-2 infection and reduce mortality. COVID-19 vaccination is effective in preventing myocardial infarction, cerebrovascular events, myopericarditis, and long COVID, all associated with CVD risk factors. Vaccination reduces mortality in patients with pre-existing CVD. Further study investigating ideal vaccination schedules for individuals with CVD should be undertaken to protect this vulnerable group and address new risks from variants of concern.
Collapse
Affiliation(s)
- Michael P Fundora
- Children's Healthcare of Atlanta Cardiology, Department of Pediatrics, Emory University, 1405 Clifton Rd NE, Atlanta, GA, 30322, USA
| | - Satoshi Kamidani
- The Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and the Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew E Oster
- Children's Healthcare of Atlanta Cardiology, Department of Pediatrics, Emory University, 1405 Clifton Rd NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
341
|
Alp Çavuş S, Çelik M, Süner AF, Güzel I, Irmak Ç, Çağlayan D, Öztürk HG, Şiyve N, Appak Ö, Işık E, Ergör G, Ergör OA, Demiral Y, Sayıner AA, Kılıç B. Pre-infection antibody levels of vaccinated healthcare workers with SARS-CoV-2 breakthrough infection: A nested case-control study. Immunol Lett 2023; 262:1-6. [PMID: 37597753 DOI: 10.1016/j.imlet.2023.08.002] [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: 04/18/2023] [Revised: 07/24/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
AIM To evaluate anti-RBD IgG antibody levels and neutralizing antibody titers between the health care workers (HCWs) with breakthrough SARS-CoV-2 infection and controls. METHODS In this nested case-case control study, we followed 548 vaccinated HCWs with homologous (only with inactivated vaccine) or heterologous (both with inactivated and BNT162b2 vaccine) vaccination for 11 months, prospectively. We obtained blood samples from the participants for quantitative anti-RBD IgG and surrogate neutralization test. The participants with SARS-CoV-2 PCR positivity (at least 14 days after the last vaccination) were considered breakthrough infection. We chose 1:2 matched controls from the cohort, according to age, sex and vaccination status. We used R version 4.0.2 for the statistical analysis. RESULTS Sixty-five cases and 130 controls were included in the study. The number of the breakthrough infections in HCWs were correlated with the pandemic waves in Türkiye and peaked during Omicron outbreak. The median age of the cases was 39 and 78.5% were female. The cases had more comorbidities than controls, significantly (p = 0.021). All cases experienced no or mild symptoms and recovered completely. Both pre-infection anti-RBD antibody and neutralizing antibody titers did not differ between cases and matched controls (p = 0.767, p = 0.628). CONCLUSION In this study, we showed that there was no comparable difference in humoral response after homologous or heterologous vaccination between the cases of breakthrough infection and matched controls. Compliance with infection control measures should be ensured, in combination with vaccination.
Collapse
Affiliation(s)
- Sema Alp Çavuş
- Department of Infectious Diseases and Clinical Microbiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye.
| | - Muammer Çelik
- Department of Infectious Diseases and Clinical Microbiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Ahmet Furkan Süner
- Department of Public Health, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Irmak Güzel
- Department of Medical Microbiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Çağlar Irmak
- Department of Infectious Diseases and Clinical Microbiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Derya Çağlayan
- Department of Public Health, Division of Epidemiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Huriye Gamze Öztürk
- Department of Medical Microbiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Neslişah Şiyve
- Department of Public Health, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Özgür Appak
- Department of Medical Microbiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Elif Işık
- Department of Public Health, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Gül Ergör
- Department of Public Health, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Osman Alparslan Ergör
- Department of Public Health, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Yücel Demiral
- Department of Public Health, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Ayça Arzu Sayıner
- Department of Medical Microbiology, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Bülent Kılıç
- Department of Public Health, School of Medicine, Dokuz Eylul University, Izmir, Türkiye
| |
Collapse
|
342
|
Rick AM, Laurens MB, Huang Y, Yu C, Martin TCS, Rodriguez CA, Rostad CA, Maboa RM, Baden LR, El Sahly HM, Grinsztejn B, Gray GE, Gay CL, Gilbert PB, Janes HE, Kublin JG, Huang Y, Leav B, Hirsch I, Struyf F, Dunkle LM, Neuzil KM, Corey L, Goepfert PA, Walsh SR, Follmann D, Kotloff KL. Risk of COVID-19 after natural infection or vaccination. EBioMedicine 2023; 96:104799. [PMID: 37738833 PMCID: PMC10518569 DOI: 10.1016/j.ebiom.2023.104799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND While vaccines have established utility against COVID-19, phase 3 efficacy studies have generally not comprehensively evaluated protection provided by previous infection or hybrid immunity (previous infection plus vaccination). Individual patient data from US government-supported harmonized vaccine trials provide an unprecedented sample population to address this issue. We characterized the protective efficacy of previous SARS-CoV-2 infection and hybrid immunity against COVID-19 early in the pandemic over three-to six-month follow-up and compared with vaccine-associated protection. METHODS In this post-hoc cross-protocol analysis of the Moderna, AstraZeneca, Janssen, and Novavax COVID-19 vaccine clinical trials, we allocated participants into four groups based on previous-infection status at enrolment and treatment: no previous infection/placebo; previous infection/placebo; no previous infection/vaccine; and previous infection/vaccine. The main outcome was RT-PCR-confirmed COVID-19 >7-15 days (per original protocols) after final study injection. We calculated crude and adjusted efficacy measures. FINDINGS Previous infection/placebo participants had a 92% decreased risk of future COVID-19 compared to no previous infection/placebo participants (overall hazard ratio [HR] ratio: 0.08; 95% CI: 0.05-0.13). Among single-dose Janssen participants, hybrid immunity conferred greater protection than vaccine alone (HR: 0.03; 95% CI: 0.01-0.10). Too few infections were observed to draw statistical inferences comparing hybrid immunity to vaccine alone for other trials. Vaccination, previous infection, and hybrid immunity all provided near-complete protection against severe disease. INTERPRETATION Previous infection, any hybrid immunity, and two-dose vaccination all provided substantial protection against symptomatic and severe COVID-19 through the early Delta period. Thus, as a surrogate for natural infection, vaccination remains the safest approach to protection. FUNDING National Institutes of Health.
Collapse
Affiliation(s)
- Anne-Marie Rick
- Department of Pediatrics, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Matthew B Laurens
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ying Huang
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas C S Martin
- Department of Infectious Diseases and Global Public Health, University of California San Diego, San Diego, CA, USA
| | - Carina A Rodriguez
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Christina A Rostad
- Department of Pediatrics, Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Glenda E Gray
- South African Medical Research Council, Cape Town, South Africa
| | - Cynthia L Gay
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | | | | | - Yunda Huang
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Ian Hirsch
- AstraZeneca BioPharmaceuticals, Cambridge, UK
| | | | | | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Paul A Goepfert
- University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Stephen R Walsh
- Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Karen L Kotloff
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
343
|
Laughey W, Lodhi I, Pennick G, Smart L, Sanni O, Sandhu S, Charlesworth B. Ibuprofen, other NSAIDs and COVID-19: a narrative review. Inflammopharmacology 2023; 31:2147-2159. [PMID: 37603158 PMCID: PMC10518289 DOI: 10.1007/s10787-023-01309-7] [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: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/22/2023]
Abstract
At the start of the coronavirus disease 2019 (COVID-19) pandemic (March 2020), there was speculation that non-steroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen, used to manage some of the symptoms of COVID-19, could increase the susceptibility to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and negatively impact clinical outcomes. In the absence of any robust mechanistic and clinical evidence, this speculation led to confusion about the safety of ibuprofen, contributing to the so-called 'infodemic' surrounding COVID-19. A wealth of evidence has been generated in subsequent years, and this narrative review aims to consider the body of in vitro and in vivo research, observational studies, systematic reviews and meta-analyses on the use of NSAIDs, including ibuprofen, in COVID-19. Overall, the direction of evidence supports that NSAIDs do not increase susceptibility to infection, nor worsen disease outcomes in patients with COVID-19. Neither do they impact the immune response to COVID-19 vaccines. There is no basis to limit the use of NSAIDs, and doing so may deprive patients of effective self-care measures to control symptoms.
Collapse
Affiliation(s)
- William Laughey
- Reckitt Health Care UK Ltd, Hull, UK.
- Hull York Medical School, University of York, York, UK.
| | | | | | | | | | | | | |
Collapse
|
344
|
Abstract
COVID-19, the illness caused by SARS-CoV-2, became a worldwide pandemic in 2020. Initial clinical manifestations range from asymptomatic infection to mild upper respiratory illness but may progress to pulmonary involvement with hypoxemia and, in some cases, multiorgan involvement, shock, and death. Older adults, pregnant persons, those with common comorbidities, and those with immunosuppression are at greatest risk for progression. Vaccination is effective in preventing symptomatic infection and reducing risk for severe disease, hospitalization, and death. Antiviral treatment and immunomodulators have been shown to benefit certain patients. This article summarizes current recommendations on prevention, diagnosis, management, and treatment of COVID-19.
Collapse
Affiliation(s)
| | - Roy M Gulick
- Weill Cornell Medicine, New York, New York (K.M.M., R.M.G.)
| |
Collapse
|
345
|
Costa GJ, da Silva JR, da Silva CCA, de Lima TPF, Costa MM, Sousa MHO, Costa GCDS, Costa JI, Sales MJT. Risk factors for death and illness severity in vaccinated versus unvaccinated COVID-2019 inpatients: a retrospective cohort study. J Bras Pneumol 2023; 49:e20230145. [PMID: 37729337 PMCID: PMC10578947 DOI: 10.36416/1806-3756/e20230145] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/05/2023] [Indexed: 09/22/2023] Open
Abstract
OBJECTIVE To determine the clinical profile of COVID-19 inpatients who were vaccinated prior to hospitalization and to compare the risk factors for death and the 28-day survival rate of between those inpatients vaccinated with one, two, or three doses and unvaccinated COVID-19 inpatients. METHODS This was a retrospective observational cohort study involving COVID-19 patients admitted to a referral hospital in the city of Recife, Brazil, between July of 2020 and June of 2022. RESULTS The sample comprised 1,921 inpatients, 996 of whom (50.8%) were vaccinated prior to hospitalization. After adjusting the mortality risk for vaccinated patients, those undergoing invasive mechanical ventilation (IMV) had the highest mortality risk (adjusted OR [aOR] = 7.4; 95% CI, 3.8-14.1; p < 0.001), followed by patients > 80 years of age (aOR = 7.3; 95% CI, 3.4-15.4; p < 0.001), and those needing vasopressors (aOR = 5.6; 95% CI, 2.9-10.9; p < 0.001). After adjusting the mortality risk for all patients, having received three vaccine doses (aOR = 0.06; 95% CI, 0.03-0.11; p < 0.001) was the most important protective factor against death. There were progressive benefits of vaccination, reducing the frequency of ICU admissions, use for IMV, and death (respectively, from 44.9%, 39.0% and 39.9% after the first dose to 16.7%, 6.2% and 4.4% after the third dose), as well as significant improvements in survival after each subsequent dose (p < 0.001). CONCLUSIONS Vaccines were effective in reducing illness severity and death in this cohort of COVID-19 inpatients, and the administration of additional doses conferred them with accumulative vaccine protection.
Collapse
Affiliation(s)
| | - José Roberto da Silva
- . Departamento de Ensino e Pesquisa, Instituto de Medicina Integral Professor Fernando Figueira - IMIP - Recife (PE) Brasil
| | - Caio Cesar Arruda da Silva
- . Departamento de Ensino e Pesquisa, Instituto de Medicina Integral Professor Fernando Figueira - IMIP - Recife (PE) Brasil
| | | | - Mariana Menezes Costa
- . Departamento de Ensino e Pesquisa, Instituto de Medicina Integral Professor Fernando Figueira - IMIP - Recife (PE) Brasil
| | - Marcos Henrique Oliveira Sousa
- . Departamento de Ensino e Pesquisa, Instituto de Medicina Integral Professor Fernando Figueira - IMIP - Recife (PE) Brasil
| | | | - José Iran Costa
- . Departamento de Ensino e Pesquisa, Hospital Alfa, Recife (PE) Brasil
| | - Mozart Júlio Tabosa Sales
- . Departamento de Ensino e Pesquisa, Instituto de Medicina Integral Professor Fernando Figueira - IMIP - Recife (PE) Brasil
| |
Collapse
|
346
|
Quirk GE, Schoenle MV, Peyton KL, Uhrlaub JL, Lau B, Burgess JL, Ellingson K, Beitel S, Romine J, Lutrick K, Fowlkes A, Britton A, Tyner HL, Caban-Martinez AJ, Naleway A, Gaglani M, Yoon S, Edwards L, Olsho L, Dake M, LaFleur BJ, Nikolich JŽ, Sprissler R, Worobey M, Bhattacharya D. Determinants of de novo B cell responses to drifted epitopes in post-vaccination SARS-CoV-2 infections. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.12.23295384. [PMID: 37745498 PMCID: PMC10516057 DOI: 10.1101/2023.09.12.23295384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Vaccine-induced immunity may impact subsequent de novo responses to drifted epitopes in SARS-CoV-2 variants, but this has been difficult to quantify due to the challenges in recruiting unvaccinated control groups whose first exposure to SARS-CoV-2 is a primary infection. Through local, statewide, and national SARS-CoV-2 testing programs, we were able to recruit cohorts of individuals who had recovered from either primary or post-vaccination infections by either the Delta or Omicron BA.1 variants. Regardless of variant, we observed greater Spike-specific and neutralizing antibody responses in post-vaccination infections than in those who were infected without prior vaccination. Through analysis of variant-specific memory B cells as markers of de novo responses, we observed that Delta and Omicron BA.1 infections led to a marked shift in immunodominance in which some drifted epitopes elicited minimal responses, even in primary infections. Prior immunity through vaccination had a small negative impact on these de novo responses, but this did not correlate with cross-reactive memory B cells, arguing against competitive inhibition of naïve B cells. We conclude that dampened de novo B cell responses against drifted epitopes are mostly a function of altered immunodominance hierarchies that are apparent even in primary infections, with a more modest contribution from pre-existing immunity, perhaps due to accelerated antigen clearance.
Collapse
Affiliation(s)
- Grace E Quirk
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
| | - Marta V Schoenle
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Kameron L Peyton
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Branden Lau
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Jefferey L Burgess
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA
| | - Katherine Ellingson
- Department of Epidemiology and Biostatistics, Zuckerman College of Public Health, University of Arizona, Tucson
| | - Shawn Beitel
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA
| | - James Romine
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA
| | - Karen Lutrick
- College of Medicine-Tucson, University of Arizona, Tucson, Arizona, USA
| | - Ashley Fowlkes
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA
| | - Amadea Britton
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA
| | - Harmony L Tyner
- St. Luke's Regional Health Care System, Duluth, Minnesota, USA
| | | | - Allison Naleway
- Kaiser Permanente Northwest Center for Health Research, Portland, Oregon, USA
| | - Manjusha Gaglani
- Baylor Scott & White Health and Texas A&M University College of Medicine, Temple, Texas, USA
| | - Sarang Yoon
- Rocky Mountain Center for Occupational and Environmental Health, Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, USA
| | | | | | - Michael Dake
- Office of the Senior Vice-President for Health Sciences, University of Arizona, Tucson, AZ, USA
| | | | - Janko Ž Nikolich
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Ryan Sprissler
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Michael Worobey
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
347
|
Curtis NC, Shin S, Hederman AP, Connor RI, Wieland-Alter WF, Ionov S, Boylston J, Rose J, Sakharkar M, Dorman DB, Dessaint JA, Gwilt LL, Crowley AR, Feldman J, Hauser BM, Schmidt AG, Ashare A, Walker LM, Wright PF, Ackerman ME, Lee J. Characterization of SARS-CoV-2 Convalescent Patients' Serological Repertoire Reveals High Prevalence of Iso-RBD Antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556349. [PMID: 37745524 PMCID: PMC10515772 DOI: 10.1101/2023.09.08.556349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
While our understanding of SARS-CoV-2 pathogenesis and antibody responses following infection and vaccination has improved tremendously since the outbreak in 2019, the sequence identities and relative abundances of the individual constituent antibody molecules in circulation remain understudied. Using Ig-Seq, we proteomically profiled the serological repertoire specific to the whole ectodomain of SARS-CoV-2 prefusion-stabilized spike (S) as well as to the receptor binding domain (RBD) over a 6-month period in four subjects following SARS-CoV-2 infection before SARS-CoV-2 vaccines were available. In each individual, we identified between 59 and 167 unique IgG clonotypes in serum. To our surprise, we discovered that ∼50% of serum IgG specific for RBD did not recognize prefusion-stabilized S (referred to as iso-RBD antibodies), suggesting that a significant fraction of serum IgG targets epitopes on RBD inaccessible on the prefusion-stabilized conformation of S. On the other hand, the abundance of iso-RBD antibodies in nine individuals who received mRNA-based COVID-19 vaccines encoding prefusion-stabilized S was significantly lower (∼8%). We expressed a panel of 12 monoclonal antibodies (mAbs) that were abundantly present in serum from two SARS-CoV-2 infected individuals, and their binding specificities to prefusion-stabilized S and RBD were all in agreement with the binding specificities assigned based on the proteomics data, including 1 iso-RBD mAb which bound to RBD but not to prefusion-stabilized S. 2 of 12 mAbs demonstrated neutralizing activity, while other mAbs were non-neutralizing. 11 of 12 mAbs also bound to S (B.1.351), but only 1 maintained binding to S (B.1.1.529). This particular mAb binding to S (B.1.1.529) 1) represented an antibody lineage that comprised 43% of the individual's total S-reactive serum IgG binding titer 6 months post-infection, 2) bound to the S from a related human coronavirus, HKU1, and 3) had a high somatic hypermutation level (10.9%), suggesting that this antibody lineage likely had been elicited previously by pre-pandemic coronavirus and was re-activated following the SARS-CoV-2 infection. All 12 mAbs demonstrated their ability to engage in Fc-mediated effector function activities. Collectively, our study provides a quantitative overview of the serological repertoire following SARS-CoV-2 infection and the significant contribution of iso-RBD antibodies, demonstrating how vaccination strategies involving prefusion-stabilized S may have reduced the elicitation of iso-RBD serum antibodies which are unlikely to contribute to protection.
Collapse
|
348
|
Cabanillas-Bernal O, Valdovinos-Navarro BJ, Cervantes-Luevano KE, Sanchez-Campos N, Licea-Navarro AF. Unleashing the power of shark variable single domains (VNARs): broadly neutralizing tools for combating SARS-CoV-2. Front Immunol 2023; 14:1257042. [PMID: 37753081 PMCID: PMC10518403 DOI: 10.3389/fimmu.2023.1257042] [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: 07/11/2023] [Accepted: 08/18/2023] [Indexed: 09/28/2023] Open
Abstract
The pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) generated a joint global effort to develop vaccines and other treatments that could mitigate the negative effects and the rapid spread of the virus. Single-domain antibodies derived from various sources, including cartilaginous fish, camelids, and humans, have gained attention as promising therapeutic tools against coronavirus disease 2019. Shark-derived variable new antigen receptors (VNARs) have emerged as the smallest naturally occurring antigen-binding molecules. Here, we compile and review recent published studies on VNARs with the capacity to recognize and/or neutralize SARS-CoV-2. We found a close balance between the use of natural immune libraries and synthetic VNAR libraries for the screening against SARS-CoV-2, with phage display being the preferred display technology for the selection of VNARs against this virus. In addition, we discuss potential modifications and engineering strategies employed to improve the neutralization potential of VNARs, such as exploring fusion with the Fc domain of human Immunoglobulin G (IgG) to increase avidity and therapeutic potential. This research highlights the potential of VNARs as powerful molecular tools in the fight against infectious diseases.
Collapse
Affiliation(s)
| | | | | | | | - Alexei F. Licea-Navarro
- Biomedical Innovation Department, Centro de Investigación Científica y Educación Superior de Ensenada, (CICESE), Ensenada, Baja California, Mexico
| |
Collapse
|
349
|
Roper KJ, Thomas J, Albalawi W, Maddocks E, Dobson S, Alshehri A, Barone FG, Baltazar M, Semple MG, Ho A, Turtle L, Paxton WA, Pollakis G. Quantifying neutralising antibody responses against SARS-CoV-2 in dried blood spots (DBS) and paired sera. Sci Rep 2023; 13:15014. [PMID: 37697014 PMCID: PMC10495436 DOI: 10.1038/s41598-023-41928-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
The ongoing SARS-CoV-2 pandemic was initially managed by non-pharmaceutical interventions such as diagnostic testing, isolation of positive cases, physical distancing and lockdowns. The advent of vaccines has provided crucial protection against SARS-CoV-2. Neutralising antibody (nAb) responses are a key correlate of protection, and therefore measuring nAb responses is essential for monitoring vaccine efficacy. Fingerstick dried blood spots (DBS) are ideal for use in large-scale sero-surveillance because they are inexpensive, offer the option of self-collection and can be transported and stored at ambient temperatures. Such advantages also make DBS appealing to use in resource-limited settings and in potential future pandemics. In this study, nAb responses in sera, venous blood and fingerstick blood stored on filter paper were measured. Samples were collected from SARS-CoV-2 acutely infected individuals, SARS-CoV-2 convalescent individuals and SARS-CoV-2 vaccinated individuals. Good agreement was observed between the nAb responses measured in eluted DBS and paired sera. Stability of nAb responses was also observed in sera stored on filter paper at room temperature for 28 days. Overall, this study provides support for the use of filter paper as a viable sample collection method to study nAb responses.
Collapse
Affiliation(s)
- Kelly J Roper
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Wejdan Albalawi
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Emily Maddocks
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Susan Dobson
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Abdullateef Alshehri
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Francesco G Barone
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, L69 3BX, UK
| | - Murielle Baltazar
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Malcolm G Semple
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Respiratory Medicine, Alder Hey Children's Hospital, Institute in The Park, University of Liverpool, Liverpool, UK
| | - Antonia Ho
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow, G61 1QH, UK
| | - Lance Turtle
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - William A Paxton
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
| | - Georgios Pollakis
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK.
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK.
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK.
| |
Collapse
|
350
|
Kopplin N, Garcia A, Reczek A, Wilkinson K, Yekkaluri S, Murphy CC, Tiro J, Muthukumar AR, Masica A, Singal AG. Post-acute sequelae of COVID-19 and longitudinal antibody levels in a community-based cohort. PLoS One 2023; 18:e0291259. [PMID: 37682916 PMCID: PMC10490864 DOI: 10.1371/journal.pone.0291259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) infection invokes variable immune responses and poses a risk of post-acute sequelae SARS-CoV-2 infection (PASC) symptoms; however, most data on natural history are derived from patients with severe infection. Further data are needed among patients with mild infection, who comprise most cases. METHODS The Dallas Fort-Worth (DFW) COVID-19 Prevalence Study included 21,597 community-dwelling adults (ages 18-89) who underwent COVID-19 PCR and anti-nucleocapsid antibody testing between July 2020 and March 2021. We invited participants with positive COVID-19 results (cases) and a subset with negative results (controls), matched on age, sex, race/ethnicity, and ZIP code, to complete a follow-up questionnaire for PASC symptoms and repeat anti-nucleocapsid testing, and anti-spike antibody testing between July and December 2021. RESULTS Of 3,917 adults invited to participate, 2260 (57.7%) completed the questionnaire- 1150 cases and 1110 controls. Persistent symptoms were reported in 21.1% of cases, with the most common being shortness of breath, fatigue, and loss of taste or smell. Among 292 cases with asymptomatic infection, >15% reported new fatigue and 8-10% reported new loss of taste/smell, myalgias, or headache. Median anti-nucleocapsid levels in cases decreased from 3.5U to 0.7U over a median follow-up of 8.6 months. Anti-spike antibody levels at 6-7 months post-vaccination in cases were similar to that of controls. CONCLUSIONS More than 1 in 5 patients with COVID-19 infection, including those with mild infection, reported persistent symptoms during follow-up. Both nucleocapsid and spike protein antibody levels decreased within six months following a COVID-19 infection and vaccination.
Collapse
Affiliation(s)
- Noa Kopplin
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Angie Garcia
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Annika Reczek
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Kate Wilkinson
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Sruthi Yekkaluri
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Caitlin C. Murphy
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Jasmin Tiro
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Alagar R. Muthukumar
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Andrew Masica
- Texas Health Resources, Fort Worth, TX, United States of America
| | - Amit G. Singal
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| |
Collapse
|