1201
|
Stasi C, Meoni B, Voller F, Silvestri C. SARS-CoV-2 Vaccination and the Bridge between First and Fourth Dose: Where Are We? Vaccines (Basel) 2022; 10:444. [PMID: 35335075 PMCID: PMC8953092 DOI: 10.3390/vaccines10030444] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/24/2022] [Accepted: 03/09/2022] [Indexed: 12/23/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has induced the explosion of vaccine research. Currently, according to the data of the World Health Organization, there are several vaccines in clinical (145) and preclinical (195) stages, while at least 10 are already in clinical phase 4 (post-marketing). Vaccines have proven to be safe, effective, and able to reduce the spread of SARS-CoV-2 infection and its variants, as well as the clinical consequences of the development of coronavirus disease-19 (COVID-19). In the two-dose primary vaccination, different time intervals between the two doses have been used. Recently, special attention has been paid to assessing the immunogenicity following booster administration. The third dose of the vaccine against COVID-19 may be administered at least 8 weeks after the second dose. In Israel, a fourth dose has already been approved in immunocompromised groups. The main objective of this review is to describe the principal results of studies on the effectiveness of first-to-fourth dose vaccination to reduce reinfection by variants and the incidence of severe disease/death caused by COVID-19. Vaccines have shown a high level of protection from symptomatic infection and reinfection by variants after a third dose. Accelerating mass third-dose vaccination could potentially induce immunogenicity against variants.
Collapse
Affiliation(s)
- Cristina Stasi
- Epidemiology Unit, Tuscany Regional Health Agency, Via Pietro Dazzi, 1, 50141 Florence, Italy; (B.M.); (F.V.); (C.S.)
- MASVE Interdepartmental Hepatology Center, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Center for Research and Innovation CRIA-MASVE, Careggi University Hospital, 50134 Florence, Italy
| | - Barbara Meoni
- Epidemiology Unit, Tuscany Regional Health Agency, Via Pietro Dazzi, 1, 50141 Florence, Italy; (B.M.); (F.V.); (C.S.)
| | - Fabio Voller
- Epidemiology Unit, Tuscany Regional Health Agency, Via Pietro Dazzi, 1, 50141 Florence, Italy; (B.M.); (F.V.); (C.S.)
| | - Caterina Silvestri
- Epidemiology Unit, Tuscany Regional Health Agency, Via Pietro Dazzi, 1, 50141 Florence, Italy; (B.M.); (F.V.); (C.S.)
| |
Collapse
|
1202
|
Hofmeyer KA, Bianchi KM, Wolfe DN. Utilization of Viral Vector Vaccines in Preparing for Future Pandemics. Vaccines (Basel) 2022; 10:436. [PMID: 35335068 PMCID: PMC8950656 DOI: 10.3390/vaccines10030436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
As the global response to COVID-19 continues, government stakeholders and private partners must keep an eye on the future for the next emerging viral threat with pandemic potential. Many of the virus families considered to be among these threats currently cause sporadic outbreaks of unpredictable size and timing. This represents a major challenge in terms of both obtaining sufficient funding to develop vaccines, and the ability to evaluate clinical efficacy in the field. However, this also presents an opportunity in which vaccines, along with robust diagnostics and contact tracing, can be utilized to respond to outbreaks as they occur, and limit the potential for further spread of the disease in question. While mRNA-based vaccines have proven, during the COVID-19 response, to be an effective and safe solution in terms of providing a rapid response to vaccine development, virus vector-based vaccines represent a class of vaccines that can offer key advantages in certain performance characteristics with regard to viruses of pandemic potential. Here, we will discuss some of the key pros and cons of viral vector vaccines in the context of preparing for future pandemics.
Collapse
Affiliation(s)
| | | | - Daniel N. Wolfe
- US Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, Washington, DC 20201, USA; (K.A.H.); (K.M.B.)
| |
Collapse
|
1203
|
Sikora D, Rzymski P. COVID-19 Vaccination and Rates of Infections, Hospitalizations, ICU Admissions, and Deaths in the European Economic Area during Autumn 2021 Wave of SARS-CoV-2. Vaccines (Basel) 2022; 10:437. [PMID: 35335069 PMCID: PMC8955952 DOI: 10.3390/vaccines10030437] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 01/14/2023] Open
Abstract
The COVID-19 vaccination campaigns were met with a varying level of vaccine hesitancy in Europe. We analyzed the potential relationships between COVID-19 vaccine coverage in different countries of the European Economic Area and rates of infection, hospitalizations, admissions to intensive care units (ICU), and deaths during the autumn 2021 SARS-CoV-2 wave (September-December). Significant negative correlations between infection rates and the percentage of fully vaccinated individuals were found during September, October, and November, but not December. The loss of this protective effect in December is likely due to the emergence of the omicron (B.1.1.529) variant, better adapted to evade vaccine-induced humoral immunity. For every considered month, the negative linear associations between the vaccine coverage and mean number of hospitalizations (r= -0.61 to -0.88), the mean number of ICU admissions (r= -0.62 to -0.81), and death rate (r= -0.64 to -0.84) were observed. The results highlight that vaccines provided significant benefits during autumn 2021. The vaccination of unvaccinated individuals should remain the primary strategy to decrease the hospital overloads, severe consequences of COVID-19, and deaths.
Collapse
Affiliation(s)
- Dominika Sikora
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
- Doctoral School, Poznan University of Medical Sciences, Fredry St. 10, 61-701 Poznań, Poland
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), 60-806 Poznań, Poland
| |
Collapse
|
1204
|
Majhen D. Human adenovirus type 26 basic biology and its usage as vaccine vector. Rev Med Virol 2022; 32:e2338. [PMID: 35278248 DOI: 10.1002/rmv.2338] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 11/10/2022]
Abstract
Due to their nature, adenoviruses have been recognised as promising candidates for vaccine vector development. Since they mimic natural infection, recombinant adenovirus vectors have been proven as ideal shuttles to deliver foreign transgenes aiming at inducing both humoral and cellular immune response. In addition, a potent adjuvant effect can be exerted due to the adenovirus inherent stimulation of various elements of innate and adaptive immunity. Due to its low seroprevalence in humans as well as induction of favourable immune response to inserted transgene, human adenovirus type 26 (HAdV-D26) has been recognised as a promising platform for vaccine vector development and is studied in number of completed or ongoing clinical studies. Very recently HAdV-D26 based Ebola and Covid-19 vaccines were approved for medical use. In this review, current state of the art regarding HAdV-D26 basic biology and its usage as vaccine vector will be discussed.
Collapse
Affiliation(s)
- Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
1205
|
Tomic A, Skelly DT, Ogbe A, O'Connor D, Pace M, Adland E, Alexander F, Ali M, Allott K, Azim Ansari M, Belij-Rammerstorfer S, Bibi S, Blackwell L, Brown A, Brown H, Cavell B, Clutterbuck EA, de Silva T, Eyre D, Lumley S, Flaxman A, Grist J, Hackstein CP, Halkerston R, Harding AC, Hill J, James T, Jay C, Johnson SA, Kronsteiner B, Lie Y, Linder A, Longet S, Marinou S, Matthews PC, Mellors J, Petropoulos C, Rongkard P, Sedik C, Silva-Reyes L, Smith H, Stockdale L, Taylor S, Thomas S, Tipoe T, Turtle L, Vieira VA, Wrin T, Pollard AJ, Lambe T, Conlon CP, Jeffery K, Travis S, Goulder P, Frater J, Mentzer AJ, Stafford L, Carroll MW, James WS, Klenerman P, Barnes E, Dold C, Dunachie SJ. Divergent trajectories of antiviral memory after SARS-CoV-2 infection. Nat Commun 2022; 13:1251. [PMID: 35273178 PMCID: PMC8913789 DOI: 10.1038/s41467-022-28898-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/17/2022] [Indexed: 12/17/2022] Open
Abstract
The trajectories of acquired immunity to severe acute respiratory syndrome coronavirus 2 infection are not fully understood. We present a detailed longitudinal cohort study of UK healthcare workers prior to vaccination, presenting April-June 2020 with asymptomatic or symptomatic infection. Here we show a highly variable range of responses, some of which (T cell interferon-gamma ELISpot, N-specific antibody) wane over time, while others (spike-specific antibody, B cell memory ELISpot) are stable. We use integrative analysis and a machine-learning approach (SIMON - Sequential Iterative Modeling OverNight) to explore this heterogeneity. We identify a subgroup of participants with higher antibody responses and interferon-gamma ELISpot T cell responses, and a robust trajectory for longer term immunity associates with higher levels of neutralising antibodies against the infecting (Victoria) strain and also against variants B.1.1.7 (alpha) and B.1.351 (beta). These variable trajectories following early priming may define subsequent protection from severe disease from novel variants.
Collapse
Affiliation(s)
- Adriana Tomic
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Donal T Skelly
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Nuffield Dept of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Matthew Pace
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Emily Adland
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Frances Alexander
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Kirk Allott
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - M Azim Ansari
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | | | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Luke Blackwell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Helen Brown
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Breeze Cavell
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
| | | | - Thushan de Silva
- The Florey Institute for Host-Pathogen Interactions and Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
| | - David Eyre
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Big Data Institute, Nuffield Dept. of Population Health, University of Oxford, Oxford, UK
| | - Sheila Lumley
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Amy Flaxman
- Jenner Institute, University of Oxford, Oxford, UK
| | - James Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK
| | - Carl-Philipp Hackstein
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Rachel Halkerston
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
| | - Adam C Harding
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Tim James
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Cecilia Jay
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Síle A Johnson
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Oxford University Medical School, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford Centre For Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Yolanda Lie
- Monogram Biosciences LabCorp, San Francisco, CA, USA
| | - Aline Linder
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Stephanie Longet
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Spyridoula Marinou
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Philippa C Matthews
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jack Mellors
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
| | | | - Patpong Rongkard
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Cynthia Sedik
- Monogram Biosciences LabCorp, San Francisco, CA, USA
| | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Holly Smith
- Jenner Institute, University of Oxford, Oxford, UK
| | - Lisa Stockdale
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Stephen Taylor
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
| | - Stephen Thomas
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
| | - Timothy Tipoe
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Lance Turtle
- HPRU in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust (a member of Liverpool Health Partners), Liverpool, UK
| | - Vinicius Adriano Vieira
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Terri Wrin
- Monogram Biosciences LabCorp, San Francisco, CA, USA
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Teresa Lambe
- Jenner Institute, University of Oxford, Oxford, UK
| | - Chris P Conlon
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katie Jeffery
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Simon Travis
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, UK
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alex J Mentzer
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lizzie Stafford
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Miles W Carroll
- United Kingdom Health Security Agency, Porton Down, Wiltshire, England
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - William S James
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK.
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Susanna J Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Oxford Centre For Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| |
Collapse
|
1206
|
Sablerolles RSG, Rietdijk WJR, Goorhuis A, Postma DF, Visser LG, Geers D, Schmitz KS, Garcia Garrido HM, Koopmans MPG, Dalm VASH, Kootstra NA, Huckriede ALW, Lafeber M, van Baarle D, GeurtsvanKessel CH, de Vries RD, van der Kuy PHM. Immunogenicity and Reactogenicity of Vaccine Boosters after Ad26.COV2.S Priming. N Engl J Med 2022; 386:951-963. [PMID: 35045226 PMCID: PMC8796791 DOI: 10.1056/nejmoa2116747] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The Ad26.COV2.S vaccine, which was approved as a single-shot immunization regimen, has been shown to be effective against severe coronavirus disease 2019. However, this vaccine induces lower severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein (S)-specific antibody levels than those induced by messenger RNA (mRNA)-based vaccines. The immunogenicity and reactogenicity of a homologous or heterologous booster in persons who have received an Ad26.COV2.S priming dose are unclear. METHODS In this single-blind, multicenter, randomized, controlled trial involving health care workers who had received a priming dose of Ad26.COV2.S vaccine, we assessed immunogenicity and reactogenicity 28 days after a homologous or heterologous booster vaccination. The participants were assigned to receive no booster, an Ad26.COV2.S booster, an mRNA-1273 booster, or a BNT162b2 booster. The primary end point was the level of S-specific binding antibodies, and the secondary end points were the levels of neutralizing antibodies, S-specific T-cell responses, and reactogenicity. A post hoc analysis was performed to compare mRNA-1273 boosting with BNT162b2 boosting. RESULTS Homologous or heterologous booster vaccination resulted in higher levels of S-specific binding antibodies, neutralizing antibodies, and T-cell responses than a single Ad26.COV2.S vaccination. The increase in binding antibodies was significantly larger with heterologous regimens that included mRNA-based vaccines than with the homologous booster. The mRNA-1273 booster was most immunogenic and was associated with higher reactogenicity than the BNT162b2 and Ad26.COV2.S boosters. Local and systemic reactions were generally mild to moderate in the first 2 days after booster administration. CONCLUSIONS The Ad26.COV2.S and mRNA boosters had an acceptable safety profile and were immunogenic in health care workers who had received a priming dose of Ad26.COV2.S vaccine. The strongest responses occurred after boosting with mRNA-based vaccines. Boosting with any available vaccine was better than not boosting. (Funded by the Netherlands Organization for Health Research and Development ZonMw; SWITCH ClinicalTrials.gov number, NCT04927936.).
Collapse
Affiliation(s)
- Roos S G Sablerolles
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Wim J R Rietdijk
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Abraham Goorhuis
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Douwe F Postma
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Leo G Visser
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Daryl Geers
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Katharina S Schmitz
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Hannah M Garcia Garrido
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Marion P G Koopmans
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Virgil A S H Dalm
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Neeltje A Kootstra
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Anke L W Huckriede
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Melvin Lafeber
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Debbie van Baarle
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Corine H GeurtsvanKessel
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - Rory D de Vries
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| | - P Hugo M van der Kuy
- From the Departments of Internal Medicine (R.S.G.S., M.L.), Hospital Pharmacy (R.S.G.S., W.J.R.R., P.H.M.K.), and Viroscience (D.G., K.S.S., M.P.G.K., C.H.G., R.D.V.) and the Department of Internal Medicine, Division of Allergy and Clinical Immunology, and Department of Immunology (V.A.S.H.D.), Erasmus University Medical Center, Rotterdam, the Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases (A.G., H.M.G.G.), and the Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam (N.A.K.), Amsterdam, the Department of Internal Medicine and Infectious Diseases (D.F.P.), and the Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen (A.L.W.H., D.B.), Groningen, the Department of Infectious Diseases, Leiden University Medical Center, Leiden (L.G.V.), and the Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven (D.B.) - all in the Netherlands
| |
Collapse
|
1207
|
Zhang H, Jia Y, Ji Y, Cong X, Liu Y, Yang R, Kong X, Shi Y, Zhu L, Wang Z, Wang W, Fei R, Liu F, Lu F, Chen H, Rao H. Inactivated Vaccines Against SARS-CoV-2: Neutralizing Antibody Titers in Vaccine Recipients. Front Microbiol 2022; 13:816778. [PMID: 35359732 PMCID: PMC8960924 DOI: 10.3389/fmicb.2022.816778] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/14/2022] [Indexed: 01/01/2023] Open
Abstract
Background Although effective vaccines have been developed against coronavirus disease 2019 (COVID-19), the level of neutralizing antibodies (NAbs) induced after vaccination in the real world is still unknown. The aim of this work was to evaluate the level and persistence of NAbs induced by two inactivated COVID-19 vaccines in China. Methods Serum samples were collected from 1,335 people aged 18 years and over who were vaccinated with an inactivated COVID-19 vaccine at Peking University People’s Hospital from January 19 to June 23, 2021, for the detection of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies. Results The positive rate for NAbs against SARS-CoV-2 was 79–91% from the first month to the second month after the second vaccine dose. The gradual decline in positivity rate for NAb response was observed from 78% at 3 months post-vaccination to 0% at 12 months post-vaccination. When there was a 21-day interval between the two doses of vaccine, the NAb positivity rate was 0% 6 months after the second dose. NAb levels were significantly higher when the interval between two doses were 3–8 weeks than when it was 0–3 weeks (χ2 = 14.04, p < 0.001). There was a linear correlation between NAbs and IgG antibodies in 1,335 vaccinated patients. NAb levels decreased in 31 patients (81.6%) and increased in 7 patients (18.4%) over time in the series of 38 patients after the second vaccination. The NAb positivity rate was significantly higher in 18- to 40-year-old subjects than in 41- to 60-year-old subjects (t = −1.959, p < 0.01; t = 0.839, p < 0.01). Conclusion The NAb positivity rate was the highest at the first and second month after the second dose of vaccine, and gradually decreased over time. With a 21-day interval between two doses of vaccine, neutralizing antibody levels persisted for only 6 months after the second dose of vaccine. Therefore, a third vaccine dose is recommended. Our results suggest that in cases in which NAbs cannot be detected, IgM/IgG antibodies can be detected instead. The level of NAbs produced after vaccination was affected by age but not by sex. Our results suggest that an interval of 21 to 56 days between shots is suitable for vaccination.
Collapse
|
1208
|
Spies R, Potter M, Govender S, Kirk L, Rauch S, Black J. SARS-CoV-2 infection in public hospital medical doctors in an Eastern Cape metro. S Afr J Infect Dis 2022; 37:335. [PMID: 35399560 PMCID: PMC8991316 DOI: 10.4102/sajid.v37i1.335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/22/2021] [Indexed: 01/08/2023] Open
Affiliation(s)
- Ruan Spies
- Department of Medicine, Port Elizabeth Hospital Complex, Gqeberha, South Africa
| | - Matthew Potter
- Department of Medicine, Port Elizabeth Hospital Complex, Gqeberha, South Africa
| | - Sudarshan Govender
- Department of Medicine, Port Elizabeth Hospital Complex, Gqeberha, South Africa
| | - Luke Kirk
- Department of Medicine, Port Elizabeth Hospital Complex, Gqeberha, South Africa
| | - Simon Rauch
- Department of Medicine, Port Elizabeth Hospital Complex, Gqeberha, South Africa
| | - John Black
- Department of Infectious Diseases, Livingstone Hospital, Gqeberha, South Africa
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
1209
|
van der Velden YU, Grobben M, Caniels TG, Burger JA, Poniman M, Oomen M, Rijnstra ESV, Tejjani K, Guerra D, Kempers R, Stegmann T, van Gils MJ, Sanders RW. A SARS-CoV-2 Wuhan spike virosome vaccine induces superior neutralization breadth compared to one using the Beta spike. Sci Rep 2022; 12:3884. [PMID: 35273217 PMCID: PMC8913678 DOI: 10.1038/s41598-022-07590-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Current SARS-CoV-2 vaccines are effective, but long-term protection is threatened by the emergence of virus variants. We generated a virosome vaccine containing the Beta spike protein and compared its immunogenicity in mice to a virosome vaccine containing the original Wuhan spike. Two administrations of the virosomes induced potent SARS-CoV-2 neutralizing antibodies in both vaccine groups. The level of autologous neutralization in Beta-vaccinated mice was similar to the level of autologous neutralization in Wuhan-vaccinated mice. However, heterologous neutralization to the Wuhan strain in Beta-vaccinated mice was 4.7-fold lower than autologous neutralization, whereas heterologous neutralization to the Beta strain in Wuhan-vaccinated mice was reduced by only 1.9-fold compared to autologous neutralization levels. In addition, neutralizing activity against the D614G, Alpha and Delta variants was also significantly lower after Beta spike vaccination than after Wuhan spike vaccination. Our results show that Beta spike vaccination induces inferior neutralization breadth. These results are informative for programs aimed to develop broadly active SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Yme U van der Velden
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Marloes Grobben
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Tom G Caniels
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Judith A Burger
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Meliawati Poniman
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Melissa Oomen
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Esther Siteur-van Rijnstra
- Experimental Immunology, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Khadija Tejjani
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Denise Guerra
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Ronald Kempers
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Toon Stegmann
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Marit J van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Rogier W Sanders
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
1210
|
Lin DY, Gu Y, Wheeler B, Young H, Holloway S, Sunny SK, Moore Z, Zeng D. Effectiveness of Covid-19 Vaccines over a 9-Month Period in North Carolina. N Engl J Med 2022; 386:933-941. [PMID: 35020982 PMCID: PMC8781317 DOI: 10.1056/nejmoa2117128] [Citation(s) in RCA: 231] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The duration of protection afforded by coronavirus disease 2019 (Covid-19) vaccines in the United States is unclear. Whether the increase in postvaccination infections during the summer of 2021 was caused by declining immunity over time, the emergence of the B.1.617.2 (delta) variant, or both is unknown. METHODS We extracted data regarding Covid-19-related vaccination and outcomes during a 9-month period (December 11, 2020, to September 8, 2021) for approximately 10.6 million North Carolina residents by linking data from the North Carolina Covid-19 Surveillance System and the Covid-19 Vaccine Management System. We used a Cox regression model to estimate the effectiveness of the BNT162b2 (Pfizer-BioNTech), mRNA-1273 (Moderna), and Ad26.COV2.S (Johnson & Johnson-Janssen) vaccines in reducing the current risks of Covid-19, hospitalization, and death, as a function of time elapsed since vaccination. RESULTS For the two-dose regimens of messenger RNA (mRNA) vaccines BNT162b2 (30 μg per dose) and mRNA-1273 (100 μg per dose), vaccine effectiveness against Covid-19 was 94.5% (95% confidence interval [CI], 94.1 to 94.9) and 95.9% (95% CI, 95.5 to 96.2), respectively, at 2 months after the first dose and decreased to 66.6% (95% CI, 65.2 to 67.8) and 80.3% (95% CI, 79.3 to 81.2), respectively, at 7 months. Among early recipients of BNT162b2 and mRNA-1273, effectiveness decreased by approximately 15 and 10 percentage points, respectively, from mid-June to mid-July, when the delta variant became dominant. For the one-dose regimen of Ad26.COV2.S (5 × 1010 viral particles), effectiveness against Covid-19 was 74.8% (95% CI, 72.5 to 76.9) at 1 month and decreased to 59.4% (95% CI, 57.2 to 61.5) at 5 months. All three vaccines maintained better effectiveness in preventing hospitalization and death than in preventing infection over time, although the two mRNA vaccines provided higher levels of protection than Ad26.COV2.S. CONCLUSIONS All three Covid-19 vaccines had durable effectiveness in reducing the risks of hospitalization and death. Waning protection against infection over time was due to both declining immunity and the emergence of the delta variant. (Funded by a Dennis Gillings Distinguished Professorship and the National Institutes of Health.).
Collapse
Affiliation(s)
- Dan-Yu Lin
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| | - Yu Gu
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| | - Bradford Wheeler
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| | - Hayley Young
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| | - Shannon Holloway
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| | - Shadia-Khan Sunny
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| | - Zack Moore
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| | - Donglin Zeng
- From the Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill (D.-Y.L., Y.G., D.Z.), and the North Carolina Department of Health and Human Services (B.W., H.Y., Z.M.), North Carolina State University (S.H.), and the CDC Foundation, North Carolina Department of Health and Human Services (S.-K.S.), Raleigh
| |
Collapse
|
1211
|
Marcelin JR, Pettifor A, Janes H, Brown ER, Kublin JG, Stephenson KE. COVID-19 Vaccines and SARS-CoV-2 Transmission in the Era of New Variants: A Review and Perspective. Open Forum Infect Dis 2022; 9:ofac124. [PMID: 35493113 PMCID: PMC8992234 DOI: 10.1093/ofid/ofac124] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 11/22/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines have yielded definitive prevention and major reductions in morbidity and mortality from severe acute respiratory syndrome coronavirus 2 infection, even in the context of emerging and persistent variants of concern. Newer variants have revealed less vaccine protection against infection and attenuation of vaccine effects on transmission. COVID-19 vaccines still likely reduce transmission compared with not being vaccinated at all, even with variants of concern; however, determining the magnitude of transmission reduction is constrained by the challenges of performing these studies, requiring accurate linkage of infections to vaccine status and timing thereof, particularly within households. In this review, we synthesize the currently available data on the impact of COVID-19 vaccines on infection, serious illness, and transmission; we also identify the challenges and opportunities associated with policy development based on this data.
Collapse
Affiliation(s)
- Jasmine R Marcelin
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha Nebraska, USA
| | | | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth R Brown
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
1212
|
Salleh MZ, Norazmi MN, Deris ZZ. Immunogenicity mechanism of mRNA vaccines and their limitations in promoting adaptive protection against SARS-CoV-2. PeerJ 2022; 10:e13083. [PMID: 35287350 PMCID: PMC8917804 DOI: 10.7717/peerj.13083] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/16/2022] [Indexed: 01/12/2023] Open
Abstract
Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19) in late 2019, hundreds of millions of people have been infected worldwide. There have been unprecedented efforts in acquiring effective vaccines to confer protection against the disease. mRNA vaccines have emerged as promising alternatives to conventional vaccines due to their high potency with the capacity for rapid development and low manufacturing costs. In this review, we summarize the currently available vaccines against SARS-CoV-2 in development, with the focus on the concepts of mRNA vaccines, their antigen selection, delivery and optimization to increase the immunostimulatory capability of mRNA as well as its stability and translatability. We also discuss the host immune responses to the SARS-CoV-2 infection and expound in detail, the adaptive immune response upon immunization with mRNA vaccines, in which high levels of spike-specific IgG and neutralizing antibodies were detected after two-dose vaccination. mRNA vaccines have been shown to induce a robust CD8+T cell response, with a balanced CD4+ TH1/TH2 response. We further discuss the challenges and limitations of COVID-19 mRNA vaccines, where newly emerging variants of SARS-CoV-2 may render currently deployed vaccines less effective. Imbalanced and inappropriate inflammatory responses, resulting from hyper-activation of pro-inflammatory cytokines, which may lead to vaccine-associated enhanced respiratory disease (VAERD) and rare cases of myocarditis and pericarditis also are discussed.
Collapse
Affiliation(s)
- Mohd Zulkifli Salleh
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bahru, Kelantan, Malaysia
| | - Mohd Nor Norazmi
- School of Health Sciences, Universiti Sains Malaysia, Kota Bahru, Kelantan, Malaysia
| | - Zakuan Zainy Deris
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bahru, Kelantan, Malaysia
| |
Collapse
|
1213
|
Gallo K, Goede A, Mura C, Abel R, Moahamed B, Preissner S, Nahles S, Heiland M, Bourne PE, Preissner R, Mallach M. A Comparative Analysis of COVID-19 Vaccines Based on over 580,000 Cases from the Vaccination Adverse Event Reporting System. Vaccines (Basel) 2022; 10:vaccines10030408. [PMID: 35335040 PMCID: PMC8950485 DOI: 10.3390/vaccines10030408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/23/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
Background: The COVID-19 pandemic is being battled via the largest vaccination campaign in history, with more than eight billion doses administered thus far. Therefore, discussions about potentially adverse reactions, and broader safety concerns, are critical. The U.S. Vaccination Adverse Event Reporting System (VAERS) has recorded vaccination side effects for over 30 years. About 580,000 events have been filed for COVID-19 thus far, primarily for the Johnson & Johnson (New Jersey, USA), Pfizer/BioNTech (Mainz, Germany), and Moderna (Cambridge, USA) vaccines. Methods: Using available databases, we evaluated these three vaccines in terms of the occurrence of four generally-noticed adverse reactions—namely, cerebral venous sinus thrombosis, Guillain−Barré syndrome (a severe paralytic neuropathy), myocarditis, and pericarditis. Our statistical analysis also included a calculation of odds ratios (ORs) based on total vaccination numbers, accounting for incidence rates in the general population. Results: ORs for a number of adverse events and patient groups were (largely) increased, most notably for the occurrence of cerebral venous sinus thrombosis after vaccination with the Johnson & Johnson vaccine. The overall population OR of 10 increases to 12.5 when limited to women, and further yet (to 14.4) among women below age 50 yrs. In addition, elevated risks were found (i) for Guillain−Barré syndrome (OR of 11.6) and (ii) for myocarditis/pericarditis (ORs of 5.3/4.1, respectively) among young men (<25 yrs) vaccinated with the Pfizer/BioNTech vaccine. Conclusions: Any conclusions from such a retrospective, real-world data analysis must be drawn cautiously, and should be confirmed by prospective double-blinded clinical trials. In addition, we emphasize that the adverse events reported here are not specific side effects of COVID vaccines, and the significant, well-established benefits of COVID-19 vaccination outweigh the potential complications surveyed here.
Collapse
Affiliation(s)
- Kathleen Gallo
- Institute of Physiology and Science IT, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (K.G.); (A.G.); (R.A.); (B.M.); (M.M.)
| | - Andrean Goede
- Institute of Physiology and Science IT, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (K.G.); (A.G.); (R.A.); (B.M.); (M.M.)
| | - Cameron Mura
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA; (C.M.); (P.E.B.)
| | - Renata Abel
- Institute of Physiology and Science IT, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (K.G.); (A.G.); (R.A.); (B.M.); (M.M.)
| | - Barbara Moahamed
- Institute of Physiology and Science IT, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (K.G.); (A.G.); (R.A.); (B.M.); (M.M.)
| | - Saskia Preissner
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (S.P.); (S.N.); (M.H.)
| | - Susanne Nahles
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (S.P.); (S.N.); (M.H.)
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (S.P.); (S.N.); (M.H.)
| | - Philip E. Bourne
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA; (C.M.); (P.E.B.)
| | - Robert Preissner
- Institute of Physiology and Science IT, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (K.G.); (A.G.); (R.A.); (B.M.); (M.M.)
- Correspondence: ; Tel.: +49-30-45065-5208
| | - Michael Mallach
- Institute of Physiology and Science IT, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (K.G.); (A.G.); (R.A.); (B.M.); (M.M.)
| |
Collapse
|
1214
|
Tada T, Zhou H, Samanovic MI, Dcosta BM, Cornelius A, Herati RS, Mulligan MJ, Landau NR. Neutralization of SARS-CoV-2 Variants by mRNA and Adenoviral Vector Vaccine-Elicited Antibodies. Front Immunol 2022; 13:797589. [PMID: 35350781 PMCID: PMC8957851 DOI: 10.3389/fimmu.2022.797589] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/09/2022] [Indexed: 01/19/2023] Open
Abstract
The increasing prevalence of SARS-CoV-2 variants has raised concerns regarding possible decreases in vaccine effectiveness. Here, neutralizing antibody titers elicited by mRNA-based and adenoviral vector-based vaccines against variant pseudotyped viruses were measured. BNT162b2 and mRNA-1273-elicited antibodies showed modest neutralization resistance against Beta, Delta, Delta plus and Lambda variants whereas Ad26.COV2.S-elicited antibodies from a significant fraction of vaccinated individuals had less neutralizing titer (IC50 <50). The data underscore the importance of surveillance for breakthrough infections that result in severe COVID-19 and suggest a potential benefit by second immunization following Ad26.COV2.S to increase protection from current and future variants.
Collapse
Affiliation(s)
- Takuya Tada
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States
| | - Hao Zhou
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States
| | - Marie I. Samanovic
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, United States
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Belinda M. Dcosta
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States
| | - Amber Cornelius
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, United States
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Ramin S. Herati
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, United States
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Mark J. Mulligan
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, United States
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Nathaniel R. Landau
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
1215
|
Zhou P, Song G, He WT, Beutler N, Tse LV, Martinez DR, Schäfer A, Anzanello F, Yong P, Peng L, Dueker K, Musharrafieh R, Callaghan S, Capozzola T, Yuan M, Liu H, Limbo O, Parren M, Garcia E, Rawlings SA, Smith DM, Nemazee D, Jardine JG, Wilson IA, Safonova Y, Rogers TF, Baric RS, Gralinski LE, Burton DR, Andrabi R. Broadly neutralizing anti-S2 antibodies protect against all three human betacoronaviruses that cause severe disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.04.479488. [PMID: 35291291 PMCID: PMC8923106 DOI: 10.1101/2022.03.04.479488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Pan-betacoronavirus neutralizing antibodies may hold the key to developing broadly protective vaccines against coronaviruses that cause severe disease, for anticipating novel pandemic-causing viruses, and to respond more effectively to SARS-CoV-2 variants. The emergence of the Omicron variant of SARS-CoV-2 has illustrated the limitations of solely targeting the receptor binding domain (RBD) of the envelope Spike (S)-protein. Here, we isolated a large panel of broadly neutralizing antibodies (bnAbs) from SARS-CoV-2 recovered-vaccinated donors that target a conserved S2 region in the fusion machinery on betacoronavirus spikes. Select bnAbs show broad in vivo protection against all three pathogenic betacoronaviruses, SARS-CoV-1, SARS-CoV-2 and MERS-CoV, that have spilled over into humans in the past 20 years to cause severe disease. The bnAbs provide new opportunities for antibody-based interventions and key insights for developing pan-betacoronavirus vaccines.
Collapse
Affiliation(s)
- Panpan Zhou
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wan-ting He
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Longping V. Tse
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R. Martinez
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexandra Schäfer
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Fabio Anzanello
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peter Yong
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Linghang Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Katharina Dueker
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rami Musharrafieh
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean Callaghan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tazio Capozzola
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Oliver Limbo
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mara Parren
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Stephen A. Rawlings
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Davey M. Smith
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Joseph G. Jardine
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A. Wilson
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yana Safonova
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Thomas F. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Ralph S. Baric
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Departments of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lisa E. Gralinski
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
1216
|
Association of severity and mortality of Covid-19 cases among acute kidney injury and sexual dimorphism. Mol Biol Rep 2022; 49:6753-6762. [PMID: 35249167 PMCID: PMC8898193 DOI: 10.1007/s11033-022-07308-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/24/2022] [Indexed: 11/26/2022]
Abstract
Introduction The outbreak of coronavirus disease 2019 (Covid-19) severely impacted global health and economic status. The native receptor-ligand interaction of Angiotensin-converting enzyme 2 (ACE2) and S protein induces host cell pathogenesis via immunosuppression. Material and Methods The emerging evidence reports the sex disparity in Covid-19 induced mortality rate which affects abundantly men population. Although the biological interaction of Covid-19 with receptor upregulates the viral genome protein interactions and initiates the predictive multiorgan failure followed by acute kidney injury (AKI) in Covid-19 infected male population. Conclusion Besides, the knowledge and lessons learned from the study depict that cellular and molecular links may explain the risk and severity of Covid-19 and AKI in the male population and lead to management of Covid-19 induced AKI. Therefore, this review explored the pathways associated with the pathogenesis of two diseased conditions with sex disparity.
Collapse
|
1217
|
Sadoff J, Gray G, Vandebosch A, Cárdenas V, Shukarev G, Grinsztejn B, Goepfert PA, Truyers C, Van Dromme I, Spiessens B, Vingerhoets J, Custers J, Scheper G, Robb ML, Treanor J, Ryser MF, Barouch DH, Swann E, Marovich MA, Neuzil KM, Corey L, Stoddard J, Hardt K, Ruiz-Guiñazú J, Le Gars M, Schuitemaker H, Van Hoof J, Struyf F, Douoguih M. Final Analysis of Efficacy and Safety of Single-Dose Ad26.COV2.S. N Engl J Med 2022; 386:847-860. [PMID: 35139271 PMCID: PMC8849184 DOI: 10.1056/nejmoa2117608] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The Ad26.COV2.S vaccine was highly effective against severe-critical coronavirus disease 2019 (Covid-19), hospitalization, and death in the primary phase 3 efficacy analysis. METHODS We conducted the final analysis in the double-blind phase of our multinational, randomized, placebo-controlled trial, in which adults were assigned in a 1:1 ratio to receive single-dose Ad26.COV2.S (5×1010 viral particles) or placebo. The primary end points were vaccine efficacy against moderate to severe-critical Covid-19 with onset at least 14 days after administration and at least 28 days after administration in the per-protocol population. Safety and key secondary and exploratory end points were also assessed. RESULTS Median follow-up in this analysis was 4 months; 8940 participants had at least 6 months of follow-up. In the per-protocol population (39,185 participants), vaccine efficacy against moderate to severe-critical Covid-19 at least 14 days after administration was 56.3% (95% confidence interval [CI], 51.3 to 60.8; 484 cases in the vaccine group vs. 1067 in the placebo group); at least 28 days after administration, vaccine efficacy was 52.9% (95% CI, 47.1 to 58.1; 433 cases in the vaccine group vs. 883 in the placebo group). Efficacy in the United States, primarily against the reference strain (B.1.D614G) and the B.1.1.7 (alpha) variant, was 69.7% (95% CI, 60.7 to 76.9); efficacy was reduced elsewhere against the P.1 (gamma), C.37 (lambda), and B.1.621 (mu) variants. Efficacy was 74.6% (95% CI, 64.7 to 82.1) against severe-critical Covid-19 (with only 4 severe-critical cases caused by the B.1.617.2 [delta] variant), 75.6% (95% CI, 54.3 to 88.0) against Covid-19 leading to medical intervention (including hospitalization), and 82.8% (95% CI, 40.5 to 96.8) against Covid-19-related death, with protection lasting 6 months or longer. Efficacy against any severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection was 41.7% (95% CI, 36.3 to 46.7). Ad26.COV2.S was associated with mainly mild-to-moderate adverse events, and no new safety concerns were identified. CONCLUSIONS A single dose of Ad26.COV2.S provided 52.9% protection against moderate to severe-critical Covid-19. Protection varied according to variant; higher protection was observed against severe Covid-19, medical intervention, and death than against other end points and lasted for 6 months or longer. (Funded by Janssen Research and Development and others; ENSEMBLE ClinicalTrials.gov number, NCT04505722.).
Collapse
Affiliation(s)
- Jerald Sadoff
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Glenda Gray
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - An Vandebosch
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Vicky Cárdenas
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Georgi Shukarev
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Beatriz Grinsztejn
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Paul A Goepfert
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Carla Truyers
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Ilse Van Dromme
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Bart Spiessens
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Johan Vingerhoets
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Jerome Custers
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Gert Scheper
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Merlin L Robb
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - John Treanor
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Martin F Ryser
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Dan H Barouch
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Edith Swann
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Mary A Marovich
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Kathleen M Neuzil
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Lawrence Corey
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Jeffrey Stoddard
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Karin Hardt
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Javier Ruiz-Guiñazú
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Mathieu Le Gars
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Hanneke Schuitemaker
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Johan Van Hoof
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Frank Struyf
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| | - Macaya Douoguih
- From Janssen Vaccines and Prevention, Leiden, the Netherlands (J. Sadoff, G. Shukarev, J.C., G. Scheper, M.L.G., H.S., J.V.H., M.D.); the South African Research Council, Cape Town, South Africa (G.G.); Janssen Research and Development, Beerse, Belgium (A.V., C.T., I.V.D., B.S., J.V., M.F.R., K.H., J.R.-G., F.S.); Janssen Research and Development, Spring House, PA (V.C.); Evandro Chagas National Institute of Infectious Diseases-Fiocruz, Rio de Janeiro (B.G.); the University of Alabama at Birmingham, Birmingham (P.A.G.); Walter Reed Army Institute of Research, Silver Spring (M.L.R.), the National Institute of Allergy and Infectious Diseases, Rockville (E.S., M.A.M.), and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (K.M.N.) - all in Maryland; the Biomedical Advanced Research and Development Authority, Washington, DC (J.T.); the Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston (D.H.B.); the Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle (L.C.); and Janssen Research and Development, Raritan, NJ (J. Stoddard)
| |
Collapse
|
1218
|
Hatcher SM, Endres-Dighe SM, Angulo FJ, Srivastava A, Nguyen JL, Khan F, Martin C, Swerdlow DL, McLaughlin JM, Ubaka-Blackmore N, Brown LM. COVID-19 Vaccine Effectiveness: A Review of the First 6 Months of COVID-19 Vaccine Availability (1 January-30 June 2021). Vaccines (Basel) 2022; 10:393. [PMID: 35335025 PMCID: PMC8951318 DOI: 10.3390/vaccines10030393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 02/07/2023] Open
Abstract
Observational studies are needed to demonstrate real-world vaccine effectiveness (VE) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outcomes. Our objective was to conduct a review of published SARS-CoV-2 VE articles, supplemented by preprints, during the first 6 months of COVID-19 vaccine availability. This review compares the effectiveness of completing the primary COVID-19 vaccination series against multiple SARS-CoV-2 disease presentations and disease severity outcomes in three population groups (general population, frontline workers, and older adults). Four hundred and seventy-one published articles and 47 preprints were identified. After title and abstract screening and full article review, 50 studies (28 published articles, 22 preprints) were included. VE results were reported for five COVID-19 vaccines and four combinations of COVID-19 vaccines. VE results for BNT162b2 were reported in 70.6% of all studies. Seventeen studies reported variant specific VE estimates; Alpha was the most common. This comprehensive review demonstrates that COVID-19 vaccination is an important tool for preventing COVID-19 morbidity and mortality among fully vaccinated persons aged 16 years and older and serves as an important baseline from which to follow future trends in COVID-19 evolution and effectiveness of new and updated vaccines.
Collapse
Affiliation(s)
- Sarah M. Hatcher
- RTI International, Research Triangle Park, NC 27709, USA; (S.M.H.); (S.M.E.-D.); (N.U.-B.); (L.M.B.)
| | - Stacy M. Endres-Dighe
- RTI International, Research Triangle Park, NC 27709, USA; (S.M.H.); (S.M.E.-D.); (N.U.-B.); (L.M.B.)
| | - Frederick J. Angulo
- Pfizer, Inc., New York, NY 10017, USA; (A.S.); (J.L.N.); (F.K.); (C.M.); (D.L.S.); (J.M.M.)
| | - Amit Srivastava
- Pfizer, Inc., New York, NY 10017, USA; (A.S.); (J.L.N.); (F.K.); (C.M.); (D.L.S.); (J.M.M.)
| | - Jennifer L. Nguyen
- Pfizer, Inc., New York, NY 10017, USA; (A.S.); (J.L.N.); (F.K.); (C.M.); (D.L.S.); (J.M.M.)
| | - Farid Khan
- Pfizer, Inc., New York, NY 10017, USA; (A.S.); (J.L.N.); (F.K.); (C.M.); (D.L.S.); (J.M.M.)
| | - Catherine Martin
- Pfizer, Inc., New York, NY 10017, USA; (A.S.); (J.L.N.); (F.K.); (C.M.); (D.L.S.); (J.M.M.)
| | - David L. Swerdlow
- Pfizer, Inc., New York, NY 10017, USA; (A.S.); (J.L.N.); (F.K.); (C.M.); (D.L.S.); (J.M.M.)
| | - John M. McLaughlin
- Pfizer, Inc., New York, NY 10017, USA; (A.S.); (J.L.N.); (F.K.); (C.M.); (D.L.S.); (J.M.M.)
| | - Nneka Ubaka-Blackmore
- RTI International, Research Triangle Park, NC 27709, USA; (S.M.H.); (S.M.E.-D.); (N.U.-B.); (L.M.B.)
| | - Linda Morris Brown
- RTI International, Research Triangle Park, NC 27709, USA; (S.M.H.); (S.M.E.-D.); (N.U.-B.); (L.M.B.)
| |
Collapse
|
1219
|
Afkhami S, D'Agostino MR, Zhang A, Stacey HD, Marzok A, Kang A, Singh R, Bavananthasivam J, Ye G, Luo X, Wang F, Ang JC, Zganiacz A, Sankar U, Kazhdan N, Koenig JFE, Phelps A, Gameiro SF, Tang S, Jordana M, Wan Y, Mossman KL, Jeyanathan M, Gillgrass A, Medina MFC, Smaill F, Lichty BD, Miller MS, Xing Z. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell 2022; 185:896-915.e19. [PMID: 35180381 PMCID: PMC8825346 DOI: 10.1016/j.cell.2022.02.005] [Citation(s) in RCA: 206] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/16/2021] [Accepted: 02/02/2022] [Indexed: 12/28/2022]
Abstract
The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered intramuscularly and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. We show that single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to intramuscular immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.
Collapse
Affiliation(s)
- Sam Afkhami
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ali Zhang
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Hannah D Stacey
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Art Marzok
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alisha Kang
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ramandeep Singh
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Jegarubee Bavananthasivam
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Gluke Ye
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Xiangqian Luo
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Fuan Wang
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Jann C Ang
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Uma Sankar
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Natallia Kazhdan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Joshua F E Koenig
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Allyssa Phelps
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Steven F Gameiro
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Shangguo Tang
- Department of Pathology and Molecular Medicine, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Manel Jordana
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Yonghong Wan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Karen L Mossman
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Amy Gillgrass
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Maria Fe C Medina
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Fiona Smaill
- Department of Pathology and Molecular Medicine, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Brian D Lichty
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Matthew S Miller
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Zhou Xing
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
1220
|
Frontera JA, Tamborska AA, Doheim MF, Garcia‐Azorin D, Gezegen H, Guekht A, Yusof Khan AHK, Santacatterina M, Sejvar J, Thakur KT, Westenberg E, Winkler AS, Beghi E, contributors from the Global COVID‐19 Neuro Research Coalition. Neurological Events Reported after COVID-19 Vaccines: An Analysis of VAERS. Ann Neurol 2022; 91:756-771. [PMID: 35233819 PMCID: PMC9082459 DOI: 10.1002/ana.26339] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To identify the rates of neurological events following administration of mRNA (Pfizer, Moderna) or adenovirus vector (Janssen) vaccines in the U.S.. METHODS We utilized publicly available data from the U.S. Vaccine Adverse Event Reporting System (VAERS) collected between January 1, 2021-June 14, 2021. All free text symptoms that were reported within 42 days of vaccine administration were manually reviewed and grouped into 36 individual neurological diagnostic categories. Post-vaccination neurological event rates were compared between vaccine types and to age-matched baseline incidence rates in the U.S. and rates of neurological events following COVID. RESULTS Of 306,907,697 COVID vaccine doses administered during the study timeframe, 314,610 (0.1%) people reported any adverse event and 105,214 (0.03%) reported neurological adverse events in a median of 1 day (IQR0-3) from inoculation. Guillain-Barre Syndrome (GBS), and cerebral venous thrombosis (CVT) occurred in fewer than 1 per 1,000,000 doses. Significantly more neurological adverse events were reported following Janssen (Ad26.COV2.S) vaccination compared to either Pfizer-BioNtech (BNT162b2) or Moderna (mRNA-1273; 0.15% versus 0.03% versus 0.03% of doses, respectively,P<0.0001). The observed-to-expected ratios for GBS, CVT and seizure following Janssen vaccination were ≥1.5-fold higher than background rates. However, the rate of neurological events after acute SARS-CoV-2 infection was up to 617-fold higher than after COVID vaccination. INTERPRETATION Reports of serious neurological events following COVID vaccination are rare. GBS, CVT and seizure may occur at higher than background rates following Janssen vaccination. Despite this, rates of neurological complications following acute SARS-CoV-2 infection are up to 617-fold higher than after COVID vaccination. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jennifer A. Frontera
- Department of NeurologyNew York University Grossman School of MedicineNew YorkNY
| | - Arina A. Tamborska
- Institute of InfectionVeterinary and Ecological Sciences, University of LiverpoolLiverpoolUK
- National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic InfectionsUniversity of LiverpoolLiverpoolUK
- The Walton Centre NHS Foundation TrustLiverpoolUK
| | | | - David Garcia‐Azorin
- Department of NeurologyHospital Clínico Universitario de ValladolidValladolidSpain
| | - Hasim Gezegen
- Faculty of MedicineDepartment of Neurology, Istanbul UniversityIstanbulTurkey
| | - Alla Guekht
- Moscow Research and Clinical Center for NeuropsychiatryMoscowRussia
- Pirogov Russian National Research Medical UniversityMoscowRussia
| | | | | | - James Sejvar
- Division of High‐Consequence Pathogens and PathologyNational Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and PreventionAtlantaGA
| | - Kiran T. Thakur
- Department of NeurologyColumbia University Irving Medical Center/New York Presbyterian HospitalNew YorkNY
| | - Erica Westenberg
- Department of NeurologyCenter for Global Health, Klinikum rechts der Isar, Technical University of MunichMunichGermany
| | - Andrea S. Winkler
- Department of NeurologyCenter for Global Health, Klinikum rechts der Isar, Technical University of MunichMunichGermany
| | - Ettore Beghi
- Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSMilanItaly
| | | |
Collapse
|
1221
|
Use of analgesics/antipyretics in the management of symptoms associated with COVID-19 vaccination. NPJ Vaccines 2022; 7:31. [PMID: 35236842 PMCID: PMC8891349 DOI: 10.1038/s41541-022-00453-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
COVID-19 vaccines are effective and important to control the ongoing pandemic, but vaccine reactogenicity may contribute to poor uptake. Analgesics or antipyretic medications are often used to alleviate vaccine side effects, but their effect on immunogenicity remains uncertain. Few studies have assessed the effect of analgesics/antipyretics on vaccine immunogenicity and reactogenicity. Some studies revealed changes in certain immune response parameters post-vaccination when analgesics/antipyretics were used either prophylactically or therapeutically. Still, there is no evidence that these changes impact vaccine efficacy. Specific data on the impact of analgesic/antipyretic medications on immunogenicity of COVID-19 vaccines are limited. However, available data from clinical trials of licensed vaccines, along with recommendations from public health bodies around the world, should provide reassurance to both healthcare professionals and vaccine recipients that short-term use of analgesics/antipyretics at non-prescription doses is unlikely to affect vaccine-induced immunity.
Collapse
|
1222
|
Friedrich M, Pfeifer G, Binder S, Aigner A, Vollmer Barbosa P, Makert GR, Fertey J, Ulbert S, Bodem J, König EM, Geiger N, Schambach A, Schilling E, Buschmann T, Hauschildt S, Koehl U, Sewald K. Selection and Validation of siRNAs Preventing Uptake and Replication of SARS-CoV-2. Front Bioeng Biotechnol 2022; 10:801870. [PMID: 35309990 PMCID: PMC8925020 DOI: 10.3389/fbioe.2022.801870] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/07/2022] [Indexed: 12/16/2022] Open
Abstract
In 2019, the novel highly infectious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak rapidly led to a global pandemic with more than 346 million confirmed cases worldwide, resulting in 5.5 million associated deaths (January 2022). Entry of all SARS-CoV-2 variants is mediated by the cellular angisin-converting enzyme 2 (ACE2). The virus abundantly replicates in the epithelia of the upper respiratory tract. Beyond vaccines for immunization, there is an imminent need for novel treatment options in COVID-19 patients. So far, only a few drugs have found their way into the clinics, often with modest success. Specific gene silencing based on small interfering RNA (siRNA) has emerged as a promising strategy for therapeutic intervention, preventing/limiting SARS-CoV-2 entry into host cells or interfering with viral replication. Here, we pursued both strategies. We designed and screened nine siRNAs (siA1-9) targeting the viral entry receptor ACE2. SiA1, (siRNA against exon1 of ACE2 mRNA) was most efficient, with up to 90% knockdown of the ACE2 mRNA and protein for at least six days. In vitro, siA1 application was found to protect Vero E6 and Huh-7 cells from infection with SARS-CoV-2 with an up to ∼92% reduction of the viral burden indicating that the treatment targets both the endosomal and the viral entry at the cytoplasmic membrane. Since the RNA-encoded genome makes SARS-CoV-2 vulnerable to RNA interference (RNAi), we designed and analysed eight siRNAs (siV1-8) directly targeting the Orf1a/b region of the SARS-CoV-2 RNA genome, encoding for non-structural proteins (nsp). As a significant hallmark of this study, we identified siV1 (siRNA against leader protein of SARS-CoV-2), which targets the nsp1-encoding sequence (a.k.a. ‘host shutoff factor’) as particularly efficient. SiV1 inhibited SARS-CoV-2 replication in Vero E6 or Huh-7 cells by more than 99% or 97%, respectively. It neither led to toxic effects nor induced type I or III interferon production. Of note, sequence analyses revealed the target sequence of siV1 to be highly conserved in SARS-CoV-2 variants. Thus, our results identify the direct targeting of the viral RNA genome (ORF1a/b) by siRNAs as highly efficient and introduce siV1 as a particularly promising drug candidate for therapeutic intervention.
Collapse
Affiliation(s)
- Maik Friedrich
- Institute of Clinical Immunology, Faculty of Leipzig University of Leipzig, Max-Bürger-Forschungszentrum (MBFZ), Leipzig, Germany
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- *Correspondence: Maik Friedrich,
| | - Gabriele Pfeifer
- Institute of Clinical Immunology, Faculty of Leipzig University of Leipzig, Max-Bürger-Forschungszentrum (MBFZ), Leipzig, Germany
| | - Stefanie Binder
- Institute of Clinical Immunology, Faculty of Leipzig University of Leipzig, Max-Bürger-Forschungszentrum (MBFZ), Leipzig, Germany
| | - Achim Aigner
- Rudolf Boehm Institute for Pharmacology and Toxicology, Clinical Pharmacology, Leipzig University, Faculty of Medicine, Leipzig, Germany
| | | | - Gustavo R. Makert
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Jasmin Fertey
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Sebastian Ulbert
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Jochen Bodem
- Institute of Virology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Eva-Maria König
- Institute of Virology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Nina Geiger
- Institute of Virology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Erik Schilling
- Institute of Clinical Immunology, Faculty of Leipzig University of Leipzig, Max-Bürger-Forschungszentrum (MBFZ), Leipzig, Germany
| | - Tilo Buschmann
- Institute of Clinical Immunology, Faculty of Leipzig University of Leipzig, Max-Bürger-Forschungszentrum (MBFZ), Leipzig, Germany
| | | | - Ulrike Koehl
- Institute of Clinical Immunology, Faculty of Leipzig University of Leipzig, Max-Bürger-Forschungszentrum (MBFZ), Leipzig, Germany
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Institute for Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Katherina Sewald
- Fraunhofer Institute of Toxicology and Experimental Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH) of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
1223
|
Akelew Y, Andualem H, Ebrahim E, Atnaf A, Hailemichael W. Immunomodulation of COVID‐19 severity by helminth co‐infection: Implications for COVID‐19 vaccine efficacy. Immun Inflamm Dis 2022; 10:e573. [PMID: 34861106 PMCID: PMC8926508 DOI: 10.1002/iid3.573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/07/2021] [Accepted: 11/24/2021] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), an emerging virus in late 2019 causing coronavirus disease 2019 (COVID‐19), has caused a catastrophic effect, resulting in an unprecedented global crisis. The immunopathology of COVID‐19 appears to be clearly associated with a dysregulated immune response leading to organ failure and death. Similarly, over two billion people worldwide are infected with helminth, with those living in low‐middle‐income countries disproportionately affected. Helminth infections have been shown to possess immunomodulatory effects in several conditions. Helminth co‐infection in COVID‐19 patients is one of the potential reasons for global attention to answer why COVID‐19 severity is still lower in helminth endemic countries. Recent studies have shown that helminth endemic countries showed fewer cases and deaths so far and helminth co‐infection might reduce the severity of COVID‐19. Moreover, lessons from other diseases with helminth co‐infection have been shown to substantially reduce vaccine efficacy that could also be implicated for COVID‐19. This immunomodulatory effect of helminth has intended and unintended consequences, both advantageous and disadvantageous which could decrease the severity of COVID‐19 and COVID‐19 vaccine efficacy respectively. Herewith, we discuss the overview of COVID‐19 immune response, immunomodulatory effects of helminth co‐infections in COVID‐19, lessons from other diseases, and perspectives on the efficacy of COVID‐19 vaccines.
Collapse
Affiliation(s)
- Yibeltal Akelew
- Immunology and Molecular Biology, Medical Laboratory Sciences, College of Health Sciences Debre Markos University Debre Markos Ethiopia
| | - Henok Andualem
- Immunology and Molecular Biology, Medical Laboratory Sciences, College of Health Sciences Debre Tabor University Debre Tabor Ethiopia
| | - Endris Ebrahim
- Immunology and Molecular Biology, Medical Laboratory Sciences, College of Health Sciences Wollo University Dessie Ethiopia
| | - Aytenew Atnaf
- Hematology and Immunohematology, Medical Laboratory Sciences, College of Health Sciences Debre Markos University Debre Markos Ethiopia
| | - Wasihun Hailemichael
- Immunology and Molecular Biology, Medical Laboratory Sciences, College of Health Sciences Debre Tabor University Debre Tabor Ethiopia
| |
Collapse
|
1224
|
Mahmud MS, Kamrujjaman M, Adan MMIY, Hossain MA, Rahman MM, Islam MS, Mohebujjaman M, Molla MM. Vaccine efficacy and SARS-CoV-2 control in California and U.S. during the session 2020-2026: A modeling study. Infect Dis Model 2022; 7:62-81. [PMID: 34869959 PMCID: PMC8627016 DOI: 10.1016/j.idm.2021.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/01/2021] [Accepted: 11/09/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Besides maintaining health precautions, vaccination has been the only prevention from SARS-CoV-2, though no clinically proved 100% effective vaccine has been developed till date. At this stage, to withhold the debris of this pandemic-experts need to know the impact of the vaccine efficacy rates, the threshold level of vaccine effectiveness and how long this pandemic may extent with vaccines that have different efficacy rates. In this article, a mathematical model study has been done on the importance of vaccination and vaccine efficiency rate during an ongoing pandemic. METHODS We simulated a five compartment mathematical model to analyze the pandemic scenario in both California, and whole U.S. We considered four vaccines, Pfizer (95%), Moderna (94%), AstraZeneca (79%), and Johnson & Johnson (72%), which are being used rigorously to control the SARS-CoV-2 pandemic, in addition with two special cases: a vaccine with 100% efficacy rate and no vaccine under use. SARS-CoV-2 related data of California, and U.S. were used in this study. FINDINGS Both the infection and death rates are very high in California. Our model suggests that the pandemic situation in California will be under control in the last quartile of the year 2023 if vaccination program is continued with the Pfizer vaccine. During this time, six waves may happen from the beginning of the immunization where the case fatality and recovery rates will be 1.697% and 98.30%, respectively. However, according to the considered model, this period might be extended to the mid of 2024 when vaccines with lower efficacy rates are used. On the other hand, the daily cases and deaths in the U.S. will be under control at the end of 2026 with multiple waves. Although the number of susceptible people will fall down to none in the beginning of 2027, there is less chance to stop the vaccination program if vaccinated with a vaccine other than a 100% effective vaccine or Pfizer, and at that case vaccination program must run till the mid of 2028. According to this study, the unconfirmed-infectious and infected cases will be under control at the end of 2027 and at the mid of 2028, respectively. INTERPRETATION The more effective a vaccine is, the less people suffer from this malign infection. Vaccines which are less than 90% effective do not have notable contribution to control the pandemic besides hard immunity. Furthermore, specific groups of people are getting prioritized initially, mass vaccination and quick responses are required to control the spread of this disease.
Collapse
Affiliation(s)
- Md Shahriar Mahmud
- Department of Computer Science and Engineering, State University of Bangladesh, Dhaka, 1205, Bangladesh
| | - Md Kamrujjaman
- Department of Mathematics, University of Dhaka, Dhaka, 1000, Bangladesh
- Department of Mathematics and Statistics, University of Calgary, Calgary, AB, Canada
| | | | - Md Alamgir Hossain
- Computational Biology Research Lab (CBRL), Department of Pharmacy, Jagannath University, Dhaka, 1100, Bangladesh
| | - Md Mizanur Rahman
- Hitotsubashi Institute for Advanced Study, Hitotsubashi University, Naka Kunitachi, Tokyo, 186-8601, Japan
| | - Md Shahidul Islam
- Department of Mathematics, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Muhammad Mohebujjaman
- Department of Mathematics and Physics, Texas A&M International University, Laredo, TX, 78 041, USA
| | - Md Mamun Molla
- Department of Mathematics & Physics, North South University, Dhaka, 1229, Bangladesh
- Center for Applied Scientific Computing (CASC), North South University, Dhaka, 1229, Bangladesh
| |
Collapse
|
1225
|
Liu J, Chandrashekar A, Sellers D, Barrett J, Jacob-Dolan C, Lifton M, McMahan K, Sciacca M, VanWyk H, Wu C, Yu J, Collier ARY, Barouch DH. Vaccines elicit highly conserved cellular immunity to SARS-CoV-2 Omicron. Nature 2022; 603:493-496. [PMID: 35102312 PMCID: PMC8930761 DOI: 10.1038/s41586-022-04465-y] [Citation(s) in RCA: 328] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/25/2022] [Indexed: 11/18/2022]
Abstract
The highly mutated SARS-CoV-2 Omicron (B.1.1.529) variant has been shown to evade a substantial fraction of neutralizing antibody responses elicited by current vaccines that encode the WA1/2020 spike protein1. Cellular immune responses, particularly CD8+ T cell responses, probably contribute to protection against severe SARS-CoV-2 infection2-6. Here we show that cellular immunity induced by current vaccines against SARS-CoV-2 is highly conserved to the SARS-CoV-2 Omicron spike protein. Individuals who received the Ad26.COV2.S or BNT162b2 vaccines demonstrated durable spike-specific CD8+ and CD4+ T cell responses, which showed extensive cross-reactivity against both the Delta and the Omicron variants, including in central and effector memory cellular subpopulations. Median Omicron spike-specific CD8+ T cell responses were 82-84% of the WA1/2020 spike-specific CD8+ T cell responses. These data provide immunological context for the observation that current vaccines still show robust protection against severe disease with the SARS-CoV-2 Omicron variant despite the substantially reduced neutralizing antibody responses7,8.
Collapse
Affiliation(s)
- Jinyan Liu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | - Julia Barrett
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Catherine Jacob-Dolan
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | | | | | - Haley VanWyk
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Cindy Wu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jingyou Yu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Dan H Barouch
- Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
1226
|
Kahn R, Holmdahl I, Reddy S, Jernigan J, Mina MJ, Slayton RB. Mathematical Modeling to Inform Vaccination Strategies and Testing Approaches for Coronavirus Disease 2019 (COVID-19) in Nursing Homes. Clin Infect Dis 2022; 74:597-603. [PMID: 34086877 PMCID: PMC8244782 DOI: 10.1093/cid/ciab517] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Nursing home residents and staff were included in the first phase of coronavirus disease 2019 vaccination in the United States. Because the primary trial endpoint was vaccine efficacy (VE) against symptomatic disease, there are limited data on the extent to which vaccines protect against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the ability to infect others (infectiousness). Assumptions about VE against infection and infectiousness have implications for changes to infection prevention guidance for vaccinated populations, including testing strategies. METHODS We use a stochastic agent-based Susceptible-Exposed-Infectious (Asymptomatic/Symptomatic)-Recovered model of a nursing home to simulate SARS-CoV-2 transmission. We model 3 scenarios, varying VE against infection, infectiousness, and symptoms, to understand the expected impact of vaccination in nursing homes, increasing staff vaccination coverage, and different screening testing strategies under each scenario. RESULTS Increasing vaccination coverage in staff decreases total symptomatic cases in the nursing home (among staff and residents combined) in each VE scenario. In scenarios with 50% and 90% VE against infection and infectiousness, increasing staff coverage reduces symptomatic cases among residents. If vaccination only protects against symptoms, and asymptomatic cases remain infectious, increased staff coverage increases symptomatic cases among residents. However, this is outweighed by the reduction in symptomatic cases among staff. Higher frequency testing-more than once weekly-is needed to reduce total symptomatic cases if the vaccine has lower efficacy against infection and infectiousness, or only protects against symptoms. CONCLUSIONS Encouraging staff vaccination is not only important for protecting staff, but might also reduce symptomatic cases in residents if a vaccine confers at least some protection against infection or infectiousness.
Collapse
Affiliation(s)
- Rebecca Kahn
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Inga Holmdahl
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Sujan Reddy
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - John Jernigan
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael J Mina
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rachel B Slayton
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
1227
|
Swamy S, Koch CA, Hannah-Shmouni F, Schiffrin EL, Klubo-Gwiezdzinska J, Gubbi S. Hypertension and COVID-19: Updates from the era of vaccines and variants. J Clin Transl Endocrinol 2022; 27:100285. [PMID: 34900602 PMCID: PMC8645507 DOI: 10.1016/j.jcte.2021.100285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Accepted: 11/27/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen responsible for coronavirus disease 2019 (COVID-19) has been a major cause of morbidity and mortality globally. Older age, and the presence of certain components of metabolic syndrome, including hypertension have been associated with increased risk for severe disease and death in COVID-19 patients. The role of antihypertensive agents in the pathogenesis of COVID-19 has been extensively studied since the onset of the pandemic. This review discusses the potential pathophysiologic interactions between hypertension and COVID-19 and provides an up-to-date information on the implications of newly emerging SARS-CoV-2 variants, and vaccines on patients with hypertension.
Collapse
Affiliation(s)
- Sowmya Swamy
- Department of Internal Medicine, George Washington University Medical Center, Washington, DC, USA
| | | | | | - Ernesto L. Schiffrin
- Department of Medicine, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Joanna Klubo-Gwiezdzinska
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sriram Gubbi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
1228
|
Peng Q, Zhou R, Wang Y, Zhao M, Liu N, Li S, Huang H, Yang D, Au KK, Wang H, Man K, Yuen KY, Chen Z. Waning immune responses against SARS-CoV-2 variants of concern among vaccinees in Hong Kong. EBioMedicine 2022; 77:103904. [PMID: 35248996 PMCID: PMC8893246 DOI: 10.1016/j.ebiom.2022.103904] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/07/2022] [Accepted: 02/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Nearly 4 billion doses of the BNT162b2-mRNA and CoronaVac-inactivated vaccines have been administrated globally, yet different vaccine-induced immunity against SARS-CoV-2 variants of concern (VOCs) remain incompletely investigated. METHODS We compare the immunogenicity and durability of these two vaccines among fully vaccinated Hong Kong people. FINDINGS Standard BNT162b2 and CoronaVac vaccinations were tolerated and induced neutralizing antibody (NAb) (100% and 85.7%) and spike-specific CD4 T cell responses (96.7% and 82.1%), respectively. The geometric mean NAb IC50 and median frequencies of reactive CD4 subsets were consistently lower among CoronaVac-vaccinees than BNT162b2-vaccinees. CoronaVac did not induce measurable levels of nucleocapsid protein-specific IFN-γ+ CD4+ T or IFN-γ+ CD8+ T cells compared with unvaccinated. Against VOCs, NAb response rates and geometric mean IC50 titers against B.1.617.2 (Delta) and B.1.1.529 (Omicron) were significantly lower for CoronaVac (50%, 23.2 and 7.1%, <20) than BNT162b2 (94.1%, 131 and 58.8%, 35.0), respectively. Three months after vaccinations, NAbs to VOCs dropped near to detection limit, along with waning memory T cell responses, mainly among CoronaVac-vaccinees. INTERPRETATION Our results indicate that vaccinees especially CoronaVac-vaccinees with significantly reduced NAbs may probably face higher risk to pandemic VOCs breakthrough infection. FUNDING This study was supported by the Hong Kong Research Grants Council Collaborative Research Fund (C7156-20GF and C1134-20GF); the Wellcome Trust (P86433); the National Program on Key Research Project of China (Grant 2020YFC0860600, 2020YFA0707500 and 2020YFA0707504); Shenzhen Science and Technology Program (JSGG20200225151410198 and JCYJ20210324131610027); HKU Development Fund and LKS Faculty of Medicine Matching Fund to AIDS Institute; Hong Kong Innovation and Technology Fund, Innovation and Technology Commission and generous donation from the Friends of Hope Education Fund. Z.C.'s team was also partly supported by the Theme-Based Research Scheme (T11-706/18-N).
Collapse
Affiliation(s)
- Qiaoli Peng
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China; National Clinical Research Center for Infectious Diseases, HKU AIDS Institute Shenzhen Research laboratory, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, People's Republic of China
| | - Runhong Zhou
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China
| | - Yuewen Wang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Meiqing Zhao
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China
| | - Na Liu
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China
| | - Shuang Li
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China
| | - Haode Huang
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China
| | - Dawei Yang
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China
| | - Ka-Kit Au
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China
| | - Hui Wang
- National Clinical Research Center for Infectious Diseases, HKU AIDS Institute Shenzhen Research laboratory, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, People's Republic of China
| | - Kwan Man
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Kwok-Yung Yuen
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China; State Key Laboratory for Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Zhiwei Chen
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Hong Kong Special Administrative Region, People's Republic of China; National Clinical Research Center for Infectious Diseases, HKU AIDS Institute Shenzhen Research laboratory, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, People's Republic of China; State Key Laboratory for Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
1229
|
Mabrouk MT, Huang W, Martinez‐Sobrido L, Lovell JF. Advanced Materials for SARS-CoV-2 Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107781. [PMID: 34894000 PMCID: PMC8957524 DOI: 10.1002/adma.202107781] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/28/2021] [Indexed: 05/09/2023]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), has killed untold millions worldwide and has hurtled vaccines into the spotlight as a go-to approach to mitigate it. Advances in virology, genomics, structural biology, and vaccine technologies have enabled a rapid and unprecedented rollout of COVID-19 vaccines, although much of the developing world remains unvaccinated. Several new vaccine platforms have been developed or deployed against SARS-CoV-2, with most targeting the large viral Spike immunogen. Those that safely induce strong and durable antibody responses at low dosages are advantageous, as well are those that can be rapidly produced at a large scale. Virtually all COVID-19 vaccines and adjuvants possess nanoscale or microscale dimensions and represent diverse and unique biomaterials. Viral vector vaccine platforms, lipid nanoparticle mRNA vaccines and multimeric display technologies for subunit vaccines have received much attention. Nanoscale vaccine adjuvants have also been used in combination with other vaccines. To deal with the ongoing pandemic, and to be ready for potential future ones, advanced vaccine technologies will continue to be developed in the near future. Herein, the recent use of advanced materials used for developing COVID-19 vaccines is summarized.
Collapse
Affiliation(s)
- Moustafa T. Mabrouk
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Wei‐Chiao Huang
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Luis Martinez‐Sobrido
- Division of Disease Intervention and PreventionTexas Biomedical Research InstituteSan AntonioTX78227USA
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| |
Collapse
|
1230
|
Botton J, Semenzato L, Jabagi MJ, Baricault B, Weill A, Dray-Spira R, Zureik M. Effectiveness of Ad26.COV2.S Vaccine vs BNT162b2 Vaccine for COVID-19 Hospitalizations. JAMA Netw Open 2022; 5:e220868. [PMID: 35234883 PMCID: PMC8892225 DOI: 10.1001/jamanetworkopen.2022.0868] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
This comparative effectiveness research study examines the effectiveness of full vaccination with Ad26.COV2.S vs BNT162b2 against COVID-19–related hospitalization.
Collapse
Affiliation(s)
- Jérémie Botton
- EPI-PHARE Scientific Interest Group in Epidemiology of Health Products, French National Agency for the Safety of Medicines and Health Products, French National Health Insurance, Saint-Denis, France
| | - Laura Semenzato
- EPI-PHARE Scientific Interest Group in Epidemiology of Health Products, French National Agency for the Safety of Medicines and Health Products, French National Health Insurance, Saint-Denis, France
| | - Marie-Joëlle Jabagi
- EPI-PHARE Scientific Interest Group in Epidemiology of Health Products, French National Agency for the Safety of Medicines and Health Products, French National Health Insurance, Saint-Denis, France
| | - Bérangère Baricault
- EPI-PHARE Scientific Interest Group in Epidemiology of Health Products, French National Agency for the Safety of Medicines and Health Products, French National Health Insurance, Saint-Denis, France
| | - Alain Weill
- EPI-PHARE Scientific Interest Group in Epidemiology of Health Products, French National Agency for the Safety of Medicines and Health Products, French National Health Insurance, Saint-Denis, France
| | - Rosemary Dray-Spira
- EPI-PHARE Scientific Interest Group in Epidemiology of Health Products, French National Agency for the Safety of Medicines and Health Products, French National Health Insurance, Saint-Denis, France
| | - Mahmoud Zureik
- EPI-PHARE Scientific Interest Group in Epidemiology of Health Products, French National Agency for the Safety of Medicines and Health Products, French National Health Insurance, Saint-Denis, France
| |
Collapse
|
1231
|
Tanislav C, Rosenbauer J, Zingel R, Kostev K. No increased incidence of venous thrombosis or pulmonary embolism after SARS-CoV-2 vaccination in Germany. Public Health 2022; 207:14-18. [PMID: 35461122 PMCID: PMC8923878 DOI: 10.1016/j.puhe.2022.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/26/2022]
Abstract
Objectives Vaccination is one of the most effective measures to combat the COVID-19 pandemic. The main reason for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination hesitancy is the potential side-effects. This study aimed to investigate the incidence of venous thrombosis and pulmonary embolism in patients who received SARS-CoV-2 vaccination. Study design This was a retrospective cohort study. Methods Individuals aged ≥18 years who received an initial vaccination for COVID-19 in one of 1134 general practices in Germany between April and June 2021 were included in the study. Vaccinated patients were matched to unvaccinated individuals by age, sex, index month (April to June 2020 [unvaccinated cohort] or April to June 2021 [vaccinated cohort]) and diagnoses that may be associated with an increased incidence of thrombosis documented within 12 months before the index date. The incidences of thrombosis and non-fatal pulmonary embolism as a function of COVID-19 vaccination were analysed. Results The present study included 326,833 individuals who were vaccinated against COVID-19 and 326,833 matched unvaccinated individuals. During the follow-up period, 406 vaccinated patients and 342 individuals in the control group received a diagnosis of thrombosis or non-fatal pulmonary embolism. This resulted in an incidence rate of 11.9 vs 11.3 cases per 1000 patient-years for vaccinated vs unvaccinated individuals, respectively, and a non-significant overall incidence rate ratio (IRR: 1.06; 95% confidence interval [CI]: 0.93–1.22). The highest IRR was observed in the 41–60 years age group (IRR: 1.30; 95% CI: 0.98–1.73), and the lowest IRR was seen in the 18–40 years age group (IRR: 0.6; 95% CI: 0.0–1.05); however, none of the individual age group incidence rates was significant. Conclusions The results indicate that the occurrence of thrombosis or pulmonary embolism after COVID-19 vaccination is a coincidental finding rather than a consequence of vaccination.
Collapse
|
1232
|
Kundi M, Montgomery S, Mao S, Asghar S. Not All That Is Droopy Post Ad26.COV2.S (JNJ) Vaccine Is Bell's Palsy: A Rare Case of Isolated Dorsal Pontine Stroke Causing Ipsilateral Complete Hemi-Facial Palsy. Cureus 2022; 14:e23195. [PMID: 35444866 PMCID: PMC9010170 DOI: 10.7759/cureus.23195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2022] [Indexed: 11/19/2022] Open
Abstract
The Ad26.COV2.S vaccine, developed by Janssen (Beerse, Belgium), the pharmaceutical wing of Johnson & Johnson (JNJ), is one of the three vaccines approved for use against coronavirus disease 2019 (COVID-19) infection in the United States. We present a case of a 66-year-old female who presented to the emergency department with a one-day history of nausea, vomiting, room-spinning vertigo, and complete right facial weakness immediately after getting vaccinated with Ad26.COV2.S. Initial workup focused on uncovering a possible association between the vaccine and Bell's palsy. However, her prior history of stroke, presence of predisposing risk factors, and additional symptoms of nausea, vomiting, and vertigo prompted further neurological testing, which revealed an isolated right pontine lacunar infarct involving the right facial colliculus, mimicking Bell's palsy. Isolated dorsal pontine lesion presenting as hemifacial palsy is very rare and can be easily missed by clinicians. Relevant history and thorough neurological examination can help guide appropriate diagnostic testing and prevent potential biases. It is crucial for clinicians to know the distinguishing features between true Bell's palsy and acute brainstem infarction masquerading as Bell's.
Collapse
Affiliation(s)
- Maryam Kundi
- Internal Medicine, Coliseum Medical Centers, Macon, USA
| | | | - Shirley Mao
- Emergency Medicine, Coliseum Medical Centers, Macon, USA
| | - Saleha Asghar
- Internal Medicine, Coliseum Medical Centers, Macon, USA
| |
Collapse
|
1233
|
Ahn JY, Lee J, Suh YS, Song YG, Choi YJ, Lee KH, Seo SH, Song M, Oh JW, Kim M, Seo HY, Kwak JE, Youn JW, Woo JW, Shin EC, Sung YC, Park SH, Choi JY. Safety and immunogenicity of two recombinant DNA COVID-19 vaccines containing the coding regions of the spike or spike and nucleocapsid proteins: an interim analysis of two open-label, non-randomised, phase 1 trials in healthy adults. THE LANCET. MICROBE 2022; 3:e173-e183. [PMID: 35156068 PMCID: PMC8824525 DOI: 10.1016/s2666-5247(21)00358-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND We assessed the safety and immunogenicity of two recombinant DNA vaccines for COVID-19: GX-19 containing plasmid DNA encoding the SARS-CoV-2 spike protein, and GX-19N containing plasmid DNA encoding the SARS-CoV-2 receptor-binding domain (RBD) foldon, nucleocapsid protein, and plasmid DNA encoding the spike protein. METHODS Two open-label non-randomised phase 1 trials, one of GX-19 and the other of GX-19N were done at two hospitals in South Korea. We enrolled healthy adults aged 19-49 years for the GX-19 trial and healthy adults aged 19-54 years for the GX-19N trial. Participants who tested positive by serological testing for SARS-CoV-2 were excluded. At 4-week intervals, the GX-19 trial participants received two vaccine doses (either 1·5 mg or 3·0 mg), and the GX-19N trial participants received two 3·0 mg doses. The vaccines were delivered intramuscularly using an electroporator. The participants were followed up for 52 weeks after first vaccination. Data collected up to day 57 after first vaccination were analysed in this interim analysis. The primary outcome was safety within 28 days after each vaccination measured in the intention-to-treat population. The secondary outcome was vaccine immunogenicity using blood samples collected on day 43 or 57 after first vaccination measured in the intention-to-treat population. The GX-19 (NCT044445389) and GX-19N (NCT04715997) trials are registered with ClinicalTrials.gov. FINDINGS Between June 17 and July 30, 2020, we screened 97 individuals, of whom 40 (41%) participants were enrolled in the GX-19 trial (20 [50%] in the 1·5 mg group and 20 [50%] in the 3·0 mg group). Between Dec 28 and 31, 2020, we screened 23 participants, of whom 21 (91%) participants were enrolled on the GX-19N trial. 32 (52%) of 61 participants reported 80 treatment-emergent adverse events after vaccination. All solicited adverse events were mild except one (2%) case of moderate fatigue in the 1·5 mg GX-19 group; no serious vaccine-related adverse events were detected. Binding antibody responses increased after second dose of vaccination in all groups (p=0·0002 in the 1·5 mg GX-19 group; p<0·0001 in the 3·0 mg GX-19; and p=0·0004 for the spike protein and p=0·0001 for the RBD in the 3·0 mg GX-19N group). INTERPRETATION GX-19 and GX-19N are safe and well tolerated. GX-19N induces humoral and broad SARS-CoV-2-specific T-cell responses. GX-19N shows lower neutralising antibody responses and needs improvement to enhance immunogenicity. FUNDING The Korea Drug Development Fund, funded by the Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare.
Collapse
Affiliation(s)
- Jin Young Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei University, Seoul, South Korea
| | - Jeongsoo Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - You Suk Suh
- Genexine, Korea Bio Park, Seongnam, Gyeonggi-do, South Korea
| | - Young Goo Song
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei University, Seoul, South Korea
| | - Yoon-Jeong Choi
- Genexine, Korea Bio Park, Seongnam, Gyeonggi-do, South Korea
| | - Kyoung Hwa Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei University, Seoul, South Korea
| | - Sang Hwan Seo
- Science Unit, International Vaccine Institute, Seoul, South Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul, South Korea
| | - Jong-Won Oh
- Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Minwoo Kim
- Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Han Yeong Seo
- Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Jeong-Eun Kwak
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jin Won Youn
- Genexine, Korea Bio Park, Seongnam, Gyeonggi-do, South Korea
| | - Jung Won Woo
- Genexine, Korea Bio Park, Seongnam, Gyeonggi-do, South Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
- The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Young Chul Sung
- Genexine, Korea Bio Park, Seongnam, Gyeonggi-do, South Korea
- Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
- The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jun Yong Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei University, Seoul, South Korea
| |
Collapse
|
1234
|
Mattila JP, Amaro A, Longo M, Antaki J, Koirala S, Gandini A. RapidQ: A reader-free microfluidic platform for the quantitation of antibodies against the SARS-CoV-2 spike protein. BIOMICROFLUIDICS 2022; 16:024105. [PMID: 35356130 PMCID: PMC8933056 DOI: 10.1063/5.0079054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/04/2022] [Indexed: 06/14/2023]
Abstract
We describe RapidQ, a fast, disposable, easy-to-use microfluidic assay for the quantitation of the anti-SARS-CoV-2 spike (S) protein IgG in plasma samples. The assay utilizes antigen-coated paramagnetic microbeads, which are induced to aggregate inside the RapidQ microfluidic device in the presence of the target antibody. Aggregation occurs via interaction between the biotinylated detection antibody and polymeric streptavidin. The mobility of the beads inside the two microchannels of the device depends on their aggregation state, with larger clusters moving at higher velocities under a given liquid flow rate. One of the microchannels incorporates a permanent magnet that captures arriving beads and forms a localized constriction that retards liquid flow. Since the constriction grows faster when the beads are more aggregated, the length of the liquid column accumulated downstream from the constriction relative to that of the unconstricted control channel is proportional to the sample antibody concentration. The assay demonstrates a detection limit of 4 μg/ml of monoclonal anti-S protein antibody diluted in plasma with CV ≤ 13%, as well as negative and positive percent agreements of 100% (95% CI: 92.75%-100%) and 100% (95% CI: 80.5%-100%), respectively, when compared to a nucleic acid amplification test used to identify COVID-19 positive individuals, whose samples were collected ≥17 d from a positive PCR test. Finally, the RapidQ assay was used to monitor the kinetics of antibody responses to COVID-19 vaccination in a small study cohort.
Collapse
Affiliation(s)
| | - Arlene Amaro
- Accel Diagnostics, LLC, Houston, Texas 77057, USA
| | - Monica Longo
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - James Antaki
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | | | | |
Collapse
|
1235
|
Schmidt AL, Labaki C, Hsu CY, Bakouny Z, Balanchivadze N, Berg SA, Blau S, Daher A, El Zarif T, Friese CR, Griffiths EA, Hawley JE, Hayes-Lattin B, Karivedu V, Latif T, Mavromatis BH, McKay RR, Nagaraj G, Nguyen RH, Panagiotou OA, Portuguese AJ, Puc M, Santos Dutra M, Schroeder BA, Thakkar A, Wulff-Burchfield EM, Mishra S, Farmakiotis D, Shyr Y, Warner JL, Choueiri TK. COVID-19 vaccination and breakthrough infections in patients with cancer. Ann Oncol 2022; 33:340-346. [PMID: 34958894 PMCID: PMC8704021 DOI: 10.1016/j.annonc.2021.12.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Vaccination is an important preventive health measure to protect against symptomatic and severe COVID-19. Impaired immunity secondary to an underlying malignancy or recent receipt of antineoplastic systemic therapies can result in less robust antibody titers following vaccination and possible risk of breakthrough infection. As clinical trials evaluating COVID-19 vaccines largely excluded patients with a history of cancer and those on active immunosuppression (including chemotherapy), limited evidence is available to inform the clinical efficacy of COVID-19 vaccination across the spectrum of patients with cancer. PATIENTS AND METHODS We describe the clinical features of patients with cancer who developed symptomatic COVID-19 following vaccination and compare weighted outcomes with those of contemporary unvaccinated patients, after adjustment for confounders, using data from the multi-institutional COVID-19 and Cancer Consortium (CCC19). RESULTS Patients with cancer who develop COVID-19 following vaccination have substantial comorbidities and can present with severe and even lethal infection. Patients harboring hematologic malignancies are over-represented among vaccinated patients with cancer who develop symptomatic COVID-19. CONCLUSIONS Vaccination against COVID-19 remains an essential strategy in protecting vulnerable populations, including patients with cancer. Patients with cancer who develop breakthrough infection despite full vaccination, however, remain at risk of severe outcomes. A multilayered public health mitigation approach that includes vaccination of close contacts, boosters, social distancing, and mask-wearing should be continued for the foreseeable future.
Collapse
Affiliation(s)
- A L Schmidt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - C Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - C-Y Hsu
- Department of Biostatistics, Vanderbilt University, Nashville, USA
| | - Z Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - N Balanchivadze
- Hematology and Oncology Fellowship Program, Henry Ford Cancer Institute, Detroit, USA
| | - S A Berg
- Department of Internal Medicine and Cancer Biology, Division of Hematology and Oncology, Cardinal Bernardin Cancer Centre, Loyola University Chicago, Maywood, USA
| | - S Blau
- Division of Oncology, Northwest Medical Specialties, Tacoma, USA; Division of Hematology, University of Washington, Seattle, USA
| | - A Daher
- Hartford HealthCare Medical Group, Hartford, USA
| | - T El Zarif
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - C R Friese
- University of Michigan School of Nursing, School of Public Health, and Rogel Cancer Centre, Ann Arbor, USA
| | - E A Griffiths
- Leukemia Section, Roswell Park Comprehensive Cancer Centre, Buffalo, USA
| | - J E Hawley
- Herbert Irving Comprehensive Cancer Centre, Columbia University Irving Medical Centre, New York, USA; University of Washington/Fred Hutchinson Cancer Research Center, Seattle, USA
| | - B Hayes-Lattin
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, USA
| | - V Karivedu
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Wexner Medical Centre, Columbus, USA
| | - T Latif
- Division of Hematology/Medical Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, USA
| | - B H Mavromatis
- Department of Cancer, Oncology, Hematology, UPMC Western Maryland, Cumberland, USA
| | - R R McKay
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, San Diego, USA
| | - G Nagaraj
- Division of Medical Oncology & Hematology, Department of Medicine, Loma Linda University Cancer Centre, Loma Linda, USA
| | - R H Nguyen
- Department of Medicine, Division of Hematology and Oncology, University of Illinois at Chicago, Chicago, USA
| | - O A Panagiotou
- Department of Health Services, Policy & Practice, Brown University School of Public Health, Providence, USA
| | - A J Portuguese
- Division of Hematology, University of Washington, Seattle, USA
| | - M Puc
- Department of Surgery, Section of Thoracic Surgery, Virtua Health, Marlton, USA
| | - M Santos Dutra
- Segal Cancer Centre of the Jewish General Hospital, Montréal, Canada
| | | | - A Thakkar
- Division of Oncology, Montefiore Medical Centre, Bronx, USA
| | - E M Wulff-Burchfield
- Department of Medicine, Divisions of Medical Oncology and Palliative Medicine, The University of Kansas Health System, Westwood, USA
| | - S Mishra
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, USA
| | - D Farmakiotis
- Department of Medicine, Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, USA
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University, Nashville, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, USA
| | - J L Warner
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, USA; Department of Medicine, Division of Hematology/Oncology, Vanderbilt University, Nashville, USA; Department of Biomedical Informatics, Vanderbilt University, Nashville, USA.
| | - T K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA.
| |
Collapse
|
1236
|
Jiang L, Driedonks TA, Jong WS, Dhakal S, Bart van den Berg van Saparoea H, Sitaras I, Zhou R, Caputo C, Littlefield K, Lowman M, Chen M, Lima G, Gololobova O, Smith B, Mahairaki V, Riley Richardson M, Mulka KR, Lane AP, Klein SL, Pekosz A, Brayton C, Mankowski JL, Luirink J, Villano JS, Witwer KW. A bacterial extracellular vesicle-based intranasal vaccine against SARS-CoV-2 protects against disease and elicits neutralizing antibodies to wild-type and Delta variants. J Extracell Vesicles 2022; 11:e12192. [PMID: 35289114 PMCID: PMC8920961 DOI: 10.1002/jev2.12192] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Several vaccines have been introduced to combat the coronavirus infectious disease-2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Current SARS-CoV-2 vaccines include mRNA-containing lipid nanoparticles or adenoviral vectors that encode the SARS-CoV-2 Spike (S) protein of SARS-CoV-2, inactivated virus, or protein subunits. Despite growing success in worldwide vaccination efforts, additional capabilities may be needed in the future to address issues such as stability and storage requirements, need for vaccine boosters, desirability of different routes of administration, and emergence of SARS-CoV-2 variants such as the Delta variant. Here, we present a novel, well-characterized SARS-CoV-2 vaccine candidate based on extracellular vesicles (EVs) of Salmonella typhimurium that are decorated with the mammalian cell culture-derived Spike receptor-binding domain (RBD). RBD-conjugated outer membrane vesicles (RBD-OMVs) were used to immunize the golden Syrian hamster (Mesocricetus auratus) model of COVID-19. Intranasal immunization resulted in high titres of blood anti-RBD IgG as well as detectable mucosal responses. Neutralizing antibody activity against wild-type and Delta variants was evident in all vaccinated subjects. Upon challenge with live virus, hamsters immunized with RBD-OMV, but not animals immunized with unconjugated OMVs or a vehicle control, avoided body mass loss, had lower virus titres in bronchoalveolar lavage fluid, and experienced less severe lung pathology. Our results emphasize the value and versatility of OMV-based vaccine approaches.
Collapse
Affiliation(s)
- Linglei Jiang
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Tom A.P. Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | | | - Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | | | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Ruifeng Zhou
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Christopher Caputo
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Maggie Lowman
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mengfei Chen
- Department of Otolaryngology‐Head and Neck SurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Gabriela Lima
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Barbara Smith
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Vasiliki Mahairaki
- Department of Genetic Medicine and The Richman Family Precision Medicine Centre of Excellence in Alzheimer's Disease Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - M. Riley Richardson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kathleen R. Mulka
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Andrew P. Lane
- Department of Otolaryngology‐Head and Neck SurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Cory Brayton
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Joseph L. Mankowski
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Neurology and NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Joen Luirink
- Abera Bioscience ABUppsalaSweden
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS)Vrije UniversiteitAmsterdamThe Netherlands
| | - Jason S. Villano
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Genetic Medicine and The Richman Family Precision Medicine Centre of Excellence in Alzheimer's Disease Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Neurology and NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
1237
|
Kremsner PG, Ahuad Guerrero RA, Arana-Arri E, Aroca Martinez GJ, Bonten M, Chandler R, Corral G, De Block EJL, Ecker L, Gabor JJ, Garcia Lopez CA, Gonzales L, Granados González MA, Gorini N, Grobusch MP, Hrabar AD, Junker H, Kimura A, Lanata CF, Lehmann C, Leroux-Roels I, Mann P, Martinez-Reséndez MF, Ochoa TJ, Poy CA, Reyes Fentanes MJ, Rivera Mejia LM, Ruiz Herrera VV, Sáez-Llorens X, Schönborn-Kellenberger O, Schunk M, Sierra Garcia A, Vergara I, Verstraeten T, Vico M, Oostvogels L, Lovesio L, Diez F, Grazziani F, Ganaha MC, Zalatnik VJ, Dittrich RJ, Espínola L, Lambert S, Longhi A, Vecchio C, Mastruzzo M, Fernandez A, Borchowiek S, Potito R, Ahuad Guerrero RA, Guardiani FM, Castella S, Foccoli M, Pedernera A, Braida A, Durigan V, Martella C, Bobat A, Boggia BE, Nemi SA, Tartaglione JG, Piedimonte FC, De Bie J, Reynales Londoño H, Rodríguez Ordoñez PA, García Cruz JM, Bautista Toloza L, Ladino González MC, Zambrano Ochoa AP, Prieto Pradera I, Torres Hernandez D, Mazo Elorza DP, Collazos Lennis MF, Vanegas Dominguez B, Solano Mosquera LM, Fendel R, Fleischmann WA, Koehne E, Kreidenweiss A, Köhler C, Esen M, Horn C, Eberts S, Kroidl A, Huber K, Thiel V, Mazara Rosario S, Reyes G, Rivera L, Donastorg Y, Lantigua F, Torres Almanzar D, Candelario R, Peña Mendez L, Rosario Gomez N, Portolés-Pérez A, Ascaso del Río A, Laredo Velasco L, Bustinduy Odriozola MJ, et alKremsner PG, Ahuad Guerrero RA, Arana-Arri E, Aroca Martinez GJ, Bonten M, Chandler R, Corral G, De Block EJL, Ecker L, Gabor JJ, Garcia Lopez CA, Gonzales L, Granados González MA, Gorini N, Grobusch MP, Hrabar AD, Junker H, Kimura A, Lanata CF, Lehmann C, Leroux-Roels I, Mann P, Martinez-Reséndez MF, Ochoa TJ, Poy CA, Reyes Fentanes MJ, Rivera Mejia LM, Ruiz Herrera VV, Sáez-Llorens X, Schönborn-Kellenberger O, Schunk M, Sierra Garcia A, Vergara I, Verstraeten T, Vico M, Oostvogels L, Lovesio L, Diez F, Grazziani F, Ganaha MC, Zalatnik VJ, Dittrich RJ, Espínola L, Lambert S, Longhi A, Vecchio C, Mastruzzo M, Fernandez A, Borchowiek S, Potito R, Ahuad Guerrero RA, Guardiani FM, Castella S, Foccoli M, Pedernera A, Braida A, Durigan V, Martella C, Bobat A, Boggia BE, Nemi SA, Tartaglione JG, Piedimonte FC, De Bie J, Reynales Londoño H, Rodríguez Ordoñez PA, García Cruz JM, Bautista Toloza L, Ladino González MC, Zambrano Ochoa AP, Prieto Pradera I, Torres Hernandez D, Mazo Elorza DP, Collazos Lennis MF, Vanegas Dominguez B, Solano Mosquera LM, Fendel R, Fleischmann WA, Koehne E, Kreidenweiss A, Köhler C, Esen M, Horn C, Eberts S, Kroidl A, Huber K, Thiel V, Mazara Rosario S, Reyes G, Rivera L, Donastorg Y, Lantigua F, Torres Almanzar D, Candelario R, Peña Mendez L, Rosario Gomez N, Portolés-Pérez A, Ascaso del Río A, Laredo Velasco L, Bustinduy Odriozola MJ, Larrea Arranz I, Martínez Alcorta LI, Durán Laviña MI, Imaz-Ayo N, Meijide S, García-de-Vicuña A, Santorcuato A, Gallego M, Aguirre-García GM, Olmos Vega J, González Limón P, Vázquez Villar A, Chávez Barón J, Arredondo Saldaña F, Luján Palacios JDD, Camacho Choza LJ, Vázquez Saldaña EG, Ortega Dominguez SJ, Vega Orozco KS, Torres Quiroz IA, Martinez Avendaño A, Herrera Sanchez J, Guzman E, Castro Castrezana L, Ruiz Palacios y Santos GM, de Winter RFJ, de Jonge HK, Schnyder JL, Boersma W, Hessels L, Djamin R, van der Sar S, DeAntonio R, Peña M, Rebollon G, Rojas M, Escobar J, Hammerschlag Icaza B, Wong T DY, Barrera Perigault P, Ruiz S, Chan M, Arias Hoo DJ, Gil AI, Celis CR, Balmaceda MP, Flores O, Ochoa M, Peña B, de la Flor C, Webb CM, Cornejo E, Sanes F, Mayorga V, Valdiviezo G, Ramírez Lamas SP, Grandez Castillo GA, Lama JR, Matta Aguirre ME, Arancibia Luna LA, Carbajal Paulet Ó, Zambrano Ortiz J, Camara A, Guzman Quintanilla F, Diaz-Parra C, Morales-Oliva J, Cornejo RE, Ricalde SA, Vidal J, Rios Nogales L, Cheatham-Seitz D, Gregoraci G, Brecx A, Walz L, Vahrenhorst D, Seibel T, Quintini G. Efficacy and safety of the CVnCoV SARS-CoV-2 mRNA vaccine candidate in ten countries in Europe and Latin America (HERALD): a randomised, observer-blinded, placebo-controlled, phase 2b/3 trial. THE LANCET. INFECTIOUS DISEASES 2022; 22:329-340. [PMID: 34826381 PMCID: PMC8610426 DOI: 10.1016/s1473-3099(21)00677-0] [Show More Authors] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/02/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Additional safe and efficacious vaccines are needed to control the COVID-19 pandemic. We aimed to analyse the efficacy and safety of the CVnCoV SARS-CoV-2 mRNA vaccine candidate. METHODS HERALD is a randomised, observer-blinded, placebo-controlled, phase 2b/3 clinical trial conducted in 47 centres in ten countries in Europe and Latin America. By use of an interactive web response system and stratification by country and age group (18-60 years and ≥61 years), adults with no history of virologically confirmed COVID-19 were randomly assigned (1:1) to receive intramuscularly either two 0·6 mL doses of CVnCoV containing 12 μg of mRNA or two 0·6 mL doses of 0·9% NaCl (placebo) on days 1 and 29. The primary efficacy endpoint was the occurrence of a first episode of virologically confirmed symptomatic COVID-19 of any severity and caused by any strain from 15 days after the second dose. For the primary endpoint, the trial was considered successful if the lower limit of the CI was greater than 30%. Key secondary endpoints were the occurrence of a first episode of virologically confirmed moderate-to-severe COVID-19, severe COVID-19, and COVID-19 of any severity by age group. Primary safety outcomes were solicited local and systemic adverse events within 7 days after each dose and unsolicited adverse events within 28 days after each dose in phase 2b participants, and serious adverse events and adverse events of special interest up to 1 year after the second dose in phase 2b and phase 3 participants. Here, we report data up to June 18, 2021. The study is registered at ClinicalTrials.gov, NCT04652102, and EudraCT, 2020-003998-22, and is ongoing. FINDINGS Between Dec 11, 2020, and April 12, 2021, 39 680 participants were enrolled and randomly assigned to receive either CVnCoV (n=19 846) or placebo (n=19 834), of whom 19 783 received at least one dose of CVnCoV and 19 746 received at least one dose of placebo. After a mean observation period of 48·2 days (SE 0·2), 83 cases of COVID-19 occurred in the CVnCoV group (n=12 851) in 1735·29 person-years and 145 cases occurred in the placebo group (n=12 211) in 1569·87 person-years, resulting in an overall vaccine efficacy against symptomatic COVID-19 of 48·2% (95·826% CI 31·0-61·4; p=0·016). Vaccine efficacy against moderate-to-severe COVID-19 was 70·7% (95% CI 42·5-86·1; CVnCoV 12 cases in 1735·29 person-years, placebo 37 cases in 1569·87 person-years). In participants aged 18-60 years, vaccine efficacy against symptomatic disease was 52·5% (95% CI 36·2-64·8; CVnCoV 71 cases in 1591·47 person-years, placebo, 136 cases in 1449·23 person-years). Too few cases occurred in participants aged 61 years or older (CVnCoV 12, placebo nine) to allow meaningful assessment of vaccine efficacy. Solicited adverse events, which were mostly systemic, were more common in CVnCoV recipients (1933 [96·5%] of 2003) than in placebo recipients (1344 [67·9%] of 1978), with 542 (27·1%) CVnCoV recipients and 61 (3·1%) placebo recipients reporting grade 3 solicited adverse events. The most frequently reported local reaction after any dose in the CVnCoV group was injection-site pain (1678 [83·6%] of 2007), with 22 grade 3 reactions, and the most frequently reported systematic reactions were fatigue (1603 [80·0%] of 2003) and headache (1541 [76·9%] of 2003). 82 (0·4%) of 19 783 CVnCoV recipients reported 100 serious adverse events and 66 (0·3%) of 19 746 placebo recipients reported 76 serious adverse events. Eight serious adverse events in five CVnCoV recipients and two serious adverse events in two placebo recipients were considered vaccination-related. None of the fatal serious adverse events reported (eight in the CVnCoV group and six in the placebo group) were considered to be related to study vaccination. Adverse events of special interest were reported for 38 (0·2%) participants in the CVnCoV group and 31 (0·2%) participants in the placebo group. These events were considered to be related to the trial vaccine for 14 (<0·1%) participants in the CVnCoV group and for five (<0·1%) participants in the placebo group. INTERPRETATION CVnCoV was efficacious in the prevention of COVID-19 of any severity and had an acceptable safety profile. Taking into account the changing environment, including the emergence of SARS-CoV-2 variants, and timelines for further development, the decision has been made to cease activities on the CVnCoV candidate and to focus efforts on the development of next-generation vaccine candidates. FUNDING German Federal Ministry of Education and Research and CureVac.
Collapse
|
1238
|
JOSEPH NAIMAT, MILLER EMILYS. Obstetric Outpatient Management During the COVID-19 Pandemic: Prevention, Treatment of Mild Disease, and Vaccination. Clin Obstet Gynecol 2022; 65:161-178. [PMID: 35045038 PMCID: PMC8767923 DOI: 10.1097/grf.0000000000000670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The majority of patients with coronavirus disease 2019 will have mild or asymptomatic disease, however, obstetric patients are uniquely at risk for disease progression and adverse outcomes. Preventive strategies including masking, physical distancing, vaccination, and chemoprophylaxis have been well studied, are critical to disease mitigation, and can be used in the pregnant population. High-quality data are needed to assess safety and effectiveness of therapeutics and vaccination in pregnancy, as well as long-term data on maternal and newborn outcomes.
Collapse
Affiliation(s)
- NAIMA T. JOSEPH
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - EMILY S. MILLER
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
1239
|
Hyer RN. Insights for Oncology Trials Garnered From the Rapid Development of an mRNA COVID-19 Vaccine. Cancer J 2022; 28:146-150. [PMID: 35333501 PMCID: PMC8974181 DOI: 10.1097/ppo.0000000000000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT The sudden emergence of the coronavirus disease 2019 (COVID-19) pandemic in early 2020 stimulated unprecedented scientific initiatives to rapidly develop effective treatments and vaccines. One example was the development of vaccines based on messenger RNA platforms, which received emergency use authorization in the United States less than 1 year after the primary sequence of the severe acute respiratory syndrome coronavirus 2 virus was published. Novel practices arose from the collaborative efforts and inclusive clinical studies that facilitated the vaccines' rapid development and clinical testing. I describe insights gained from the experience of mRNA-1273 vaccine development that may be applied to or adapted for oncology research. These insights include clinical study design, diversity and inclusion initiatives, speed, and real-world evidence generation, as well as close partnership among regulatory agencies, government, and pharmaceutical companies.
Collapse
|
1240
|
Del Cura-Bilbao A, López-Mendoza H, Chaure-Pardos A, Vergara-Ugarriza A, Guimbao-Bescós J. Effectiveness of 3 COVID-19 Vaccines in Preventing SARS-CoV-2 Infections, January–May 2021, Aragon, Spain. Emerg Infect Dis 2022; 28:591-598. [PMID: 35195514 PMCID: PMC8888243 DOI: 10.3201/eid2803.212027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Reducing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission is a worldwide challenge; widespread vaccination could be one strategy for control. We conducted a prospective, population-based cohort study of 964,258 residents of Aragon, Spain, during December 2020–May 2021. We used the Cox proportional-hazards model with vaccination status as the exposure condition to estimate the effectiveness of 3 coronavirus disease vaccines in preventing SARS-CoV-2 infection. Pfizer-BioNTech had 20.8% (95% CI 11.6%–29.0%) vaccine effectiveness (VE) against infection after 1 dose and 70.0% (95% CI 65.3%–74.1%) after 2 doses, Moderna had 52.8% (95% CI 30.7%–67.8%) VE after 1 dose and 70.3% (95% CI 52.2%–81.5%) after 2 doses, and Oxford-AstraZeneca had 40.3% (95% CI 31.8%–47.7%) VE after 1 dose. All estimates were lower than those from previous studies. Results imply that, although high vaccination coverage remains critical to protect people from disease, it will be difficult to effectively minimize transmission opportunities.
Collapse
|
1241
|
Haroz EE, Kemp CG, O'Keefe VM, Pocock K, Wilson DR, Christensen L, Walls M, Barlow A, Hammitt L. Nurturing Innovation at the Roots: The Success of COVID-19 Vaccination in American Indian and Alaska Native Communities. Am J Public Health 2022; 112:383-387. [PMID: 35196058 PMCID: PMC8887173 DOI: 10.2105/ajph.2021.306635] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2021] [Indexed: 11/04/2022]
Affiliation(s)
- Emily E Haroz
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - Christopher G Kemp
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - Victoria M O'Keefe
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - Katherine Pocock
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - David R Wilson
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - Loretta Christensen
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - Melissa Walls
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - Allison Barlow
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| | - Laura Hammitt
- Emily E. Haroz, Christopher G. Kemp, Victoria M. O'Keefe, Melissa Walls, Allison Barlow, and Laura Hammitt are with the Center for American Indian Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD. Katherine Pocock is with the Whiteriver Indian Hospital, Whiteriver, AZ. David R. Wilson is with the Tribal Health Research Office, National Institutes of Health, Bethesda, MD. Loretta Christensen is with the Indian Health Service, Rockville, MD
| |
Collapse
|
1242
|
Winkelman TNA, Rai NK, Bodurtha PJ, Chamberlain AM, DeSilva M, Jeruzal J, Johnson SG, Kharbanda A, Klyn N, Mink PJ, Muscoplat M, Waring S, Yu Y, Drawz PE. Trends in COVID-19 Vaccine Administration and Effectiveness Through October 2021. JAMA Netw Open 2022; 5:e225018. [PMID: 35357452 PMCID: PMC8972031 DOI: 10.1001/jamanetworkopen.2022.5018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
IMPORTANCE COVID-19 vaccines are effective, but inequities in vaccine administration and waning immunity may limit vaccine effectiveness. OBJECTIVES To report statewide trends in vaccine administration and vaccine effectiveness in Minnesota. DESIGN, SETTING, AND PARTICIPANTS This cohort study used COVID-19 vaccine data from the Minnesota Immunization Information Connection from October 25, 2020, through October 30, 2021 that were linked with electronic health record (EHR) data from health systems collaborating as part of the Minnesota EHR Consortium (MNEHRC). Participants included individuals who were seen at a participating health system in Minnesota. EXPOSURES Individuals were considered fully vaccinated in the second week after receipt of a second dose of a BNT162b2 or mRNA-1273 vaccine or a single dose of an Ad26.COV.2.S vaccine. MAIN OUTCOMES AND MEASURES A completed vaccination series and vaccine breakthrough, defined as either a positive SARS-CoV-2 polymerase chain reaction (PCR) test or a hospital admission the same week or within the 3 weeks following a positive SARS-CoV-2 PCR test. A test-negative design and incident rate ratio were used to evaluate COVID-19 vaccine effectiveness separately for the BNT162b2, mRNA-1273, and Ad26.COV.2.S vaccines. Rurality and social vulnerability index were assessed at the area level. RESULTS This study included 4 431 190 unique individuals at participating health systems, and 3 013 704 (68%) of the individuals were fully vaccinated. Vaccination rates were lowest among Minnesotans who identified as Hispanic (116 422 of 217 019 [54%]), multiracial (30 066 of 57 412 [52%]), American Indian or Alaska Native (22 190 of 41 437 [54%]), and Black or African American (158 860 of 326 595 [49%]) compared with Minnesotans who identified as Asian or Pacific Islander (159 999 of 210 994 [76%]) or White (2 402 928 of 3 391 747 [71%]). Among individuals aged 19 to 64 years, vaccination rates were lower in rural areas (196 479 of 308 047 [64%]) compared with urban areas (151 541 of 1 951 265 [77%]) and areas with high social vulnerability (544 433 of 774 952 [70%]) compared with areas with low social vulnerability (571 613 of 724 369 [79%]). In the 9 weeks ending October 30, 2021, vaccine effectiveness as assessed by a test-negative design was 33% (95% CI, 30%-37%) for Ad26.COV.2.S; 53% (95% CI, 52%-54%) for BNT162b2; and 66% (95% CI, 65%-67%) for mRNA-1273. For SARS-CoV-2-related hospitalizations, vaccine effectiveness in the 9 weeks ending October 30, 2021, was 78% (95% CI, 75%-81%) for Ad26.COV.2.S; 81% (95% CI, 79%-82%) for BNT162b2; and 81% (95% CI, 79%-82%) for mRNA-1273. CONCLUSIONS AND RELEVANCE This cohort study of data from a Minnesota statewide consortium suggests disparities in vaccine administration and effectiveness. Vaccine effectiveness against infection was lower for Ad26.COV.2.S and BNT162b2 but was associated with protection against SARS-CoV-2-related hospitalizations despite the increased prevalence of the Delta variant in Minnesota.
Collapse
Affiliation(s)
- Tyler N A Winkelman
- Division of General Internal Medicine, Department of Medicine, Hennepin Healthcare, Minneapolis, Minnesota
- Health, Homelessness, and Criminal Justice Lab, Hennepin Healthcare Research Institute, Minneapolis, Minnesota
| | - Nayanjot K Rai
- Division of Nephrology and Hypertension, University of Minnesota Medical School, Minneapolis
| | - Peter J Bodurtha
- Health, Homelessness, and Criminal Justice Lab, Hennepin Healthcare Research Institute, Minneapolis, Minnesota
| | | | | | - Jessica Jeruzal
- Health, Homelessness, and Criminal Justice Lab, Hennepin Healthcare Research Institute, Minneapolis, Minnesota
| | - Steven G Johnson
- Institute for Health Informatics, University of Minnesota, Minneapolis
| | - Anupam Kharbanda
- Department of Pediatric Emergency Medicine, Children's Minnesota, Minneapolis
| | - Niall Klyn
- Department of Information Services, Essentia Health, Duluth, Minnesota
- School of Communication, Northwestern University, Evanston, Illinois
| | - Pamela J Mink
- Health Economics Program, Health Policy Division, Minnesota Department of Health, Saint Paul
| | - Miriam Muscoplat
- Division of Infectious Disease, Epidemiology, Prevention, and Control, Minnesota Department of Health, Saint Paul
| | | | - Yue Yu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Paul E Drawz
- Division of Nephrology and Hypertension, University of Minnesota Medical School, Minneapolis
| |
Collapse
|
1243
|
Yi S, Choe YJ, Kim J, Kim YY, Kim RK, Jang EJ, Lim DS, Byeon HR, Lee S, Park E, Kim SJ, Park YJ. SARS-CoV-2 Breakthrough Infections after introduction of 4 COVID-19 Vaccines, South Korea, 2021. Emerg Infect Dis 2022; 28:753-756. [PMID: 35202529 PMCID: PMC8888240 DOI: 10.3201/eid2803.212210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
We conducted a nationwide retrospective cohort study to estimate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infection among recipients of 4 different vaccines in South Korea. Age-adjusted breakthrough infection rate per month was highest for Janssen (42.6/100,000 population), followed by AstraZeneca (21.7/100,000 population), Pfizer-BioNTech (8.5/100,000 population), and Moderna (1.8/100,000 population).
Collapse
|
1244
|
Ullah M, Mubashir M, Atique H, Aslam F, Tahir M, Naqvi M. COVID-19 Infection in Vaccinated Healthcare Professionals. Cureus 2022; 14:e23386. [PMID: 35475110 PMCID: PMC9022484 DOI: 10.7759/cureus.23386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND There are different types of Coronavirus disease (COVID-19) vaccines available presently, and their emergency use has been approved by WHO worldwide on a mass scale. COVID-19 vaccine breakthrough infections have been reported worldwide. In Pakistan, there are limited data on COVID-19 vaccine breakthrough infections and their clinical course, especially in healthcare professionals (HCPs). Our study aims to investigate COVID-19 infections among vaccinated HCPs. METHODS A prospective study was conducted on 425 healthcare professionals. Data collected from healthcare professionals included names, age, gender, number of vaccination doses, COVID-19 infection pre and post-vaccination, the severity of COVID-19 infection (if positive), and co-morbid conditions. Ethical board approval was taken. Statistical Package for Social Sciences (SPSS) version 23 (IBM Corp., Armonk, NY) was used to analyze the data. RESULTS After complete vaccination, 17.27% acquired COVID-19 infection; 2.47% had COVID-19 infection both pre and post-vaccination. The mean age was 32.46 years (n=71) with a standard deviation of ±9.376. The male to female ratio was 1.53. COVID-19 PCR was positive in 95.77%. During the course of the disease, 4.2% were asymptomatic, 92.95% had mild symptoms, 1.4% were hospitalized, and 1.4% had to be managed in the intensive care unit. None of the HCPs who had received booster doses acquired a COVID-19 infection. CONCLUSION It was found that prior COVID-19 infection and vaccination do not confer immunity from infection. However, proper vaccination limits the severity, morbidity, and mortality of COVID-19 infection.
Collapse
Affiliation(s)
- Muneeb Ullah
- General Surgery, Maroof International Hospital, Islamabad, PAK
| | - Muaz Mubashir
- Internal Medicine, Federal Government Polyclinic Hospital, Islamabad, PAK
| | - Hassan Atique
- Internal Medicine, Federal Government Polyclinic Hospital, Islamabad, PAK
| | - Farhan Aslam
- Internal Medicine, Federal Government Polyclinic Hospital, Islamabad, PAK
| | - Musfirah Tahir
- Paediatrics, Maroof International Hospital, Islamabad, PAK
| | - Mehdi Naqvi
- Internal Medicine, Federal Government Polyclinic Hospital, Islamabad, PAK
| |
Collapse
|
1245
|
Klaassen F, Chitwood MH, Cohen T, Pitzer VE, Russi M, Swartwood NA, Salomon JA, Menzies NA. Population immunity to pre-Omicron and Omicron SARS-CoV-2 variants in US states and counties through December 1, 2021. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2021.12.23.21268272. [PMID: 34981078 PMCID: PMC8722621 DOI: 10.1101/2021.12.23.21268272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prior infection and vaccination both contribute to population-level SARS-CoV-2 immunity. We used a Bayesian model to synthesize evidence and estimate population immunity to prevalent SARS-CoV-2 variants in the United States over the course of the epidemic until December 1, 2021, and how this changed with the introduction of the Omicron variant. We used daily SARS-CoV-2 infection estimates and vaccination coverage data for each US state and county. We estimated relative rates of vaccination conditional on previous infection status using the Census Bureau’s Household Pulse Survey. We used published evidence on natural and vaccine-induced immunity, including waning and immune escape. The estimated percentage of the US population with a history of SARS-CoV-2 infection or vaccination as of December 1, 2021, was 88.2% (95%CrI: 83.6%-93.5%), compared to 24.9% (95%CrI: 18.5%-34.1%) on January 1, 2021. State-level estimates for December 1, 2021, ranged between 76.9% (95%CrI: 67.6%-87.6%, West Virginia) and 94.4% (95%CrI: 91.2%-97.3%, New Mexico). Accounting for waning and immune escape, the effective protection against the Omicron variant on December 1, 2021, was 21.8% (95%CrI: 20.7%-23.4%) nationally and ranged between 14.4% (95%CrI: 13.2%-15.8%, West Virginia), to 26.4% (95%CrI: 25.3%-27.8%, Colorado). Effective protection against severe disease from Omicron was 61.2% (95%CrI: 59.1%-64.0%) nationally and ranged between 53.0% (95%CrI: 47.3%-60.0%, Vermont) and 65.8% (95%CrI: 64.9%-66.7%, Colorado). While over three-quarters of the US population had prior immunological exposure to SARS-CoV-2 via vaccination or infection on December 1, 2021, only a fifth of the population was estimated to have effective protection to infection with the immune-evading Omicron variant. Significance Both SARS-CoV-2 infection and COVID-19 vaccination contribute to population-level immunity against SARS-CoV-2. This study estimates the immunity and effective protection against future SARS-CoV-2 infection in each US state and county over 2020-2021. The estimated percentage of the US population with a history of SARS-CoV-2 infection or vaccination as of December 1, 2021, was 88.2% (95%CrI: 83.6%-93.5%). Accounting for waning and immune escape, protection against the Omicron variant was 21.8% (95%CrI: 20.7%-23.4%). Protection against infection with the Omicron variant ranged between 14.4% (95%CrI: 13.2%-15.8%%, West Virginia) and 26.4% (95%CrI: 25.3%-27.8%, Colorado) across US states. The introduction of the immune-evading Omicron variant resulted in an effective absolute increase of approximately 30 percentage points in the fraction of the population susceptible to infection.
Collapse
Affiliation(s)
- Fayette Klaassen
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Melanie H. Chitwood
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, CT
| | - Ted Cohen
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, CT
| | - Virginia E. Pitzer
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, CT
| | - Marcus Russi
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, CT
| | - Nicole A. Swartwood
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Joshua A. Salomon
- Department of Health Policy, Stanford University School of Medicine, Stanford, CA
| | - Nicolas A. Menzies
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
1246
|
Saceleanu V, Moreanu MS, Covache-Busuioc RA, Mohan AG, Ciurea AV. SARS-COV-2 - the pandemic of the XXI century, clinical manifestations - neurological implications. J Med Life 2022; 15:319-327. [PMID: 35450003 PMCID: PMC9015186 DOI: 10.25122/jml-2020-0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/02/2022] [Indexed: 11/21/2022] Open
Abstract
In December 2019, in Wuhan, China, the first cases of infection with SARS-CoV 2 responsible for COVID-19 disease were identified. SARS-CoV 2 was declared a pandemic on March 11, 2020, and since then has attracted the medical world's attention. The threat to humans' health that this emerging pandemic could leave raises awareness on the importance of understanding the mechanisms that underlie the developing conditions. The epidemiology, clinical picture, and pathogenesis of COVID-19 show that this virus presents new strategies to overcome the past defensive medicine. While all the current data has focused on the pulmonary and cardiovascular manifestations, little has been written about the neurological implications of the disease. This review updates new clinical aspects that SARS-CoV 2 expresses in humans by focusing primarily on neurological manifestations. The damage to the nervous system became more apparent - anosmia, ageusia, polyneuritis, meningitis, meningoencephalitis, stroke, acute necrotizing encephalopathy. Oxygen therapy is vital for those in critical health situations. Finally, prevention is the most important element in breaking the epidemiological chain.
Collapse
Affiliation(s)
- Vicentiu Saceleanu
- Department of Neurosurgery, Faculty of Medicine, Lucian Blaga University, Sibiu, Romania
- Department of Neurosurgery, County Emergency Hospital, Sibiu, Romania
| | - Mihai-Stelian Moreanu
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Aurel George Mohan
- Department of Neurosurgery, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
- Department of Neurosurgery, County Emergency Hospital, Oradea, Romania
| | - Alexandru-Vlad Ciurea
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Neurosurgery, Sanador Clinical Hospital, Bucharest, Romania
| |
Collapse
|
1247
|
Itaya T, Isobe Y, Suzuki S, Koike K, Nishigaki M, Yamamoto Y. The Fragility of Statistically Significant Results in Randomized Clinical Trials for COVID-19. JAMA Netw Open 2022; 5:e222973. [PMID: 35302631 PMCID: PMC8933746 DOI: 10.1001/jamanetworkopen.2022.2973] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
IMPORTANCE Interpreting results from randomized clinical trials (RCTs) for COVID-19, which have been published rapidly and in vast numbers, is challenging during a pandemic. OBJECTIVE To evaluate the robustness of statistically significant findings from RCTs for COVID-19 using the fragility index. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional study included COVID-19 trial articles that randomly assigned patients 1:1 into 2 parallel groups and reported at least 1 binary outcome as significant in the abstract. A systematic search was conducted using PubMed to identify RCTs on COVID-19 published until August 7, 2021. EXPOSURES Trial characteristics, such as type of intervention (treatment drug, vaccine, or others), number of outcome events, and sample size. MAIN OUTCOMES AND MEASURES Fragility index. RESULTS Of the 47 RCTs for COVID-19 included, 36 (77%) were studies of the effects of treatment drugs, 5 (11%) were studies of vaccines, and 6 (13%) were of other interventions. A total of 138 235 participants were included in these trials. The median (IQR) fragility index of the included trials was 4 (1-11). The medians (IQRs) of the fragility indexes of RCTs of treatment drugs, vaccines, and other interventions were 2.5 (1-6), 119 (61-139), and 4.5 (1-18), respectively. The fragility index among more than half of the studies was less than 1% of each sample size, although the fragility index as a proportion of events needing to change would be much higher. CONCLUSIONS AND RELEVANCE This cross-sectional study found a relatively small number of events (a median of 4) would be required to change the results of COVID-19 RCTs from statistically significant to not significant. These findings suggest that health care professionals and policy makers should not rely heavily on individual results of RCTs for COVID-19.
Collapse
Affiliation(s)
- Takahiro Itaya
- Department of Healthcare Epidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Yotsuha Isobe
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sayoko Suzuki
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kanako Koike
- Department of Medical Genetics, International University of Health and Welfare Graduate School, Tokyo, Japan
| | - Masakazu Nishigaki
- Department of Medical Genetics, International University of Health and Welfare Graduate School, Tokyo, Japan
| | - Yosuke Yamamoto
- Department of Healthcare Epidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| |
Collapse
|
1248
|
Woldemeskel BA, Dykema AG, Garliss CC, Cherfils S, Smith KN, Blankson JN. CD4+ T cells from COVID-19 mRNA vaccine recipients recognize a conserved epitope present in diverse coronaviruses. J Clin Invest 2022; 132:e156083. [PMID: 35061630 PMCID: PMC8884904 DOI: 10.1172/jci156083] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/19/2022] [Indexed: 11/28/2022] Open
Abstract
Recent studies have shown that vaccinated individuals harbor T cells that can cross-recognize SARS-CoV-2 and endemic human common cold coronaviruses. However, it is still unknown whether CD4+ T cells from vaccinated individuals recognize peptides from bat coronaviruses that may have the potential of causing future pandemics. In this study, we identified a SARS-CoV-2 spike protein epitope (S815-827) that is conserved in coronaviruses from different genera and subgenera, including SARS-CoV, MERS-CoV, multiple bat coronaviruses, and a feline coronavirus. Our results showed that S815-827 was recognized by 42% of vaccinated participants in our study who received the Pfizer-BioNTech (BNT162b2) or Moderna (mRNA-1273) COVID-19 vaccines. Using T cell expansion and T cell receptor sequencing assays, we demonstrated that S815-827-reactive CD4+ T cells from the majority of responders cross-recognized homologous peptides from at least 6 other diverse coronaviruses. Our results support the hypothesis that the current mRNA vaccines elicit T cell responses that can cross-recognize bat coronaviruses and thus might induce some protection against potential zoonotic outbreaks. Furthermore, our data provide important insights that inform the development of T cell-based pan-coronavirus vaccine strategies.
Collapse
Affiliation(s)
| | - Arbor G. Dykema
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | - Kellie N. Smith
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | | |
Collapse
|
1249
|
Polinski JM, Weckstein AR, Batech M, Kabelac C, Kamath T, Harvey R, Jain S, Rassen JA, Khan N, Schneeweiss S. Durability of the Single-Dose Ad26.COV2.S Vaccine in the Prevention of COVID-19 Infections and Hospitalizations in the US Before and During the Delta Variant Surge. JAMA Netw Open 2022; 5:e222959. [PMID: 35297969 PMCID: PMC8931562 DOI: 10.1001/jamanetworkopen.2022.2959] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
IMPORTANCE Vaccination against the SARS-CoV-2 virus is critical to control the pandemic. Randomized clinical trials demonstrated efficacy of the single-dose Ad26.COV2.S COVID-19 vaccine, but data on longer-term protection in clinical practice and effectiveness against variants are needed. OBJECTIVE To assess the association between receiving the Ad26.COV2.S vaccine and COVID-19-related infections and hospitalizations before and during the Delta variant surge. DESIGN, SETTING, AND PARTICIPANTS This cohort study included adults aged 18 years and older who were newly Ad26.COV2.S-vaccinated matched to as many as 10 unvaccinated individuals by date, location, age, sex, and comorbidity index. This was followed by 1:4 propensity score matching on COVID-19 risk factors. Data were collected from US insurance claims data from March 1, 2020, through August 31, 2021. EXPOSURES Vaccination with Ad26.COV2.S vs no vaccination. MAIN OUTCOMES AND MEASURES Vaccine effectiveness (VE) was estimated for recorded COVID-19 infection and COVID-19-related hospitalization, nationwide and in subgroups by age, high-risk factors, calendar time, and states with high incidences of the Delta variant. VE estimates were corrected for underrecording of vaccinations in insurance data. RESULTS Among 422 034 vaccinated individuals (mean [SD] age, 54.7 [17.4] years; 236 437 [56.0%] women) and 1 645 397 matched unvaccinated individuals (mean [SD] age, 54.5 [17.5] years; 922 937 [56.1%] women), VE was 76% (95% CI, 75%-77%) for COVID-19 infections and 81% (95% CI, 78%-82%) for COVID-19-related hospitalizations. VE was stable for at least 180 days after vaccination and over calendar time. Among states with high Delta variant incidence, VE during June to August 2021 was 74% (95% CI, 71%-77%) for infections and 81% (95% CI, 75%-86%) for hospitalizations. VE for COVID-19 was higher in individuals younger than 65 years (78%; 95% CI, 77%-79%) and lower in immunocompromised patients (64%; 95% CI, 59%-68%). All estimates were corrected for vaccination underrecording; uncorrected VE, which served as a lower bound, was 66% (95% CI, 64%-67%) for any recorded COVID-19 infection and 72% (95% CI, 69%-74%) for COVID-19-related hospitalization. CONCLUSIONS AND RELEVANCE This cohort study in US clinical practice showed stable VE of Ad26.COV2.S for at least 6 months before as well as during the time the Delta variant emerged and became dominant.
Collapse
Affiliation(s)
| | | | | | - Carly Kabelac
- Department of Science, Aetion Inc, New York, New York
| | - Tripthi Kamath
- Janssen Research and Development Data Science, Spring House, Pennsylvania
| | - Raymond Harvey
- Janssen Research and Development Data Science, Spring House, Pennsylvania
| | - Sid Jain
- Janssen Research and Development Data Science, Spring House, Pennsylvania
| | | | - Najat Khan
- Janssen Research and Development Data Science, Spring House, Pennsylvania
| | - Sebastian Schneeweiss
- Department of Science, Aetion Inc, New York, New York
- Division of Pharmacoepidemiology, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
1250
|
Hsu CM, Weiner DE, Manley HJ, Aweh GN, Ladik V, Frament J, Miskulin D, Argyropoulos C, Abreo K, Chin A, Gladish R, Salman L, Johnson D, Lacson EK. Seroresponse to SARS-CoV-2 Vaccines among Maintenance Dialysis Patients over 6 Months. Clin J Am Soc Nephrol 2022; 17:403-413. [PMID: 35144972 PMCID: PMC8975038 DOI: 10.2215/cjn.12250921] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/05/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Although most patients receiving maintenance dialysis exhibit initial seroresponse to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination, concerns exist regarding the durability of this antibody response. This study evaluated seroresponse over time. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This retrospective cohort study included patients on maintenance dialysis, from a midsize national dialysis provider, who received a complete SARS-CoV-2 vaccine series and had at least one antibody titer checked after full vaccination. IgG spike antibodies (anti-spike IgG) titers were assessed monthly with routine laboratory tests after vaccination; the semiquantitative assay reported a range between zero and ≥20 Index. Descriptive analyses compared trends over time by history of coronavirus disease 2019 (COVID-19) and vaccine type. Time-to-event analyses examined the outcome of loss of seroresponse (anti-spike IgG <1 Index or development of COVID-19). Cox regression adjusted for additional clinical characteristics. RESULTS Among 1870 patients receiving maintenance dialysis, 1569 had no prior COVID-19. Patients without prior COVID-19 had declining titers over time. Among 443 recipients of BNT162b2 (Pfizer), median (interquartile range) anti-spike IgG titer declined from ≥20 (5.89 to ≥20) in month 1 after full vaccination to 1.96 (0.60-5.88) by month 6. Among 778 recipients of mRNA-1273 (Moderna), anti-spike IgG titer declined from ≥20 (interquartile range, ≥20 to ≥20) in month 1 to 7.99 (2.61 to ≥20) by month 6. The 348 recipients of Ad26.COV2.S (Janssen) had a lower titer response than recipients of an mRNA vaccine over all time periods. In time-to-event analyses, recipients of Ad26.COV2.S and mRNA-1273 had the shortest and longest time to loss of seroresponse, respectively. The maximum titer reached in the first 2 months after full vaccination was associated with durability of the anti-spike IgG seroresponse; patients with anti-spike IgG titer 1-19.99 had a shorter time to loss of seroresponse compared with patients with anti-spike IgG titer ≥20 (hazard ratio, 15.5; 95% confidence interval, 11.7 to 20.7). CONCLUSIONS Among patients receiving maintenance dialysis, vaccine-induced seroresponse wanes over time across vaccine types. Early titers after full vaccination are associated with the durability of seroresponse.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kenneth Abreo
- Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Andrew Chin
- University of California, Davis, Sacramento, California
| | | | | | | | - Eduardo K. Lacson
- Tufts Medical Center, Boston, Massachusetts,Dialysis Clinic Inc., Nashville, Tennessee
| |
Collapse
|