101
|
Markewitz R, Pauli D, Dargvainiene J, Steinhagen K, Engel S, Herbst V, Zapf D, Krüger C, Sharifzadeh S, Schomburg B, Leypoldt F, Rupp J, Görg S, Junker R, Wandinger KP. The temporal course of T- and B-cell responses to vaccination with BNT162b2 and mRNA-1273. Clin Microbiol Infect 2022; 28:701-709. [PMID: 34547457 PMCID: PMC8450229 DOI: 10.1016/j.cmi.2021.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/31/2021] [Accepted: 09/05/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To investigate the response of the immune system (and its influencing factors) to vaccination with BNT162b2 or mRNA-1273. METHODS 531 vaccinees, recruited from healthcare professionals, donated samples before, in between, and after the administration of the two doses of the vaccine. T- and B-cell responses were examined via interferon-γ (IFN-γ) release assay, and antibodies against different epitopes of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (S1 and NCP) were detected via ELISA and surrogate neutralization assay. Results were correlated with influencing factors such as age, sex, prior infection, vaccine received (BNT162b2 or mRNA-1273), and immunosuppression. Furthermore, antinuclear antibodies (ANAs) were measured to screen for autoimmune responses following vaccination with an mRNA vaccine. RESULTS No markers of immunity against SARS-CoV-2 were found before the first vaccination. Two weeks after it, specific responses against SARS-CoV-2 were already measurable (median ± median absolute deviation (MAD): anti-S1 IgG 195.5 ± 172.7 BAU/mL; IgA 6.7 ± 4.9 OD; surrogate neutralization 39 ± 23.7%), and were significantly increased two weeks after the second dose (anti-S1 IgG 3744 ± 2571.4 BAU/mL; IgA 12 ± 0 OD; surrogate neutralization 100 ± 0%, IFN-γ 1897.2 ± 886.7 mIU/mL). Responses were stronger for younger participants (this difference decreasing after the second dose). Further influences were previous infection with SARS-CoV-2 (causing significantly stronger responses after the first dose compared to unexposed individuals (p ≤ 0.0001)) and the vaccine received (significantly stronger reactions for recipients of mRNA-1273 after both doses, p < 0.05-0.0001). Some forms of immunosuppression significantly impeded the immune response to the vaccination (with no observable immune response in three immunosuppressed participants). There was no significant induction of ANAs by the vaccination (no change in qualitative ANA results (p 0.2592) nor ANA titres (p 0.08) from pre-to post-vaccination. CONCLUSIONS Both vaccines elicit strong and specific immune responses against SARS-CoV-2 which become detectable one week (T-cell response) or two weeks (B-cell response) after the first dose.
Collapse
Affiliation(s)
- Robert Markewitz
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany.
| | - Daniela Pauli
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Justina Dargvainiene
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Katja Steinhagen
- Institute for Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | - Sarah Engel
- Department of Anesthesiology and Intensive Care, University Hospital of Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Victor Herbst
- Institute for Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | - Dorinja Zapf
- Institute for Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | - Christina Krüger
- Institute for Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | - Shahpour Sharifzadeh
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Benjamin Schomburg
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Frank Leypoldt
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany; Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Siegfried Görg
- Institute of Transfusion Medicine, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Ralf Junker
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
102
|
Sridhar S, Joaquin A, Bonaparte MI, Bueso A, Chabanon AL, Chen A, Chicz RM, Diemert D, Essink BJ, Fu B, Grunenberg NA, Janosczyk H, Keefer MC, Rivera M DM, Meng Y, Michael NL, Munsiff SS, Ogbuagu O, Raabe VN, Severance R, Rivas E, Romanyak N, Rouphael NG, Schuerman L, Sher LD, Walsh SR, White J, von Barbier D, de Bruyn G, Canter R, Grillet MH, Keshtkar-Jahromi M, Koutsoukos M, Lopez D, Masotti R, Mendoza S, Moreau C, Ceregido MA, Ramirez S, Said A, Tavares-Da-Silva F, Shi J, Tong T, Treanor J, Diazgranados CA, Savarino S. Safety and immunogenicity of an AS03-adjuvanted SARS-CoV-2 recombinant protein vaccine (CoV2 preS dTM) in healthy adults: interim findings from a phase 2, randomised, dose-finding, multicentre study. THE LANCET. INFECTIOUS DISEASES 2022; 22:636-648. [PMID: 35090638 PMCID: PMC8789245 DOI: 10.1016/s1473-3099(21)00764-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/16/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND We evaluated our SARS-CoV-2 prefusion spike recombinant protein vaccine (CoV2 preS dTM) with different adjuvants, unadjuvanted, and in a one-injection and two-injection dosing schedule in a previous phase 1-2 study. Based on interim results from that study, we selected a two-injection schedule and the AS03 adjuvant for further clinical development. However, lower than expected antibody responses, particularly in older adults, and higher than expected reactogenicity after the second vaccination were observed. In the current study, we evaluated the safety and immunogenicity of an optimised formulation of CoV2 preS dTM adjuvanted with AS03 to inform progression to phase 3 clinical trial. METHODS This phase 2, randomised, parallel-group, dose-ranging study was done in adults (≥18 years old), including those with pre-existing medical conditions, those who were immunocompromised (except those with recent organ transplant or chemotherapy) and those with a potentially increased risk for severe COVID-19, at 20 clinical research centres in the USA and Honduras. Women who were pregnant or lactating or, for those of childbearing potential, not using an effective method of contraception or abstinence, and those who had received a COVID-19 vaccine, were excluded. Participants were randomly assigned (1:1:1) using an interactive response technology system, with stratification by age (18-59 years and ≥60 years), rapid serodiagnostic test result (positive or negative), and high-risk medical conditions (yes or no), to receive two injections (day 1 and day 22) of 5 7mu;g (low dose), 10 7mu;g (medium dose), or 15 7mu;g (high dose) CoV2 preS dTM antigen with fixed AS03 content. All participants and outcome assessors were masked to group assignment; unmasked study staff involved in vaccine preparation were not involved in safety outcome assessments. All laboratory staff performing the assays were masked to treatment. The primary safety objective was to describe the safety profile in all participants, for each candidate vaccine formulation. Safety endpoints were evaluated for all randomised participants who received at least one dose of the study vaccine (safety analysis set), and are presented here for the interim study period (up to day 43). The primary immunogenicity objective was to describe the neutralising antibody titres to the D614G variant 14 days after the second vaccination (day 36) in participants who were SARS-CoV-2 naive who received both injections, provided samples at day 1 and day 36, did not have protocol deviations, and did not receive an authorised COVID-19 vaccine before day 36. Neutralising antibodies were measured using a pseudovirus neutralisation assay and are presented here up to 14 days after the second dose. As a secondary immunogenicity objective, we assessed neutralising antibodies in non-naive participants. This trial is registered with ClinicalTrials.gov (NCT04762680) and is closed to new participants for the cohort reported here. FINDINGS Of 722 participants enrolled and randomly assigned between Feb 24, 2021, and March 8, 2021, 721 received at least one injection (low dose=240, medium dose=239, and high dose=242). The proportion of participants reporting at least one solicited adverse reaction (injection site or systemic) in the first 7 days after any vaccination was similar between treatment groups (217 [91%] of 238 in the low-dose group, 213 [90%] of 237 in the medium-dose group, and 218 [91%] of 239 in the high-dose group); these adverse reactions were transient, were mostly mild to moderate in intensity, and occurred at a higher frequency and intensity after the second vaccination. Four participants reported immediate unsolicited adverse events; two (one each in the low-dose group and medium-dose group) were considered by the investigators to be vaccine related and two (one each in the low-dose and high-dose groups) were considered unrelated. Five participants reported seven vaccine-related medically attended adverse events (two in the low-dose group, one in the medium-dose group, and four in the high-dose group). No vaccine-related serious adverse events and no adverse events of special interest were reported. Among participants naive to SARS-CoV-2 at day 36, 158 (98%) of 162 in the low-dose group, 166 (99%) of 168 in the medium-dose group, and 163 (98%) of 166 in the high-dose group had at least a two-fold increase in neutralising antibody titres to the D614G variant from baseline. Neutralising antibody geometric mean titres (GMTs) at day 36 for participants who were naive were 2189 (95% CI 1744-2746) for the low-dose group, 2269 (1792-2873) for the medium-dose group, and 2895 (2294-3654) for the high-dose group. GMT ratios (day 36: day 1) were 107 (95% CI 85-135) in the low-dose group, 110 (87-140) in the medium-dose group, and 141 (111-179) in the high-dose group. Neutralising antibody titres in non-naive adults 21 days after one injection tended to be higher than titres after two injections in adults who were naive, with GMTs 21 days after one injection for participants who were non-naive being 3143 (95% CI 836-11 815) in the low-dose group, 2338 (593-9226) in the medium-dose group, and 7069 (1361-36 725) in the high-dose group. INTERPRETATION Two injections of CoV2 preS dTM-AS03 showed acceptable safety and reactogenicity, and robust immunogenicity in adults who were SARS-CoV-2 naive and non-naive. These results supported progression to phase 3 evaluation of the 10 7mu;g antigen dose for primary vaccination and a 5 7mu;g antigen dose for booster vaccination. FUNDING Sanofi Pasteur and Biomedical Advanced Research and Development Authority.
Collapse
Affiliation(s)
| | - Arnel Joaquin
- Charles R Drew University of Medicine and Science, Los Angeles, CA, USA
| | | | | | | | | | | | - David Diemert
- The George Washington School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Bo Fu
- Sanofi Pasteur, Swiftwater, PA, USA
| | | | | | - Michael C Keefer
- University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Ya Meng
- Sanofi Pasteur, Swiftwater, PA, USA
| | | | - Sonal S Munsiff
- University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Vanessa N Raabe
- New York University Grossman School of Medicine, New York, NY, USA
| | | | | | | | | | | | - Lawrence D Sher
- Peninsula Research Associates, Rolling Hills Estates, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Shelly Ramirez
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | - Jiayuan Shi
- TechData Service Company, King of Prussia, PA, USA
| | - Tina Tong
- Vaccine Translational Research Branch, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - John Treanor
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | | |
Collapse
|
103
|
Gavriatopoulou M, Terpos E, Kastritis E, Briasoulis A, Gumeni S, Ntanasis-Stathopoulos I, Sklirou AD, Malandrakis P, Eleutherakis-Papaiakovou E, Migkou M, Trougakos IP, Dimopoulos MA. Low neutralizing antibody responses in WM, CLL and NHL patients after the first dose of the BNT162b2 and AZD1222 vaccine. Clin Exp Med 2022; 22:319-323. [PMID: 34283338 PMCID: PMC8290394 DOI: 10.1007/s10238-021-00746-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/12/2021] [Indexed: 12/27/2022]
Abstract
Vaccination against SARS-CoV-2 is considered as the most important preventive strategy against COVID-19, but its efficacy in patients with hematological malignancies is largely unknown. We investigated the development of neutralizing antibodies (NAbs) against SARS-CoV-2 in patients with Waldenstrom Macroglobulinemia (WM), Chronic Lymphocytic Leukemia (CLL) and Non-Hodgkin Lymphoma (NHL). After the first dose of the vaccine, on D22, WM/CLL/NHL patients had lower NAb titers compared to controls: the median NAb inhibition titer was 17% (range 0-91%, IQR 8-27%) for WM/CLL/NHL patients versus 32% (range 2-98%, IQR 19-48%) for controls (P < 0.001). Only 8 (14%) patients versus 114 (54%) controls developed NAb titers ≥ 30% on D22 (p < 0.001). Our data indicate that the first dose of both BNT162b2 and AZD1222 leads to lower production of NAbs against SARS-CoV-2 in patients with WM/CLL/NHL compared to controls of similar age and gender and without malignant disease. Even though the response rates were not optimal, vaccination is still considered essential and if possible should be performed before treatment initiation. These patients with suboptimal responses should be considered to be prioritized for booster doses.
Collapse
Affiliation(s)
- Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece.
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| | - Alexandros Briasoulis
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| | - Aimilia D Sklirou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| | - Evangelos Eleutherakis-Papaiakovou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| | - Magdalini Migkou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 80 Vas. Sofias Avenue, 11528, Athens, Greece
| |
Collapse
|
104
|
Patil R, Shanbhag S, Shankarkumar A, Madkaikar M. Antibody profile in post-vaccinated & SARS-CoV-2 infected individuals. Indian J Med Res 2022; 155:538-545. [PMID: 36124500 PMCID: PMC9807196 DOI: 10.4103/ijmr.ijmr_3330_21] [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: 11/16/2021] [Indexed: 02/04/2023] Open
Abstract
Background & objectives During the COVID-19 pandemic it was important to assess the antibody profile in individuals vaccinated with Covaxin (BBV152) and Covishield (ChAdOx1 nCoV-19) with both 28 and 84 days gaps between two doses, those infected with SARS-CoV-2 and post-COVID-19-infected individuals vaccinated with only one dose of either of the vaccines. The present study was aimed to assess these objectives. Methods Fifty real time reverse transcription-polymerase chain reaction (qRT-PCR)-confirmed COVID-19-infected individuals, along with 90 COVID-19-naïve (BBV152 and ChAdOx1 nCov-19)-vaccinated individuals, were included in the study. Individuals who received a single dose of either vaccine with a confirmed past diagnosis of SARS-CoV-2 infection (n=15) were also included. Blood samples were collected strictly between the 4th and 5th wk after development of symptoms for SARS-CoV-2 infected individuals and after the first/second vaccination dose. Antibody profile assessment was done using whole-virus, spike-receptor binding domain (RBD) and nucleocapsid-specific ELISA kits along with neutralizing antibody kit. Results There was an overall 97.7 per cent seropositivity rate in vaccinated individuals, and a strong correlation (R2=0.8, P<0.001) between neutralizing and spike-RBD antibodies. Among individuals who received two standard doses of ChAdOx1 nCoV-19 vaccine, the spike antibody levels developed were of higher titre with a longer prime boost interval than in those with shorter intervals (P<0.01). Individuals vaccinated with two doses as well as only one dose post-SARS-CoV-2 infection had high neutralizing and spike-specific antibodies. Interpretation & conclusions High neutralizing and spike-specific antibodies were developed in individuals vaccinated only with one dose of either vaccine post-SARS-CoV-2 infection. With the main priority being vaccinating majority of the population in our country, single-dose administration to such individuals would be a sensible way to make the most of the limited supplies. Furthermore, neutralizing antibody levels observed in COVID-19-naïve vaccinees imply the need for booster vaccination.
Collapse
Affiliation(s)
- Rucha Patil
- Department of Haemostasis and Thrombosis, ICMR-National Institute of Immunohaematology, Mumbai, Maharashtra, India
| | - Sharda Shanbhag
- Department of Haemostasis and Thrombosis, ICMR-National Institute of Immunohaematology, Mumbai, Maharashtra, India
| | - Aruna Shankarkumar
- Department of Haemostasis and Thrombosis, ICMR-National Institute of Immunohaematology, Mumbai, Maharashtra, India
| | - Manisha Madkaikar
- Department of Haemostasis and Thrombosis, ICMR-National Institute of Immunohaematology, Mumbai, Maharashtra, India
| |
Collapse
|
105
|
DeGrace MM, Ghedin E, Frieman MB, Krammer F, Grifoni A, Alisoltani A, Alter G, Amara RR, Baric RS, Barouch DH, Bloom JD, Bloyet LM, Bonenfant G, Boon ACM, Boritz EA, Bratt DL, Bricker TL, Brown L, Buchser WJ, Carreño JM, Cohen-Lavi L, Darling TL, Davis-Gardner ME, Dearlove BL, Di H, Dittmann M, Doria-Rose NA, Douek DC, Drosten C, Edara VV, Ellebedy A, Fabrizio TP, Ferrari G, Fischer WM, Florence WC, Fouchier RAM, Franks J, García-Sastre A, Godzik A, Gonzalez-Reiche AS, Gordon A, Haagmans BL, Halfmann PJ, Ho DD, Holbrook MR, Huang Y, James SL, Jaroszewski L, Jeevan T, Johnson RM, Jones TC, Joshi A, Kawaoka Y, Kercher L, Koopmans MPG, Korber B, Koren E, Koup RA, LeGresley EB, Lemieux JE, Liebeskind MJ, Liu Z, Livingston B, Logue JP, Luo Y, McDermott AB, McElrath MJ, Meliopoulos VA, Menachery VD, Montefiori DC, Mühlemann B, Munster VJ, Munt JE, Nair MS, Netzl A, Niewiadomska AM, O'Dell S, Pekosz A, Perlman S, Pontelli MC, Rockx B, Rolland M, Rothlauf PW, Sacharen S, Scheuermann RH, Schmidt SD, Schotsaert M, Schultz-Cherry S, Seder RA, Sedova M, Sette A, Shabman RS, Shen X, Shi PY, Shukla M, Simon V, Stumpf S, Sullivan NJ, Thackray LB, Theiler J, Thomas PG, Trifkovic S, Türeli S, Turner SA, Vakaki MA, van Bakel H, VanBlargan LA, Vincent LR, Wallace ZS, Wang L, Wang M, Wang P, Wang W, Weaver SC, Webby RJ, Weiss CD, Wentworth DE, Weston SM, Whelan SPJ, Whitener BM, Wilks SH, Xie X, Ying B, Yoon H, Zhou B, Hertz T, Smith DJ, Diamond MS, Post DJ, Suthar MS. Defining the risk of SARS-CoV-2 variants on immune protection. Nature 2022; 605:640-652. [PMID: 35361968 PMCID: PMC9345323 DOI: 10.1038/s41586-022-04690-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/24/2022] [Indexed: 11/09/2022]
Abstract
The global emergence of many severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants jeopardizes the protective antiviral immunity induced after infection or vaccination. To address the public health threat caused by the increasing SARS-CoV-2 genomic diversity, the National Institute of Allergy and Infectious Diseases within the National Institutes of Health established the SARS-CoV-2 Assessment of Viral Evolution (SAVE) programme. This effort was designed to provide a real-time risk assessment of SARS-CoV-2 variants that could potentially affect the transmission, virulence, and resistance to infection- and vaccine-induced immunity. The SAVE programme is a critical data-generating component of the US Government SARS-CoV-2 Interagency Group to assess implications of SARS-CoV-2 variants on diagnostics, vaccines and therapeutics, and for communicating public health risk. Here we describe the coordinated approach used to identify and curate data about emerging variants, their impact on immunity and effects on vaccine protection using animal models. We report the development of reagents, methodologies, models and notable findings facilitated by this collaborative approach and identify future challenges. This programme is a template for the response to rapidly evolving pathogens with pandemic potential by monitoring viral evolution in the human population to identify variants that could reduce the effectiveness of countermeasures.
Collapse
Affiliation(s)
- Marciela M DeGrace
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Elodie Ghedin
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Systems Genomics Section, Laboratory of Parasitic Diseases, National Institutes of Health, Rockville, MD, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jesse D Bloom
- Fred Hutch Cancer Center, Howard Hughes Medical Institute, Seattle, WA, USA
| | - Louis-Marie Bloyet
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Gaston Bonenfant
- CDC COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Eli A Boritz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Debbie L Bratt
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- CAMRIS, Contractor for NIAID, Bethesda, MD, USA
| | - Traci L Bricker
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Liliana Brown
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - William J Buchser
- High Throughput Screening Center, Washington University School of Medicine, St Louis, MO, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liel Cohen-Lavi
- National Institute for Biotechnology in the Negev, Department of Industrial Engineering and Management, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Tamarand L Darling
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Meredith E Davis-Gardner
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Bethany L Dearlove
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Han Di
- CDC COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Meike Dittmann
- Microbiology Department, New York University Grossman School of Medicine, New York, NY, USA
| | - Nicole A Doria-Rose
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Daniel C Douek
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin and German Center for Infection Research (DZIF), Berlin, Germany
| | - Venkata-Viswanadh Edara
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Ali Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Thomas P Fabrizio
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Will M Fischer
- Los Alamos National Laboratory, New Mexico Consortium, Los Alamos, NM, USA
| | - William C Florence
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - John Franks
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam Godzik
- University of California Riverside School of Medicine, Riverside, CA, USA
| | - Ana Silvia Gonzalez-Reiche
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aubree Gordon
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Bart L Haagmans
- Department Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Michael R Holbrook
- National Institute of Allergy and Infectious Diseases Integrated Research Facility, Frederick, MD, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Sarah L James
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Lukasz Jaroszewski
- University of California Riverside School of Medicine, Riverside, CA, USA
| | - Trushar Jeevan
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert M Johnson
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Terry C Jones
- Institute of Virology, Charité-Universitätsmedizin and German Center for Infection Research (DZIF), Berlin, Germany
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Astha Joshi
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
| | - Lisa Kercher
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Bette Korber
- Los Alamos National Laboratory, New Mexico Consortium, Los Alamos, NM, USA
| | - Eilay Koren
- National Institute for Biotechnology in the Negev, Department of Industrial Engineering and Management, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
- The Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Richard A Koup
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Eric B LeGresley
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | | | - Mariel J Liebeskind
- High Throughput Screening Center, Washington University School of Medicine, St Louis, MO, USA
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Brandi Livingston
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - James P Logue
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yang Luo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Adrian B McDermott
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | | | - Victoria A Meliopoulos
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Vineet D Menachery
- Department of Microbiology and Immunology, Institute for Human Infection and Immunity, World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Barbara Mühlemann
- Institute of Virology, Charité-Universitätsmedizin and German Center for Infection Research (DZIF), Berlin, Germany
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Vincent J Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jenny E Munt
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Antonia Netzl
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | | | - Sijy O'Dell
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Marjorie C Pontelli
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Barry Rockx
- Department Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Morgane Rolland
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Paul W Rothlauf
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Sinai Sacharen
- National Institute for Biotechnology in the Negev, Department of Industrial Engineering and Management, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
- The Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | | | - Stephen D Schmidt
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert A Seder
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Mayya Sedova
- University of California Riverside School of Medicine, Riverside, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Reed S Shabman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Maulik Shukla
- University of Chicago Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL, USA
- Data Science and Learning Division, Argonne National Laboratory, Argonne, IL, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Spencer Stumpf
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Nancy J Sullivan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Vaccine Research Center, Bethesda, MD, USA
| | - Larissa B Thackray
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - James Theiler
- Los Alamos National Laboratory, New Mexico Consortium, Los Alamos, NM, USA
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sanja Trifkovic
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sina Türeli
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Samuel A Turner
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Maria A Vakaki
- High Throughput Screening Center, Washington University School of Medicine, St Louis, MO, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura A VanBlargan
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Leah R Vincent
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Zachary S Wallace
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA
| | - Li Wang
- CDC COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Maple Wang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Pengfei Wang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Wei Wang
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Scott C Weaver
- Department of Microbiology and Immunology, Institute for Human Infection and Immunity, World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Carol D Weiss
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - David E Wentworth
- CDC COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Stuart M Weston
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Bradley M Whitener
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Samuel H Wilks
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Baoling Ying
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Hyejin Yoon
- Los Alamos National Laboratory, New Mexico Consortium, Los Alamos, NM, USA
| | - Bin Zhou
- CDC COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Tomer Hertz
- Department of Microbiology, Immunology and Genetics Faculty of Health Sciences Ben-Gurion University of the Negev, Be'er Sheva, Israel.
| | - Derek J Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK.
| | - Michael S Diamond
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA.
| | - Diane J Post
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| | - Mehul S Suthar
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
106
|
Lechmere T, Snell LB, Graham C, Seow J, Shalim ZA, Charalampous T, Alcolea-Medina A, Batra R, Nebbia G, Edgeworth JD, Malim MH, Doores KJ. Broad Neutralization of SARS-CoV-2 Variants, Including Omicron, following Breakthrough Infection with Delta in COVID-19-Vaccinated Individuals. mBio 2022; 13:e0379821. [PMID: 35297676 PMCID: PMC9040729 DOI: 10.1128/mbio.03798-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/14/2022] [Indexed: 12/16/2022] Open
Abstract
Numerous studies have shown that a prior SARS-CoV-2 infection can greatly enhance the antibody response to COVID-19 vaccination, with this so called "hybrid immunity" leading to greater neutralization breadth against SARS-CoV-2 variants of concern. However, little is known about how breakthrough infection (BTI) in COVID-19-vaccinated individuals will impact the magnitude and breadth of the neutralizing antibody response. Here, we compared neutralizing antibody responses between unvaccinated and COVID-19-double-vaccinated individuals (including both AZD1222 and BNT162b2 vaccinees) who have been infected with the Delta (B.1.617.2) variant. Rapid production of spike-reactive IgG was observed in the vaccinated group, providing evidence of effective vaccine priming. Overall, potent cross-neutralizing activity against current SARS-CoV-2 variants of concern was observed in the BTI group compared to the infection group, including neutralization of the Omicron (B.1.1.529) variant. This study provides important insights into population immunity where transmission levels remain high and in the context of new or emerging variants of concern. IMPORTANCE COVID-19 vaccines have been vital in controlling SARS-CoV-2 infections and reducing hospitalizations. However, breakthrough SARS-CoV-2 infections (BTI) occur in some vaccinated individuals. Here, we study how BTI impacts on the potency and the breadth of the neutralizing antibody response. We show that a Delta infection in COVID-19-vaccinated individuals provides potent neutralization against the current SARS-CoV-2 variants of concern, including the Omicron variant.
Collapse
Affiliation(s)
- Thomas Lechmere
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London, United Kingdom
| | - Luke B. Snell
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Carl Graham
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London, United Kingdom
| | - Jeffrey Seow
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London, United Kingdom
| | - Zayed A. Shalim
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London, United Kingdom
| | - Themoula Charalampous
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Adela Alcolea-Medina
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Rahul Batra
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Gaia Nebbia
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Jonathan D. Edgeworth
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Michael H. Malim
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London, United Kingdom
| | - Katie J. Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
107
|
Markewitz RDH, Juhl D, Pauli D, Görg S, Junker R, Rupp J, Engel S, Steinhagen K, Herbst V, Zapf D, Krüger C, Brockmann C, Leypoldt F, Dargvainiene J, Schomburg B, Sharifzadeh SR, Salek Nejad L, Wandinger KP, Ziemann M. Differences in Immunogenicity of Three Different Homo- and Heterologous Vaccination Regimens against SARS-CoV-2. Vaccines (Basel) 2022; 10:vaccines10050649. [PMID: 35632405 PMCID: PMC9145236 DOI: 10.3390/vaccines10050649] [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: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 12/10/2022] Open
Abstract
Background: Due to findings on adverse reactions and clinical efficacy of different vaccinations against SARS-CoV-2, the administration of vaccination regimens containing both adenoviral vector vaccines and mRNA-based vaccines has become common. Data are still needed on the direct comparison of immunogenicity for these different regimens. Methods: We compared markers for immunogenicity (anti-S1 IgG/IgA, neutralizing antibodies, and T-cell response) with three different vaccination regimens (homologous ChAdOx1 nCoV-19 (n = 103), or mixture of ChAdOx1 nCoV-19 with mRNA-1273 (n = 116) or BNT162b2 (n = 105)) at two time points: the day of the second vaccination as a baseline and 14 days later. Results: All examined vaccination regimens elicited measurable immune responses that were significantly enhanced after the second dose. Homologous ChAdOx1 nCoV-19 was markedly inferior in immunogenicity to all other examined regimens after administration of the second dose. Between the heterologous regimens, mRNA-1273 as second dose induced greater antibody responses than BNT162b2, with no difference found for neutralizing antibodies and T-cell response. Discussion: While these findings allow no prediction about clinical protection, from an immunological point of view, vaccination against SARS-CoV-2 with an mRNA-based vaccine at one or both time points appears preferable to homologous vaccination with ChAdOx1 nCoV-19. Whether or not the demonstrated differences between the heterologous regimens are of clinical significance will be subject to further research.
Collapse
Affiliation(s)
- Robert Daniel Heinrich Markewitz
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
- Correspondence: ; Tel.: +49-451-00-16315
| | - David Juhl
- Institute for Transfusion Medicine, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany; (D.J.); (S.G.); (C.B.); (M.Z.)
| | - Daniela Pauli
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Siegfried Görg
- Institute for Transfusion Medicine, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany; (D.J.); (S.G.); (C.B.); (M.Z.)
| | - Ralf Junker
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany;
| | - Sarah Engel
- Department of Anesthesiology and Intensive Care, University Hospital of Schleswig-Holstein Campus Lübeck, 23562 Lübeck, Germany;
| | - Katja Steinhagen
- Institute for Experimental Immunology, EUROIMMUN AG, 23560 Lübeck, Germany; (K.S.); (V.H.); (D.Z.); (C.K.)
| | - Victor Herbst
- Institute for Experimental Immunology, EUROIMMUN AG, 23560 Lübeck, Germany; (K.S.); (V.H.); (D.Z.); (C.K.)
| | - Dorinja Zapf
- Institute for Experimental Immunology, EUROIMMUN AG, 23560 Lübeck, Germany; (K.S.); (V.H.); (D.Z.); (C.K.)
| | - Christina Krüger
- Institute for Experimental Immunology, EUROIMMUN AG, 23560 Lübeck, Germany; (K.S.); (V.H.); (D.Z.); (C.K.)
| | - Christian Brockmann
- Institute for Transfusion Medicine, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany; (D.J.); (S.G.); (C.B.); (M.Z.)
| | - Frank Leypoldt
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Justina Dargvainiene
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Benjamin Schomburg
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Shahpour Reza Sharifzadeh
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Lukas Salek Nejad
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Arnold-Heller-Straße 3, 24105 Kiel, Germany; (D.P.); (R.J.); (F.L.); (J.D.); (B.S.); (S.R.S.); (L.S.N.); (K.-P.W.)
| | - Malte Ziemann
- Institute for Transfusion Medicine, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany; (D.J.); (S.G.); (C.B.); (M.Z.)
| |
Collapse
|
108
|
van den Hoogen LL, Verheul MK, Vos ERA, van Hagen CCE, van Boven M, Wong D, Wijmenga-Monsuur AJ, Smits G, Kuijer M, van Rooijen D, Bogaard-van Maurik M, Zutt I, van Vliet J, Wolf J, van der Klis FRM, de Melker HE, van Binnendijk RS, den Hartog G. SARS-CoV-2 Spike S1-specific IgG kinetic profiles following mRNA or vector-based vaccination in the general Dutch population show distinct kinetics. Sci Rep 2022; 12:5935. [PMID: 35396570 PMCID: PMC8990276 DOI: 10.1038/s41598-022-10020-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/30/2022] [Indexed: 12/21/2022] Open
Abstract
mRNA- and vector-based vaccines are used at a large scale to prevent COVID-19. We compared Spike S1-specific (S1) IgG antibodies after vaccination with mRNA-based (Comirnaty, Spikevax) or vector-based (Janssen, Vaxzevria) vaccines, using samples from a Dutch nationwide cohort. In adults 18-64 years old (n = 2412), the median vaccination interval between the two doses was 77 days for Vaxzevria (interquartile range, IQR: 69-77), 35 days (28-35) for Comirnaty and 33 days (28-35) for Spikevax. mRNA vaccines induced faster inclines and higher S1 antibodies compared to vector-based vaccines. For all vaccines, one dose resulted in boosting of S1 antibodies in adults with a history of SARS-CoV-2 infection. For Comirnaty, two to four months following the second dose (n = 196), S1 antibodies in adults aged 18-64 years old (436 BAU/mL, IQR: 328-891) were less variable and median concentrations higher compared to those in persons ≥ 80 years old (366, 177-743), but differences were not statistically significant (p > 0.100). Nearly all participants seroconverted following COVID-19 vaccination, including the aging population. These data confirm results from controlled vaccine trials in a general population, including vulnerable groups.
Collapse
Affiliation(s)
- Lotus L van den Hoogen
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Marije K Verheul
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Eric R A Vos
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Cheyenne C E van Hagen
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Michiel van Boven
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Denise Wong
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Alienke J Wijmenga-Monsuur
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Gaby Smits
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Marjan Kuijer
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Debbie van Rooijen
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Marjan Bogaard-van Maurik
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Ilse Zutt
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Jeffrey van Vliet
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Janine Wolf
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Fiona R M van der Klis
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Hester E de Melker
- Centre for Infectious Diseases, Epidemiology and Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Robert S van Binnendijk
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands
| | - Gerco den Hartog
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands.
| |
Collapse
|
109
|
Vicenti I, Gatti F, Scaggiante R, Boccuto A, Zago D, Basso M, Dragoni F, Parisi SG, Zazzi M. The second dose of the BNT162b2 mRNA vaccine does not boost SARS-CoV-2 neutralizing antibody response in previously infected subjects. Infection 2022; 50:541-543. [PMID: 34342854 PMCID: PMC8329626 DOI: 10.1007/s15010-021-01680-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Gatti
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121, Padova, Italy
| | | | - Adele Boccuto
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Daniela Zago
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121, Padova, Italy
| | - Monica Basso
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121, Padova, Italy
| | - Filippo Dragoni
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Saverio Giuseppe Parisi
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121, Padova, Italy.
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
110
|
Magicova M, Zahradka I, Fialova M, Neskudla T, Gurka J, Modos I, Hojny M, Raska P, Smejkal P, Striz I, Viklicky O. Determinants of Immune Response to Anti-SARS-CoV-2 mRNA Vaccines in Kidney Transplant Recipients: A Prospective Cohort Study. Transplantation 2022; 106:842-852. [PMID: 34999659 PMCID: PMC8942601 DOI: 10.1097/tp.0000000000004044] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination has been recently shown to be impaired in kidney transplant recipients (KTRs), but the underlying factors affecting vaccine effectiveness need to be further elucidated. METHODS In this prospective cohort study, antibodies against S1 and S2 subunits of SARS-CoV-2 were evaluated using an immunochemiluminescent assay (cutoff 9.5 AU/mL, sensitivity 91.2%, and specificity 90.2%) in 736 KTRs, who were previously either naive or infected with SARS-CoV-2 and vaccinated before or after transplantation. Cellular response was analyzed in a subset of patients using an interferon gamma release assay (cutoff 0.15 IU/mL, sensitivity 92%, and specificity 100%). RESULTS Seroconversion was significantly more impaired in SARS-CoV-2-naive KTRs than in those previously infected (40.1% versus 97.1%; P < 0.001). Mycophenolate use (odds ratio, 0.15; 95% confidence interval, 0.09-0.24; P < 0.001) and depleting therapy in the past year (odds ratio, 0.19; 95% confidence interval, 0.05-0.8; P = 0.023) were found to be among independent factors associated with impaired humoral response. Similarly, the interferon gamma release assay tested in 50 KTRs (cutoff 0.15 IU/mL, sensitivity 92%, specificity 100%) showed that specific T-cell responses against spike protein epitopes are impaired in SARS-CoV-2-naive KTRs, as compared to previously infected KTRs (9.4% versus 90%, P < 0.001). All 35 KTRs vaccinated on the waiting list before transplantation exhibited sustained seroconversion persisting after transplantation. CONCLUSIONS Survivors of coronavirus disease 2019 and those vaccinated while on the waiting list exhibited a marked immune response to mRNA vaccines, contrary to poor response in naive KTRs vaccinated after transplantation (NCT04832841).
Collapse
Affiliation(s)
- Maria Magicova
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ivan Zahradka
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martina Fialova
- Department of Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Tomas Neskudla
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jiri Gurka
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Istvan Modos
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michal Hojny
- Institutional Pharmacy, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Raska
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Smejkal
- Division of Hygiene and Epidemiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ilja Striz
- Department of Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ondrej Viklicky
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Transplantation Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
111
|
Wang RC, Murphy CE, Kornblith AE, Hohenstein NA, Carter CM, Wong AHK, Kurtz T, Kohn MA. SARS COV-2 anti-nucleocapsid and anti-spike antibodies in an emergency department healthcare worker cohort: September 2020 – April 2021. Am J Emerg Med 2022; 54:81-86. [PMID: 35144108 PMCID: PMC8808429 DOI: 10.1016/j.ajem.2022.01.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 01/06/2023] Open
Abstract
Background Emergency department (ED) workers have an increased seroprevalence of SARS-CoV-2 antibodies. However, breakthrough infections in ED workers have led to a reduced workforce within a strained healthcare system. By measuring levels of IgG antibodies to the SARS-CoV-2 nucleocapsid and spike antigens in ED workers, we determined the incidence of infection and described the course of antibody levels. We also measured the antibody response to vaccination and examined factors associated with immunogenicity. Methods We conducted a prospective cohort study of ED workers conducted at a single ED from September 2020–April 2021. IgG antibodies to the SARS-CoV-2 nucleocapsid antigen were measured at baseline, 3, and 6 months, and IgG antibodies to the SARS-CoV-2 spike antigen were measured at 6 months. Results At baseline, we found 5 out of 139 (3.6%) participants with prior infection. At 6 months, 4 of the 5 had antibody results below the test manufacturer's positivity threshold. We identified one incident case of SARS-COV-2 infection out of 130 seronegative participants (0.8%, 95% CI 0.02–4.2%). In 131 vaccinated participants (125 BNT162b2, 6 mRNA-1273), 131 tested positive for anti-spike antibodies. We identified predictors of anti-spike antibody levels: time since vaccination, prior COVID-19 infection, age, and vaccine type. Each additional week since vaccination was associated with an 11.1% decrease in anti-spike antibody levels. (95% CI 6.2–15.8%). Conclusion ED workers experienced a low incidence of SARS-CoV-2 infection and developed antibodies in response to vaccines and prior infection. Antibody levels decreased markedly with time since infection or vaccination.
Collapse
Affiliation(s)
- Ralph C Wang
- Department of Emergency Medicine, University of California, San Francisco, CA, United States of America.
| | - Charles E Murphy
- Department of Emergency Medicine, University of California, San Francisco, CA, United States of America.
| | - Aaron E Kornblith
- Department of Emergency Medicine and Department of Pediatrics, University of California, , San Francisco, CA, United States of America.
| | - Nicole A Hohenstein
- Department of Emergency Medicine, University of California, San Francisco, CA, United States of America.
| | - Cornelius M Carter
- Department of Emergency Medicine, University of California, San Francisco, CA, United States of America.
| | - Angela H K Wong
- Department of Emergency Medicine, University of California, San Francisco, CA, United States of America.
| | - Theodore Kurtz
- Department of Laboratory Medicine, University of California, , San Francisco, CA, United States of America.
| | - Michael A Kohn
- Department of Epidemiology and Biostatistics, University of California, , San Francisco, CA, United States of America.
| |
Collapse
|
112
|
Van Rompay KKA, Olstad KJ, Sammak RL, Dutra J, Watanabe JK, Usachenko JL, Immareddy R, Roh JW, Verma A, Shaan Lakshmanappa Y, Schmidt BA, Di Germanio C, Rizvi N, Liu H, Ma ZM, Stone M, Simmons G, Dumont LJ, Allen AM, Lockwood S, Pollard RE, Ramiro de Assis R, Yee JL, Nham PB, Ardeshir A, Deere JD, Jain A, Felgner PL, Coffey LL, Iyer SS, Hartigan-O’Connor DJ, Busch MP, Reader JR. Early post-infection treatment of SARS-CoV-2 infected macaques with human convalescent plasma with high neutralizing activity had no antiviral effects but moderately reduced lung inflammation. PLoS Pathog 2022; 18:e1009925. [PMID: 35443018 PMCID: PMC9060337 DOI: 10.1371/journal.ppat.1009925] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 05/02/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
Early in the SARS-CoV-2 pandemic, there was a high level of optimism based on observational studies and small controlled trials that treating hospitalized patients with convalescent plasma from COVID-19 survivors (CCP) would be an important immunotherapy. However, as more data from controlled trials became available, the results became disappointing, with at best moderate evidence of efficacy when CCP with high titers of neutralizing antibodies was used early in infection. To better understand the potential therapeutic efficacy of CCP, and to further validate SARS-CoV-2 infection of macaques as a reliable animal model for testing such strategies, we inoculated 12 adult rhesus macaques with SARS-CoV-2 by intratracheal and intranasal routes. One day later, 8 animals were infused with pooled human CCP with a high titer of neutralizing antibodies (RVPN NT50 value of 3,003), while 4 control animals received normal human plasma. Animals were monitored for 7 days. Animals treated with CCP had detectable but low levels of antiviral antibodies after infusion. In comparison to the control animals, CCP-treated animals had similar levels of viral RNA in upper and lower respiratory tract secretions, similar detection of viral RNA in lung tissues by in situ hybridization, but lower amounts of infectious virus in the lungs. CCP-treated animals had a moderate, but statistically significant reduction in interstitial pneumonia, as measured by comprehensive lung histology. Thus overall, therapeutic benefits of CCP were marginal and inferior to results obtained earlier with monoclonal antibodies in this animal model. By highlighting strengths and weaknesses, data of this study can help to further optimize nonhuman primate models to provide proof-of-concept of intervention strategies, and guide the future use of convalescent plasma against SARS-CoV-2 and potentially other newly emerging respiratory viruses.
Collapse
Affiliation(s)
- Koen K. A. Van Rompay
- California National Primate Research Center, University of California, Davis, California, United States of America
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California, United States of America
| | - Katherine J. Olstad
- California National Primate Research Center, University of California, Davis, California, United States of America
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California, United States of America
| | - Rebecca L. Sammak
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Joseph Dutra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California, United States of America
| | - Jennifer K. Watanabe
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Jodie L. Usachenko
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Ramya Immareddy
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Jamin W. Roh
- Center for Immunology and Infectious Diseases, University of California, Davis, California, United States of America
- Graduate Group in Immunology, University of California, Davis, California, United States of America
| | - Anil Verma
- Center for Immunology and Infectious Diseases, University of California, Davis, California, United States of America
| | - Yashavanth Shaan Lakshmanappa
- Center for Immunology and Infectious Diseases, University of California, Davis, California, United States of America
| | - Brian A. Schmidt
- Center for Immunology and Infectious Diseases, University of California, Davis, California, United States of America
| | - Clara Di Germanio
- Vitalant Research Institute, San Francisco, California, United States of America
| | - Nabeela Rizvi
- Vitalant Research Institute, San Francisco, California, United States of America
| | - Hongwei Liu
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California, United States of America
| | - Zhong-Min Ma
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Mars Stone
- Vitalant Research Institute, San Francisco, California, United States of America
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, California, United States of America
| | - Larry J. Dumont
- Vitalant Research Institute, Denver, Colorado; University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - A. Mark Allen
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Sarah Lockwood
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Rachel E. Pollard
- School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Rafael Ramiro de Assis
- Vaccine Research and Development Center, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California, United States of America
| | - JoAnn L. Yee
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Peter B. Nham
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - Jesse D. Deere
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California, United States of America
| | - Aarti Jain
- Vaccine Research and Development Center, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California, United States of America
| | - Philip L. Felgner
- Vaccine Research and Development Center, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California, United States of America
| | - Lark L. Coffey
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California, United States of America
| | - Smita S. Iyer
- California National Primate Research Center, University of California, Davis, California, United States of America
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California, United States of America
- Center for Immunology and Infectious Diseases, University of California, Davis, California, United States of America
| | - Dennis J. Hartigan-O’Connor
- California National Primate Research Center, University of California, Davis, California, United States of America
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California, United States of America
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, California, United States of America
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - J. Rachel Reader
- California National Primate Research Center, University of California, Davis, California, United States of America
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California, United States of America
| |
Collapse
|
113
|
Modeling of waning immunity after SARS-CoV-2 vaccination and influencing factors. Nat Commun 2022; 13:1614. [PMID: 35347129 PMCID: PMC8960902 DOI: 10.1038/s41467-022-29225-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 03/04/2022] [Indexed: 11/25/2022] Open
Abstract
SARS-CoV-2 vaccines are crucial in controlling COVID-19, but knowledge of which factors determine waning immunity is limited. We examined antibody levels and T-cell gamma-interferon release after two doses of BNT162b2 vaccine or a combination of ChAdOx1-nCoV19 and BNT162b2 vaccines for up to 230 days after the first dose. Generalized mixed models with and without natural cubic splines were used to determine immunity over time. Antibody responses were influenced by natural infection, sex, and age. IgA only became significant in naturally infected. A one-year IgG projection suggested an initial two-phase response in those given the second dose delayed (ChAdOx1/BNT162b2) followed by a more rapid decrease of antibody levels. T-cell responses correlated significantly with IgG antibody responses. Our results indicate that IgG levels will drop at different rates depending on prior infection, age, sex, T-cell response, and the interval between vaccine injections. Only natural infection mounted a significant and lasting IgA response. This study investigates the dynamics of immunological markers after first SARS-CoV-2 vaccination dose in cohort of healthcare professionals in Denmark. Natural infection was associated with higher antibody responses, and IgG decline varied by age, sex, T-cell response, previous infection, and interval between vaccine doses.
Collapse
|
114
|
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: 6.5] [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
|
115
|
Ganczak M, Korzeń M, Sobieraj E, Goławski J, Pasek O, Biesiada D. COVID-19 Vaccination within the Context of Reactogenicity and Immunogenicity of ChAdOx1 Vaccine Administered to Teachers in Poland. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:3111. [PMID: 35270806 PMCID: PMC8910564 DOI: 10.3390/ijerph19053111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023]
Abstract
In February 2021, Polish teachers were offered the ChAdOx1-S vaccine as a priority group. However, there have been concerns among educators regarding the efficacy of this vaccine, as compared to the other types of vaccines (e.g., mRNA). The objective of this study was to investigate the reactogenicity and the immunogenicity of this vaccine. Participants, specifically teachers, were invited for serological testing ≥ 4 weeks post-vaccination. Antibodies against the receptor-binding domain (RBD) were measured. Of the 192 participants, the mean age was 50.5 ± 8.3 years and the mean (range) dosing interval was 69.6 ± (25−111) days. Adverse reactions included feeling feverish (44.8%), headache (41.7%), malaise/chills (38.0%), and injection-site tenderness (37.5%); these were reported more frequently after the first dose (84.9%). Fewer males than females (54.8% vs. 80.1%) and fewer older participants (65.7% vs. 90.4%) reported side effects (p < 0.002; p < 0.0001, respectively). All participants presented detectable anti-RBD IgG; the median (range) reading was 525.0 BAU/mL (20.6−5680.0); 1008.02 BAU/mL (115.3−5680.0) in those with prior SARS-CoV-2 infection; and 381.42 BAU/mL (20.6−3108.8) in those without (p = 0.001). In 27.6%, the anti-RBD IgG level was >500 BAU/mL. A multivariate logistic regression revealed that previous infection and longer dose intervals were predictors of higher immunologic responses (p < 0.0001; p = 0.01, respectively). The results demonstrated good tolerability and immunogenicity of the ChAdOx1-S vaccine. Our study justified the longer dose interval to enhance a higher antibody response. Our findings may also support the prioritization of uninfected individuals in regions where COVID-19 vaccine-sparing strategies are required.
Collapse
Affiliation(s)
- Maria Ganczak
- Department of Infectious Diseases, University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Marcin Korzeń
- Department of Methods of Artificial Intelligence and Applied Mathematics, West Pomeranian Institute of Technology, 71-210 Szczecin, Poland;
| | - Ewa Sobieraj
- Student Research Group, University of Zielona Gora, 65-417 Zielona Gora, Poland; (E.S.); (J.G.); (O.P.)
| | - Jakub Goławski
- Student Research Group, University of Zielona Gora, 65-417 Zielona Gora, Poland; (E.S.); (J.G.); (O.P.)
| | - Oskar Pasek
- Student Research Group, University of Zielona Gora, 65-417 Zielona Gora, Poland; (E.S.); (J.G.); (O.P.)
| | | |
Collapse
|
116
|
Walls AC, Sprouse KR, Bowen JE, Joshi A, Franko N, Navarro MJ, Stewart C, Cameroni E, McCallum M, Goecker EA, Degli-Angeli EJ, Logue J, Greninger A, Corti D, Chu HY, Veesler D. SARS-CoV-2 breakthrough infections elicit potent, broad, and durable neutralizing antibody responses. Cell 2022; 185:872-880.e3. [PMID: 35123650 PMCID: PMC8769922 DOI: 10.1016/j.cell.2022.01.011] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/04/2022] [Accepted: 01/14/2022] [Indexed: 11/27/2022]
Abstract
Although infections among vaccinated individuals lead to milder COVID-19 symptoms relative to those in unvaccinated subjects, the specificity and durability of antibody responses elicited by breakthrough cases remain unknown. Here, we demonstrate that breakthrough infections induce serum-binding and -neutralizing antibody responses that are markedly more potent, durable, and resilient to spike mutations observed in variants than those in subjects who received only 2 doses of vaccine. However, we show that breakthrough cases, subjects who were vaccinated after infection, and individuals vaccinated three times have serum-neutralizing activity of comparable magnitude and breadth, indicating that an increased number of exposures to SARS-CoV-2 antigen(s) enhance the quality of antibody responses. Neutralization of SARS-CoV was moderate, however, underscoring the importance of developing vaccines eliciting broad sarbecovirus immunity for pandemic preparedness.
Collapse
Affiliation(s)
- Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| | - Kaitlin R Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - John E Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Anshu Joshi
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Nicholas Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Mary Jane Navarro
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Elisabetta Cameroni
- Humabs Biomed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Erin A Goecker
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Emily J Degli-Angeli
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jenni Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Alex Greninger
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Davide Corti
- Humabs Biomed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Helen Y Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
117
|
McDade TW, Sancilio A, D’Aquila R, Mustanski B, Vaught LA, Reiser NL, Velez ME, Hsieh RR, Ryan DT, Saber R, McNally EM, Demonbreun AR. Low Levels of Neutralizing Antibodies After Natural Infection With Severe Acute Respiratory Syndrome Coronavirus 2 in a Community-Based Serological Study. Open Forum Infect Dis 2022; 9:ofac055. [PMID: 35252468 PMCID: PMC8890497 DOI: 10.1093/ofid/ofac055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Confidence in natural immunity after infection with severe acute respiratory syndrome coronavirus 2 is one reason for vaccine hesitancy. METHODS We measured antibody-mediated neutralization of spike protein-ACE2 receptor binding in a large community-based sample of seropositive individuals who differed in severity of infection (N = 790). RESULTS A total of 39.8% of infections were asymptomatic, 46.5% were symptomatic with no clinical care, 13.8% were symptomatic with clinical care, and 3.7% required hospitalization. Moderate/high neutralizing activity was present after 41.3% of clinically managed infections, in comparison with 7.9% of symptomatic and 1.9% of asymptomatic infections. CONCLUSIONS Prior coronavirus disease 2019 infection does not guarantee a high level of antibody-mediated protection against reinfection in the general population.
Collapse
Affiliation(s)
- Thomas W McDade
- Department of Anthropology, Northwestern University, Evanston, IllinoisUSA,Institute for Policy Research, Northwestern University, Evanston, Illinois, USA,Correspondence: Thomas McDade, PhD, Northwestern University, 1810 Hinman Avenue, Evanston, IL 60208, USA ()
| | - Amelia Sancilio
- Department of Anthropology, Northwestern University, Evanston, IllinoisUSA,Institute for Policy Research, Northwestern University, Evanston, Illinois, USA
| | - Richard D’Aquila
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Brian Mustanski
- Institute for Sexual and Gender Minority Health and Wellbeing, Northwestern University, Chicago, Illinois, USA,Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lauren A Vaught
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nina L Reiser
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew E Velez
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ryan R Hsieh
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel T Ryan
- Institute for Sexual and Gender Minority Health and Wellbeing, Northwestern University, Chicago, Illinois, USA,Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rana Saber
- Institute for Sexual and Gender Minority Health and Wellbeing, Northwestern University, Chicago, Illinois, USA,Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
118
|
Šošić L, Paolucci M, Duda A, Hasler F, Walton SM, Kündig TM, Johansen P. Kinetics and persistence of anti-SARS-CoV-2 neutralisation and antibodies after BNT162b2 vaccination in a Swiss cohort. Immun Inflamm Dis 2022; 10:e583. [PMID: 34965032 PMCID: PMC8926495 DOI: 10.1002/iid3.583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/25/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
Introduction Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), substantial effort has been made to gain knowledge about the immunity elicited by infection or vaccination. Methods We studied the kinetics of antibodies and virus neutralisation induced by vaccination with BNT162b2 in a Swiss cohort of SARS‐CoV‐2 naïve (n = 40) and convalescent (n = 9) persons. Blood sera were analysed in a live virus neutralisation assay and specific IgG and IgA levels were measured by enzyme‐linked immunoassay and analysed by descriptive statistics. Results Virus neutralisation was detected in all individuals 2–4 weeks after the second vaccine. Both neutralisation and antibodies remained positive for >4 months. Neutralisation and antibodies showed positive correlation, but immunoglobulin G (IgG) and immunoglobulin A (IgA) seroconversion took place 2–4 weeks faster than neutralisation. Spike‐protein specific IgG levels rose significantly faster and were more stable over time than virus neutralisation titres or IgA responses. For naïve but not convalescent persons, a clear boosting effect was observed. Convalescent individuals showed faster, more robust and longer‐lasting immune responses after vaccination compared to noninfected persons. No threshold could be determined for spike protein‐specific IgG or IgA that would confer protection in the neutralisation assay, implicating the need for a better correlate of protection then antibody titres alone. Conclusions This study clearly shows the complex translation of antibody data and virus neutralisation, while supporting the evidence of a single dose being sufficient for effective antibody response in convalescent individuals.
Collapse
Affiliation(s)
- Lara Šošić
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - Marta Paolucci
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - Agathe Duda
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Fabio Hasler
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - Senta M Walton
- Department of Dermatology, University of Zurich, Zurich, Switzerland.,Research & Development, Saiba Biotech, Pfaeffikon, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University of Zurich, Zurich, Switzerland.,Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Pål Johansen
- Department of Dermatology, University of Zurich, Zurich, Switzerland.,Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
119
|
Humoral and Cellular Responses to COVID-19 Vaccines in SARS-CoV-2 Infection-Naïve and -Recovered Korean Individuals. Vaccines (Basel) 2022; 10:vaccines10020332. [PMID: 35214791 PMCID: PMC8878120 DOI: 10.3390/vaccines10020332] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
In the face of a global COVID-19 vaccine shortage, an efficient vaccination strategy is required. Therefore, the immunogenicity of single or double COVID-19 vaccination doses (ChAdOX1, BNT162b2, or mRNA-1273) of SARS-CoV-2-recovered individuals was compared to that of unvaccinated individuals with SARS-CoV-2 infection at least one year post-convalescence. Moreover, the immunogenicity of SARS-CoV-2-naïve individuals vaccinated with a complete schedule of Ad26.CoV2.S, ChAdOX1, BNT162b2, mRNA-1273, or ChAdOX1/BNT162b2 vaccines was evaluated. Anti-SARS-CoV-2 S1 IgG antibody (S1-IgG), pseudotyped virus-neutralizing antibody titer (pVNT50), and IFN-γ ELISpot counts were measured. Humoral immune responses were significantly higher in vaccinated than in unvaccinated recovered individuals, with a 43-fold increase in the mean pVNT50 values. However, there was no significant difference in the pVNT50 and IFN-γ ELISpot values between the single- and double-dose regimens. In SARS-CoV-2-naïve individuals, antibody responses varied according to the vaccine type: BNT162b2 and mRNA-1273 induced similar levels of S1-IgG to those observed in vaccinated, convalescent individuals; in contrast, pVNT50 was much lower in SARS-CoV-2-naïve vaccinees than in vaccinated recovered individuals. Therefore, a single dose of ChAdOX1, BNT162b2, or mRNA-1273 vaccines would be a good alternative for recovered individuals instead of a double-dose regimen.
Collapse
|
120
|
Azzolini E, Pozzi C, Germagnoli L, Oresta B, Carriglio N, Calleri M, Selmi C, De Santis M, Finazzi S, Carlo-Stella C, Bertuzzi A, Motta F, Ceribelli A, Mantovani A, Bonelli F, Rescigno M. mRNA COVID-19 vaccine booster fosters B- and T-cell responses in immunocompromised patients. Life Sci Alliance 2022; 5:5/6/e202201381. [PMID: 35169017 PMCID: PMC8860093 DOI: 10.26508/lsa.202201381] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/20/2022] Open
Abstract
Immunocompromised patients poorly respond to two doses of SARS-CoV-2 mRNA vaccines. However, an additional booster dose elicits a strong humoral and cellular immune response in these subjects. SARS-CoV-2 vaccination has proven effective in inducing an immune response in healthy individuals and is progressively us allowing to overcome the pandemic. Recent evidence has shown that response to vaccination in some vulnerable patients may be diminished, and it has been proposed a booster dose. We tested the kinetic of development of serum antibodies to the SARS-CoV-2 Spike protein, their neutralizing capacity, the CD4 and CD8 IFN-γ T-cell response in 328 subjects, including 131 immunocompromised individuals (cancer, rheumatologic, and hemodialysis patients), 160 health-care workers (HCW) and 37 subjects older than 75 yr, after vaccination with two or three doses of mRNA vaccines. We stratified the patients according to the type of treatment. We found that immunocompromised patients, depending on the type of treatment, poorly respond to SARS-CoV-2 mRNA vaccines. However, an additional booster dose of vaccine induced a good immune response in almost all of the patients except those receiving anti-CD20 antibody. Similarly to HCW, previously infected and vaccinated immunocompromised individuals demonstrate a stronger SARS-CoV-2–specific immune response than those who are vaccinated without prior infection.
Collapse
Affiliation(s)
- Elena Azzolini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Chiara Pozzi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Luca Germagnoli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | | | | | | | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Maria De Santis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Silvia Finazzi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Alexia Bertuzzi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Francesca Motta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Angela Ceribelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy.,William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Maria Rescigno
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele MI, Italy .,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano MI, Italy
| |
Collapse
|
121
|
Angel-Korman A, Peres E, Bryk G, Lustig Y, Indenbaum V, Amit S, Rappoport V, Katzir Z, Yagil Y, Iaina NL, Leiba A, Brosh-Nissimov T. Diminished and waning immunity to COVID-19 vaccination among hemodialysis patients in Israel: the case for a third vaccine dose. Clin Kidney J 2022; 15:226-234. [PMID: 35140934 PMCID: PMC8524605 DOI: 10.1093/ckj/sfab206] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Indexed: 01/07/2023] Open
Abstract
Background Humoral responses to coronavirus disease 2019 (COVID-19) vaccines in hemodialysis (HD) patients can direct vaccination policy. Methods We compared 409 COVID-19-naïve HD patients from 13 HD units in Israel to 148 non-dialysis-dependent COVID-19-naïve controls. Twenty-four previously infected (antinucleocapsid positive) HD patients were analysed separately. Blood samples were obtained ≥14 days post-vaccination (BNT162b2, Pfizer/BioNTech) to assess seroconversion rates and titers of anti-spike (anti-S) and neutralizing antibodies. Results The median time from vaccination to blood sample collection was 82 days [interquartile range (IAR) 64-87] and 89 days (IQR 68-96) for HD patients and controls, respectively. Seroconversion rates were lower in HD patients compared with controls for both anti-S and neutralizing antibodies (89% and 77% versus 99.3%, respectively; P < 0.0001). Antibody titers were also significantly lower in HD patients compared with controls {median 69.6 [IQR 33.2-120] versus 196.5 [IQR 118.5-246], P < 0.0001; geometric mean titer [GMT] 23.3 [95% confidence interval (CI) 18.7-29.1] versus 222.7 [95% CI 174-284], P < 0.0001, for anti-S and neutralizing antibodies, respectively}. Multivariate analysis demonstrated dialysis dependence to be strongly associated with lower antibody responses and antibody titers waning with time. Age, low serum albumin and low lymphocyte count were also associated with lower seroconversion rates and antibody titers. HD patients previously infected with sudden acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had no difference in their seroconversion rates or antibody titers compared with COVID-19-naïve patients. Conclusion This study demonstrates diminished and waning humoral responses following COVID-19 vaccination in a large and diverse cohort of HD patients, including those previously infected with SARS-CoV-2. Considering these results and reduced vaccine effectiveness against variants of concern, in addition to continued social distancing precautions, a third booster dose should be considered in this population.
Collapse
Affiliation(s)
- Avital Angel-Korman
- Nephrology and Hypertension Institute, Samson Assuta Ashdod University Hospital, Ashdod, Israel
| | - Esther Peres
- Nephrology and Hypertension Institute, Samson Assuta Ashdod University Hospital, Ashdod, Israel
| | - Gabriel Bryk
- Laboratory Division, Assuta Ashdod University Hospital, Ashdod, Israel
| | - Yaniv Lustig
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer, Israel
| | - Victoria Indenbaum
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer, Israel
| | - Sharon Amit
- Clinical Microbiology, Sheba Medical Center, Tel-Hashomer, Israel
| | - Vladimir Rappoport
- Nephrology and Hypertension Institute, Samson Assuta Ashdod University Hospital, Ashdod, Israel
| | - Zeev Katzir
- Nephrology and Hypertension Institute, Samson Assuta Ashdod University Hospital, Ashdod, Israel
| | - Yoram Yagil
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheba, Israel
| | - Nomy Levin Iaina
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheba, Israel
| | - Adi Leiba
- Nephrology and Hypertension Institute, Samson Assuta Ashdod University Hospital, Ashdod, Israel
| | - Tal Brosh-Nissimov
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
122
|
Uprichard SL, O’Brien A, Evdokimova M, Rowe CL, Joyce C, Hackbart M, Cruz-Pulido YE, Cohen CA, Rock ML, Dye JM, Kuehnert P, Ricks KM, Casper M, Linhart L, Anderson K, Kirk L, Maggiore JA, Herbert AS, Clark NM, Reid GE, Baker SC. Antibody Response to SARS-CoV-2 Infection and Vaccination in COVID-19-naïve and Experienced Individuals. Viruses 2022; 14:370. [PMID: 35215962 PMCID: PMC8878640 DOI: 10.3390/v14020370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 11/25/2022] Open
Abstract
Understanding the magnitude of responses to vaccination during the ongoing SARS-CoV-2 pandemic is essential for ultimate mitigation of the disease. Here, we describe a cohort of 102 subjects (70 COVID-19-naïve, 32 COVID-19-experienced) who received two doses of one of the mRNA vaccines (BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna)). We document that a single exposure to antigen via infection or vaccination induces a variable antibody response which is affected by age, gender, race, and co-morbidities. In response to a second antigen dose, both COVID-19-naïve and experienced subjects exhibited elevated levels of anti-spike and SARS-CoV-2 neutralizing activity; however, COVID-19-experienced individuals achieved higher antibody levels and neutralization activity as a group. The COVID-19-experienced subjects exhibited no significant increase in antibody or neutralization titer in response to the second vaccine dose (i.e., third antigen exposure). Finally, we found that COVID-19-naïve individuals who received the Moderna vaccine exhibited a more robust boost response to the second vaccine dose (p = 0.004) as compared to the response to Pfizer-BioNTech. Ongoing studies with this cohort will continue to contribute to our understanding of the range and durability of responses to SARS-CoV-2 mRNA vaccines.
Collapse
Affiliation(s)
- Susan L. Uprichard
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Amornrat O’Brien
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Monika Evdokimova
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Cynthia L. Rowe
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Cara Joyce
- Department of Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL 60153, USA;
| | - Matthew Hackbart
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Yazmin E. Cruz-Pulido
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Courtney A. Cohen
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
- The Geneva Foundation, Tacoma, WA 98042, USA
| | - Michelle L. Rock
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
- The Geneva Foundation, Tacoma, WA 98042, USA
| | - John M. Dye
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
| | - Paul Kuehnert
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD 21702, USA; (P.K.); (K.M.R.)
| | - Keersten M. Ricks
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD 21702, USA; (P.K.); (K.M.R.)
| | - Marybeth Casper
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Lori Linhart
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Katrina Anderson
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Laura Kirk
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Jack A. Maggiore
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
| | - Andrew S. Herbert
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
| | - Nina M. Clark
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Gail E. Reid
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Susan C. Baker
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
123
|
He WT, Musharrafieh R, Song G, Dueker K, Tse LV, Martinez DR, Schäfer A, Callaghan S, Yong P, Beutler N, Torres JL, Volk RM, Zhou P, Yuan M, Liu H, Anzanello F, Capozzola T, Parren M, Garcia E, Rawlings SA, Smith DM, Wilson IA, Safonova Y, Ward AB, Rogers TF, Baric RS, Gralinski LE, Burton DR, Andrabi R. Targeted isolation of panels of diverse human protective broadly neutralizing antibodies against SARS-like viruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.09.08.459480. [PMID: 35169804 PMCID: PMC8845431 DOI: 10.1101/2021.09.08.459480] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The emergence of current SARS-CoV-2 variants of concern (VOCs) and potential future spillovers of SARS-like coronaviruses into humans pose a major threat to human health and the global economy 1-7 . Development of broadly effective coronavirus vaccines that can mitigate these threats is needed 8, 9 . Notably, several recent studies have revealed that vaccination of recovered COVID-19 donors results in enhanced nAb responses compared to SARS-CoV-2 infection or vaccination alone 10-13 . Here, we utilized a targeted donor selection strategy to isolate a large panel of broadly neutralizing antibodies (bnAbs) to sarbecoviruses from two such donors. Many of the bnAbs are remarkably effective in neutralization against sarbecoviruses that use ACE2 for viral entry and a substantial fraction also show notable binding to non-ACE2-using sarbecoviruses. The bnAbs are equally effective against most SARS-CoV-2 VOCs and many neutralize the Omicron variant. Neutralization breadth is achieved by bnAb binding to epitopes on a relatively conserved face of the receptor binding domain (RBD) as opposed to strain-specific nAbs to the receptor binding site that are commonly elicited in SARS-CoV-2 infection and vaccination 14-18 . Consistent with targeting of conserved sites, select RBD bnAbs exhibited in vivo protective efficacy against diverse SARS-like coronaviruses in a prophylaxis challenge model. The generation of a large panel of potent bnAbs provides new opportunities and choices for next-generation antibody prophylactic and therapeutic applications and, importantly, provides a molecular basis for effective design of pan-sarbecovirus vaccines.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Reid M. Volk
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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 9203
| | - Yana Safonova
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Andrew B. Ward
- 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
| | - 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
|
124
|
Chahla RE, Tomas-Grau RH, Cazorla SI, Ploper D, Vera Pingitore E, López MA, Aznar P, Alcorta ME, Vélez EMDM, Stagnetto A, Ávila CL, Maldonado-Galdeano C, Socias SB, Heinze D, Navarro SA, Llapur CJ, Costa D, Flores I, Edelstein A, Kowdle S, Perandones C, Lee B, Apfelbaum G, Mostoslavsky R, Mostoslavsky G, Perdigón G, Chehín RN. Long-term analysis of antibodies elicited by SPUTNIK V: A prospective cohort study in Tucumán, Argentina. LANCET REGIONAL HEALTH. AMERICAS 2022; 6:100123. [PMID: 34841388 PMCID: PMC8604626 DOI: 10.1016/j.lana.2021.100123] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gam-COVID-Vac (SPUTNIK V) has been granted emergency use authorization in 70 nations and has been administered to millions worldwide. However, there are very few peer-reviewed studies describing its effects. Independent reports regarding safety and effectiveness could accelerate the final approval by the WHO. We aimed to study the long-term humoral immune response in naïve and previously infected volunteers who received SPUTNIK V. METHODS Humoral immune responses, assayed by anti-SARS-CoV-2-spike-RBD IgG ELISA and neutralization assays, were measured in 602 healthcare workers at 0, 14, 28, 60 and 180 days after receiving SPUTNIK V between December 2020 and July 2021 in Tucumán, Argentina. FINDINGS Seroconversion was detected in 97% of individuals after 28 days post-vaccination (dpv) (N = 405). Anti-RBD titers began to decrease after 60 dpv (N = 328), but remained detectable in 94% at 90 dpv (N = 224). At 180 dpv, anti-RDB titers persisted in 31% (N = 146). Previous infection triggered an increased immune response to the first dose and increased neutralization activity against variants of concern (VOC). Second doses in previously infected individuals further increased titers, even 90 dpv (N = 75). Basal antibody titers had more influence on post-vaccination anti-RBD responses than the time elapsed between diagnosis and vaccination (N = 274). INTERPRETATION Data presented herein provides essential knowledge regarding the kinetics of antibodies induced by SPUTNIK V up to six months after immunization, and suggests that when considering one-dose vaccination policies for individuals with previous SARS-CoV-2 infection, serological studies to determine basal titers may be important, independent of when diagnosis occurred. FUNDING Tucumán Public Health System (SIPROSA), Argentinean National Research Council (CONICET), National University of Tucumán (UNT).
Collapse
Affiliation(s)
| | - Rodrigo Hernán Tomas-Grau
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| | | | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| | - Esteban Vera Pingitore
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| | | | - Patricia Aznar
- Laboratorio de Salud Pública (LSP-SIPROSA). Tucumán, Argentina
| | | | | | - Agustín Stagnetto
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| | - César Luís Ávila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| | | | - Sergio Benjamín Socias
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| | - Dar Heinze
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine (CReM), Boston University School of Medicine. Boston, MA, United States
| | - Silvia Adriana Navarro
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| | | | - Dardo Costa
- Laboratorio de Salud Pública (LSP-SIPROSA). Tucumán, Argentina
| | - Isolina Flores
- Laboratorio de Salud Pública (LSP-SIPROSA). Tucumán, Argentina
| | - Alexis Edelstein
- Administración Nacional de Laboratorios e Institutos de Salud (ANLIS), Dr. Carlos G. Malbrán. Buenos Aires, Argentina
| | - Shreyas Kowdle
- Department of Microbiology at the Icahn School of Medicine at Mount Sinai. New York, NY, United States
| | - Claudia Perandones
- Administración Nacional de Laboratorios e Institutos de Salud (ANLIS), Dr. Carlos G. Malbrán. Buenos Aires, Argentina
| | - Benhur Lee
- Department of Microbiology at the Icahn School of Medicine at Mount Sinai. New York, NY, United States
| | - Gabriela Apfelbaum
- Facultad de Medicina, Universidad Nacional de Tucumán (UNT). Tucumán, Argentina
| | - Raúl Mostoslavsky
- The Massachusetts General Hospital Cancer Center, Harvard Medical School. Boston, MA, United States
| | - Gustavo Mostoslavsky
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine (CReM), Boston University School of Medicine. Boston, MA, United States
| | - Gabriela Perdigón
- Centro de Referencia para Lactobacilos-CERELA (CONICET). Tucumán, Argentina
| | - Rosana Nieves Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada IMMCA (UNT-CONICET-SIPROSA). Tucumán, Argentina
| |
Collapse
|
125
|
Bruxvoort KJ, Sy LS, Qian L, Ackerson BK, Luo Y, Lee GS, Tian Y, Florea A, Takhar HS, Tubert JE, Talarico CA, Tseng HF. Real-world effectiveness of the mRNA-1273 vaccine against COVID-19: Interim results from a prospective observational cohort study. LANCET REGIONAL HEALTH. AMERICAS 2022; 6:100134. [PMID: 34849505 PMCID: PMC8614600 DOI: 10.1016/j.lana.2021.100134] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Phase 3 trials found mRNA-1273 was highly effective in preventing COVID-19. We conducted a prospective cohort study at Kaiser Permanente Southern California (KPSC) to determine the real-world vaccine effectiveness (VE) of mRNA-1273 in preventing COVID-19 infection and severe disease. Methods For this planned interim analysis, individuals aged ≥18 years receiving 2 doses of mRNA-1273 ≥24 days apart (18/12/2020-31/03/2021) were 1:1 matched to randomly selected unvaccinated individuals by age, sex, and race/ethnicity, with follow-up through 30/06/2021. Outcomes were COVID-19 infection (SARS-CoV-2 positive molecular test or COVID-19 diagnosis code) or severe disease (COVID-19 hospitalization and COVID-19 hospital death). Adjusted hazard ratios (aHR) and confidence intervals (CI) for COVID-19 outcomes comparing vaccinated and unvaccinated individuals were estimated by Cox proportional hazards models accounting for multiple comparisons. Adjusted VE was calculated as (1-aHR)x100. Whole genome sequencing was performed on SARS-CoV-2 positive specimens from the KPSC population. Findings This analysis included 352,878 recipients of 2 doses of mRNA-1273 matched to 352,878 unvaccinated individuals. VE (99·3% CI) against COVID-19 infection was 87·4% (84·8–89·6%). VE against COVID-19 hospitalization and hospital death was 95·8% (90·7–98·1%) and 97·9% (66·9-99·9%), respectively. VE was higher against symptomatic (88·3% [98·3% CI: 86·1–90·2%]) than asymptomatic COVID-19 (72·7% [53·4–84·0%]), but was generally similar across age, sex, and racial/ethnic subgroups. VE among individuals with history of COVID-19 ranged from 8·2–33·6%. The most prevalent variants were Alpha (41·6%), Epsilon (17·5%), Delta (11·5%), and Gamma (9·1%), with Delta increasing to 54·0% of variants by June 2021. Interpretation These interim results provide reassuring evidence of the VE of 2 doses of mRNA-1273 across age, sex, and racial/ethnic subgroups, and against asymptomatic and symptomatic COVID-19, and severe COVID-19 outcomes. Among individuals with history of COVID-19, mRNA-1273 vaccination may offer added protection beyond immunity acquired from prior infection. Longer follow-up is needed to fully evaluate VE of mRNA-1273 against emerging SARS-CoV-2 variants. Funding Moderna Inc.
Collapse
Affiliation(s)
- Katia J Bruxvoort
- Kaiser Permanente Southern California, Pasadena, CA, USA.,University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lina S Sy
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Lei Qian
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | - Yi Luo
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Gina S Lee
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Yun Tian
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Ana Florea
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | - Julia E Tubert
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | - Hung Fu Tseng
- Kaiser Permanente Southern California, Pasadena, CA, USA.,Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| |
Collapse
|
126
|
Fiolet T, Kherabi Y, MacDonald CJ, Ghosn J, Peiffer-Smadja N. Comparing COVID-19 vaccines for their characteristics, efficacy and effectiveness against SARS-CoV-2 and variants of concern: a narrative review. Clin Microbiol Infect 2022; 28:202-221. [PMID: 34715347 PMCID: PMC8548286 DOI: 10.1016/j.cmi.2021.10.005] [Citation(s) in RCA: 540] [Impact Index Per Article: 270.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/07/2021] [Accepted: 10/16/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Vaccines are critical cost-effective tools to control the coronavirus disease 2019 (COVID-19) pandemic. However, the emergence of variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may threaten the global impact of mass vaccination campaigns. AIMS The objective of this study was to provide an up-to-date comparative analysis of the characteristics, adverse events, efficacy, effectiveness and impact of the variants of concern for 19 COVID-19 vaccines. SOURCES References for this review were identified through searches of PubMed, Google Scholar, BioRxiv, MedRxiv, regulatory drug agencies and pharmaceutical companies' websites up to 22nd September 2021. CONTENT Overall, all COVID-19 vaccines had a high efficacy against the original strain and the variants of concern, and were well tolerated. BNT162b2, mRNA-1273 and Sputnik V after two doses had the highest efficacy (>90%) in preventing symptomatic cases in phase III trials. mRNA vaccines, AZD1222, and CoronaVac were effective in preventing symptomatic COVID-19 and severe infections against Alpha, Beta, Gamma or Delta variants. Regarding observational real-life data, full immunization with mRNA vaccines and AZD1222 seems to effectively prevent SARS-CoV-2 infection against the original strain and Alpha and Beta variants but with reduced effectiveness against the Delta strain. A decline in infection protection was observed at 6 months for BNT162b2 and AZD1222. Serious adverse event rates were rare for mRNA vaccines-anaphylaxis 2.5-4.7 cases per million doses, myocarditis 3.5 cases per million doses-and were similarly rare for all other vaccines. Prices for the different vaccines varied from $2.15 to $29.75 per dose. IMPLICATIONS All vaccines appear to be safe and effective tools to prevent severe COVID-19, hospitalization, and death against all variants of concern, but the quality of evidence greatly varies depending on the vaccines considered. Questions remain regarding a booster dose and waning immunity, the duration of immunity, and heterologous vaccination. The benefits of COVID-19 vaccination outweigh the risks, despite rare serious adverse effects.
Collapse
Affiliation(s)
- Thibault Fiolet
- Paris-Saclay University, UVSQ, INSERM, Gustave Roussy, 'Exposome and Heredity' team, CESP UMR1018, Villejuif, France.
| | - Yousra Kherabi
- Université de Paris, IAME, INSERM, Paris, France; Infectious and Tropical Diseases Department, Bichat-Claude Bernard Hospital, AP-HP, Paris, France
| | - Conor-James MacDonald
- Paris-Saclay University, UVSQ, INSERM, Gustave Roussy, 'Exposome and Heredity' team, CESP UMR1018, Villejuif, France
| | - Jade Ghosn
- Université de Paris, IAME, INSERM, Paris, France; Infectious and Tropical Diseases Department, Bichat-Claude Bernard Hospital, AP-HP, Paris, France
| | - Nathan Peiffer-Smadja
- Université de Paris, IAME, INSERM, Paris, France; Infectious and Tropical Diseases Department, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College, London, UK
| |
Collapse
|
127
|
Hamady A, Lee J, Loboda ZA. Waning antibody responses in COVID-19: what can we learn from the analysis of other coronaviruses? Infection 2022; 50:11-25. [PMID: 34324165 PMCID: PMC8319587 DOI: 10.1007/s15010-021-01664-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The coronavirus disease 2019 (COVID-19), caused by the novel betacoronavirus severe acute respiratory syndrome 2 (SARS-CoV-2), was declared a pandemic in March 2020. Due to the continuing surge in incidence and mortality globally, determining whether protective, long-term immunity develops after initial infection or vaccination has become critical. METHODS/RESULTS In this narrative review, we evaluate the latest understanding of antibody-mediated immunity to SARS-CoV-2 and to other coronaviruses (SARS-CoV, Middle East respiratory syndrome coronavirus and the four endemic human coronaviruses) in order to predict the consequences of antibody waning on long-term immunity against SARS-CoV-2. We summarise their antibody dynamics, including the potential effects of cross-reactivity and antibody waning on vaccination and other public health strategies. At present, based on our comparison with other coronaviruses we estimate that natural antibody-mediated protection for SARS-CoV-2 is likely to last for 1-2 years and therefore, if vaccine-induced antibodies follow a similar course, booster doses may be required. However, other factors such as memory B- and T-cells and new viral strains will also affect the duration of both natural and vaccine-mediated immunity. CONCLUSION Overall, antibody titres required for protection are yet to be established and inaccuracies of serological methods may be affecting this. We expect that with standardisation of serological testing and studies with longer follow-up, the implications of antibody waning will become clearer.
Collapse
Affiliation(s)
- Ali Hamady
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - JinJu Lee
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Zuzanna A Loboda
- Department of Immunology and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
128
|
Wratil PR, Stern M, Priller A, Willmann A, Almanzar G, Vogel E, Feuerherd M, Cheng CC, Yazici S, Christa C, Jeske S, Lupoli G, Vogt T, Albanese M, Mejías-Pérez E, Bauernfried S, Graf N, Mijocevic H, Vu M, Tinnefeld K, Wettengel J, Hoffmann D, Muenchhoff M, Daechert C, Mairhofer H, Krebs S, Fingerle V, Graf A, Steininger P, Blum H, Hornung V, Liebl B, Überla K, Prelog M, Knolle P, Keppler OT, Protzer U. Three exposures to the spike protein of SARS-CoV-2 by either infection or vaccination elicit superior neutralizing immunity to all variants of concern. Nat Med 2022; 28:496-503. [PMID: 35090165 DOI: 10.1038/s41591-022-01715-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/25/2022] [Indexed: 11/09/2022]
Abstract
Infection-neutralizing antibody responses after SARS-CoV-2 infection or COVID-19 vaccination are an essential component of antiviral immunity. Antibody-mediated protection is challenged by the emergence of SARS-CoV-2 variants of concern (VoCs) with immune escape properties, such as omicron (B.1.1.529) that is rapidly spreading worldwide. Here, we report neutralizing antibody dynamics in a longitudinal cohort of COVID-19 convalescent and infection-naive individuals vaccinated with mRNA BNT162b2 by quantifying anti-SARS-CoV-2-spike antibodies and determining their avidity and neutralization capacity in serum. Using live-virus neutralization assays, we show that a superior infection-neutralizing capacity against all VoCs, including omicron, developed after either two vaccinations in convalescents or after a third vaccination or breakthrough infection of twice-vaccinated, naive individuals. These three consecutive spike antigen exposures resulted in an increasing neutralization capacity per anti-spike antibody unit and were paralleled by stepwise increases in antibody avidity. We conclude that an infection-plus-vaccination-induced hybrid immunity or a triple immunization can induce high-quality antibodies with superior neutralization capacity against VoCs, including omicron.
Collapse
Affiliation(s)
- Paul R Wratil
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Marcel Stern
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Alina Priller
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Annika Willmann
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Giovanni Almanzar
- Pediatric Rheumatology / Special Immunology, Pediatrics Department, University Hospital Würzburg, Würzburg, Germany
| | - Emanuel Vogel
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Martin Feuerherd
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Sarah Yazici
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Catharina Christa
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Samuel Jeske
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Gaia Lupoli
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Tim Vogt
- Pediatric Rheumatology / Special Immunology, Pediatrics Department, University Hospital Würzburg, Würzburg, Germany
| | - Manuel Albanese
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.,National Institute of Molecular Genetics (INGM), Milano, Italy
| | - Ernesto Mejías-Pérez
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Stefan Bauernfried
- Gene Center and Department of Biochemistry, LMU München, Munich, Germany
| | - Natalia Graf
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Hrvoje Mijocevic
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Martin Vu
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Kathrin Tinnefeld
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Jochen Wettengel
- German Centre for Infection Research (DZIF), Partner Site, Munich, Germany.,Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Dieter Hoffmann
- German Centre for Infection Research (DZIF), Partner Site, Munich, Germany.,Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Christopher Daechert
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Helga Mairhofer
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Volker Fingerle
- Bavarian Health and Food Safety Authority (LGL (LGL), Oberschleißheim, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, LMU München, Munich, Germany
| | - Bernhard Liebl
- Bavarian Health and Food Safety Authority (LGL (LGL), Oberschleißheim, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martina Prelog
- Pediatric Rheumatology / Special Immunology, Pediatrics Department, University Hospital Würzburg, Würzburg, Germany
| | - Percy Knolle
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Oliver T Keppler
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany. .,German Centre for Infection Research (DZIF), Partner Site, Munich, Germany.
| | - Ulrike Protzer
- German Centre for Infection Research (DZIF), Partner Site, Munich, Germany. .,Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany.
| |
Collapse
|
129
|
Fonseca MHG, Souza TDFGD, Araújo FMDC, Andrade LOMD. Dynamics of antibody response to CoronaVac vaccine. J Med Virol 2022; 94:2139-2148. [PMID: 35060174 PMCID: PMC9015561 DOI: 10.1002/jmv.27604] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/17/2022]
Abstract
CoronaVac was the first vaccine approved in Brazil for use in healthcare workers (HCWs). However, there is limited information about it, with little long‐term evidence on post‐vaccination antibody persistence. This study evaluated the antibody response to SARS‐CoV‐2 in 1237 HCWs after the first (1D), second dose (2D), and 6 months postvaccination (6mA2D) with CoronaVac. The seropositivity was 88% at 1D, increasing to 99.8% at 2D, but decreasing to 97.9% at 6mA2D, which was also observed at the analyzed antibody levels. Interestingly, the levels in females were higher than males, and we found a positive correlation with previous SARS‐CoV‐2 infection. Participants with comorbidities had lower levels suggesting the need to monitor for a potential booster dose. Our findings suggest that CoronaVac induced a robust antibody response that wanes significantly over time. Further longitudinal studies are needed to identify whether the antibodies will decline or plateau at a lower level. Our findings suggest that CoronaVac induced a robust antibody response that wanes significantly over time.
In our study, two doses of CoronaVac were capable of induction an antibody response in people ≥51 years old.
The seropositivity and the levels of antibodies were higher in females when compared to males.
We found a positive correlation with previous SARS‐CoV‐2 infection, previously infected participants had a significantly higher antibody response than previously uninfected participants.
Our findings suggest that patients with chronic diseases may need a booster shot of CoronaVac vaccine.
Individuals with immune‐mediated diseases developed a significant humoral response following the administration of two doses of CoronaVac, albeit with lower antibody titers.
Collapse
|
130
|
van Gils MJ, van Willigen HD, Wynberg E, Han AX, van der Straten K, Burger JA, Poniman M, Oomen M, Tejjani K, Bouhuijs JH, Verveen A, Lebbink R, Dijkstra M, Appelman B, Lavell AA, Caniels TG, Bontjer I, van Vught LA, Vlaar AP, Sikkens JJ, Bomers MK, Russell CA, Kootstra NA, Sanders RW, Prins M, de Bree GJ, de Jong MD. A single mRNA vaccine dose in COVID-19 patients boosts neutralizing antibodies against SARS-CoV-2 and variants of concern. Cell Rep Med 2022; 3:100486. [PMID: 35103254 PMCID: PMC8668345 DOI: 10.1016/j.xcrm.2021.100486] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/28/2021] [Accepted: 11/26/2021] [Indexed: 12/20/2022]
Abstract
The urgent need for, but limited availability of, SARS-CoV-2 vaccines worldwide has led to widespread consideration of dose-sparing strategies. Here, we evaluate the SARS-CoV-2-specific antibody responses following BNT162b2 vaccination in 150 previously SARS-CoV-2-infected individuals from a population-based cohort. One week after first vaccine dose, spike protein antibody levels are 27-fold higher and neutralizing antibody titers 12-fold higher, exceeding titers of fully vaccinated SARS-CoV-2-naive controls, with minimal additional boosting after the second dose. Neutralizing antibody titers against four variants of concern increase after vaccination; however, overall neutralization breadth does not improve. Pre-vaccination neutralizing antibody titers and time since infection have the largest positive effect on titers following vaccination. COVID-19 severity and the presence of comorbidities have no discernible impact on vaccine response. In conclusion, a single dose of BNT162b2 vaccine up to 15 months after SARS-CoV-2 infection offers higher neutralizing antibody titers than 2 vaccine doses in SARS-CoV-2-naive individuals.
Collapse
Affiliation(s)
- Marit J. van Gils
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Hugo D.G. van Willigen
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Elke Wynberg
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, the Netherlands
| | - Alvin X. Han
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Karlijn van der Straten
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Judith A. Burger
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Meliawati Poniman
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Melissa Oomen
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Khadija Tejjani
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Joey H. Bouhuijs
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Anouk Verveen
- Department of Medical Psychology, Amsterdam UMC, University of Amsterdam, Amsterdam School of Public Health, Amsterdam, the Netherlands
| | - Romy Lebbink
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, the Netherlands
| | - Maartje Dijkstra
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Brent Appelman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - A.H. Ayesha Lavell
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Tom G. Caniels
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Ilja Bontjer
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Lonneke A. van Vught
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Alexander P.J. Vlaar
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Jonne J. Sikkens
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Marije K. Bomers
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Colin A. Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Neeltje A. Kootstra
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Rogier W. Sanders
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Maria Prins
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Godelieve J. de Bree
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Menno D. de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| |
Collapse
|
131
|
Kelsen SG, Braverman AS, Aksoy MO, Hayman JA, Patel PS, Rajput C, Zhao H, Fisher SG, Ruggieri MR, Gentile NT. SARS-CoV-2 BNT162b2 vaccine-induced humoral response and reactogenicity in individuals with prior COVID-19 disease. JCI Insight 2022; 7:155889. [PMID: 35019861 PMCID: PMC8876462 DOI: 10.1172/jci.insight.155889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Most individuals with prior COVID-19 disease manifest long-term protective immune responses against reinfection. Accordingly, we tested the hypothesis that humoral immune and reactogenicity responses to a SARS-CoV-2 mRNA vaccine differ in individuals with and without prior COVID-19 disease. METHODS Health care workers (n = 61) with (n = 30) and without (n = 31) prior COVID-19 disease received two 30 μg doses of Pfizer BNT162b2 vaccine 3 weeks apart. Serum IgG antibody against the spike receptor-binding domain; serum neutralizing activity; and vaccine reactogenicity were assessed longitudinally every 2 weeks for 56 days after the first injection. RESULTS The COVID-19 group manifested more rapid increases in spike IgG antibody and serum neutralizing activity after the first vaccine dose but showed little or no increase after the second dose compared with the infection-naive group. In fact, spike IgG was at its maximum level after the first dose in 36% of the COVID-19 group versus 0% of the infection-naive group. Peak IgG antibody levels were lower but appeared to fall more slowly in the COVID-19 group versus the infection-naive group. Finally, adverse systemic reactions, e.g., fever, headache, and malaise, were more frequent and lasted longer after both the first and second injection in the COVID-19 group than in the infection-naive group. CONCLUSION Individuals with prior COVID-19 disease demonstrate a robust, accelerated humoral immune response to the first dose but an attenuated response to the second dose of BNT162b2 vaccine compared with controls. The COVID-19 group also experienced greater reactogenicity. Humoral responses and reactogenicity to BNT162b2 differ qualitatively and quantitatively in individuals with prior COVID-19 disease compared with infection-naive individuals. FUNDING This work was supported by Temple University institutional funds.
Collapse
Affiliation(s)
- Steven G Kelsen
- Departments of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Alan S Braverman
- Department of Anatomy, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Mark O Aksoy
- Departments of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Jacob A Hayman
- Departments of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Puja S Patel
- Departments of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Charu Rajput
- Departments of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Huaqing Zhao
- Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Susan G Fisher
- Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Michael R Ruggieri
- Department of Anatomy, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Nina T Gentile
- Department of Emergency Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| |
Collapse
|
132
|
Faro-Viana J, Bergman ML, Gonçalves LA, Duarte N, Coutinho TP, Borges PC, Diwo C, Castro R, Matoso P, Malheiro V, Brennand A, Kosack L, Akpogheneta O, Figueira JM, Cardoso C, Casaca AM, Alves PM, Nunes T, Penha-Gonçalves C, Demengeot J. Population homogeneity for the antibody response to COVID-19 BNT162b2/Comirnaty vaccine is only reached after the second dose across all adult age ranges. Nat Commun 2022; 13:140. [PMID: 35013258 PMCID: PMC8748650 DOI: 10.1038/s41467-021-27761-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
While mRNA vaccines are administrated worldwide in an effort to contain the COVID-19 pandemic, the heterogeneity of the humoral immune response they induce at the population scale remains unclear. Here, in a prospective, longitudinal, cohort-study, including 1245 hospital care workers and 146 nursing home residents scheduled for BNT162b2 vaccination, together covering adult ages from 19 to 99 years, we analyse seroconversion to SARS-CoV-2 spike protein and amount of spike-specific IgG, IgM and IgA before vaccination, and 3-5 weeks after each dose. We show that immunogenicity after a single vaccine dose is biased to IgG, heterogeneous and reduced with increasing age. The second vaccine dose normalizes IgG seroconversion in all age strata. These findings indicate two dose mRNA vaccines is required to reach population scale humoral immunity. The results advocate for the interval between the two doses not to be extended, and for serological monitoring of elderly and immunosuppressed vaccinees.
Collapse
Affiliation(s)
- João Faro-Viana
- CHLO, Centro Hospitalar de Lisboa Ocidental, Serviço de Patologia Clínica, Lisbon, 1449-005, Portugal
| | | | | | - Nádia Duarte
- IGC, Instituto Gulbenkian de Ciência, Oeiras, 2780-156, Portugal
| | - Teresa P Coutinho
- CIISA, Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, 1300-477, Portugal
| | | | - Christian Diwo
- IGC, Instituto Gulbenkian de Ciência, Oeiras, 2780-156, Portugal
| | - Rute Castro
- IBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, 2780-901, Portugal
| | - Paula Matoso
- IGC, Instituto Gulbenkian de Ciência, Oeiras, 2780-156, Portugal
| | - Vanessa Malheiro
- IGC, Instituto Gulbenkian de Ciência, Oeiras, 2780-156, Portugal
| | - Ana Brennand
- IGC, Instituto Gulbenkian de Ciência, Oeiras, 2780-156, Portugal
| | - Lindsay Kosack
- IGC, Instituto Gulbenkian de Ciência, Oeiras, 2780-156, Portugal
| | | | - João M Figueira
- CHLO, Centro Hospitalar de Lisboa Ocidental, Serviço de Patologia Clínica, Lisbon, 1449-005, Portugal
| | - Conceição Cardoso
- CHLO, Centro Hospitalar de Lisboa Ocidental, Serviço de Patologia Clínica, Lisbon, 1449-005, Portugal
| | - Ana M Casaca
- CHLO, Centro Hospitalar de Lisboa Ocidental, Serviço de Patologia Clínica, Lisbon, 1449-005, Portugal
| | - Paula M Alves
- IBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, 2780-901, Portugal
- ITQB NOVA, Instituto de Tecnológia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, 2780-157, Portugal
| | - Telmo Nunes
- CIISA, Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, 1300-477, Portugal
| | | | | |
Collapse
|
133
|
Mistry P, Barmania F, Mellet J, Peta K, Strydom A, Viljoen IM, James W, Gordon S, Pepper MS. SARS-CoV-2 Variants, Vaccines, and Host Immunity. Front Immunol 2022; 12:809244. [PMID: 35046961 PMCID: PMC8761766 DOI: 10.3389/fimmu.2021.809244] [Citation(s) in RCA: 167] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new beta coronavirus that emerged at the end of 2019 in the Hubei province of China. SARS-CoV-2 causes coronavirus disease 2019 (COVID-19) and was declared a pandemic by the World Health Organization (WHO) on 11 March 2020. Herd or community immunity has been proposed as a strategy to protect the vulnerable, and can be established through immunity from past infection or vaccination. Whether SARS-CoV-2 infection results in the development of a reservoir of resilient memory cells is under investigation. Vaccines have been developed at an unprecedented rate and 7 408 870 760 vaccine doses have been administered worldwide. Recently emerged SARS-CoV-2 variants are more transmissible with a reduced sensitivity to immune mechanisms. This is due to the presence of amino acid substitutions in the spike protein, which confer a selective advantage. The emergence of variants therefore poses a risk for vaccine effectiveness and long-term immunity, and it is crucial therefore to determine the effectiveness of vaccines against currently circulating variants. Here we review both SARS-CoV-2-induced host immune activation and vaccine-induced immune responses, highlighting the responses of immune memory cells that are key indicators of host immunity. We further discuss how variants emerge and the currently circulating variants of concern (VOC), with particular focus on implications for vaccine effectiveness. Finally, we describe new antibody treatments and future vaccine approaches that will be important as we navigate through the COVID-19 pandemic.
Collapse
Affiliation(s)
- Priyal Mistry
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Fatima Barmania
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Juanita Mellet
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Kimberly Peta
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Adéle Strydom
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ignatius M. Viljoen
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - William James
- James and Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Michael S. Pepper
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
134
|
Brnjarchevska Blazhevska T, Babačić H, Sibinovska O, Dobrevski B, Kirijas M, Milanovski G, Arsov T, Petlichkovski A. A single dose of BNT162b2 vaccine elicits strong humoral response in SARS-CoV-2 seropositive individuals. Allergy 2022; 77:296-298. [PMID: 34386995 PMCID: PMC8441874 DOI: 10.1111/all.15047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/01/2022]
Affiliation(s)
- Teodora Brnjarchevska Blazhevska
- Faculty of Medicine‐Skopje Institute for Immunobiology and Human Genetics Ss. Cyril and Methodius University Skopje North Macedonia
| | | | - Olgica Sibinovska
- Faculty of Medicine‐Skopje Institute for Immunobiology and Human Genetics Ss. Cyril and Methodius University Skopje North Macedonia
| | - Boban Dobrevski
- Faculty of Medicine‐Skopje Institute for Immunobiology and Human Genetics Ss. Cyril and Methodius University Skopje North Macedonia
| | - Meri Kirijas
- Faculty of Medicine‐Skopje Institute for Immunobiology and Human Genetics Ss. Cyril and Methodius University Skopje North Macedonia
| | - Gorjan Milanovski
- Faculty of Medicine‐Skopje Institute for Immunobiology and Human Genetics Ss. Cyril and Methodius University Skopje North Macedonia
| | - Todor Arsov
- Faculty of Medicine‐Skopje Institute for Immunobiology and Human Genetics Ss. Cyril and Methodius University Skopje North Macedonia
- John Curtin School of Medical Research Australian National University Canberra ACT Australia
| | - Aleksandar Petlichkovski
- Faculty of Medicine‐Skopje Institute for Immunobiology and Human Genetics Ss. Cyril and Methodius University Skopje North Macedonia
| |
Collapse
|
135
|
Abstract
Adaptive immune responses play critical roles in viral clearance and protection against re-infection, and SARS-CoV-2 is no exception. What is exceptional is the rapid characterization of the immune response to the virus performed by researchers during the first 20 months of the pandemic. This has given us a more detailed understanding of SARS-CoV-2 compared to many viruses that have been with us for a long time. Furthermore, effective COVID-19 vaccines were developed in record time, and their rollout worldwide is already making a significant difference, although major challenges remain in terms of equal access. The pandemic has engaged scientists and the public alike, and terms such as seroprevalence, neutralizing antibodies, antibody escape and vaccine certificates have become familiar to a broad community. Here, we review key findings concerning B cell and antibody (Ab) responses to SARS-CoV-2, focusing on non-severe cases and anti-spike (S) Ab responses in particular, the latter being central to protective immunity induced by infection or vaccination. The emergence of viral variants that have acquired mutations in S acutely highlights the need for continued characterization of both emerging variants and Ab responses against these during the evolving pathogen-immune system arms race.
Collapse
Affiliation(s)
- Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
136
|
Bengel CP, Kacapor R. A report of two cases of myocarditis following mRNA coronavirus disease 2019 vaccination. Eur Heart J Case Rep 2022; 6:ytac004. [PMID: 35169677 PMCID: PMC8755378 DOI: 10.1093/ehjcr/ytac004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/24/2021] [Accepted: 12/20/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Vaccination is the most important measure to control the coronavirus disease 2019 (COVID-19) pandemic. Myocarditis has been reported as a rare adverse reaction to COVID-19 vaccines. The clinical presentation of myocarditis in such cases can range from mild general symptoms to acute heart failure. CASE SUMMARY We report the cases of two young men who presented with chest pain and dyspnoea following the administration of the mRNA COVID-19 vaccine. Cardiac investigations revealed findings typical of acute myocarditis. DISCUSSION Myocarditis is a rare complication following mRNA COVID-19 vaccination. In this case series, the temporal proximity of the development of acute myocarditis and the administration of the mRNA COVID-19 vaccine was acknowledged. In the absence of other causative factors, myocarditis in these patients potentially occurred due to an adverse reaction to the mRNA COVID-19 vaccine. However, a causal relationship remains speculative. Clinical suspicion of myocarditis should be high if patients present with chest pain or dyspnoea after receiving COVID-19 vaccination.
Collapse
Affiliation(s)
- Christopher Paul Bengel
- Department of Cardiology, Kliniken Frankfurt-Main-Taunus, Bad Soden Hospital, Medizinische Klinik I, Kronberger Str. 36, 65812 Bad Soden, Germany
| | - Rifat Kacapor
- Department of Cardiology, Kliniken Frankfurt-Main-Taunus, Bad Soden Hospital, Medizinische Klinik I, Kronberger Str. 36, 65812 Bad Soden, Germany
| |
Collapse
|
137
|
Krüttgen A, Haase G, Haefner H, Imöhl M, Kleines M. Large inter-individual variability of cellular and humoral immunological responses to mRNA-1273 (Moderna) vaccination against SARS-CoV-2 in health care workers. Clin Exp Vaccine Res 2022; 11:96-103. [PMID: 35223670 PMCID: PMC8844675 DOI: 10.7774/cevr.2022.11.1.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/06/2021] [Accepted: 10/01/2021] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Studies on the immune responses to severe acute respiratory syndrome coronavirus 2 vaccines are necessary to evaluate the ongoing vaccination programs by correlating serological response data and clinical effectiveness data. We performed a longitudinal immunological profiling of health care workers vaccinated with mRNA-1273 (Moderna, Cambridge, MA, USA). Half of these vaccinees had experienced a mild coronavirus disease 2019 (COVID-19) infection in the spring of 2020 ("COVID-recovered" cohort), whereas the other half of the vaccinees had no previous COVID-19 infection ("COVID-naive" cohort). MATERIALS AND METHODS Serum was drawn at multiple time points and subjected to assays measuring anti-Spike immunoglobulin G (IgG), avidity of anti-Spike IgG, avidity of anti-receptor binding domain (RBD) IgG, virus neutralizing activity, and interferon-γ release from stimulated lymphocytes. RESULTS Between both cohorts and within each cohort, we found remarkable inter-individual differences regarding cellular and humoral immune responses to the Moderna mRNA-1273 vaccine. CONCLUSION First, our study indicates that the success of mRNA-1273 vaccinations should be verified by serological assays in order to identify "low-responders" to vaccination. Second, the kinetics of anti-S IgG and neutralizing activity correlate well with clinical effectiveness data, thus explaining incipient protection against infection 2 weeks after the first dose of mRNA-1273 in COVID-naive vaccinees. Third, our IgG-avidity data indicate that this incipient protection is mediated by low-avidity anti-RBD IgG and low-avidity anti-S IgG.
Collapse
Affiliation(s)
- Alexander Krüttgen
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Gerhard Haase
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Helga Haefner
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Matthias Imöhl
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Kleines
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
138
|
Kadali RAK, Janagama R, Yedlapati SH, Kanike N, Gajula V, Madathala RR, Poddar S, Sukka N, Jogu HR, Racherla S, Shah I. Side effects of messenger RNA vaccines and prior history of COVID-19, a cross-sectional study. Am J Infect Control 2022; 50:8-14. [PMID: 34718069 PMCID: PMC8552581 DOI: 10.1016/j.ajic.2021.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND There are concerns regarding immunogenicity with coronavirus disease 2019 (COVID-19) mRNA vaccines among persons with prior history of COVID-19 (PHC). This study was to analyze the short-term side effects of mRNA vaccines among health care workers (HCWs) with and without PHC. METHODS A cross-sectional study was performed using an independent online survey questionnaire that gathered responses from HCWs. RESULTS Among 1,475 HCWs, 1268 (85.97%) completed the survey, 102/1268 (44/447 in Moderna group and 58/821 in Pfizer-BioNTech group) reported PHC during pre-vaccination period. Symptoms of flushing/P = .05, brain fogging/P= .005, vertigo/P= .041, numbness/P= .023, diarrhea/P= .047, hives/P= .028, itching/P= .028, swelling of lips/mouth/P= .001, shortness of breath/P= .022, and anxiety/P= .048 have greater occurrence among Pfizer-BioNtech group with PHC when compared to Pfizer-BioNtech group with no PHC. Symptoms of chills/P= .027, flushing/P= .045, tremor/P= .05, muscle spasm/P= .039, vomiting/P= .031, diarrhea/P= .015, and cough/P= .011 have higher occurrence among Moderna group with PHC when compared to Moderna group with no PHC. CONCLUSIONS Few short-term side effects among mRNA vaccine recipients with PHC may have necessitated transient time-off from work. The PHC can be considered as a predictor for severity of side effects. While the vaccination program continues in the United States, a future COVID legislation that mandates vaccination among employees along with paid time off provision may help with higher compliance and acceptance.
Collapse
Affiliation(s)
- Renuka Ananth Kalyan Kadali
- Department of Internal Medicine, Harnett Hospitalist Group, Campbell University Jerry M. Wallace School of Osteopathic Medicine, Lillington, NC.
| | | | - Siva H Yedlapati
- Department of Internal Medicine, Erie County Medical Center, Buffalo, NY
| | - Neelakanta Kanike
- Division of Neonatology, Department of Pediatrics, Indiana University School of Medicine, Newburgh, IN
| | - Viswanath Gajula
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, The University of Mississippi Medical Center, Jackson, MS
| | | | - Swati Poddar
- Psychiatry Program, California Health Care Facility, Stockton, CA
| | - Neelakanta Sukka
- Department of Internal Medicine, Vidant medical group, Lenoir Memorial hospital, Kinston, NC
| | - Hanumantha R Jogu
- Department of Internal Medicine, Vidant medical group, Greenville, NC
| | - Shailaja Racherla
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA
| | - Isha Shah
- Rheumatology Program, Kent Hospital, Warwick, Rhode Island, USA
| |
Collapse
|
139
|
Serraino C, Melchio R, Badinella Martini M, Gerbaudo L, Fenoglio L. Response to BNT162b2 mRNA COVID-19 vaccine among healthcare workers in Italy: a 3-month follow-up: comment. Intern Emerg Med 2022; 17:311-312. [PMID: 35006536 PMCID: PMC8744016 DOI: 10.1007/s11739-021-02907-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022]
Affiliation(s)
- Cristina Serraino
- Department of Internal Medicine, Santa Croce e Carle" Hospital, Via Michele Coppino 26, Cuneo, Italy
| | - Remo Melchio
- Department of Internal Medicine, Santa Croce e Carle" Hospital, Via Michele Coppino 26, Cuneo, Italy
| | - Marco Badinella Martini
- Department of Internal Medicine, Santa Croce e Carle" Hospital, Via Michele Coppino 26, Cuneo, Italy.
| | - Laura Gerbaudo
- Occupational Health Service, "Santa Croce e Carle" Hospital, Cuneo, Italy
| | - Luigi Fenoglio
- Department of Internal Medicine, Santa Croce e Carle" Hospital, Via Michele Coppino 26, Cuneo, Italy
| |
Collapse
|
140
|
Moyet J, Helle F, Bourdenet G, Joseph C, Gubler B, Deschasse G, Defouilloy I, Slovenski T, François C, Liabeuf S, Schmit JL, Lanoix JP, Castelain S, Bloch F, Brochot E. Kinetics of SARS-CoV-2-Neutralising Antibodies of Residents of Long-Term Care Facilities. J Nutr Health Aging 2022; 26:57-63. [PMID: 35067704 PMCID: PMC8683825 DOI: 10.1007/s12603-021-1713-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/15/2021] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Elderly residents of nursing homes (NHs) and long-term care units (LTCUs) have been shown to have a high risk of mortality and morbidity in cases of SARS-CoV-2 infection. The objective of this study was to examine the kinetics of neutralizing antibodies (NAbs) directed against the SARS-CoV-2 virus in residents of the NH and LTCU units of our University Hospital who were identified with positive serology after the first epidemic outbreak. MATERIALS AND METHODS The participants included were sampled every three months for qualitative serological testing, as well as quantitative testing by neutralization tests using retroviral particles containing the S glycoprotein of SARS-CoV-2. Vaccination using the Comirnaty (Pfizer BNT162b2) vaccine begun before the last serological follow-up. RESULTS The median NAb titer in June 2020 was 80 [40; 60] versus 40 [40; 160] three months later, showing a statistically significant decline (p < 0.007), but remained stable between the three- and six-month timepoints (p = 0.867). By nine months after vaccination, we observed a significant difference between vaccinated residents known to have positive serology before vaccination (SERO+, Vacc+) and those vaccinated without having previously shown COVID-19 seroconversion (SERO-, Vacc+), the latter group showing similar titers to the SERO+, Vacc- participants (p=0.166). The median antibody titer in SERO+, Vacc+ patients increased 15-fold following vaccination. DISCUSSION Humoral immunity against SARS-CoV-2 appears to be persistent in elderly institutionalized patients, with a good post-vaccination response by residents who had already shown seroconversion but a notably diminished response by those who were seronegative before vaccination. To evaluate immunity in its entirety and elaborate a sound vaccination strategy, the cellular immune response via T cells specific to SARS-CoV-2 merits analysis, as this response is susceptible to being affected by immunosenescence.
Collapse
Affiliation(s)
- J Moyet
- Pr Frédéric Bloch, Department of Gerontology, University Hospital Amiens-Picardie - Hôpital Nord, Place Victor Pauchet - 80054 Amiens Cedex 1, France, Phone / Fax: +333.22.45.57.20 / +333. 22.45.53.30, E-mail: , ORCID identifier : 0000-0002-6046-7097
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Blain H, Tuaillon E, Gamon L, Pisoni A, Miot S, Rolland Y, Picot M, Bousquet J. Antibody response after one and two jabs of the BNT162b2 vaccine in nursing home residents: The CONsort-19 study. Allergy 2022; 77:271-281. [PMID: 34286856 PMCID: PMC8441741 DOI: 10.1111/all.15007] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022]
Abstract
Background The humoral immune response following COVID‐19 vaccination in nursing home residents is poorly known. A longitudinal study compared levels of IgG antibodies against the spike protein (S‐RBD IgG) (S‐RDB protein IgG) after one and two BNT162b2/Pfizer jabs in residents with and without prior COVID‐19. Methods In 22 French nursing homes, COVID‐19 was diagnosed with real‐time reverse‐transcriptase polymerase chain reaction (RT‐PCR) for SARS‐CoV‐2. Blood S‐RDB‐protein IgG and nucleocapsid (N) IgG protein (N‐protein IgG) were measured 21–24 days after the first jab (1,004 residents) and 6 weeks after the second (820 residents). Results In 735 residents without prior COVID‐19, 41.7% remained seronegative for S‐RDB‐protein IgG after the first jab vs. 2.1% of the 270 RT‐PCR‐positive residents (p < 0.001). After the second jab, 3% of the 586 residents without prior COVID‐19 remained seronegative. However, 26.5% had low S‐RDB‐protein IgG levels (50–1050 UA/ml) vs. 6.4% of the 222 residents with prior COVID‐19. Residents with an older infection (first wave), or with N‐protein IgG at the time of vaccination, had the highest S‐RDB‐protein IgG levels. Residents with a prior COVID‐19 infection had higher S‐RDB‐protein IgG levels after one jab than those without after two jabs. Interpretation A single vaccine jab is sufficient to reach a high humoral immune response in residents with prior COVID‐19. Most residents without prior COVID‐19 are seropositive for S‐RDB‐protein IgG after the second jab, but around 30% have low levels. Whether residents with no or low post‐vaccine S‐RDB protein IgG are at higher risk of symptomatic COVID‐19 requires further analysis.
Collapse
Affiliation(s)
- Hubert Blain
- Department of Internal Medicine and Geriatrics MUSE University Montpellier France
| | | | - Lucie Gamon
- Clinical research and epidemiology unit University hospital Montpellier France
| | | | - Stéphanie Miot
- Department of Internal Medicine and Geriatrics MUSE University Montpellier France
| | - Yves Rolland
- INSERM 1027 Gérontopôle de Toulouse Toulouse France
| | | | - Jean Bousquet
- Clinical research and epidemiology unit University hospital Montpellier France
- Department of Dermatology and Allergy Universitätsmedizin Berlin Germany
| |
Collapse
|
142
|
Sookaromdee P, Wiwanitkit V. Previous COVID-19 infection, previous cross infection, and post-COVID-19 vaccination adverse effect. MEDICAL JOURNAL OF DR. D.Y. PATIL VIDYAPEETH 2022. [DOI: 10.4103/mjdrdypu.mjdrdypu_348_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
143
|
Spicer KB, Glick C, Cavanaugh AM, Thoroughman D. Protective Immunity after Natural Infection with Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) - Kentucky, USA, 2020. Int J Infect Dis 2022; 114:21-28. [PMID: 34649001 PMCID: PMC8506664 DOI: 10.1016/j.ijid.2021.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As vaccine supply and access remain limited in many parts of the world, understanding the duration of protection from reinfection after natural infection is important. METHODS Distinct individuals testing positive and negative for SARS-CoV-2 between March 6, 2020, and August 31, 2020, in Kentucky, USA, were identified using the Kentucky National Electronic Disease Surveillance System. Individuals were followed for occurrence of a positive test for SARS-CoV-2 from 91 days after their initial test result through December 31, 2020. Protection from reinfection provided by a prior infection was calculated and additional analyses evaluated impact of age, sex, symptom status, long-term care facility connection, testing occurrence and frequency, and time from initial infection. RESULTS The protective effect from prior infection was 80.3% (95% CI, 78.2%-82.2%) for those aged 20-59 years and 67.4% (95% CI, 62.8%-71.4%) for those aged ≥60 years. At 30-day time periods through 270 days (with limited exceptions), protection was estimated to be >75% for those aged 20-59 years and >65% for those aged ≥60 years. Factors associated with repeat positive testing included a connection to a long-term care facility, duration of potential exposure, and absence of symptoms during initial infection. CONCLUSIONS Natural infection provides substantial and persistent immunologic protection for a period of several months for most individuals, although subpopulations may be at greater risk of repeat positive testing and potential poor outcomes associated with reinfection. These subgroups include individuals aged ≥60 years, residents and staff of long-term care facilities, and those who have mild or asymptomatic illness with initial infection. Continued emphasis on vaccination and infection prevention and control strategies remains critically important in reducing the risk of reinfection and associated severe outcomes for these groups.
Collapse
Affiliation(s)
- Kevin B Spicer
- Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention (CDC), USA; Kentucky Department for Public Health, USA.
| | | | - Alyson M Cavanaugh
- Kentucky Department for Public Health, USA; Epidemic Intelligence Service, CDC, USA
| | - Douglas Thoroughman
- Kentucky Department for Public Health, USA; Career Epidemiology Field Officer Program, Center for Preparedness and Response, CDC, USA
| |
Collapse
|
144
|
Du J, Zhang D, Pathakamuri JA, Kuebler D, Yang Y, Loginova Y, Chu E, Madej R, Neves JV, Singer B, Radke H, Spencer N, Rizk E, Zhang A, Lu CM, Sha MY. A New Testing Platform Using Fingerstick Blood for Quantitative Antibody Response Evaluation after SARS-CoV-2 Vaccination. Emerg Microbes Infect 2021; 11:250-259. [PMID: 34951566 PMCID: PMC8745372 DOI: 10.1080/22221751.2021.2023328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Testing and vaccination have been major components of the strategy for combating the ongoing COVID-19 pandemic. In this study, we have developed a quantitative anti-SARS-CoV-2 spike (S1) IgG antibody assay using a fingerstick dried blood sample. We evaluated the feasibility of using this high-throughput and quantitative anti-SARS-CoV-2 spike (S1) IgG antibody testing assay in vaccinated individuals. Fingerstick blood samples were collected and analyzed from 137 volunteers before and after receiving the Moderna or Pfizer mRNA vaccine. Anti-SARS-CoV-2 S1 IgG antibody could not be detected within the first 7 days after receiving the first vaccine dose, however, the assay reliably detected antibodies from day 14 onwards. In addition, no anti-SARS-CoV-2 nucleocapsid (N) protein IgG antibody was detected in any of the vaccinated or healthy participants, indicating that the anti-SARS-CoV-2 S1 IgG assay is specific for the mRNA vaccine-induced antibodies. The S1 IgG levels detected in fingerstick samples correlated with the levels found in venous blood plasma samples and with the efficacy of venous blood plasma samples in the plaque reduction neutralization test (PRNT). The assay displayed a limit of quantification (LOQ) of 0.59 μg/mL and was found to be linear in the range of 0.51-1000 μg/mL. Finally, its clinical performance displayed a Positive Percent Agreement (PPA) of 100% (95% CI: 0.89-1.00) and a Negative Percent Agreement (NPA) of 100% (95% CI: 0.93-1.00). In summary, the assay described here represents a sensitive, precise, accurate, and simple method for the quantitative detection and monitoring of post-vaccination anti-SARS-CoV-2 spike IgG responses.
Collapse
Affiliation(s)
- Jinwei Du
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| | - Dayu Zhang
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| | | | - Daniel Kuebler
- Franciscan University of Steubenville, Steubenville, OH. 43952
| | - Ying Yang
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| | - Yulia Loginova
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| | - Eric Chu
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| | - Roberta Madej
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| | - Jocelyn V Neves
- Franciscan University of Steubenville, Steubenville, OH. 43952
| | - Brianna Singer
- Franciscan University of Steubenville, Steubenville, OH. 43952
| | - Holly Radke
- Franciscan University of Steubenville, Steubenville, OH. 43952
| | - Naomi Spencer
- Franciscan University of Steubenville, Steubenville, OH. 43952
| | - Elizabeth Rizk
- Franciscan University of Steubenville, Steubenville, OH. 43952
| | - Aiguo Zhang
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| | - Chuanyi M Lu
- University of California and VA Health Care System, San Francisco, CA
| | - Michael Y Sha
- DiaCarta Inc., 4385 Hopyard Rd. suite 100, Pleasanton, CA 94588, US
| |
Collapse
|
145
|
Weigert A, Bergman ML, Gonçalves LA, Godinho I, Duarte N, Abrantes R, Borges P, Brennand A, Malheiro V, Matoso P, Akpogheneta O, Kosack L, Cruz P, Nogueira E, Pereira M, Ferreira A, Marques M, Nunes T, Faro-Viana J, Demengeot J, Penha-Gonçalves C. Longitudinal Analysis of Antibody Responses to the mRNA BNT162b2 Vaccine in Patients Undergoing Maintenance Hemodialysis: A 6-Month Follow-Up. Front Med (Lausanne) 2021; 8:796676. [PMID: 35004771 PMCID: PMC8740691 DOI: 10.3389/fmed.2021.796676] [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: 10/17/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Patients on hemodialysis (HD) are at higher risk for COVID-19, overall are poor responders to vaccines, and were prioritized in the Portuguese vaccination campaign. Objective: This work aimed at evaluating in HD patients the immunogenicity of BTN162b2 after the two doses induction phase, the persistence of specific antibodies along time, and factors predicting these outcomes. Methods: We performed a prospective, 6-month long longitudinal cohort analysis of 156 HD patients scheduled to receive BTN162b2. ELISA quantified anti-spike IgG, IgM, and IgA levels in sera were collected every 3 weeks during the induction phase (t0 before vaccine; t1, d21 post first dose; and t2 d21 post second dose), and every 3-4 months during the waning phase (t3, d140, and t4, d180 post first dose). The age-matched control cohort was similarly analyzed from t0 to t2. Results: Upon exclusion of participants identified as previously exposed to SARS-CoV-2, seroconversion at t1 was lower in patients than controls (29 and 50%, respectively, p = 0.0014), while the second vaccine dose served as a boost in both cohorts (91 and 95% positivity, respectively, at t2, p = 0.2463). Lower response in patients than controls at t1 was a singularity of the participants ≤ 70 years (p = 2.01 × 10-05), associated with immunosuppressive therapies (p = 0.013), but not with lack of responsiveness to hepatitis B. Anti-spike IgG, IgM, and IgA levels decreased at t3, with IgG levels further waning at t4 and resulting in >30% seronegativity. Anti-spike IgG levels at t1 and t4 were correlated (ρ = 0.65, p < 2.2 × 10-16). Conclusions: While most HD patients seroconvert upon 2 doses of BNT162b2 vaccination, anti-spike antibodies levels wane over the following 4 months, leading to early seroreversion in a sizeable fraction of the patients. These findings warrant close monitoring of COVID-19 infection in vaccinated HD patients, and advocate for further studies following reinforced vaccination schedules.
Collapse
Affiliation(s)
- André Weigert
- DaVita Óbidos, Óbidos, Portugal
- Serviço de Nefrologia, Centro Hospitalar de Lisboa Ocidental, Hospital Santa Cruz, Carnaxide, Portugal
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Iolanda Godinho
- DaVita Óbidos, Óbidos, Portugal
- Serviço de Nefrologia e Transplantação Renal, Centro Hospitalar de Lisboa Norte EPE, Lisboa, Portugal
| | - Nádia Duarte
- IGC, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Rita Abrantes
- DaVita Óbidos, Óbidos, Portugal
- Serviço de Nefrologia, Centro Hospitalar do Médio Tejo, Torres Novas, Portugal
| | | | - Ana Brennand
- IGC, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Paula Matoso
- IGC, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Pedro Cruz
- DaVita Óbidos, Óbidos, Portugal
- Serviço de Nefrologia, Hospital Das Forças Armadas, Lisboa, Portugal
| | - Estela Nogueira
- DaVita Óbidos, Óbidos, Portugal
- Serviço de Nefrologia e Transplantação Renal, Centro Hospitalar de Lisboa Norte EPE, Lisboa, Portugal
| | | | | | | | - Telmo Nunes
- CIISA, Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
| | - João Faro-Viana
- Serviço de Patologia Clínica, Centro Hospitalar de Lisboa Ocidental EPE, Carnaxide, Portugal
| | | | | |
Collapse
|
146
|
Spike Antibody Titers Evaluation after a 2-Dose Regimen of BNT162b2 Vaccination in Healthcare Workers Previously Infected with SARS-CoV-2. Microbiol Spectr 2021; 9:e0103621. [PMID: 34756062 PMCID: PMC8579918 DOI: 10.1128/spectrum.01036-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
147
|
Amatu A, Pani A, Patelli G, Gagliardi OM, Loparco M, Piscazzi D, Cassingena A, Tosi F, Ghezzi S, Campisi D, Grifantini R, Abrignani S, Siena S, Scaglione F, Sartore-Bianchi A. Impaired seroconversion after SARS-CoV-2 mRNA vaccines in patients with solid tumours receiving anticancer treatment. Eur J Cancer 2021; 163:16-25. [PMID: 35032813 PMCID: PMC8692068 DOI: 10.1016/j.ejca.2021.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 12/19/2022]
Abstract
Background Patients with solid tumours have high COVID-19 mortality. Limited and heterogeneous data are available regarding the immunogenicity of SARS-CoV-2 mRNA vaccines in this population. Methods and findings This is a prospective, single-centre cohort study aiming at evaluating seroconversion in terms of anti-spike antibodies in a population of patients with solid tumours undergoing cancer therapy within 2 months before the second vaccine dose, as compared with a cohort of controls. Subjects who were not SARS-CoV-2 naïve were excluded, and 171 patients were included in the final study population (150 vaccinated with BNT162b2, 87.7%; 21 with mRNA-1273, 12.3%) and compared with 2406 controls. The median follow-up time from the second dose of vaccination was 30 days (12–42; IQR: 26–34). Most patients had metastatic disease (138, 80.7%). Seroconversion rate was significantly lower in cancer patients than in controls (94.2% versus 99.8%, p < 0.001). At univariate logistic regression analysis, Odds ratio (OR) for seroconversion was also reduced in older individuals (>70 years). A multivariate logistic model confirmed cancer as the only significant variable in impairing seroconversion (OR 0.03, p < 0.001). In the cancer population, a multivariate analysis among clinical variables, including the type of cancer treatment, showed ECOG PS > 2 as the only one of impact (OR 0.07, p = 0.012). Conclusions There is a fraction of 6% of patients with solid tumours undergoing cancer treatment, mainly with poorer performance status, who fail to obtain seroconversion after SARS-CoV-2 mRNA vaccines. These patients should be considered for enhanced vaccination strategies and carefully monitored for SARS-CoV-2 infection during cancer treatment.
Collapse
Affiliation(s)
- Alessio Amatu
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Arianna Pani
- SC Analisi Chimico Cliniche e Microbiologia, Dipartimento Medicina di Laboratorio, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy
| | - Giorgio Patelli
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy
| | - Oscar M Gagliardi
- SC Analisi Chimico Cliniche e Microbiologia, Dipartimento Medicina di Laboratorio, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy
| | - Marina Loparco
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy
| | - Daniele Piscazzi
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy
| | - Andrea Cassingena
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Silvia Ghezzi
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Daniela Campisi
- SC Analisi Chimico Cliniche e Microbiologia, Dipartimento Medicina di Laboratorio, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Renata Grifantini
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy; INGM, Istituto Nazionale Genetica Molecolare 'Romeo Ed Enrica Invernizzi', Milan, Italy
| | - Sergio Abrignani
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy; INGM, Istituto Nazionale Genetica Molecolare 'Romeo Ed Enrica Invernizzi', Milan, Italy
| | - Salvatore Siena
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy
| | - Francesco Scaglione
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy.
| | - Andrea Sartore-Bianchi
- SC Oncologia Falck, Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Dipartimento di Oncologia Ed Emato-Oncologia, Università Degli Studi di Milano (La Statale), Milan, Italy.
| |
Collapse
|
148
|
Bowen JE, Walls AC, Joshi A, Sprouse KR, Stewart C, Tortorici MA, Franko NM, Logue JK, Mazzitelli IG, Tiles SW, Ahmed K, Shariq A, Snell G, Iqbal NT, Geffner J, Bandera A, Gori A, Grifantini R, Chu HY, Van Voorhis WC, Corti D, Veesler D. SARS-CoV-2 spike conformation determines plasma neutralizing activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.12.19.473391. [PMID: 34981060 PMCID: PMC8722597 DOI: 10.1101/2021.12.19.473391] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Numerous safe and effective COVID-19 vaccines have been developed that utilize various delivery technologies and engineering strategies. The influence of the SARS-CoV-2 spike (S) glycoprotein conformation on antibody responses induced by vaccination or infection in humans remains unknown. To address this question, we compared plasma antibodies elicited by six globally-distributed vaccines or infection and observed markedly higher binding titers for vaccines encoding a prefusion-stabilized S relative to other groups. Prefusion S binding titers positively correlated with plasma neutralizing activity, indicating that physical stabilization of the prefusion conformation enhances protection against SARS-CoV-2. We show that almost all plasma neutralizing activity is directed to prefusion S, in particular the S 1 subunit, and that variant cross-neutralization is mediated solely by RBD-specific antibodies. Our data provide a quantitative framework for guiding future S engineering efforts to develop vaccines with higher resilience to the emergence of variants and longer durability than current technologies.
Collapse
Affiliation(s)
- John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Anshu Joshi
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Kaitlin R. Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | - Nicholas M. Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Jennifer K. Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Ignacio G. Mazzitelli
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - Sasha W Tiles
- Center for Emerging and Re-emerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Kumail Ahmed
- Department of Paediatrics and Child Health, and Biological & Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Asefa Shariq
- Department of Paediatrics and Child Health, and Biological & Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | | | - Najeeha Talat Iqbal
- Department of Paediatrics and Child Health, and Biological & Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - Alessandra Bandera
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, 20122 Milan, Italy
| | - Helen Y. Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Wesley C. Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
149
|
Wolszczak-Biedrzycka B, Bieńkowska A, Dorf J. Assessment of Post-Vaccination Antibody Response Eight Months after the Administration of BNT1622b2 Vaccine to Healthcare Workers with Particular Emphasis on the Impact of Previous COVID-19 Infection. Vaccines (Basel) 2021; 9:1508. [PMID: 34960254 PMCID: PMC8704861 DOI: 10.3390/vaccines9121508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022] Open
Abstract
At the end of 2020, COVID-19 vaccination programs were initiated in many countries, including Poland. The first vaccine approved in Poland was the BNT162b2 mRNA preparation (Pfizer/BioNTech), and the first vaccinated group were healthcare workers. The aim of the present study was to evaluate post-vaccine antibody titers 8 months after the second vaccine dose had been administered to a group of employees of the Hospital of the Ministry of the Interior and Administration in Olsztyn (Poland). The employees were divided into two groups: persons who had COVID-19 in the fourth quarter of 2020 and were vaccinated in January-February 2021, and persons without a history of COVID-19 who were vaccinated during the same period. The analyzed material was venous blood serum collected from 100 hospital employees on 23-28 September 2021. The level of anti-SARS-CoV-2 S antibodies was measured with a Roche Cobas e411 analyzer using the electrochemiluminescence (ECLIA) method. The study demonstrated that persons with a history of SARS-CoV-2 infection had significantly higher antibody levels (taking into account gender, age, type of work performed, and severity of post-vaccination symptoms) than employees without a history of COVID-19. The study also revealed that the type of work, age, gender, and the course of SARS-CoV-2 infection can influence the humoral immune response. The presented results may prove helpful in the context of administering additional vaccine doses.
Collapse
Affiliation(s)
- Blanka Wolszczak-Biedrzycka
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland;
- The Oncology Center of the Region of Warmia and Mazury in Olsztyn, Hospital of the Ministry of the Interior and Administration, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| | - Anna Bieńkowska
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland;
- The Oncology Center of the Region of Warmia and Mazury in Olsztyn, Hospital of the Ministry of the Interior and Administration, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| | - Justyna Dorf
- Department of Clinical Laboratory Diagnostics, Medical University of Białystok, Waszyngtona 15A, 15-269 Bialystok, Poland;
| |
Collapse
|
150
|
Costiniuk CT, Singer J, Langlois MA, Kulic I, Needham J, Burchell A, Jenabian MA, Walmsley S, Ostrowski M, Kovacs C, Tan D, Harris M, Hull M, Brumme Z, Brockman M, Margolese S, Mandarino E, Angel JB, Routy JP, Anis AH, Cooper C. CTN 328: immunogenicity outcomes in people living with HIV in Canada following vaccination for COVID-19 (HIV-COV): protocol for an observational cohort study. BMJ Open 2021; 11:e054208. [PMID: 34916326 PMCID: PMC8678543 DOI: 10.1136/bmjopen-2021-054208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Most existing vaccines require higher or additional doses or adjuvants to provide similar protection for people living with HIV (PLWH) compared with HIV-uninfected individuals. Additional research is necessary to inform COVID-19 vaccine use in PLWH. METHODS AND ANALYSIS This multicentred observational Canadian cohort study will enrol 400 PLWH aged >16 years from Montreal, Ottawa, Toronto and Vancouver. Subpopulations of PLWH of interest will include individuals: (1) >55 years of age; (2) with CD4 counts <350 cells/mm3; (3) with multimorbidity (>2 comorbidities) and (4) 'stable' or 'reference' PLWH (CD4 T cells >350 cells/mm3, suppressed viral load for >6 months and <1 comorbidity). Data for 1000 HIV-negative controls will be obtained via a parallel cohort study (Stop the Spread Ottawa), using similar time points and methods. Participants receiving >1 COVID-19 vaccine will attend five visits: prevaccination; 1 month following the first vaccine dose; and at 3, 6 and 12 months following the second vaccine dose. The primary end point will be the percentage of PLWH with COVID-19-specific antibodies at 6 months following the second vaccine dose. Humoral and cell-mediated immune responses, and the interplay between T cell phenotypes and inflammatory markers, will be described. Regression techniques will be used to compare COVID-19-specific immune responses to determine whether there are differences between the 'unstable' PLWH group (CD4 <350 cells/mm3), the stable PLWH cohort and the HIV-negative controls, adjusting for factors believed to be associated with immune response. Unadjusted analyses will reveal whether there are differences in driving factors associated with group membership. ETHICS AND DISSEMINATION Research ethics boards at all participating institutions have granted ethics approval for this study. Written informed consent will be obtained from all study participants prior to enrolment. The findings will inform the design of future COVID-19 clinical trials, dosing strategies aimed to improve immune responses and guideline development for PLWH. TRIAL REGISTRATION NUMBER NCT04894448.
Collapse
Affiliation(s)
- Cecilia T Costiniuk
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Joel Singer
- Canadian Institutes of Health Research (CIHR)--Canadian HIV Trials Network and Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Iva Kulic
- Canadian Institutes of Health Research (CIHR)--Canadian HIV Trials Network and Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Judy Needham
- Canadian Institutes of Health Research (CIHR)--Canadian HIV Trials Network and Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Ann Burchell
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Family and Community Medicine, St Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Ontario, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, Quebec, Canada
| | - Sharon Walmsley
- Department of Medicine, Division of Infectious Diseases, Toronto General Hospital, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Clinical Sciences Division and Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Division of Infectious Diseases, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Colin Kovacs
- Maple Leaf Medical Clinic, Toronto, Ontario, Canada
| | - Darrell Tan
- Department of Medicine, Division of Infectious Diseases, University of Toronto, Montreal, Ontario, Canada
- MAP Centre for Urban Health Solutions, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Marianne Harris
- Brisith Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Mark Hull
- Brisith Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Zabrina Brumme
- Brisith Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mark Brockman
- Brisith Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Shari Margolese
- Community Advisory Committee, CIHR Canadian HIV Trials Network, Vancouver, British Columbia, Canada
| | - Enrico Mandarino
- Community Advisory Committee, CIHR Canadian HIV Trials Network, Vancouver, British Columbia, Canada
| | - Jonathan B Angel
- Department of Medicine, Division of Infectious Diseases, The Ottawa Hospital, The Ottawa Hospital Research Institute and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean-Pierre Routy
- Department of Medicine, Division of Hematology and Chronic Viral Illness Service, McGill University Health Centre, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Aslam H Anis
- Canadian Institutes of Health Research (CIHR)--Canadian HIV Trials Network and Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Curtis Cooper
- Department of Medicine, Division of Infectious Diseases, The Ottawa Hospital, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|