201
|
Tan AT, Lim JM, Le Bert N, Kunasegaran K, Chia A, Qui MD, Tan N, Chia WN, de Alwis R, Ying D, Sim JX, Ooi EE, Wang LF, Chen MIC, Young BE, Hsu LY, Low JG, Lye DC, Bertoletti A. Rapid measurement of SARS-CoV-2 spike T cells in whole blood from vaccinated and naturally infected individuals. J Clin Invest 2021; 131:152379. [PMID: 34623327 DOI: 10.1172/jci152379] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/20/2021] [Indexed: 12/26/2022] Open
Abstract
Defining the correlates of protection necessary to manage the COVID-19 pandemic requires the analysis of both antibody and T cell parameters, but the complexity of traditional tests limits virus-specific T cell measurements. We tested the sensitivity and performance of a simple and rapid SARS-CoV-2 spike protein-specific T cell test based on the stimulation of whole blood with peptides covering the SARS-CoV-2 spike protein, followed by cytokine (IFN-γ, IL-2) measurement in different cohorts including BNT162b2-vaccinated individuals (n = 112), convalescent asymptomatic and symptomatic COVID-19 patients (n = 130), and SARS-CoV-1-convalescent individuals (n = 12). The sensitivity of this rapid test is comparable to that of traditional methods of T cell analysis (ELISPOT, activation-induced marker). Using this test, we observed a similar mean magnitude of T cell responses between the vaccinees and SARS-CoV-2 convalescents 3 months after vaccination or virus priming. However, a wide heterogeneity of the magnitude of spike-specific T cell responses characterized the individual responses, irrespective of the time of analysis. The magnitude of these spike-specific T cell responses cannot be predicted from the neutralizing antibody levels. Hence, both humoral and cellular spike-specific immunity should be tested after vaccination to define the correlates of protection necessary to evaluate current vaccine strategies.
Collapse
Affiliation(s)
- Anthony T Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Joey Me Lim
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Kamini Kunasegaran
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Adeline Chia
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Martin Dc Qui
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Nicole Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Wan Ni Chia
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Ruklanthi de Alwis
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Ding Ying
- National Centre for Infectious Diseases (NCID), Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Singapore
| | - Jean Xy Sim
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Eng Eong Ooi
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Barnaby E Young
- National Centre for Infectious Diseases (NCID), Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Singapore
| | - Li Yang Hsu
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Jenny Gh Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - David C Lye
- National Centre for Infectious Diseases (NCID), Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Singapore.,Yong Loo Lin School of Medicine, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Singapore Immunology Network, A*STAR, Singapore
| |
Collapse
|
202
|
Liu W, Russell RM, Bibollet-Ruche F, Skelly AN, Sherrill-Mix S, Freeman DA, Stoltz R, Lindemuth E, Lee FH, Sterrett S, Bar KJ, Erdmann N, Gouma S, Hensley SE, Ketas T, Cupo A, Cruz Portillo VM, Moore JP, Bieniasz PD, Hatziioannou T, Massey G, Minyard MB, Saag MS, Davis RS, Shaw GM, Britt WJ, Leal SM, Goepfert P, Hahn BH. Predictors of Nonseroconversion after SARS-CoV-2 Infection. Emerg Infect Dis 2021; 27:2454-2458. [PMID: 34193339 PMCID: PMC8386781 DOI: 10.3201/eid2709.211042] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Not all persons recovering from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection develop SARS-CoV-2-specific antibodies. We show that nonseroconversion is associated with younger age and higher reverse transcription PCR cycle threshold values and identify SARS-CoV-2 viral loads in the nasopharynx as a major correlate of the systemic antibody response.
Collapse
|
203
|
Vaccine against SARS-CoV-2 in previously infected health care workers. EBioMedicine 2021; 71:103556. [PMID: 34454402 PMCID: PMC8387027 DOI: 10.1016/j.ebiom.2021.103556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 11/29/2022] Open
|
204
|
Lozano-Ojalvo D, Camara C, Lopez-Granados E, Nozal P, Del Pino-Molina L, Bravo-Gallego LY, Paz-Artal E, Pion M, Correa-Rocha R, Ortiz A, Lopez-Hoyos M, Iribarren ME, Portoles J, Rojo-Portoles MP, Ojeda G, Cervera I, Gonzalez-Perez M, Bodega-Mayor I, Montes-Casado M, Portoles P, Perez-Olmeda M, Oteo J, Sanchez-Tarjuelo R, Pothula V, Schwarz M, Brahmachary M, Tan AT, Le Bert N, Berin C, Bertoletti A, Guccione E, Ochando J. Differential effects of the second SARS-CoV-2 mRNA vaccine dose on T cell immunity in naive and COVID-19 recovered individuals. Cell Rep 2021; 36:109570. [PMID: 34390647 PMCID: PMC8332924 DOI: 10.1016/j.celrep.2021.109570] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/15/2021] [Accepted: 07/29/2021] [Indexed: 11/17/2022] Open
Abstract
The rapid development of mRNA-based vaccines against the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) led to the design of accelerated vaccination schedules that have been extremely effective in naive individuals. While a two-dose immunization regimen with the BNT162b2 vaccine has been demonstrated to provide a 95% efficacy in naive individuals, the effects of the second vaccine dose in individuals who have previously recovered from natural SARS-CoV-2 infection has not been investigated in detail. In this study, we characterize SARS-CoV-2 spike-specific humoral and cellular immunity in naive and previously infected individuals during and after two doses of BNT162b2 vaccination. Our results demonstrate that, while the second dose increases both the humoral and cellular immunity in naive individuals, COVID-19 recovered individuals reach their peak of immunity after the first dose. These results suggests that a second dose, according to the current standard regimen of vaccination, may be not necessary in individuals previously infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Daniel Lozano-Ojalvo
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carmen Camara
- Department of Immunology, Hospital La Paz, 28046 Madrid, Spain
| | - Eduardo Lopez-Granados
- Department of Immunology, Hospital La Paz, 28046 Madrid, Spain; Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute of Biomedical Research (IdiPAZ), 28046 Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U767), 28046 Madrid, Spain
| | - Pilar Nozal
- Department of Immunology, Hospital La Paz, 28046 Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U767), 28046 Madrid, Spain; Complement Research Group, La Paz Institute of Biomedical Research (IdiPAZ), 28046 Madrid, Spain
| | - Lucía Del Pino-Molina
- Department of Immunology, Hospital La Paz, 28046 Madrid, Spain; Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute of Biomedical Research (IdiPAZ), 28046 Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U767), 28046 Madrid, Spain
| | - Luz Yadira Bravo-Gallego
- Department of Immunology, Hospital La Paz, 28046 Madrid, Spain; Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute of Biomedical Research (IdiPAZ), 28046 Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U767), 28046 Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, Hospital 12 de Octubre, 28041 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Marjorie Pion
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology, IIS-Fundación Jimenez Díaz, 28040 Madrid, Spain
| | - Marcos Lopez-Hoyos
- Department of Immunology, Hospital Universitario Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain
| | | | - Jose Portoles
- Department of Nephrology, Hospital Puerta de Hierro, 28220 Madrid, Spain
| | | | - Gloria Ojeda
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Isabel Cervera
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Maria Gonzalez-Perez
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Irene Bodega-Mayor
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Maria Montes-Casado
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Pilar Portoles
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain; Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Mayte Perez-Olmeda
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Jesus Oteo
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Rodrigo Sanchez-Tarjuelo
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Venu Pothula
- Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Megan Schwarz
- Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Manisha Brahmachary
- Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Anthony Tanoto Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169547, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169547, Singapore
| | - Cecilia Berin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169547, Singapore
| | - Ernesto Guccione
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Jordi Ochando
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
205
|
Goel RR, Painter MM, Apostolidis SA, Mathew D, Meng W, Rosenfeld AM, Lundgreen KA, Reynaldi A, Khoury DS, Pattekar A, Gouma S, Kuri-Cervantes L, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Baxter AE, Oldridge DA, Giles JR, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Drapeau EM, Dougherty J, Long S, D'Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, Frank I, Betts MR, Vella LA, Grifoni A, Weiskopf D, Sette A, Hensley SE, Davenport MP, Bates P, Luning Prak ET, Greenplate AR, Wherry EJ. mRNA Vaccination Induces Durable Immune Memory to SARS-CoV-2 with Continued Evolution to Variants of Concern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.08.23.457229. [PMID: 34462751 PMCID: PMC8404899 DOI: 10.1101/2021.08.23.457229] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SARS-CoV-2 mRNA vaccines have shown remarkable efficacy, especially in preventing severe illness and hospitalization. However, the emergence of several variants of concern and reports of declining antibody levels have raised uncertainty about the durability of immune memory following vaccination. In this study, we longitudinally profiled both antibody and cellular immune responses in SARS-CoV-2 naïve and recovered individuals from pre-vaccine baseline to 6 months post-mRNA vaccination. Antibody and neutralizing titers decayed from peak levels but remained detectable in all subjects at 6 months post-vaccination. Functional memory B cell responses, including those specific for the receptor binding domain (RBD) of the Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2) variants, were also efficiently generated by mRNA vaccination and continued to increase in frequency between 3 and 6 months post-vaccination. Notably, most memory B cells induced by mRNA vaccines were capable of cross-binding variants of concern, and B cell receptor sequencing revealed significantly more hypermutation in these RBD variant-binding clones compared to clones that exclusively bound wild-type RBD. Moreover, the percent of variant cross-binding memory B cells was higher in vaccinees than individuals who recovered from mild COVID-19. mRNA vaccination also generated antigen-specific CD8+ T cells and durable memory CD4+ T cells in most individuals, with early CD4+ T cell responses correlating with humoral immunity at later timepoints. These findings demonstrate robust, multi-component humoral and cellular immune memory to SARS-CoV-2 and current variants of concern for at least 6 months after mRNA vaccination. Finally, we observed that boosting of pre-existing immunity with mRNA vaccination in SARS-CoV-2 recovered individuals primarily increased antibody responses in the short-term without significantly altering antibody decay rates or long-term B and T cell memory. Together, this study provides insights into the generation and evolution of vaccine-induced immunity to SARS-CoV-2, including variants of concern, and has implications for future booster strategies.
Collapse
Affiliation(s)
- Rishi R Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Mark M Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sokratis A Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Wenzhao Meng
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aaron M Rosenfeld
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kendall A Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip Hicks
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Dysinger
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amanda Hicks
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Harsh Sharma
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sarah Herring
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Scott Korte
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Amy E Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Josephine R Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Madison E Weirick
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christopher M McAllister
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Moses Awofolaju
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicole Tanenbaum
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth M Drapeau
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sherea Long
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kurt D'Andrea
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jacob T Hamilton
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maura McLaughlin
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Justine C Williams
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Sharon Adamski
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ian Frank
- Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael R Betts
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura A Vella
- Division of Infectious Disease, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Scott E Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
206
|
Trougakos IP, Terpos E, Zirou C, Sklirou AD, Apostolakou F, Gumeni S, Charitaki I, Papanagnou ED, Bagratuni T, Liacos CI, Scorilas A, Korompoki E, Papassotiriou I, Kastritis E, Dimopoulos MA. Comparative kinetics of SARS-CoV-2 anti-spike protein RBD IgGs and neutralizing antibodies in convalescent and naïve recipients of the BNT162b2 mRNA vaccine versus COVID-19 patients. BMC Med 2021; 19:208. [PMID: 34420521 PMCID: PMC8380479 DOI: 10.1186/s12916-021-02090-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/09/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Coronavirus SARS-CoV-2, the causative agent of COVID-19, has caused a still evolving global pandemic. Given the worldwide vaccination campaign, the understanding of the vaccine-induced versus COVID-19-induced immunity will contribute to adjusting vaccine dosing strategies and speeding-up vaccination efforts. METHODS Anti-spike-RBD IgGs and neutralizing antibodies (NAbs) titers were measured in BNT162b2 mRNA vaccinated participants (n = 250); we also investigated humoral and cellular immune responses in vaccinated individuals (n = 21) of this cohort 5 months post-vaccination and assayed NAbs levels in COVID-19 hospitalized patients (n = 60) with moderate or severe disease, as well as in COVID-19 recovered patients (n = 34). RESULTS We found that one (boosting) dose of the BNT162b2 vaccine triggers robust immune (i.e., anti-spike-RBD IgGs and NAbs) responses in COVID-19 convalescent healthy recipients, while naïve recipients require both priming and boosting shots to acquire high antibody titers. Severe COVID-19 triggers an earlier and more intense (versus moderate disease) immune response in hospitalized patients; in all cases, however, antibody titers remain at high levels in COVID-19 recovered patients. Although virus infection promotes an earlier and more intense, versus priming vaccination, immune response, boosting vaccination induces antibody titers significantly higher and likely more durable versus COVID-19. In support, high anti-spike-RBD IgGs/NAbs titers along with spike (vaccine encoded antigen) specific T cell clones were found in the serum and peripheral blood mononuclear cells, respectively, of vaccinated individuals 5 months post-vaccination. CONCLUSIONS These findings support vaccination efficacy, also suggesting that vaccination likely offers more protection than natural infection.
Collapse
Affiliation(s)
- Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | | | - Aimilia D Sklirou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Filia Apostolakou
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Charitaki
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni-Dimitra Papanagnou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Tina Bagratuni
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christine-Ivy Liacos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Korompoki
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
207
|
Nersisyan S, Zhiyanov A, Shkurnikov M, Tonevitsky A. T-CoV: a comprehensive portal of HLA-peptide interactions affected by SARS-CoV-2 mutations. Nucleic Acids Res 2021; 50:D883-D887. [PMID: 34396391 PMCID: PMC8385993 DOI: 10.1093/nar/gkab701] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
Rapidly appearing SARS-CoV-2 mutations can affect T cell epitopes, which can help the virus to evade either CD8 or CD4 T-cell responses. We developed T-cell COVID-19 Atlas (T-CoV, https://t-cov.hse.ru) – the comprehensive web portal, which allows one to analyze how SARS-CoV-2 mutations alter the presentation of viral peptides by HLA molecules. The data are presented for common virus variants and the most frequent HLA class I and class II alleles. Binding affinities of HLA molecules and viral peptides were assessed with accurate in silico methods. The obtained results highlight the importance of taking HLA alleles diversity into account: mutation-mediated alterations in HLA-peptide interactions were highly dependent on HLA alleles. For example, we found that the essential number of peptides tightly bound to HLA-B*07:02 in the reference Wuhan variant ceased to be tight binders for the Indian (Delta) and the UK (Alpha) variants. In summary, we believe that T-CoV will help researchers and clinicians to predict the susceptibility of individuals with different HLA genotypes to infection with variants of SARS-CoV-2 and/or forecast its severity.
Collapse
Affiliation(s)
- Stepan Nersisyan
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | - Anton Zhiyanov
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Maxim Shkurnikov
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
208
|
Gancino M, Santiago Vispo N. Hybrid immunity: the immune response of COVID-19 survivors to vaccination. BIONATURA 2021. [DOI: 10.21931/rb/2021.06.03.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Hybrid immunity can become a turning-point opportunity to defeat the infection in context with the current pandemic scenario. Adopting policies aligned with the data mentioned above may be particularly beneficial for regions like Latin America. While this region shares only ~5% of the world population, it accumulates +16% of the ~200 million COVID-19 total cases, so far reported1. Acknowledging the reduced diagnostic capabilities of these countries, this entire area is probably one of the most affected globally, leading to one of the more numerous populations of convalescent people. From both an economic and pharmacological perspective, applying a one-dose regime of pertinent vaccines to convalescent individuals is sustainable5. As COVID-19 survivors may need just one vaccination to achieve high levels of protective immunity, massive antibody screening for SARS-CoV-2 spike antibodies could help prioritize and free up doses, optimize vaccine supply efficiency, and surpass problems linked to the current vaccine manufacturing bottleneck
Collapse
Affiliation(s)
- Marlon Gancino
- Faculty of Health, NANOMED EMJMD, University of Paris, France
| | - Nelson Santiago Vispo
- Yachay Tech University, School of Biological Sciences and engineering, Hda. San José s/n y Proyecto Yachay, 100119, Urcuquí, . Ecuador
| |
Collapse
|
209
|
Havervall S, Marking U, Greilert-Norin N, Ng H, Gordon M, Salomonsson AC, Hellström C, Pin E, Blom K, Mangsbo S, Phillipson M, Klingström J, Hober S, Nilsson P, Åberg M, Thålin C. Antibody responses after a single dose of ChAdOx1 nCoV-19 vaccine in healthcare workers previously infected with SARS-CoV-2. EBioMedicine 2021; 70:103523. [PMID: 34391088 PMCID: PMC8357428 DOI: 10.1016/j.ebiom.2021.103523] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recent reports demonstrate robust serological responses to a single dose of messenger RNA (mRNA) vaccines in individuals previously infected with SARS-CoV-2. Data on immune responses following a single-dose adenovirus-vectored vaccine expressing the SARS-CoV-2 spike protein (ChAdOx1 nCoV-19) in individuals with previous SARS-CoV-2 infection are however limited, and current guidelines recommend a two-dose regimen regardless of preexisting immunity. METHODS We compared RBD-specific IgG and RBD-ACE2 blocking antibodies against SARS-CoV-2 wild type and variants of concern following two doses of the mRNA vaccine BNT162b2 in SARS-CoV-2 naïve healthcare workers (n=65) and a single dose of the adenovector vaccine ChAdOx1 nCoV-19 in 82 healthcare workers more than (n=45) and less than (n=37) 11 months post mild SARS-CoV-2 infection at time of vaccination. FINDINGS The post-vaccine levels of RBD-specific IgG and neutralizing antibodies against the SARS-CoV-2 wild type and variants of concern including Delta lineage 1.617.2 were similar or higher in participants receiving a single dose of ChAdOx1 nCoV-19 vaccine post SARS-CoV-2 infection (both more than and less than 11 months post infection) compared to SARS-CoV-2 naïve participants who received two doses of BNT162b2 vaccine. INTERPRETATION Our data support that a single dose ChAdOx1 nCoV-19 vaccine that is administered up to at least 11 months post SARS-CoV-2 infection serves as an effective immune booster. This provides a possible rationale for a single-dose vaccine regimen. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Sebastian Havervall
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Ulrika Marking
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Nina Greilert-Norin
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Henry Ng
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden; Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Max Gordon
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | | | - Cecilia Hellström
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Elisa Pin
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Kim Blom
- Public Health Agency of Sweden, Solna, Sweden
| | - Sara Mangsbo
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jonas Klingström
- Public Health Agency of Sweden, Solna, Sweden; Centre for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Sophia Hober
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Peter Nilsson
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry. Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden.
| |
Collapse
|
210
|
Urbanowicz RA, Tsoleridis T, Jackson HJ, Cusin L, Duncan JD, Chappell JG, Tarr AW, Nightingale J, Norrish AR, Ikram A, Marson B, Craxford SJ, Kelly A, Aithal GP, Vijay A, Tighe PJ, Ball JK, Valdes AM, Ollivere BJ. Two doses of the SARS-CoV-2 BNT162b2 vaccine enhance antibody responses to variants in individuals with prior SARS-CoV-2 infection. Sci Transl Med 2021; 13:eabj0847. [PMID: 34376569 PMCID: PMC9835846 DOI: 10.1126/scitranslmed.abj0847] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Understanding the impact of prior infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on the response to vaccination is a priority for responding to the coronavirus disease 2019 (COVID-19) pandemic. In particular, it is necessary to understand how prior infection plus vaccination can modulate immune responses against variants of concern. To address this, we sampled 20 individuals with and 25 individuals without confirmed previous SARS-CoV-2 infection from a large cohort of health care workers followed serologically since April 2020. All 45 individuals had received two doses of the Pfizer-BioNTech BNT162b2 vaccine with a delayed booster at 10 weeks. Absolute and neutralizing antibody titers against wild-type SARS-CoV-2 and variants were measured using enzyme immunoassays and pseudotype neutralization assays. We observed antibody reactivity against lineage A, B.1.351, and P.1 variants with increasing antigenic exposure, through either vaccination or natural infection. This improvement was further confirmed in neutralization assays using fixed dilutions of serum samples. The impact of antigenic exposure was more evident in enzyme immunoassays measuring SARS-CoV-2 spike protein–specific IgG antibody concentrations. Our data show that multiple exposures to SARS-CoV-2 spike protein in the context of a delayed booster expand the neutralizing breadth of the antibody response to neutralization-resistant SARS-CoV-2 variants. This suggests that additional vaccine boosts may be beneficial in improving immune responses against future SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
- Richard A. Urbanowicz
- Wolfson Centre for Global Virus Research, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,School of Life Sciences, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool Science Park IC2, 146 Brownlow Hill, Liverpool L3 5RF, UK
| | - Theocharis Tsoleridis
- Wolfson Centre for Global Virus Research, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,School of Life Sciences, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Hannah J. Jackson
- School of Life Sciences, University of Nottingham, Life Sciences Building, University Park Campus, Nottingham NG7 2RD, UK
| | - Lola Cusin
- School of Life Sciences, University of Nottingham, Life Sciences Building, University Park Campus, Nottingham NG7 2RD, UK
| | - Joshua D. Duncan
- Wolfson Centre for Global Virus Research, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,School of Life Sciences, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Joseph G. Chappell
- Wolfson Centre for Global Virus Research, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,School of Life Sciences, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Alexander W. Tarr
- Wolfson Centre for Global Virus Research, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,School of Life Sciences, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Jessica Nightingale
- Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Trauma and Orthopaedics, University Hospitals Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Alan R. Norrish
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Adeel Ikram
- Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Trauma and Orthopaedics, University Hospitals Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Ben Marson
- Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Trauma and Orthopaedics, University Hospitals Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Simon J. Craxford
- Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Trauma and Orthopaedics, University Hospitals Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Anthony Kelly
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Guruprasad P. Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Amrita Vijay
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK
| | - Patrick J. Tighe
- School of Life Sciences, University of Nottingham, Life Sciences Building, University Park Campus, Nottingham NG7 2RD, UK
| | - Jonathan K. Ball
- Wolfson Centre for Global Virus Research, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,School of Life Sciences, University of Nottingham, A Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Corresponding author. (J.K.B.); (A.M.V.); (B.J.O.)
| | - Ana M. Valdes
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Corresponding author. (J.K.B.); (A.M.V.); (B.J.O.)
| | - Benjamin J. Ollivere
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Injury, Inflammation & Recovery Sciences, School of Medicine, University of Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Trauma and Orthopaedics, University Hospitals Nottingham, C Floor, West Block, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, UK.,Corresponding author. (J.K.B.); (A.M.V.); (B.J.O.)
| |
Collapse
|
211
|
Zollner A, Watschinger C, Rössler A, Farcet MR, Penner A, Böhm V, Kiechl SJ, Stampfel G, Hintenberger R, Tilg H, Koch R, Antlanger M, Kreil TR, Kimpel J, Moschen AR. B and T cell response to SARS-CoV-2 vaccination in health care professionals with and without previous COVID-19. EBioMedicine 2021; 70:103539. [PMID: 34391087 PMCID: PMC8358275 DOI: 10.1016/j.ebiom.2021.103539] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In recent months numerous health care professional acquired COVID-19 at the workplace resulting in significant shortages in medical and nursing staff. We investigated how prior COVID-19 affects SARS-CoV-2 vaccination and how such knowledge could facilitate frugal vaccination strategies. METHODS In a cohort of 41 healthcare professionals with (n=14) and without (n=27) previous SARS-CoV-2 infection, we assessed the immune status before, during and after vaccination with BNT162b2. The humoral immune response was assessed by receptor binding domain ELISA and different SARS-CoV-2 neutralisation assays using wildtype and pseudo-typed viruses. T cell immunity against SARS-CoV-2 surface and nucleocapsid peptides were studied using interferon-γ release assays and intracellular flow cytometry. Vaccine-related side effects were captured. FINDINGS Prior COVID-19 resulted in improved vaccine responses both in the B and T cell compartment. In vaccine recipients with prior COVID-19, the first vaccine dose induced high antibody concentrations comparable to seronegative vaccine recipients after two injections. This translated into more efficient neutralisation of virus particles, even more pronounced than expected from the RBD ELISA results. Furthermore, T cell responses were stronger in convalescents and particularly strong against the SARS-CoV-2 nucleocapsid protein. INTERPRETATION Herein, we corroborate recent findings suggesting that in convalescents a single vaccine dose is sufficient to boost adequate in vitro neutralisation of SARS-CoV-2 and therefore may be sufficient to induce adequate protection against severe COVID-19. New spike mutated virus variants render the highly conserved nucleocapsid protein - eliciting strong SARS-CoV-2 specific T cell immunity - an interesting additional vaccine target. FUNDING Christian Doppler Research Association, Johannes Kepler University Linz.
Collapse
Affiliation(s)
- Andreas Zollner
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Department of Medicine, Division of Internal Medicine 1 (Gastroenterology and Hepatology, Endocrinology and Metabolism), Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Watschinger
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Department of Internal Medicine 2 (Gastroenterology and Hepatology, Endocrinology and Metabolism, Nephrology, Rheumatology), Johannes Kepler University Linz, Linz, Austria
| | - Annika Rössler
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck Austria
| | - Maria R Farcet
- Global Pathogen Safety, Baxter AG (part of Takeda), Vienna, Austria
| | - Agnes Penner
- Department of Internal Medicine 2 (Gastroenterology and Hepatology, Endocrinology and Metabolism, Nephrology, Rheumatology), Johannes Kepler University Linz, Linz, Austria
| | - Vincent Böhm
- Department of Internal Medicine 2 (Gastroenterology and Hepatology, Endocrinology and Metabolism, Nephrology, Rheumatology), Johannes Kepler University Linz, Linz, Austria
| | - Sophia J Kiechl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria; VASCage, Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Gerald Stampfel
- Department of Internal Medicine 2 (Gastroenterology and Hepatology, Endocrinology and Metabolism, Nephrology, Rheumatology), Johannes Kepler University Linz, Linz, Austria
| | - Rainer Hintenberger
- Department of Internal Medicine 2 (Gastroenterology and Hepatology, Endocrinology and Metabolism, Nephrology, Rheumatology), Johannes Kepler University Linz, Linz, Austria
| | - Herbert Tilg
- Department of Medicine, Division of Internal Medicine 1 (Gastroenterology and Hepatology, Endocrinology and Metabolism), Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Koch
- Department of Medicine, Division of Internal Medicine 1 (Gastroenterology and Hepatology, Endocrinology and Metabolism), Medical University of Innsbruck, Innsbruck, Austria
| | - Marlies Antlanger
- Department of Internal Medicine 2 (Gastroenterology and Hepatology, Endocrinology and Metabolism, Nephrology, Rheumatology), Johannes Kepler University Linz, Linz, Austria
| | - Thomas R Kreil
- Global Pathogen Safety, Baxter AG (part of Takeda), Vienna, Austria
| | - Janine Kimpel
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck Austria
| | - Alexander R Moschen
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Department of Medicine, Division of Internal Medicine 1 (Gastroenterology and Hepatology, Endocrinology and Metabolism), Medical University of Innsbruck, Innsbruck, Austria; Department of Internal Medicine 2 (Gastroenterology and Hepatology, Endocrinology and Metabolism, Nephrology, Rheumatology), Johannes Kepler University Linz, Linz, Austria.
| |
Collapse
|
212
|
Tretyn A, Szczepanek J, Skorupa M, Jarkiewicz-Tretyn J, Sandomierz D, Dejewska J, Ciechanowska K, Jarkiewicz-Tretyn A, Koper W, Pałgan K. Differences in the Concentration of Anti-SARS-CoV-2 IgG Antibodies Post-COVID-19 Recovery or Post-Vaccination. Cells 2021; 10:1952. [PMID: 34440721 PMCID: PMC8391384 DOI: 10.3390/cells10081952] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/20/2022] Open
Abstract
At the end of 2020, population-based vaccination programs with new generation mRNA-based vaccines began almost all over the world. The aim of the study was to evaluate the titer of anti-SARS-CoV-2 IgG antibodies against the S1 subunit of the virus's spike protein as a marker of the humoral response in 477 patients and the concentration of interferon-gamma as an indicator of cellular response in 28 individuals. In our studies, we used serological enzyme-linked immunosorbent assays. IgG was measured in weeks 2 and 3 after the first dose and 1-5 weeks after the second dose of an mRNA vaccine in seropositive and seronegative individuals as well as in symptomatic and asymptomatic convalescents. High levels of antibodies were observed in 98% of our vaccinated cohort, and the presence of protective T cells was confirmed in the blood samples of all participants. The humoral immune response is diversified and is visible as early as 2-3 weeks after the first dose of the mRNA vaccine. The level of protection increased significantly after the second dose, with the increase being much greater in pre-vaccine healthy subjects and less in convalescents. In the second and third weeks after the second dose, the concentration of IgG antibodies was the highest, and in the following weeks, it decreased gradually. Regular serological measurements on eight subjects show that antibody titers are lower four months after vaccination than before the second dose.
Collapse
Affiliation(s)
- Andrzej Tretyn
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland; (A.T.); (M.S.); (J.D.)
| | - Joanna Szczepanek
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, ul. Wilenska 4, 87-100 Torun, Poland
| | - Monika Skorupa
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland; (A.T.); (M.S.); (J.D.)
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, ul. Wilenska 4, 87-100 Torun, Poland
| | - Joanna Jarkiewicz-Tretyn
- Non-Public Health Care Centre, Cancer Genetics Laboratory, 87-100 Torun, Poland; (J.J.-T.); (D.S.); (K.C.); (A.J.-T.)
| | - Dorota Sandomierz
- Non-Public Health Care Centre, Cancer Genetics Laboratory, 87-100 Torun, Poland; (J.J.-T.); (D.S.); (K.C.); (A.J.-T.)
| | - Joanna Dejewska
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland; (A.T.); (M.S.); (J.D.)
- Non-Public Health Care Centre, Cancer Genetics Laboratory, 87-100 Torun, Poland; (J.J.-T.); (D.S.); (K.C.); (A.J.-T.)
| | - Karolina Ciechanowska
- Non-Public Health Care Centre, Cancer Genetics Laboratory, 87-100 Torun, Poland; (J.J.-T.); (D.S.); (K.C.); (A.J.-T.)
| | - Aleksander Jarkiewicz-Tretyn
- Non-Public Health Care Centre, Cancer Genetics Laboratory, 87-100 Torun, Poland; (J.J.-T.); (D.S.); (K.C.); (A.J.-T.)
- Polish-Japanese Academy of Information Technology, 02-008 Warszawa, Poland
| | - Wojciech Koper
- The Voivodeship Sanitary-Epidemiological Station in Bydgoszcz, 85-031 Bydgoszcz, Poland;
| | - Krzysztof Pałgan
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland;
| |
Collapse
|
213
|
Taborska P, Lastovicka J, Stakheev D, Strizova Z, Bartunkova J, Smrz D. SARS-CoV-2 spike glycoprotein-reactive T cells can be readily expanded from COVID-19 vaccinated donors. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1452-1467. [PMID: 34314576 PMCID: PMC8427053 DOI: 10.1002/iid3.496] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/02/2021] [Accepted: 07/13/2021] [Indexed: 12/16/2022]
Abstract
Introduction The COVID‐19 vaccine was designed to provide protection against infection by the severe respiratory coronavirus 2 (SARS‐CoV‐2) and coronavirus disease 2019 (COVID‐19). However, the vaccine's efficacy can be compromised in patients with immunodeficiencies or the vaccine‐induced immunoprotection suppressed by other comorbidity treatments, such as chemotherapy or immunotherapy. To enhance the protective role of the COVID‐19 vaccine, we have investigated a combination of the COVID‐19 vaccination with ex vivo enrichment and large‐scale expansion of SARS‐CoV‐2 spike glycoprotein‐reactive CD4+ and CD8+ T cells. Methods SARS‐CoV‐2‐unexposed donors were vaccinated with two doses of the BNT162b2 SARS‐CoV‐2 vaccine. The peripheral blood mononuclear cells of the vaccinated donors were cell culture‐enriched with T cells reactive to peptides derived from SARS‐CoV‐2 spike glycoprotein. The enriched cell cultures were large‐scale expanded using the rapid expansion protocol (REP) and the peptide‐reactive T cells were evaluated. Results We show that vaccination with the SARS‐CoV‐2 spike glycoprotein‐based mRNA COVID‐19 vaccine‐induced humoral response against SARS‐CoV‐2 spike glycoprotein in all tested healthy SARS‐CoV‐2‐unexposed donors. This humoral response was found to correlate with the ability of the donors' PBMCs to become enriched with SARS‐CoV‐2 spike glycoprotein‐reactive CD4+ and CD8+ T cells. Using an 11‐day REP, the enriched cell cultures were expanded nearly 1000‐fold, and the proportions of the SARS‐CoV‐2 spike glycoprotein‐reactive T cells increased. Conclusion These findings show for the first time that the combination of the COVID‐19 vaccination and ex vivo T cell large‐scale expansion of SARS‐CoV‐2‐reactive T cells could be a powerful tool for developing T cell‐based adoptive cellular immunotherapy of COVID‐19.
Collapse
Affiliation(s)
- Pavla Taborska
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan Lastovicka
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Dmitry Stakheev
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jirina Bartunkova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Daniel Smrz
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
214
|
Wall EC, Wu M, Harvey R, Kelly G, Warchal S, Sawyer C, Daniels R, Adams L, Hobson P, Hatipoglu E, Ngai Y, Hussain S, Ambrose K, Hindmarsh S, Beale R, Riddell A, Gamblin S, Howell M, Kassiotis G, Libri V, Williams B, Swanton C, Gandhi S, Bauer DL. AZD1222-induced neutralising antibody activity against SARS-CoV-2 Delta VOC. Lancet 2021; 398:207-209. [PMID: 34197809 PMCID: PMC8238446 DOI: 10.1016/s0140-6736(21)01462-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Emma C Wall
- Francis Crick Institute, London NW1 1AT, UK; National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, UK; NIHR UCLH Clinical Research Facility, London, UK
| | - Mary Wu
- Francis Crick Institute, London NW1 1AT, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | - Rupert Beale
- Francis Crick Institute, London NW1 1AT, UK; NIHR UCLH Clinical Research Facility, London, UK; University College London, London, UK
| | | | | | | | - George Kassiotis
- Francis Crick Institute, London NW1 1AT, UK; Department of Infectious Disease, St Mary's Hospital, Imperial College London, London, UK
| | - Vincenzo Libri
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, UK; NIHR UCLH Clinical Research Facility, London, UK; University College London, London, UK
| | - Bryan Williams
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, UK; NIHR UCLH Clinical Research Facility, London, UK; University College London, London, UK
| | - Charles Swanton
- Francis Crick Institute, London NW1 1AT, UK; University College London, London, UK
| | - Sonia Gandhi
- Francis Crick Institute, London NW1 1AT, UK; University College London, London, UK
| | | |
Collapse
|
215
|
Abstract
The rapid and remarkably successful development, manufacture, and deployment of several effective severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is now tempered by three key challenges. First, reducing virus transmission will require prevention of asymptomatic and mild infections in addition to severe symptomatic infections. Second, the emergence of variants of concern with mutations in the S protein's receptor binding domain increases the likelihood that vaccines will have to be updated because some of these mutations render variants less optimally targeted by current vaccines. This will require coordinated global SARS-CoV-2 surveillance to link genotypes to phenotypes, potentially using the WHO's global influenza surveillance program as a guide. Third, concerns about the longevity of vaccine-induced immunity highlight the potential need for re-vaccination, depending on the extent to which the virus has been controlled and whether re-vaccination can target those at greatest risk of severe illness. Fortunately, as I discuss in this review, these challenges can be addressed.
Collapse
Affiliation(s)
- Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza; Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| |
Collapse
|
216
|
High Prevalence of Anti-PF4 Antibodies Following ChAdOx1 nCov-19 (AZD1222) Vaccination Even in the Absence of Thrombotic Events. Vaccines (Basel) 2021; 9:vaccines9070712. [PMID: 34358129 PMCID: PMC8309977 DOI: 10.3390/vaccines9070712] [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: 05/24/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
It is unclear whether the ChAdOx1 nCov-19 vaccine can induce the development of anti-PF4 antibodies in vaccinated individuals who have not developed thrombosis. The aim of this prospective study was to evaluate the presence of antibodies against heparin/PF4 in adults who received a first dose of the ChAdOx1 nCov-19 vaccine, and correlate them with clinical data and antibody responses to the vaccine. We detected non-platelet activating anti-PF4 antibodies in 67% (29/43) of the vaccinated individuals on day 22 following the first dose of the ChAdOx1 nCov-19 vaccine, though these were detected in low titers. Furthermore, there was no correlation between the presence of anti-PF4 IgG antibodies and the baseline clinical characteristics of the patients. Our findings suggest that the ChAdOx1 nCov-19 vaccine can elicit anti-PF4 antibody production even in recipients without a clinical manifestation of thrombosis. The presence of anti-PF4 antibodies was not sufficient to provoke clinically evident thrombosis. Our results offer an important insight into the ongoing investigations regarding the underlying multifactorial pathophysiology of thrombotic events induced by the ChAdOx1 nCov-19 vaccine.
Collapse
|
217
|
Wang Z, Muecksch F, Schaefer-Babajew D, Finkin S, Viant C, Gaebler C, Hoffmann HH, Barnes CO, Cipolla M, Ramos V, Oliveira TY, Cho A, Schmidt F, Da Silva J, Bednarski E, Aguado L, Yee J, Daga M, Turroja M, Millard KG, Jankovic M, Gazumyan A, Zhao Z, Rice CM, Bieniasz PD, Caskey M, Hatziioannou T, Nussenzweig MC. Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection. Nature 2021; 595:426-431. [PMID: 34126625 PMCID: PMC8277577 DOI: 10.1038/s41586-021-03696-9] [Citation(s) in RCA: 518] [Impact Index Per Article: 172.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/04/2021] [Indexed: 02/05/2023]
Abstract
More than one year after its inception, the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains difficult to control despite the availability of several working vaccines. Progress in controlling the pandemic is slowed by the emergence of variants that appear to be more transmissible and more resistant to antibodies1,2. Here we report on a cohort of 63 individuals who have recovered from COVID-19 assessed at 1.3, 6.2 and 12 months after SARS-CoV-2 infection, 41% of whom also received mRNA vaccines3,4. In the absence of vaccination, antibody reactivity to the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number of RBD-specific memory B cells remain relatively stable between 6 and 12 months after infection. Vaccination increases all components of the humoral response and, as expected, results in serum neutralizing activities against variants of concern similar to or greater than the neutralizing activity against the original Wuhan Hu-1 strain achieved by vaccination of naive individuals2,5-8. The mechanism underlying these broad-based responses involves ongoing antibody somatic mutation, memory B cell clonal turnover and development of monoclonal antibodies that are exceptionally resistant to SARS-CoV-2 RBD mutations, including those found in the variants of concern4,9. In addition, B cell clones expressing broad and potent antibodies are selectively retained in the repertoire over time and expand markedly after vaccination. The data suggest that immunity in convalescent individuals will be very long lasting and that convalescent individuals who receive available mRNA vaccines will produce antibodies and memory B cells that should be protective against circulating SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Zijun Wang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | | | - Shlomo Finkin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Charlotte Viant
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Christian Gaebler
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Hans- Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Christopher O Barnes
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Melissa Cipolla
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Victor Ramos
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Alice Cho
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Fabian Schmidt
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Justin Da Silva
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Eva Bednarski
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Lauren Aguado
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Jim Yee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mridushi Daga
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Martina Turroja
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Katrina G Millard
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Mila Jankovic
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Zhen Zhao
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA.
| | | | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
218
|
Abstract
COVID-19 vaccine responses provide insights into how the immune system perceives threats
Collapse
Affiliation(s)
- Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), 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
| |
Collapse
|
219
|
Wang Z, Muecksch F, Schaefer-Babajew D, Finkin S, Viant C, Gaebler C, Hoffmann HH, Barnes CO, Cipolla M, Ramos V, Oliveira TY, Cho A, Schmidt F, da Silva J, Bednarski E, Aguado L, Yee J, Daga M, Turroja M, Millard KG, Jankovic M, Gazumyan A, Zhao Z, Rice CM, Bieniasz PD, Caskey M, Hatziioannou T, Nussenzweig MC. Naturally enhanced neutralizing breadth to SARS-CoV-2 after one year. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34100013 DOI: 10.1101/2021.05.07.443175] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Over one year after its inception, the coronavirus disease-2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remains difficult to control despite the availability of several excellent vaccines. Progress in controlling the pandemic is slowed by the emergence of variants that appear to be more transmissible and more resistant to antibodies 1,2 . Here we report on a cohort of 63 COVID-19-convalescent individuals assessed at 1.3, 6.2 and 12 months after infection, 41% of whom also received mRNA vaccines 3,4 . In the absence of vaccination antibody reactivity to the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number of RBD-specific memory B cells remain relatively stable from 6 to 12 months. Vaccination increases all components of the humoral response, and as expected, results in serum neutralizing activities against variants of concern that are comparable to or greater than neutralizing activity against the original Wuhan Hu-1 achieved by vaccination of naïve individuals 2,5-8 . The mechanism underlying these broad-based responses involves ongoing antibody somatic mutation, memory B cell clonal turnover, and development of monoclonal antibodies that are exceptionally resistant to SARS-CoV-2 RBD mutations, including those found in variants of concern 4,9 . In addition, B cell clones expressing broad and potent antibodies are selectively retained in the repertoire over time and expand dramatically after vaccination. The data suggest that immunity in convalescent individuals will be very long lasting and that convalescent individuals who receive available mRNA vaccines will produce antibodies and memory B cells that should be protective against circulating SARS-CoV-2 variants.
Collapse
|
220
|
Pegu A, O’Connell S, Schmidt SD, O’Dell S, Talana CA, Lai L, Albert J, Anderson E, Bennett H, Corbett KS, Flach B, Jackson L, Leav B, Ledgerwood JE, Luke CJ, Makowski M, Roberts PC, Roederer M, Rebolledo PA, Rostad CA, Rouphael NG, Shi W, Wang L, Widge AT, Yang ES, Beigel JH, Graham BS, Mascola JR, Suthar MS, McDermott A, Doria-Rose NA. Durability of mRNA-1273-induced antibodies against SARS-CoV-2 variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.13.444010. [PMID: 34031659 PMCID: PMC8142657 DOI: 10.1101/2021.05.13.444010] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SARS-CoV-2 mutations may diminish vaccine-induced protective immune responses, and the durability of such responses has not been previously reported. Here, we present a comprehensive assessment of the impact of variants B.1.1.7, B.1.351, P.1, B.1.429, and B.1.526 on binding, neutralizing, and ACE2-blocking antibodies elicited by the vaccine mRNA-1273 over seven months. Cross-reactive neutralizing responses were rare after a single dose of mRNA-1273. At the peak of response to the second dose, all subjects had robust responses to all variants. Binding and functional antibodies against variants persisted in most subjects, albeit at low levels, for 6 months after the primary series of mRNA-1273. Across all assays, B.1.351 had the greatest impact on antibody recognition, and B.1.1.7 the least. These data complement ongoing studies of clinical protection to inform the potential need for additional boost vaccinations. ONE-SENTENCE SUMMARY Most mRNA-1273 vaccinated individuals maintained binding and functional antibodies against SARS-CoV-2 variants for 6 months.
Collapse
Affiliation(s)
- Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Sarah O’Connell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Stephen D Schmidt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Sijy O’Dell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Chloe A. Talana
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Lilin Lai
- Department of Medicine, Center for Childhood Infections and Vaccines (CCIV) of Children’s Healthcare of Atlanta, Emory Vaccine Center, and Emory University Department of Pediatrics, Emory University School of Medicine; Atlanta, GA, USA
| | | | - Evan Anderson
- Department of Medicine, Center for Childhood Infections and Vaccines (CCIV) of Children’s Healthcare of Atlanta, Emory Vaccine Center, and Emory University Department of Pediatrics, Emory University School of Medicine; Atlanta, GA, USA
| | | | - Kizzmekia S. Corbett
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Britta Flach
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Lisa Jackson
- Kaiser Permanente Washington Health Research Institute; Seattle, WA, USA
| | | | - Julie E. Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Catherine J. Luke
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD, USA
| | | | - Paul C. Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Paulina A. Rebolledo
- Hope Clinic, Department of Medicine, Emory University School of Medicine; Decatur, GA, USA
| | - Christina A. Rostad
- Department of Medicine, Center for Childhood Infections and Vaccines (CCIV) of Children’s Healthcare of Atlanta, Emory Vaccine Center, and Emory University Department of Pediatrics, Emory University School of Medicine; Atlanta, GA, USA
| | - Nadine G. Rouphael
- Hope Clinic, Department of Medicine, Emory University School of Medicine; Decatur, GA, USA
| | - Wei Shi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Alicia T. Widge
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | | | - John H. Beigel
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Mehul S. Suthar
- Department of Medicine, Center for Childhood Infections and Vaccines (CCIV) of Children’s Healthcare of Atlanta, Emory Vaccine Center, and Emory University Department of Pediatrics, Emory University School of Medicine; Atlanta, GA, USA
| | - Adrian McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| | - Nicole A. Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda MD, USA
| |
Collapse
|