301
|
Mak WA, Koeleman JG, van der Vliet M, Keuren F, Ong DS. SARS-CoV-2 antibody and T cell responses one year after COVID-19 and the booster effect of vaccination: A prospective cohort study. J Infect 2022; 84:171-178. [PMID: 34896516 PMCID: PMC8656179 DOI: 10.1016/j.jinf.2021.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/04/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES First, to describe SARS-CoV-2 T cell and antibody responses in a prospective cohort of healthcare workers that suffered from mild to moderate COVID-19 approximately one year ago. Second, to assess COVID-19 vaccine-induced immune responses in these prior-infected individuals. METHODS SARS-CoV-2-specific T cell and anti-SARS-CoV-2-Spike-RBD immunoglobulin G (IgG) responses in blood were determined before COVID-19 vaccination with mRNA-1273, BNT162b2, Ad26.CoV2-S or ChAdOx1-S, two weeks after first vaccination, and after second vaccination. RESULTS 55 prior SARS-CoV-2 infected and seroconverted individuals were included. S1-specific T cell responses and anti-RBD IgG were detectable one year post SARS-CoV-2 infection: 24 spot-forming cells per 106 peripheral blood mononuclear cells (SFCs/106 PBMCs) after S1 stimulation and anti-RBD IgG concentration of 74 (IQR 36-158) IU/mL. Responses after the first and second vaccination were comparable with S1-specfic T cell responses of 198 (IQR 137-359) and 180 (IQR 103-347) SFCs/106 PBMCs, and IgG concentrations of 6792 (IQR 3386-15,180) and 6326 (IQR 2336-13,440) IU/mL, respectively. These responses retained up to four months after vaccination. CONCLUSIONS Both T cell and IgG responses against SARS-CoV-2 persist for up to one year after COVID-19. A second COVID-19 vaccination in prior-infected individuals did not further increase immune responses in comparison to one vaccination.
Collapse
Affiliation(s)
- Willem A. Mak
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - Johannes G.M. Koeleman
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - Marijke van der Vliet
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - Frans Keuren
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - David S.Y. Ong
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Universiteitsweg 100, Utrecht 3584 GC, the Netherlands,Corresponding author at: Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| |
Collapse
|
302
|
Minervina AA, Pogorelyy MV, Kirk AM, Crawford JC, Allen EK, Chou CH, Mettelman RC, Allison KJ, Lin CY, Brice DC, Zhu X, Vegesana K, Wu G, Trivedi S, Kottapalli P, Darnell D, McNeely S, Olsen SR, Schultz-Cherry S, Estepp JH, McGargill MA, Wolf J, Thomas PG. SARS-CoV-2 antigen exposure history shapes phenotypes and specificity of memory CD8 T cells. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2021.07.12.21260227. [PMID: 34341799 PMCID: PMC8328067 DOI: 10.1101/2021.07.12.21260227] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although mRNA vaccine efficacy against severe COVID-19 remains high, variant emergence and breakthrough infections have changed vaccine policy to include booster immunizations. However, the effect of diverse and repeated antigen exposures on SARS-CoV-2 memory T cells is poorly understood. Here, we utilize DNA-barcoded MHC-multimers combined with scRNAseq and scTCRseq to capture the ex vivo profile of SARS-CoV-2-responsive T cells within a cohort of individuals with one, two, or three antigen exposures, including vaccination, primary infection, and breakthrough infection. We found that the order of exposure determined the relative distribution between spike- and non-spike-specific responses, with vaccination after infection leading to further expansion of spike-specific T cells and differentiation to a CCR7-CD45RA+ effector phenotype. In contrast, individuals experiencing a breakthrough infection mount vigorous non-spike-specific responses. In-depth analysis of over 4,000 epitope-specific T cell receptor sequences demonstrates that all types of exposures elicit diverse repertoires characterized by shared, dominant TCR motifs, with no evidence for repertoire narrowing from repeated exposure. Our findings suggest that breakthrough infections diversify the T cell memory repertoire and that current vaccination protocols continue to expand and differentiate spike-specific memory responses.
Collapse
Affiliation(s)
| | - Mikhail V. Pogorelyy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Allison M. Kirk
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | | | - E. Kaitlynn Allen
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Ching-Heng Chou
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Robert C. Mettelman
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Kim J. Allison
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Chun-Yang Lin
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - David C. Brice
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Xun Zhu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Kasi Vegesana
- Information Services, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Sanchit Trivedi
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Pratibha Kottapalli
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Daniel Darnell
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Suzanne McNeely
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Scott R. Olsen
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Jeremie H. Estepp
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN USA
| | | | - Maureen A. McGargill
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Joshua Wolf
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| |
Collapse
|
303
|
Mazzoni A, Vanni A, Spinicci M, Capone M, Lamacchia G, Salvati L, Coppi M, Antonelli A, Carnasciali A, Farahvachi P, Giovacchini N, Aiezza N, Malentacchi F, Zammarchi L, Liotta F, Rossolini GM, Bartoloni A, Cosmi L, Maggi L, Annunziato F. SARS-CoV-2 Spike-Specific CD4+ T Cell Response Is Conserved Against Variants of Concern, Including Omicron. Front Immunol 2022; 13:801431. [PMID: 35154116 PMCID: PMC8826050 DOI: 10.3389/fimmu.2022.801431] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/10/2022] [Indexed: 12/16/2022] Open
Abstract
Although accumulating data have investigated the effect of SARS-CoV-2 mutations on antibody neutralizing activity, less is known about T cell immunity. In this work, we found that the ancestral (Wuhan strain) Spike protein can efficaciously reactivate CD4+ T cell memory in subjects with previous Alpha variant infection. This finding has practical implications, as in many countries only one vaccine dose is currently administered to individuals with previous COVID-19, independently of which SARS-CoV-2 variant was responsible of the infection. We also found that only a minority of Spike-specific CD4+ T cells targets regions mutated in Alpha, Beta and Delta variants, both after natural infection and vaccination. Finally, we found that the vast majority of Spike-specific CD4+ T cell memory response induced by natural infection or mRNA vaccination is conserved also against Omicron variant. This is of importance, as this newly emerged strain is responsible for a sudden rise in COVID-19 cases worldwide due to its increased transmissibility and ability to evade antibody neutralization. Collectively, these observations suggest that most of the memory CD4+ T cell response is conserved against SARS-CoV-2 variants of concern, providing an efficacious line of defense that can protect from the development of severe forms of COVID-19.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Anna Vanni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Disease Unit, Careggi University Hospital, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giulia Lamacchia
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Marco Coppi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alberto Antonelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alberto Carnasciali
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Parham Farahvachi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Nicla Giovacchini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Noemi Aiezza
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Disease Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Disease Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| |
Collapse
|
304
|
Murugesan K, Jagannathan P, Altamirano J, Maldonado YA, Bonilla HF, Jacobson KB, Parsonnet J, Andrews JR, Shi RZ, Boyd S, Pinsky BA, Singh U, Banaei N. Long-Term Accuracy of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Interferon-γ Release Assay and Its Application in Household Investigation. Clin Infect Dis 2022; 75:e314-e321. [PMID: 35079772 PMCID: PMC8807306 DOI: 10.1093/cid/ciac045] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND An immunodiagnostic assay that sensitively detects a cell-mediated immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is needed for epidemiological investigation and for clinical assessment of T- cell-mediated immune response to vaccines, particularly in the context of emerging variants that might escape antibody responses. METHODS The performance of a whole blood interferon-gamma (IFN-γ) release assay (IGRA) for the detection of SARS-CoV-2 antigen-specific T cells was evaluated in coronavirus disease 2019 (COVID-19) convalescents tested serially up to 10 months post-infection and in healthy blood donors. SARS-CoV-2 IGRA was applied in contacts of households with index cases. Freshly collected blood in the lithium heparin tube was left unstimulated, stimulated with a SARS-CoV-2 peptide pool, and stimulated with mitogen. RESULTS The overall sensitivity and specificity of IGRA were 84.5% (153/181; 95% confidence interval [CI]: 79.0-89.0) and 86.6% (123/142; 95% CI: 80.0-91.2), respectively. The sensitivity declined from 100% (16/16; 95% CI: 80.6-100) at 0.5-month post-infection to 79.5% (31/39; 95% CI: 64.4-89.2) at 10 months post-infection (P < .01). The IFN-γ response remained relatively robust at 10 months post-infection (3.8 vs 1.3 IU/mL, respectively). In 14 households, IGRA showed a positivity rate of 100% (12/12) and 65.2% (15/23), and IgG of 50.0% (6/12) and 43.5% (10/23) in index cases and contacts, respectively, exhibiting a difference of + 50% (95% CI: +25.4 to +74.6) and +21.7% (95% CI: +9.23 to +42.3), respectively. Either IGRA or IgG was positive in 100% (12/12) of index cases and 73.9% (17/23) of contacts. CONCLUSIONS The SARS-CoV-2 IGRA is a useful clinical diagnostic tool for assessing cell-mediated immune response to SARS-CoV-2.
Collapse
Affiliation(s)
- Kanagavel Murugesan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Prasanna Jagannathan
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan Altamirano
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yvonne A Maldonado
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Hector F Bonilla
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Karen B Jacobson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Julie Parsonnet
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Run-Zhang Shi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Scott Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin A Pinsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA,Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA,Clinical Virology Laboratory, Stanford Health Care, Stanford, CA, USA
| | - Upinder Singh
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA,Department of Microbiology and Immunology
| | - Niaz Banaei
- Corresponding Author: Niaz Banaei MD , 3375 Hillview Ave, Rm. 1602, Palo Alto, Ca 94304 USA, Phone 650-736-8052,
| |
Collapse
|
305
|
Hyun YS, Lee YH, Jo HA, Baek IC, Kim SM, Sohn HJ, Kim TG. Comprehensive Analysis of CD4 + T Cell Response Cross-Reactive to SARS-CoV-2 Antigens at the Single Allele Level of HLA Class II. Front Immunol 2022; 12:774491. [PMID: 35069546 PMCID: PMC8770530 DOI: 10.3389/fimmu.2021.774491] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Common human coronaviruses have been circulating undiagnosed worldwide. These common human coronaviruses share partial sequence homology with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); therefore, T cells specific to human coronaviruses are also cross-reactive with SARS-CoV-2 antigens. Herein, we defined CD4+ T cell responses that were cross-reactive with SARS-CoV-2 antigens in blood collected in 2016–2018 from healthy donors at the single allele level using artificial antigen-presenting cells (aAPC) expressing a single HLA class II allotype. We assessed the allotype-restricted responses in the 42 individuals using the aAPCs matched 22 HLA-DR alleles, 19 HLA-DQ alleles, and 13 HLA-DP alleles. The response restricted by the HLA-DR locus showed the highest magnitude, and that by HLA-DP locus was higher than that by HLA-DQ locus. Since two alleles of HLA-DR, -DQ, and -DP loci are expressed co-dominantly in an individual, six different HLA class II allotypes can be used to the cross-reactive T cell response. Of the 16 individuals who showed a dominant T cell response, five, one, and ten showed a dominant response by a single allotype of HLA-DR, -DQ, and -DP, respectively. The single allotype-restricted T cells responded to only one antigen in the five individuals and all the spike, membrane, and nucleocapsid proteins in the six individuals. In individuals heterozygous for the HLA-DPA and HLA-DPB loci, four combinations of HLA-DP can be expressed, but only one combination showed a dominant response. These findings demonstrate that cross-reactive T cells to SARS-CoV-2 respond with single-allotype dominance.
Collapse
Affiliation(s)
- You-Seok Hyun
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yong-Hun Lee
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyeong-A Jo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - In-Cheol Baek
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sun-Mi Kim
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyun-Jung Sohn
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Tai-Gyu Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
306
|
Rüthrich MM, Giesen N, Mellinghoff SC, Rieger CT, von Lilienfeld-Toal M. Cellular Immune Response after Vaccination in Patients with Cancer-Review on Past and Present Experiences. Vaccines (Basel) 2022; 10:182. [PMID: 35214642 PMCID: PMC8875094 DOI: 10.3390/vaccines10020182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 01/27/2023] Open
Abstract
Patients with cancer are at particular risk for infection but also have diminished vaccine responses, usually quantified by the level of specific antibodies. Nonetheless, vaccines are specifically recommended in this vulnerable patient group. Here, we discuss the cellular part of the vaccine response in patients with cancer. We summarize the experience with vaccines prior to and during the SARS-CoV-2 pandemic in different subgroups, and we discuss why, especially in patients with cancer, T cells may be the more reliable correlate of protection. Finally, we provide a brief outlook on options to improve the cellular response to vaccines.
Collapse
Affiliation(s)
- Maria Madeleine Rüthrich
- Department of Internal Medicine II, Hematology and Medical Oncology, Universitätsklinikum Jena, Am Klinikum 1, 07747 Jena, Germany;
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institut, Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| | - Nicola Giesen
- Department of Haematology and Oncology, Internal Medicine V, University Hospital Heidelberg, 69115 Heidelberg, Germany;
| | - Sibylle C. Mellinghoff
- Centre for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, University of Cologne, 50923 Cologne, Germany;
| | - Christina T. Rieger
- Hemato-Oncology Germering & Interdisciplinary Tumorcenter, Ludwig-Maximilians-University Munich, 81377 Munich, Germany;
| | - Marie von Lilienfeld-Toal
- Department of Internal Medicine II, Hematology and Medical Oncology, Universitätsklinikum Jena, Am Klinikum 1, 07747 Jena, Germany;
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institut, Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| |
Collapse
|
307
|
Synthetic multiantigen MVA vaccine COH04S1 protects against SARS-CoV-2 in Syrian hamsters and non-human primates. NPJ Vaccines 2022; 7:7. [PMID: 35064109 PMCID: PMC8782996 DOI: 10.1038/s41541-022-00436-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022] Open
Abstract
Second-generation COVID-19 vaccines could contribute to establish protective immunity against SARS-CoV-2 and its emerging variants. We developed COH04S1, a synthetic multiantigen modified vaccinia Ankara-based SARS-CoV-2 vaccine that co-expresses spike and nucleocapsid antigens. Here, we report COH04S1 vaccine efficacy in animal models. We demonstrate that intramuscular or intranasal vaccination of Syrian hamsters with COH04S1 induces robust Th1-biased antigen-specific humoral immunity and cross-neutralizing antibodies (NAb) and protects against weight loss, lower respiratory tract infection, and lung injury following intranasal SARS-CoV-2 challenge. Moreover, we demonstrate that single-dose or two-dose vaccination of non-human primates with COH04S1 induces robust antigen-specific binding antibodies, NAb, and Th1-biased T cells, protects against both upper and lower respiratory tract infection following intranasal/intratracheal SARS-CoV-2 challenge, and triggers potent post-challenge anamnestic antiviral responses. These results demonstrate COH04S1-mediated vaccine protection in animal models through different vaccination routes and dose regimens, complementing ongoing investigation of this multiantigen SARS-CoV-2 vaccine in clinical trials.
Collapse
|
308
|
Stieber F, Allen N, Carpenter K, Howard J, Alagna R, Manissero D, Nikolayevskyy V. Accuracy of Interferon Gamma Release Assays for the COVID-19 immunity assessment. J Virol Methods 2022; 302:114472. [PMID: 35065949 PMCID: PMC8772062 DOI: 10.1016/j.jviromet.2022.114472] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 01/29/2023]
Abstract
Emerging evidence suggests that T-cells play a significant role in COVID-19 immunity both in the context of natural infection and vaccination. Easy to use IGRA assays including QFN SARS are considered attractive alternatives to more “traditional” but laborious methods for detection of SARS-CoV-2-specific T-cell responses. In our Letter we are proposing explanations to an apparently lower than expected T-cell responses (44 % reactive individuals) reported by Krüttgen et al in a small cohort of healthy double vaccinated individuals. These results could have been affected by reporting raw optical density values instead of calculated Interferon-ɣ concentrations which is supported by unexpectedly low mitogen responses in healthy individuals. This study highlights an importance of adhering to good laboratory practice principles as well as overall importance of accurate T-cell immunity assessment using IGRA assays.
Collapse
Affiliation(s)
- Francis Stieber
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Nadia Allen
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Kara Carpenter
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Jenny Howard
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | | | - Davide Manissero
- QIAGEN Manchester, Citylabs 2.0 Hathersage Road, Manchester, M130BH, United Kingdom
| | | |
Collapse
|
309
|
Hirabara SM, Serdan TDA, Gorjao R, Masi LN, Pithon-Curi TC, Covas DT, Curi R, Durigon EL. SARS-COV-2 Variants: Differences and Potential of Immune Evasion. Front Cell Infect Microbiol 2022; 11:781429. [PMID: 35118007 PMCID: PMC8805732 DOI: 10.3389/fcimb.2021.781429] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022] Open
Abstract
The structural spike (S) glycoprotein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) plays an essential role in infection and is an important target for neutralizing antibody recognition. Mutations in the S gene can generate variants of concern (VOCs), which improve “viral fitness” through selective or survival advantages, such as increased ACE-2 receptor affinity, infectivity, viral replication, higher transmissibility, resistance to neutralizing antibodies and immune escape, increasing disease severity and reinfection risk. Five VOCs have been recognized and include B.1.1.7 (U.K.), B.1.351 (South Africa), P.1 (Brazil), B.1.617.2 (India), and B.1.1.529 (multiple countries). In this review, we addressed the following critical points concerning VOCs: a) characteristics of the SARS-CoV-2 VOCs with mutations in the S gene; b) possible evasion of variants from neutralizing antibodies generated through vaccination, previous infection, or immune therapies; c) potential risk of new pandemic waves induced by the variants worldwide; and d) perspectives for further studies and actions aimed at preventing or reducing the impact of new variants during the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Sandro M. Hirabara
- Interdisciplinary Program of Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
- *Correspondence: Sandro M. Hirabara,
| | - Tamires D. A. Serdan
- Department of Molecular Pathobiology, New York University, New York, NY, United States
| | - Renata Gorjao
- Interdisciplinary Program of Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Laureane N. Masi
- Interdisciplinary Program of Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Tania C. Pithon-Curi
- Interdisciplinary Program of Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Dimas T. Covas
- Butantan Institute, São Paulo, Brazil
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rui Curi
- Interdisciplinary Program of Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
- Immunobiological Production Section, Bioindustrial Center, Butantan Institute, São Paulo, Brazil
| | - Edison L. Durigon
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Scientific Platform Pasteur University of São Paulo, São Paulo, Brazil
| |
Collapse
|
310
|
Tarabini RF, Rigo MM, Faustino Fonseca A, Rubin F, Bellé R, Kavraki LE, Ferreto TC, Amaral Antunes D, de Souza APD. Large-Scale Structure-Based Screening of Potential T Cell Cross-Reactivities Involving Peptide-Targets From BCG Vaccine and SARS-CoV-2. Front Immunol 2022; 12:812176. [PMID: 35095907 PMCID: PMC8793865 DOI: 10.3389/fimmu.2021.812176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/21/2021] [Indexed: 12/22/2022] Open
Abstract
Although not being the first viral pandemic to affect humankind, we are now for the first time faced with a pandemic caused by a coronavirus. The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been responsible for the COVID-19 pandemic, which caused more than 4.5 million deaths worldwide. Despite unprecedented efforts, with vaccines being developed in a record time, SARS-CoV-2 continues to spread worldwide with new variants arising in different countries. Such persistent spread is in part enabled by public resistance to vaccination in some countries, and limited access to vaccines in other countries. The limited vaccination coverage, the continued risk for resistant variants, and the existence of natural reservoirs for coronaviruses, highlight the importance of developing additional therapeutic strategies against SARS-CoV-2 and other coronaviruses. At the beginning of the pandemic it was suggested that countries with Bacillus Calmette-Guérin (BCG) vaccination programs could be associated with a reduced number and/or severity of COVID-19 cases. Preliminary studies have provided evidence for this relationship and further investigation is being conducted in ongoing clinical trials. The protection against SARS-CoV-2 induced by BCG vaccination may be mediated by cross-reactive T cell lymphocytes, which recognize peptides displayed by class I Human Leukocyte Antigens (HLA-I) on the surface of infected cells. In order to identify potential targets of T cell cross-reactivity, we implemented an in silico strategy combining sequence-based and structure-based methods to screen over 13,5 million possible cross-reactive peptide pairs from BCG and SARS-CoV-2. Our study produced (i) a list of immunogenic BCG-derived peptides that may prime T cell cross-reactivity against SARS-CoV-2, (ii) a large dataset of modeled peptide-HLA structures for the screened targets, and (iii) new computational methods for structure-based screenings that can be used by others in future studies. Our study expands the list of BCG peptides potentially involved in T cell cross-reactivity with SARS-CoV-2-derived peptides, and identifies multiple high-density "neighborhoods" of cross-reactive peptides which could be driving heterologous immunity induced by BCG vaccination, therefore providing insights for future vaccine development efforts.
Collapse
Affiliation(s)
- Renata Fioravanti Tarabini
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Health Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | | | - André Faustino Fonseca
- Antunes Lab, Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Felipe Rubin
- School of Technology - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rafael Bellé
- Laboratório de alto desempenho – Centro de Apoio ao desenvolvimento cientifico e tecnológico da (IDEIA), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Lydia E Kavraki
- Kavraki Lab, Department of Computer Science, Rice University, Houston, TX, United States
| | - Tiago Coelho Ferreto
- School of Technology - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,Laboratório de alto desempenho – Centro de Apoio ao desenvolvimento cientifico e tecnológico da (IDEIA), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Dinler Amaral Antunes
- Antunes Lab, Department of Biology and Biochemistry, University of Houston, Houston, TX, United States,*Correspondence: Ana Paula Duarte de Souza, ; Dinler Amaral Antunes,
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Health Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,*Correspondence: Ana Paula Duarte de Souza, ; Dinler Amaral Antunes,
| |
Collapse
|
311
|
Abstract
The process of adaptation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to humans probably had started decades ago, when its ancestor diverged from the bat coronavirus. The adaptive process comprises strategies the virus uses to overcome the respiratory tract defense barriers and replicate and shed in the host cells. These strategies include the impairment of interferon production, hiding immunogenic motifs, avoiding viral RNA detection, manipulating cell autophagy, triggering host cell death, inducing lymphocyte exhaustion and depletion, and finally, mutation and escape from immunity. In addition, SARS-CoV-2 employs strategies to take advantage of host cell resources for its benefits, such as inhibiting the ubiquitin-proteasome system, hijacking mitochondria functions, and usage of enhancing antibodies. It may be anticipated that as the tradeoffs of adaptation progress, the virus destructive burden will gradually subside. Some evidence suggests that SARS-CoV-2 will become part of the human respiratory virome, as had occurred with other coronaviruses, and coevolve with its host.
Collapse
Affiliation(s)
- Eduardo Tosta
- Universidade de Brasília, Faculdade de Medicina, Brasília, DF, Brasil
| |
Collapse
|
312
|
Siracusano G, Ruggiero A, Bisoffi Z, Piubelli C, Carbonare LD, Valenti MT, Mayora-Neto M, Temperton N, Lopalco L, Zipeto D. Different decay of antibody response and VOC sensitivity in naïve and previously infected subjects at 15 weeks following vaccination with BNT162b2. J Transl Med 2022; 20:22. [PMID: 34998405 PMCID: PMC8742572 DOI: 10.1186/s12967-021-03208-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/17/2021] [Indexed: 02/01/2023] Open
Abstract
Background COVID-19 vaccines have demonstrated effectiveness in reducing SARS-CoV-2 mild and severe outcomes. In vaccinated subjects with SARS-CoV-2 history, RBD-specific IgG and pseudovirus neutralization titers were rapidly recalled by a single BTN162b2 vaccine dose to higher levels than those in naïve recipients after the second dose, irrespective of waning immunity. In this study, we inspected the long-term kinetic and neutralizing responses of S-specific IgG induced by two administrations of BTN162b2 vaccine in infection-naïve subjects and in subjects previously infected with SARS-CoV-2. Methods Twenty-six naïve and 9 previously SARS-CoV-2 infected subjects during the second wave of the pandemic in Italy were enrolled for this study. The two groups had comparable demographic and clinical characteristics. By means of ELISA and pseudotyped-neutralization assays, we investigated the kinetics of developed IgG-RBD and their neutralizing activity against both the ancestral D614G and the SARS-CoV-2 variants of concern emerged later, respectively. The Wilcoxon matched pair signed rank test and the Kruskal–Wallis test with Dunn’s correction for multiple comparison were applied when needed. Results Although after 15 weeks from vaccination IgG-RBD dropped in all participants, naïve subjects experienced a more dramatic decline than those with previous SARS-CoV-2 infection. Neutralizing antibodies remained higher in subjects with SARS-CoV-2 history and conferred broad-spectrum protection. Conclusions These data suggest that hybrid immunity to SARS-CoV-2 has a relevant impact on the development of IgG-RBD upon vaccination. However, the rapid decay of vaccination-elicited antibodies highlights that the administration of a third dose is expected to boost the response and acquire high levels of cross-neutralizing antibodies. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03208-3.
Collapse
Affiliation(s)
- Gabriel Siracusano
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy.
| | - Alessandra Ruggiero
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Zeno Bisoffi
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital Negrar, Verona, Italy.,Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Chiara Piubelli
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital Negrar, Verona, Italy
| | | | | | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, ME7, 47B, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, ME7, 47B, UK
| | - Lucia Lopalco
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Donato Zipeto
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
313
|
Gagne M, Corbett KS, Flynn BJ, Foulds KE, Wagner DA, Andrew SF, Todd JPM, Honeycutt CC, McCormick L, Nurmukhambetova ST, Davis-Gardner ME, Pessaint L, Bock KW, Nagata BM, Minai M, Werner AP, Moliva JI, Tucker C, Lorang CG, Zhao B, McCarthy E, Cook A, Dodson A, Teng IT, Mudvari P, Roberts-Torres J, Laboune F, Wang L, Goode A, Kar S, Boyoglu-Barnum S, Yang ES, Shi W, Ploquin A, Doria-Rose N, Carfi A, Mascola JR, Boritz EA, Edwards DK, Andersen H, Lewis MG, Suthar MS, Graham BS, Roederer M, Moore IN, Nason MC, Sullivan NJ, Douek DC, Seder RA. Protection from SARS-CoV-2 Delta one year after mRNA-1273 vaccination in rhesus macaques coincides with anamnestic antibody response in the lung. Cell 2022; 185:113-130.e15. [PMID: 34921774 PMCID: PMC8639396 DOI: 10.1016/j.cell.2021.12.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
mRNA-1273 vaccine efficacy against SARS-CoV-2 Delta wanes over time; however, there are limited data on the impact of durability of immune responses on protection. Here, we immunized rhesus macaques and assessed immune responses over 1 year in blood and upper and lower airways. Serum neutralizing titers to Delta were 280 and 34 reciprocal ID50 at weeks 6 (peak) and 48 (challenge), respectively. Antibody-binding titers also decreased in bronchoalveolar lavage (BAL). Four days after Delta challenge, the virus was unculturable in BAL, and subgenomic RNA declined by ∼3-log10 compared with control animals. In nasal swabs, sgRNA was reduced by 1-log10, and the virus remained culturable. Anamnestic antibodies (590-fold increased titer) but not T cell responses were detected in BAL by day 4 post-challenge. mRNA-1273-mediated protection in the lungs is durable but delayed and potentially dependent on anamnestic antibody responses. Rapid and sustained protection in upper and lower airways may eventually require a boost.
Collapse
Affiliation(s)
- Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kizzmekia S Corbett
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danielle A Wagner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shayne F Andrew
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John-Paul M Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lauren McCormick
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Saule T Nurmukhambetova
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Meredith E Davis-Gardner
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Kevin W Bock
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Anne P Werner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Courtney Tucker
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cynthia G Lorang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bingchun Zhao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth McCarthy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Prakriti Mudvari
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jesmine Roberts-Torres
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Farida Laboune
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Shi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aurélie Ploquin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eli A Boritz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
314
|
Naranbhai V, Nathan A, Kaseke C, Berrios C, Khatri A, Choi S, Getz MA, Tano-Menka R, Ofoman O, Gayton A, Senjobe F, Denis KJS, Lam EC, Garcia-Beltran WF, Balazs AB, Walker BD, Iafrate AJ, Gaiha GD. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all prior infected and vaccinated individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.01.04.21268586. [PMID: 35018386 PMCID: PMC8750712 DOI: 10.1101/2022.01.04.21268586] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The SARS-CoV-2 Omicron variant (B.1.1.529) contains mutations that mediate escape from infection and vaccine-induced antibody responses, although the extent to which these substitutions in spike and non-spike proteins affect T cell recognition is unknown. Here we show that T cell responses in individuals with prior infection, vaccination, both prior infection and vaccination, and boosted vaccination are largely preserved to Omicron spike and non-spike proteins. However, we also identify a subset of individuals (∼21%) with a >50% reduction in T cell reactivity to the Omicron spike. Evaluation of functional CD4 + and CD8 + memory T cell responses confirmed these findings and reveal that reduced recognition to Omicron spike is primarily observed within the CD8 + T cell compartment. Booster vaccination substantially enhanced T cell responses to Omicron spike. In contrast to neutralizing immunity, these findings suggest preservation of T cell responses to the Omicron variant, although with reduced reactivity in some individuals.
Collapse
|
315
|
Boedecker-Lips SC, Lautem A, Runkel S, Klimpke P, Kraus D, Keil P, Holtz S, Tomalla V, Marczynski P, Boedecker CB, Galle PR, Koch M, Weinmann-Menke J. Six-Month Follow-Up after Vaccination with BNT162b2: SARS-CoV-2 Antigen-Specific Cellular and Humoral Immune Responses in Hemodialysis Patients and Kidney Transplant Recipients. Pathogens 2022; 11:67. [PMID: 35056015 PMCID: PMC8780885 DOI: 10.3390/pathogens11010067] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/30/2021] [Accepted: 01/02/2022] [Indexed: 02/01/2023] Open
Abstract
Hemodialysis patients (HDP) and kidney transplant recipients (KTR) have a high risk of infection with SARS-CoV-2 with poor clinical outcomes. Because of this, vaccination of these groups of patients against SARS-CoV-2 is particularly important. However, immune responses may be impaired in immunosuppressed and chronically ill patients. Here, our aim was to compare the efficacy of an mRNA-based vaccine in HDP, KTR, and healthy subjects. DESIGN In this prospective observational cohort study, the humoral and cellular response of prevalent 192 HDP, 50 KTR, and 28 healthy controls (HC) was assessed 1, 2, and 6 months after the first immunization with the BNT162b2 mRNA vaccine. RESULTS After 6 months, 97.5% of HDP, 37.9% of KTR, and 100% of HC had an antibody response. Median antibody levels were 1539.7 (±3355.8), 178.5 (±369.5), and 2657.8 (±2965.8) AU/mL in HDP, KTR, and HC, respectively (p ≤ 0.05). A SARS-CoV-2 antigen-specific cell response to vaccination was found in 68.8% of HDP, 64.5% of KTR, and 90% of HC. CONCLUSION The humoral response rates to mRNA-based vaccination of HDPs are comparable to HCs, but antibody titers are lower. Furthermore, HDPs have weaker T-cell response to vaccination than HCs. KTRs have very low humoral and antigen-specific cellular response rates and antibody titers, which requires other vaccination strategies in addition to booster vaccination.
Collapse
Affiliation(s)
- Simone Cosima Boedecker-Lips
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
| | - Anja Lautem
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (A.L.); (C.B.B.); (M.K.)
| | - Stefan Runkel
- Blood Transfusion Center, University Medical Center Mainz, Johannes-Gutenberg University, D 55131 Mainz, Germany;
| | - Pascal Klimpke
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
| | - Daniel Kraus
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
| | - Philipp Keil
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
| | - Stefan Holtz
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
| | - Vanessa Tomalla
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
| | - Paul Marczynski
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
| | - Christian Benedikt Boedecker
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (A.L.); (C.B.B.); (M.K.)
| | - Peter Robert Galle
- Department of Internal Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany;
| | - Martina Koch
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (A.L.); (C.B.B.); (M.K.)
| | - Julia Weinmann-Menke
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany; (S.C.B.-L.); (P.K.); (D.K.); (P.K.); (S.H.); (V.T.); (P.M.)
- Research Center of Immunotherapy (FZI), University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| |
Collapse
|
316
|
Ahmed SF, Quadeer AA, McKay MR. SARS-CoV-2 T Cell Responses Elicited by COVID-19 Vaccines or Infection Are Expected to Remain Robust against Omicron. Viruses 2022; 14:79. [PMID: 35062283 PMCID: PMC8781795 DOI: 10.3390/v14010079] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/26/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
Omicron, the most recent SARS-CoV-2 variant of concern (VOC), harbours multiple mutations in the spike protein that were not observed in previous VOCs. Initial studies suggest Omicron to substantially reduce the neutralizing capability of antibodies induced from vaccines and previous infection. However, its effect on T cell responses remains to be determined. Here, we assess the effect of Omicron mutations on known T cell epitopes and report data suggesting T cell responses to remain broadly robust against this new variant.
Collapse
Affiliation(s)
- Syed Faraz Ahmed
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China;
| | - Ahmed Abdul Quadeer
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China;
| | - Matthew R. McKay
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China;
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Department of Electrical and Electronic Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| |
Collapse
|
317
|
Kwok SL, Cheng SM, Leung JN, Leung K, Lee CK, Peiris JM, Wu JT. Waning antibody levels after COVID-19 vaccination with mRNA Comirnaty and inactivated CoronaVac vaccines in blood donors, Hong Kong, April 2020 to October 2021. Euro Surveill 2022; 27:2101197. [PMID: 35027105 PMCID: PMC8759113 DOI: 10.2807/1560-7917.es.2022.27.2.2101197] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/13/2022] [Indexed: 04/09/2023] Open
Abstract
The mRNA vaccine Comirnaty and the inactivated vaccine CoronaVac are both available in Hong Kong's COVID-19 vaccination programme. We observed waning antibody levels in 850 fully vaccinated (at least 14 days passed after second dose) blood donors using ELISA and surrogate virus neutralisation test. The Comirnaty-vaccinated group's (n = 593) antibody levels remained over the ELISA and sVNT positive cut-offs within the first 6 months. The CoronaVac-vaccinated group's (n = 257) median antibody levels began to fall below the cut-offs 4 months after vaccination.
Collapse
Affiliation(s)
- Shirley Ll Kwok
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Samuel Ms Cheng
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection (C2i), Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Jennifer Ns Leung
- Hong Kong Red Cross Blood Transfusion Service, Hospital Authority, Hong Kong Special Administrative Region, China
| | - Kathy Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Cheuk-Kwong Lee
- Hong Kong Red Cross Blood Transfusion Service, Hospital Authority, Hong Kong Special Administrative Region, China
| | - Js Malik Peiris
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection (C2i), Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Joseph T Wu
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science Park, Hong Kong Special Administrative Region, China
| |
Collapse
|
318
|
Simnani FZ, Singh D, Kaur R. COVID-19 phase 4 vaccine candidates, effectiveness on SARS-CoV-2 variants, neutralizing antibody, rare side effects, traditional and nano-based vaccine platforms: a review. 3 Biotech 2022; 12:15. [PMID: 34926119 PMCID: PMC8665991 DOI: 10.1007/s13205-021-03076-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic has endangered world health and the economy. As the number of cases is increasing, different companies have started developing potential vaccines using both traditional and nano-based platforms to overcome the pandemic. Several countries have approved a few vaccine candidates for emergency use authorization (EUA), showing significant effectiveness and inducing a robust immune response. Oxford-AstraZeneca, Pfizer-BioNTech's BNT162, Moderna's mRNA-1273, Sinovac's CoronaVac, Johnson & Johnson, Sputnik-V, and Sinopharm's vaccine candidates are leading the race. However, the SARS-CoV-2 is constantly mutating, making the vaccines less effective, possibly by escaping immune response for some variants. Besides, some EUA vaccines have been reported to induce rare side effects such as blood clots, cardiac injury, anaphylaxis, and some neurological effects. Although the COVID-19 vaccine candidates promise to overcome the pandemic, a more significant and clear understanding is needed. In this review, we brief about the clinical trial of some leading candidates, their effectiveness, and their neutralizing effect on SARS-CoV-2 variants. Further, we have discussed the rare side effects, different traditional and nano-based platforms to understand the scope of future development.
Collapse
Affiliation(s)
| | - Dibyangshee Singh
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024 India
| | - Ramneet Kaur
- Department of Life Sciences, RIMT University, Ludhiana, Punjab India
| |
Collapse
|
319
|
Rowe LA, Beddingfield BJ, Goff K, Killeen SZ, Chirichella NR, Melton A, Roy CJ, Maness NJ. Intra-Host SARS-CoV-2 Evolution in the Gut of Mucosally-Infected Chlorocebus aethiops (African Green Monkeys). Viruses 2022; 14:77. [PMID: 35062281 PMCID: PMC8777858 DOI: 10.3390/v14010077] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 02/04/2023] Open
Abstract
In recent months, several SARS-CoV-2 variants have emerged that enhance transmissibility and escape host humoral immunity. Hence, the tracking of viral evolutionary trajectories is clearly of great importance. Little is known about SARS-CoV-2 evolution in nonhuman primate models used to test vaccines and therapies and to model human disease. Viral RNA was sequenced from rectal swabs from Chlorocebus aethiops (African green monkeys) after experimental respiratory SARS-CoV-2 infection. Two distinct patterns of viral evolution were identified that were shared between all collected samples. First, mutations in the furin cleavage site that were initially present in the virus as a consequence of VeroE6 cell culture adaptation were not detected in viral RNA recovered in rectal swabs, confirming the necessity of this motif for viral infection in vivo. Three amino acid changes were also identified; ORF 1a S2103F, and spike D215G and H655Y, which were detected in rectal swabs from all sampled animals. These findings are demonstrative of intra-host SARS-CoV-2 evolution and may identify a host-adapted variant of SARS-CoV-2 that would be useful in future primate models involving SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lori A. Rowe
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
| | - Brandon J. Beddingfield
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
| | - Stephanie Z. Killeen
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
| | - Nicole R. Chirichella
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
| | - Alexandra Melton
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
| | - Chad J. Roy
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, LA 70433, USA; (L.A.R.); (B.J.B.); (K.G.); (S.Z.K.); (N.R.C.); (A.M.); (C.J.R.)
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
320
|
Mukhopadhyay L, Gupta N, Yadav PD, Aggarwal N. Neutralization assays for SARS-CoV-2: Implications for assessment of protective efficacy of COVID-19 vaccines. Indian J Med Res 2022; 155:105-122. [PMID: 35859437 PMCID: PMC9552365 DOI: 10.4103/ijmr.ijmr_2544_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: 08/16/2021] [Indexed: 11/04/2022] Open
Abstract
The WHO emergency use-listed (EUL) COVID-19 vaccines were developed against early strains of SARS-CoV-2. With the emergence of SARS-CoV-2 variants of concern (VOCs) - Alpha, Beta, Gamma, Delta and Omicron, it is necessary to assess the neutralizing activity of these vaccines against the VOCs. PubMed and preprint platforms were searched for literature on neutralizing activity of serum from WHO EUL vaccine recipients, against the VOCs, using appropriate search terms till November 30, 2021. Our search yielded 91 studies meeting the inclusion criteria. The analysis revealed a drop of 0-8.9-fold against Alpha variant, 0.3-42.4-fold against Beta variant, 0-13.8-fold against Gamma variant and 1.35-20-fold against Delta variant in neutralization titres of serum from the WHO EUL COVID-19 vaccine recipients, as compared to early SARS-CoV-2 isolates. The wide range of variability was due to differences in the choice of virus strains selected for neutralization assays (pseudovirus or live virus), timing of serum sample collection after the final dose of vaccine (day 0 to 8 months) and sample size (ranging from 5 to 470 vaccinees). The reasons for this variation have been discussed and the possible way forward to have uniformity across neutralization assays in different laboratories have been described, which will generate reliable data. Though in vitro neutralization studies are a valuable tool to estimate the performance of vaccines against the backdrop of emerging variants, the results must be interpreted with caution and corroborated with field-effectiveness studies.
Collapse
Affiliation(s)
- Labanya Mukhopadhyay
- Virology Unit, Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India
| | - Nivedita Gupta
- Virology Unit, Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India
| | - Pragya D. Yadav
- Maximum Containment Laboratory, Indian Council of Medical Research-National Institute of Virology, Pune, Maharashtra, India
| | - Neeraj Aggarwal
- Virology Unit, Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India
| |
Collapse
|
321
|
Stuart ASV, Shaw RH, Liu X, Greenland M, Aley PK, Andrews NJ, Cameron JC, Charlton S, Clutterbuck EA, Collins AM, Darton T, Dinesh T, Duncan CJA, England A, Faust SN, Ferreira DM, Finn A, Goodman AL, Green CA, Hallis B, Heath PT, Hill H, Horsington BM, Lambe T, Lazarus R, Libri V, Lillie PJ, Mujadidi YF, Payne R, Plested EL, Provstgaard-Morys S, Ramasamy MN, Ramsay M, Read RC, Robinson H, Screaton GR, Singh N, Turner DPJ, Turner PJ, Vichos I, White R, Nguyen-Van-Tam JS, Snape MD. Immunogenicity, safety, and reactogenicity of heterologous COVID-19 primary vaccination incorporating mRNA, viral-vector, and protein-adjuvant vaccines in the UK (Com-COV2): a single-blind, randomised, phase 2, non-inferiority trial. Lancet 2022; 399:36-49. [PMID: 34883053 PMCID: PMC8648333 DOI: 10.1016/s0140-6736(21)02718-5] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Given the importance of flexible use of different COVID-19 vaccines within the same schedule to facilitate rapid deployment, we studied mixed priming schedules incorporating an adenoviral-vectored vaccine (ChAdOx1 nCoV-19 [ChAd], AstraZeneca), two mRNA vaccines (BNT162b2 [BNT], Pfizer-BioNTech, and mRNA-1273 [m1273], Moderna) and a nanoparticle vaccine containing SARS-CoV-2 spike glycoprotein and Matrix-M adjuvant (NVX-CoV2373 [NVX], Novavax). METHODS Com-COV2 is a single-blind, randomised, non-inferiority trial in which adults aged 50 years and older, previously immunised with a single dose of ChAd or BNT in the community, were randomly assigned (in random blocks of three and six) within these cohorts in a 1:1:1 ratio to receive a second dose intramuscularly (8-12 weeks after the first dose) with the homologous vaccine, m1273, or NVX. The primary endpoint was the geometric mean ratio (GMR) of serum SARS-CoV-2 anti-spike IgG concentrations measured by ELISA in heterologous versus homologous schedules at 28 days after the second dose, with a non-inferiority criterion of the GMR above 0·63 for the one-sided 98·75% CI. The primary analysis was on the per-protocol population, who were seronegative at baseline. Safety analyses were done for all participants who received a dose of study vaccine. The trial is registered with ISRCTN, number 27841311. FINDINGS Between April 19 and May 14, 2021, 1072 participants were enrolled at a median of 9·4 weeks after receipt of a single dose of ChAd (n=540, 47% female) or BNT (n=532, 40% female). In ChAd-primed participants, geometric mean concentration (GMC) 28 days after a boost of SARS-CoV-2 anti-spike IgG in recipients of ChAd/m1273 (20 114 ELISA laboratory units [ELU]/mL [95% CI 18 160 to 22 279]) and ChAd/NVX (5597 ELU/mL [4756 to 6586]) was non-inferior to that of ChAd/ChAd recipients (1971 ELU/mL [1718 to 2262]) with a GMR of 10·2 (one-sided 98·75% CI 8·4 to ∞) for ChAd/m1273 and 2·8 (2·2 to ∞) for ChAd/NVX, compared with ChAd/ChAd. In BNT-primed participants, non-inferiority was shown for BNT/m1273 (GMC 22 978 ELU/mL [95% CI 20 597 to 25 636]) but not for BNT/NVX (8874 ELU/mL [7391 to 10 654]), compared with BNT/BNT (16 929 ELU/mL [15 025 to 19 075]) with a GMR of 1·3 (one-sided 98·75% CI 1·1 to ∞) for BNT/m1273 and 0·5 (0·4 to ∞) for BNT/NVX, compared with BNT/BNT; however, NVX still induced an 18-fold rise in GMC 28 days after vaccination. There were 15 serious adverse events, none considered related to immunisation. INTERPRETATION Heterologous second dosing with m1273, but not NVX, increased transient systemic reactogenicity compared with homologous schedules. Multiple vaccines are appropriate to complete primary immunisation following priming with BNT or ChAd, facilitating rapid vaccine deployment globally and supporting recognition of such schedules for vaccine certification. FUNDING UK Vaccine Task Force, Coalition for Epidemic Preparedness Innovations (CEPI), and National Institute for Health Research. NVX vaccine was supplied for use in the trial by Novavax.
Collapse
Affiliation(s)
- Arabella S V Stuart
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Robert H Shaw
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Xinxue Liu
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Melanie Greenland
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Nick J Andrews
- Statistics, Modelling and Economics Department, UK Health Security Agency, London, UK; Immunisation and Countermeasures Division, National Infection Service, UK Health Security Agency, London, UK
| | - J C Cameron
- Public Health Scotland, Glasgow, Scotland, UK
| | - Sue Charlton
- UK Health Security Agency, Porton Down, Salisbury, UK
| | | | | | - Tom Darton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK; Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Tanya Dinesh
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Christopher J A Duncan
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Anna England
- UK Health Security Agency, Porton Down, Salisbury, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | | | - Adam Finn
- School of Population Health Sciences, and School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Anna L Goodman
- Department of Infection, and NIHR BRC, Guy's and St Thomas' NHS Foundation Trust, London, UK; MRC Clinical Trials Unit, University College London, London, UK
| | - Christopher A Green
- NIHR/Wellcome Trust Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Bassam Hallis
- UK Health Security Agency, Porton Down, Salisbury, UK
| | - Paul T Heath
- The Vaccine Institute, St George's University of London, London, UK
| | - Helen Hill
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Bryn M Horsington
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical, Science Oxford Institute, University of Oxford, Oxford, UK
| | | | - Vincenzo Libri
- NIHR UCLH Clinical Research Facility and NIHR UCLH Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | - Patrick J Lillie
- Infection Research Group, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Yama F Mujadidi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Ruth Payne
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK; Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Emma L Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Maheshi N Ramasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Mary Ramsay
- Immunisation and Countermeasures Division, National Infection Service, UK Health Security Agency, London, UK
| | - Robert C Read
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Hannah Robinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Gavin R Screaton
- Chinese Academy of Medical, Science Oxford Institute, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nisha Singh
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - David P J Turner
- University of Nottingham, Nottingham, UK; Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Paul J Turner
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Iason Vichos
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Rachel White
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Jonathan S Nguyen-Van-Tam
- Division of Epidemiology and Public Health, University of Nottingham School of Medicine, Nottingham, UK
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Oxford NIHR-Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
322
|
Mattoo SUS, Myoung J. A Promising Vaccination Strategy against COVID-19 on the Horizon: Heterologous Immunization. J Microbiol Biotechnol 2021; 31:1601-1614. [PMID: 34949742 PMCID: PMC9705928 DOI: 10.4014/jmb.2111.11026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022]
Abstract
To overcome the ongoing COVID-19 pandemic, vaccination campaigns are the highest priority of majority of countries. Limited supply and worldwide disproportionate availability issues for the approved vaccines, together with concerns about rare side-effects have recently initiated the switch to heterologous vaccination, commonly known as mixing of vaccines. The COVID-19 vaccines are highly effective in the general population. However, none of the vaccines is 100% efficacious or effective, with variants posing more challenges, resulting in breakthrough cases. This review summarizes the current knowledge of immune responses to variants of concern (VOC) and breakthrough infections. Furthermore, we discuss the scope of heterologous vaccination and future strategies to tackle the COVID-19 pandemic, including fractionation of vaccine doses and alternative route of vaccination.
Collapse
Affiliation(s)
- Sameer-ul-Salam Mattoo
- Korea Zoonosis Research Institute, Department of Bioactive Material Science and Genetic Engineering Research Institute, Jeonbuk National University, Jeonju 54531, Republic of Korea
| | - Jinjong Myoung
- Korea Zoonosis Research Institute, Department of Bioactive Material Science and Genetic Engineering Research Institute, Jeonbuk National University, Jeonju 54531, Republic of Korea,Corresponding author Phone: +82-63-9004055 Fax: +82-63-9004012 E-mail:
| |
Collapse
|
323
|
Nguyen THO, Cohen CA, Rowntree LC, Bull MB, Hachim A, Kedzierska K, Valkenburg SA. T Cells Targeting SARS-CoV-2: By Infection, Vaccination, and Against Future Variants. Front Med (Lausanne) 2021; 8:793102. [PMID: 35004764 PMCID: PMC8739267 DOI: 10.3389/fmed.2021.793102] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 01/09/2023] Open
Abstract
T cell responses are a key cornerstone to viral immunity to drive high-quality antibody responses, establishing memory for recall and for viral clearance. Inefficient recruitment of T cell responses plays a role in the development of severe COVID-19 and is also represented by reduced cellular responses in men, children, and diversity compared with other epitope-specific subsets and available T cell receptor diversity. SARS-CoV-2-specific T cell responses are elicited by multiple vaccine formats and augmented by prior infection for hybrid immunity. Epitope conservation is relatively well-maintained leading to T cell crossreactivity for variants of concern that have diminished serological responses.
Collapse
Affiliation(s)
- Thi H. O. Nguyen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Carolyn A. Cohen
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Maireid B. Bull
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Sophie A. Valkenburg
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
324
|
Benning L, Morath C, Bartenschlager M, Reineke M, Töllner M, Nusshag C, Kälble F, Reichel P, Schaier M, Schnitzler P, Zeier M, Süsal C, Bartenschlager R, Speer C. Natural SARS-CoV-2 infection results in higher neutralization response against variants of concern compared to two-dose BNT162b2 vaccination in kidney transplant recipients. Kidney Int 2021; 101:639-642. [PMID: 34954213 PMCID: PMC8695512 DOI: 10.1016/j.kint.2021.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/18/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Louise Benning
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany.
| | - Christian Morath
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Marie Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Marvin Reineke
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | | | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Florian Kälble
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Paula Reichel
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Matthias Schaier
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Paul Schnitzler
- Department of Virology, University of Heidelberg, Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Caner Süsal
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany; Transplant Immunology Research Center of Excellence, Koç University Hospital, Istanbul, Turkey
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany; German Center for Infection Research, Partner Site Heidelberg, Heidelberg, Germany; Division Virus-Associated Carcinogenesis, German Cancer Research Center, Heidelberg, Germany
| | - Claudius Speer
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany; Department of Molecular Medicine Partnership Unit Heidelberg, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
325
|
Bushman M, Kahn R, Taylor BP, Lipsitch M, Hanage WP. Population impact of SARS-CoV-2 variants with enhanced transmissibility and/or partial immune escape. Cell 2021; 184:6229-6242.e18. [PMID: 34910927 PMCID: PMC8603072 DOI: 10.1016/j.cell.2021.11.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
SARS-CoV-2 variants of concern exhibit varying degrees of transmissibility and, in some cases, escape from acquired immunity. Much effort has been devoted to measuring these phenotypes, but understanding their impact on the course of the pandemic-especially that of immune escape-has remained a challenge. Here, we use a mathematical model to simulate the dynamics of wild-type and variant strains of SARS-CoV-2 in the context of vaccine rollout and nonpharmaceutical interventions. We show that variants with enhanced transmissibility frequently increase epidemic severity, whereas those with partial immune escape either fail to spread widely or primarily cause reinfections and breakthrough infections. However, when these phenotypes are combined, a variant can continue spreading even as immunity builds up in the population, limiting the impact of vaccination and exacerbating the epidemic. These findings help explain the trajectories of past and present SARS-CoV-2 variants and may inform variant assessment and response in the future.
Collapse
Affiliation(s)
- Mary Bushman
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Rebecca Kahn
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bradford P Taylor
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marc Lipsitch
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - William P Hanage
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
326
|
Munro APS, Janani L, Cornelius V, Aley PK, Babbage G, Baxter D, Bula M, Cathie K, Chatterjee K, Dodd K, Enever Y, Gokani K, Goodman AL, Green CA, Harndahl L, Haughney J, Hicks A, van der Klaauw AA, Kwok J, Lambe T, Libri V, Llewelyn MJ, McGregor AC, Minassian AM, Moore P, Mughal M, Mujadidi YF, Murira J, Osanlou O, Osanlou R, Owens DR, Pacurar M, Palfreeman A, Pan D, Rampling T, Regan K, Saich S, Salkeld J, Saralaya D, Sharma S, Sheridan R, Sturdy A, Thomson EC, Todd S, Twelves C, Read RC, Charlton S, Hallis B, Ramsay M, Andrews N, Nguyen-Van-Tam JS, Snape MD, Liu X, Faust SN. Safety and immunogenicity of seven COVID-19 vaccines as a third dose (booster) following two doses of ChAdOx1 nCov-19 or BNT162b2 in the UK (COV-BOOST): a blinded, multicentre, randomised, controlled, phase 2 trial. Lancet 2021; 398:2258-2276. [PMID: 34863358 PMCID: PMC8639161 DOI: 10.1016/s0140-6736(21)02717-3] [Citation(s) in RCA: 454] [Impact Index Per Article: 151.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Few data exist on the comparative safety and immunogenicity of different COVID-19 vaccines given as a third (booster) dose. To generate data to optimise selection of booster vaccines, we investigated the reactogenicity and immunogenicity of seven different COVID-19 vaccines as a third dose after two doses of ChAdOx1 nCov-19 (Oxford-AstraZeneca; hereafter referred to as ChAd) or BNT162b2 (Pfizer-BioNtech, hearafter referred to as BNT). METHODS COV-BOOST is a multicentre, randomised, controlled, phase 2 trial of third dose booster vaccination against COVID-19. Participants were aged older than 30 years, and were at least 70 days post two doses of ChAd or at least 84 days post two doses of BNT primary COVID-19 immunisation course, with no history of laboratory-confirmed SARS-CoV-2 infection. 18 sites were split into three groups (A, B, and C). Within each site group (A, B, or C), participants were randomly assigned to an experimental vaccine or control. Group A received NVX-CoV2373 (Novavax; hereafter referred to as NVX), a half dose of NVX, ChAd, or quadrivalent meningococcal conjugate vaccine (MenACWY)control (1:1:1:1). Group B received BNT, VLA2001 (Valneva; hereafter referred to as VLA), a half dose of VLA, Ad26.COV2.S (Janssen; hereafter referred to as Ad26) or MenACWY (1:1:1:1:1). Group C received mRNA1273 (Moderna; hereafter referred to as m1273), CVnCov (CureVac; hereafter referred to as CVn), a half dose of BNT, or MenACWY (1:1:1:1). Participants and all investigatory staff were blinded to treatment allocation. Coprimary outcomes were safety and reactogenicity and immunogenicity of anti-spike IgG measured by ELISA. The primary analysis for immunogenicity was on a modified intention-to-treat basis; safety and reactogenicity were assessed in the intention-to-treat population. Secondary outcomes included assessment of viral neutralisation and cellular responses. This trial is registered with ISRCTN, number 73765130. FINDINGS Between June 1 and June 30, 2021, 3498 people were screened. 2878 participants met eligibility criteria and received COVID-19 vaccine or control. The median ages of ChAd/ChAd-primed participants were 53 years (IQR 44-61) in the younger age group and 76 years (73-78) in the older age group. In the BNT/BNT-primed participants, the median ages were 51 years (41-59) in the younger age group and 78 years (75-82) in the older age group. In the ChAd/ChAD-primed group, 676 (46·7%) participants were female and 1380 (95·4%) were White, and in the BNT/BNT-primed group 770 (53·6%) participants were female and 1321 (91·9%) were White. Three vaccines showed overall increased reactogenicity: m1273 after ChAd/ChAd or BNT/BNT; and ChAd and Ad26 after BNT/BNT. For ChAd/ChAd-primed individuals, spike IgG geometric mean ratios (GMRs) between study vaccines and controls ranged from 1·8 (99% CI 1·5-2·3) in the half VLA group to 32·3 (24·8-42·0) in the m1273 group. GMRs for wild-type cellular responses compared with controls ranged from 1·1 (95% CI 0·7-1·6) for ChAd to 3·6 (2·4-5·5) for m1273. For BNT/BNT-primed individuals, spike IgG GMRs ranged from 1·3 (99% CI 1·0-1·5) in the half VLA group to 11·5 (9·4-14·1) in the m1273 group. GMRs for wild-type cellular responses compared with controls ranged from 1·0 (95% CI 0·7-1·6) for half VLA to 4·7 (3·1-7·1) for m1273. The results were similar between those aged 30-69 years and those aged 70 years and older. Fatigue and pain were the most common solicited local and systemic adverse events, experienced more in people aged 30-69 years than those aged 70 years or older. Serious adverse events were uncommon, similar in active vaccine and control groups. In total, there were 24 serious adverse events: five in the control group (two in control group A, three in control group B, and zero in control group C), two in Ad26, five in VLA, one in VLA-half, one in BNT, two in BNT-half, two in ChAd, one in CVn, two in NVX, two in NVX-half, and one in m1273. INTERPRETATION All study vaccines boosted antibody and neutralising responses after ChAd/ChAd initial course and all except one after BNT/BNT, with no safety concerns. Substantial differences in humoral and cellular responses, and vaccine availability will influence policy choices for booster vaccination. FUNDING UK Vaccine Taskforce and National Institute for Health Research.
Collapse
Affiliation(s)
- Alasdair P S Munro
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Leila Janani
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | | | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Gavin Babbage
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Marcin Bula
- NIHR Liverpool and Broadgreen Clinical Research Facility, Liverpool, UK
| | - Katrina Cathie
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Krishna Chatterjee
- NIHR Cambridge Clinical Research Facility, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kate Dodd
- NIHR Liverpool and Broadgreen Clinical Research Facility, Liverpool, UK
| | | | - Karishma Gokani
- NIHR/Wellcome Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Anna L Goodman
- Department of Infection, Guy's and St Thomas' NHS Foundation Trust, London, UK; MRC Clinical Trials Unit, University College London, London, UK
| | - Christopher A Green
- NIHR/Wellcome Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Linda Harndahl
- Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | - John Haughney
- Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, Glasgow, UK
| | | | - Agatha A van der Klaauw
- Wellcome-MRC Institute of Metabolic Science, Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK
| | - Jonathan Kwok
- Cancer Research UK Oxford Centre, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Vincenzo Libri
- NIHR UCLH Clinical Research Facility and NIHR UCLH Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | | | - Alastair C McGregor
- Department of Infectious Diseases and Tropical Medicine, London Northwest University Healthcare, London, UK
| | - Angela M Minassian
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Yama F Mujadidi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Jennifer Murira
- NIHR Leeds Clinical Research Facility, Leeds Teaching Hospitals Trust and University of Leeds, Leeds, UK
| | - Orod Osanlou
- North Wales Clinical Research Facility, Betsi Cadwaladr University Health Board and Bangor University, Bangor, UK
| | - Rostam Osanlou
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Daniel R Owens
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Mihaela Pacurar
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Adrian Palfreeman
- University Hospitals of Leicester NHS Trust, University of Leicester, Leicester, UK
| | - Daniel Pan
- University Hospitals of Leicester NHS Trust, University of Leicester, Leicester, UK
| | - Tommy Rampling
- NIHR UCLH Clinical Research Facility and NIHR UCLH Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | - Karen Regan
- Bradford Institute for Health Research and Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Stephen Saich
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jo Salkeld
- Department of Infection, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Dinesh Saralaya
- Bradford Institute for Health Research and Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Sunil Sharma
- University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Ray Sheridan
- Royal Devon and Exeter Hospital NHS Foundation Trust, Exeter, UK
| | - Ann Sturdy
- Department of Infectious Diseases and Tropical Medicine, London Northwest University Healthcare, London, UK
| | - Emma C Thomson
- Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, Glasgow, UK; MRC University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Shirley Todd
- Royal Devon and Exeter Hospital NHS Foundation Trust, Exeter, UK
| | - Chris Twelves
- NIHR Leeds Clinical Research Facility, Leeds Teaching Hospitals Trust and University of Leeds, Leeds, UK
| | - Robert C Read
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | | | | | - Mary Ramsay
- UK Health Security Agency, Colindale, London, UK
| | - Nick Andrews
- UK Health Security Agency, Colindale, London, UK
| | - Jonathan S Nguyen-Van-Tam
- Division of Epidemiology and Public Health, University of Nottingham School of Medicine, Nottingham, UK
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Xinxue Liu
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
327
|
Horiuchi S, Oishi K, Carrau L, Frere J, Møller R, Panis M, tenOever BR. Immune memory from SARS-CoV-2 infection in hamsters provides variant-independent protection but still allows virus transmission. Sci Immunol 2021; 6:eabm3131. [PMID: 34699266 DOI: 10.1126/sciimmunol.abm3131] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Shu Horiuchi
- Department of Microbiology, New York University, New York, NY 10016, USA
| | - Kohei Oishi
- Department of Microbiology, New York University, New York, NY 10016, USA
| | - Lucia Carrau
- Department of Microbiology, New York University, New York, NY 10016, USA
| | - Justin Frere
- Department of Microbiology, New York University, New York, NY 10016, USA
| | - Rasmus Møller
- Department of Microbiology, New York University, New York, NY 10016, USA
| | - Maryline Panis
- Department of Microbiology, New York University, New York, NY 10016, USA
| | | |
Collapse
|
328
|
Lu Z, Laing ED, Pena DaMata J, Pohida K, Tso MS, Samuels EC, Epsi NJ, Dorjbal B, Lake C, Richard SA, Maves RC, Lindholm DA, Rozman JS, English C, Huprikar N, Mende K, Colombo RE, Colombo CJ, Broder CC, Ganesan A, Lanteri CA, Agan BK, Tribble D, Simons MP, Dalgard CL, Blair PW, Chenoweth J, Pollett SD, Snow AL, Burgess TH, Malloy AMW. Durability of SARS-CoV-2-Specific T-Cell Responses at 12 Months Postinfection. J Infect Dis 2021; 224:2010-2019. [PMID: 34673956 PMCID: PMC8672777 DOI: 10.1093/infdis/jiab543] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/19/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Characterizing the longevity and quality of cellular immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enhances understanding of coronavirus disease 2019 (COVID-19) immunity that influences clinical outcomes. Prior studies suggest SARS-CoV-2-specific T cells are present in peripheral blood 10 months after infection. Analysis of the function, durability, and diversity of cellular response long after natural infection, over a range of ages and disease phenotypes, is needed to identify preventative and therapeutic interventions. METHODS We identified participants in our multisite longitudinal, prospective cohort study 12 months after SARS-CoV-2 infection representing a range of disease severity. We investigated function, phenotypes, and frequency of T cells specific for SARS-CoV-2 using intracellular cytokine staining and spectral flow cytometry, and compared magnitude of SARS-CoV-2-specific antibodies. RESULTS SARS-CoV-2-specific antibodies and T cells were detected 12 months postinfection. Severe acute illness was associated with higher frequencies of SARS-CoV-2-specific CD4 T cells and antibodies at 12 months. In contrast, polyfunctional and cytotoxic T cells responsive to SARS-CoV-2 were identified in participants over a wide spectrum of disease severity. CONCLUSIONS SARS-CoV-2 infection induces polyfunctional memory T cells detectable at 12 months postinfection, with higher frequency noted in those who experienced severe disease.
Collapse
Affiliation(s)
- Zhongyan Lu
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Eric D Laing
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Jarina Pena DaMata
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Katherine Pohida
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Marana S Tso
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Emily C Samuels
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Nusrat J Epsi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Batsukh Dorjbal
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Camille Lake
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Stephanie A Richard
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Ryan C Maves
- Naval Medical Center San Diego, San Diego, California, USA
| | - David A Lindholm
- Brooke Army Medical Center, Joint Base San Antonio-Fort Sam Houston, San Antonio, Texas, USA
| | - Julia S Rozman
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Caroline English
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Nikhil Huprikar
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Katrin Mende
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Brooke Army Medical Center, Joint Base San Antonio-Fort Sam Houston, San Antonio, Texas, USA
| | - Rhonda E Colombo
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Madigan Army Medical Center, Tacoma, Washington, USA
| | | | - Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Anuradha Ganesan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Charlotte A Lanteri
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Brian K Agan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - David Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Mark P Simons
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Paul W Blair
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Austere Environments Consortium for Enhanced Sepsis Outcomes, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Josh Chenoweth
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Austere Environments Consortium for Enhanced Sepsis Outcomes, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Simon D Pollett
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Timothy H Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Allison M W Malloy
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
329
|
Jiang W, Shi L, Cai L, Wang X, Li J, Li H, Liang J, Gu Q, Ji G, Li J, Liu L, Sun M. A two-adjuvant multiantigen candidate vaccine induces superior protective immune responses against SARS-CoV-2 challenge. Cell Rep 2021; 37:110112. [PMID: 34863353 PMCID: PMC8610932 DOI: 10.1016/j.celrep.2021.110112] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/06/2021] [Accepted: 11/18/2021] [Indexed: 01/22/2023] Open
Abstract
An ideal vaccine against SARS-CoV-2 is expected to elicit broad immunity to prevent viral infection and disease, with efficient viral clearance in the upper respiratory tract (URT). Here, the N protein and prefusion-full S protein (SFLmut) are combined with flagellin (KF) and cyclic GMP-AMP (cGAMP) to generate a candidate vaccine, and this vaccine elicits stronger systemic and mucosal humoral immunity than vaccines containing other forms of the S protein. Furthermore, the candidate vaccine administered via intranasal route can enhance local immune responses in the respiratory tract. Importantly, human ACE2 transgenic mice given the candidate vaccine are protected against lethal SARS-CoV-2 challenge, with superior protection in the URT compared with that in mice immunized with an inactivated vaccine. In summary, the developed vaccine can elicit a multifaceted immune response and induce robust viral clearance in the URT, which makes it a potential vaccine for preventing disease and infection of SARS-CoV-2.
Collapse
Affiliation(s)
- Wenwen Jiang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Li Shi
- Laboratory of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Lukui Cai
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Xiaoyu Wang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Jingyan Li
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Heng Li
- Laboratory of Respiratory Infection, Kunming National High-level Biosafety Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Jiangli Liang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Qin Gu
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Guang Ji
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Jing Li
- Laboratory of Respiratory Infection, Kunming National High-level Biosafety Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Longding Liu
- Laboratory of Respiratory Infection, Kunming National High-level Biosafety Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China.
| | - Mingbo Sun
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China; Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China.
| |
Collapse
|
330
|
Carmen JM, Shrivastava S, Lu Z, Anderson A, Morrison EB, Sankhala RS, Chen WH, Chang WC, Bolton JS, Matyas GR, Michael NL, Joyce MG, Modjarrad K, Currier JR, Bergmann-Leitner E, Malloy AMW, Rao M. SARS-CoV-2 ferritin nanoparticle vaccine induces robust innate immune activity driving polyfunctional spike-specific T cell responses. NPJ Vaccines 2021; 6:151. [PMID: 34903722 PMCID: PMC8668928 DOI: 10.1038/s41541-021-00414-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/09/2021] [Indexed: 12/23/2022] Open
Abstract
The emergence of variants of concern, some with reduced susceptibility to COVID-19 vaccines underscores consideration for the understanding of vaccine design that optimizes induction of effective cellular and humoral immune responses. We assessed a SARS-CoV-2 spike-ferritin nanoparticle (SpFN) immunogen paired with two distinct adjuvants, Alhydrogel® or Army Liposome Formulation containing QS-21 (ALFQ) for unique vaccine evoked immune signatures. Recruitment of highly activated multifaceted antigen-presenting cells to the lymph nodes of SpFN+ALFQ vaccinated mice was associated with an increased frequency of polyfunctional spike-specific memory CD4+ T cells and Kb spike-(539-546)-specific long-lived memory CD8+ T cells with effective cytolytic function and distribution to the lungs. The presence of this epitope in SARS-CoV, suggests that generation of cross-reactive T cells may be induced against other coronavirus strains. Our study reveals that a nanoparticle vaccine, combined with a potent adjuvant that effectively engages innate immune cells, enhances SARS-CoV-2-specific durable adaptive immune T cell responses.
Collapse
Affiliation(s)
- Joshua M Carmen
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Shikha Shrivastava
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Zhongyan Lu
- Department of Pediatrics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alexander Anderson
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Oak Ridge Institute of Science and Education, Oak Ridge, TN, USA
| | - Elaine B Morrison
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Rajeshwer S Sankhala
- Emerging Infectious Diseases Branch, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Emerging Infectious Diseases Branch, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Wei-Hung Chen
- Emerging Infectious Diseases Branch, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Emerging Infectious Diseases Branch, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - William C Chang
- Emerging Infectious Diseases Branch, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Emerging Infectious Diseases Branch, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jessica S Bolton
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Nelson L Michael
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - M Gordon Joyce
- Emerging Infectious Diseases Branch, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Emerging Infectious Diseases Branch, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Elke Bergmann-Leitner
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Allison M W Malloy
- Department of Pediatrics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Mangala Rao
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| |
Collapse
|
331
|
Zhuang Z, Liu D, Sun J, Li F, Zhao J. Immune responses to human respiratory coronaviruses infection in mouse models. Curr Opin Virol 2021; 52:102-111. [PMID: 34906757 PMCID: PMC8665230 DOI: 10.1016/j.coviro.2021.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022]
Abstract
Human respiratory coronaviruses (HCoVs), including the recently emerged SARS-CoV-2, the causative agent of the coronavirus disease 2019 (COVID-19) pandemic, potentially cause severe lung infections and multiple organ damages, emphasizing the urgent need for antiviral therapeutics and vaccines against HCoVs. Small animal models, especially mice, are ideal tools for deciphering the pathogenesis of HCoV infections as well as virus-induced immune responses, which is critical for antiviral drug development and vaccine design. In this review, we focus on the antiviral innate immune response, antibody response and T cell response in HCoV infected mouse models, and discuss the potential implications for understanding the anti-HCoV immunity and fighting the COVID-19 pandemic.
Collapse
Affiliation(s)
- Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Donglan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Fang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China; Guangzhou Laboratory, Bio-Island, Guangzhou, Guangdong 510320, China.
| |
Collapse
|
332
|
Humoral Immune Response after the Third SARS-CoV-2 mRNA Vaccination in CD20 Depleted People with Multiple Sclerosis. Vaccines (Basel) 2021; 9:vaccines9121470. [PMID: 34960216 PMCID: PMC8707582 DOI: 10.3390/vaccines9121470] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/25/2021] [Accepted: 12/09/2021] [Indexed: 01/14/2023] Open
Abstract
CD20 depletion is a risk factor for unfavorable outcomes of COVID-19 in people with MS (pwMS). Evidence suggests that protective IgG response to mRNA-based vaccines in B cell-depleted individuals is limited. We studied the seroconversion after the third mRNA SARS-CoV-2 vaccine in B cell-depleted pwMS with limited or no IgG response after the standard immunization. Sixteen pwMS treated with ocrelizumab or rituximab received a third homologous SARS-CoV-2 mRNA vaccine, either the Moderna mRNA-1273 or Pfizer-BioNTech’s BNT162b2 vaccine. We quantified the response of IgG antibodies against the spike receptor-binding domain of SARS-CoV-2 four weeks later. An antibody titer of 100 AU/mL or more was considered clinically relevant. The median time between the last infusion of the anti-CD20 treatment and the third vaccination was 22.9 weeks (range 15.1–31.3). After the third vaccination, one out of 16 patients showed an IgG titer deemed clinically relevant. Only the seroconverted patient had measurable B-cell counts at the time of the third vaccination. The development of a humoral immune response remains rare in pwMS on anti-CD20 therapy, even after third dose of the homologous SARS-CoV-2 mRNA vaccine. It remains to be determined whether T-cell responses can compensate for the lack of seroconversion and provide sufficient protection against CoV-2 infections.
Collapse
|
333
|
SARS-COV-2 Memory B and T Cells Profile in Mild COVID-19 Convalescents subjects. Int J Infect Dis 2021; 115:208-214. [PMID: 34896265 PMCID: PMC8653411 DOI: 10.1016/j.ijid.2021.12.309] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES . Antiviral adaptive immunity involves memory B-(MBC) and T-cells (MTC), however their dynamics in SARS-CoV-2 convalescents warrant further investigation. METHODS . In the cross-sectional and longitudinal study, we evaluated blood-derived MBC- and MTC-responses in 68 anti-spike IgG-positive mild-COVID-19 convalescents at visit 1 between 1-7 months (median 4.1 months) after disease onset. SARS-CoV-2 anti-spike IgG was performed by ELISA, MBC by SARS-COV-2 specific receptor binding domain (RBD) Elispot and Interferon gamma (IFNg), interleukin 2 (IL2) and IFNg+IL2 secreting MTC by IFNg and IL2 SARS-CoV-2 FluoroSpot. For 24 patients sampled at first visit, the IgG, MBC and MTC analysis were also performed 3 months later at second visit. RESULTS . Seventy two percent were both MBC- and MTC-positive, 18 % - MBC-positive and MTC-negative, and 10% - MTC-positive and MBC-negative. The peak of MBC-response level was detected at 3 months after COVID-19 onset and persisted up to 7 months post infection. A significant MTC-levels were detected one month after onset in response to S1, S2_N and SNMO peptide pools. The frequency and magnitude of MTC response to SNMO was higher than to S1 and S2_N. Longitudinal analysis demonstrated that even when specific humoral immunity declined, the cellular immunity persisted. CONCLUSION . Our findings demonstrate durability of adaptive cellular immunity at least for 7-months after SARS-CoV-2 infection that suggest long-lasting protection.
Collapse
|
334
|
Dennehy KM, Löll E, Dhillon C, Classen JM, Warm TD, Schuierer L, Hyhlik-Dürr A, Römmele C, Gosslau Y, Kling E, Hoffmann R. Comparison of the Development of SARS-Coronavirus-2-Specific Cellular Immunity, and Central Memory CD4+ T-Cell Responses Following Infection versus Vaccination. Vaccines (Basel) 2021; 9:1439. [PMID: 34960185 PMCID: PMC8707815 DOI: 10.3390/vaccines9121439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/22/2021] [Accepted: 12/03/2021] [Indexed: 12/03/2022] Open
Abstract
Memory T-cell responses following infection with coronaviruses are reportedly long-lived and provide long-term protection against severe disease. Whether vaccination induces similar long-lived responses is not yet clear since, to date, there are limited data comparing memory CD4+ T-cell responses induced after SARS-CoV-2 infection versus following vaccination with BioNTech/Pfizer BNT162b2. We compared T-cell immune responses over time after infection or vaccination using ELISpot, and memory CD4+ T-cell responses three months after infection/vaccination using activation-induced marker flow cytometric assays. Levels of cytokine-producing T-cells were remarkably stable between three and twelve months after infection, and were comparable to IFNγ+ and IFNγ+IL-2+ T-cell responses but lower than IL-2+ T-cell responses at three months after vaccination. Consistent with this finding, vaccination and infection elicited comparable levels of SARS-CoV-2 specific CD4+ T-cells after three months in addition to comparable proportions of specific central memory CD4+ T-cells. By contrast, the proportions of specific effector memory CD4+ T-cells were significantly lower, whereas specific effector CD4+ T-cells were higher after infection than after vaccination. Our results suggest that T-cell responses-as measured by cytokine expression-and the frequencies of SARS-CoV-2-specific central memory CD4+T-cells-indicative of the formation of the long-lived memory T-cell compartment-are comparably induced after infection and vaccination.
Collapse
Affiliation(s)
- Kevin M. Dennehy
- Institute for Laboratory Medicine and Microbiology, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (E.L.); (L.S.); (E.K.); (R.H.)
| | - Eva Löll
- Institute for Laboratory Medicine and Microbiology, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (E.L.); (L.S.); (E.K.); (R.H.)
| | - Christine Dhillon
- Department of Pathology, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany;
| | - Johanna-Maria Classen
- Internal Medicine III-Gastroenterology and Infectious Diseases, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (J.-M.C.); (C.R.)
| | - Tobias D. Warm
- Clinic for Vascular Surgery, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (T.D.W.); (A.H.-D.); (Y.G.)
| | - Lukas Schuierer
- Institute for Laboratory Medicine and Microbiology, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (E.L.); (L.S.); (E.K.); (R.H.)
| | - Alexander Hyhlik-Dürr
- Clinic for Vascular Surgery, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (T.D.W.); (A.H.-D.); (Y.G.)
| | - Christoph Römmele
- Internal Medicine III-Gastroenterology and Infectious Diseases, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (J.-M.C.); (C.R.)
| | - Yvonne Gosslau
- Clinic for Vascular Surgery, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (T.D.W.); (A.H.-D.); (Y.G.)
| | - Elisabeth Kling
- Institute for Laboratory Medicine and Microbiology, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (E.L.); (L.S.); (E.K.); (R.H.)
| | - Reinhard Hoffmann
- Institute for Laboratory Medicine and Microbiology, Medical Faculty, University Hospital Augsburg, 86156 Augsburg, Germany; (E.L.); (L.S.); (E.K.); (R.H.)
| |
Collapse
|
335
|
Arslan F, Isık Goren B, Baysal B, Vahaboğlu H. Is vaccination necessary for COVID-19 patients? A retrospective cohort study investigating reinfection rates and symptomatology in a tertiary hospital. Expert Rev Vaccines 2021; 21:249-252. [PMID: 34839763 DOI: 10.1080/14760584.2022.2012457] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Durability of immune response by the COVID-19 natural infection and the necessity of vaccines in recovered patients are important inquiries for the healthcare provider. RESEARCH DESIGN AND METHODS Here, we investigated the characteristics and the rate of cases with reinfection that have been admitted to our tertiary hospital. RESULTS A total of 119985 patients were applied between March 2020 and May 2021. Of these patients, 32607 (27%, 32,607/119985) tested positive. A total of 27 (0.08%, 27/32607) patients were found to be reinfected beyond 90 days. Only one of these reinfected patients (0.003, 1/32607) had novel COVID-19 pneumonia and was hospitalized for the second time. Other 26 reinfected patients were followed up as outpatients. CONCLUSIONS COVID-19 reinfection is extremely rare. However, the reinfection may be severe in patients with immune deficiency. Healthcare providers may prioritize uninfected and immune-compromised patients for vaccination.
Collapse
Affiliation(s)
- Ferhat Arslan
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Burcu Isık Goren
- Department of Infectious Diseases and Clinical Microbiology, Prof. Dr. Suleyman Yalçın, Goztepe City Hospital, Istanbul, Turkey
| | - Begumhan Baysal
- Department of Radiology, Prof. Dr. Suleyman Yalçın, Goztepe City Hospital, Istanbul, Turkey
| | - Haluk Vahaboğlu
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Medeniyet University, Istanbul, Turkey
| |
Collapse
|
336
|
Fumagalli MJ, Castro-Jorge LA, Fraga-Silva TFDC, de Azevedo PO, Capato CF, Rattis BAC, Hojo-Souza NS, Floriano VG, de Castro JT, Ramos SG, da Fonseca BAL, Bonato VLD, Gazzinelli RT, Figueiredo LTM. Protective Immunity against Gamma and Zeta Variants after Inactivated SARS-CoV-2 Virus Immunization. Viruses 2021; 13:2440. [PMID: 34960708 PMCID: PMC8707686 DOI: 10.3390/v13122440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 01/08/2023] Open
Abstract
The persistent circulation of SARS-CoV-2 represents an ongoing global threat due to the emergence of new viral variants that can sometimes evade the immune system of previously exposed or vaccinated individuals. We conducted a follow-up study of adult individuals that had received an inactivated SARS-CoV-2 vaccine, evaluating antibody production and neutralizing activity over a period of 6 months. In addition, we performed mice immunization with inactivated SARS-CoV-2, and evaluated the immune response and pathological outcomes against Gamma and Zeta variant infection. Vaccinated individuals produced high levels of antibodies with robust neutralizing activity, which was significantly reduced against Gamma and Zeta variants. Production of IgG anti-S antibodies and neutralizing activity robustly reduced after 6 months of vaccination. Immunized mice demonstrated cellular response against Gamma and Zeta variants, and after viral infection, reduced viral loads, IL-6 expression, and histopathological outcome in the lungs. TNF levels were unchanged in immunized or not immunized mice after infection with the Gamma variant. Furthermore, serum neutralization activity rapidly increases after infection with the Gamma and Zeta variants. Our data suggest that immunization with inactivated WT SARS-CoV-2 induces a promptly responsive cross-reactive immunity response against the Gamma and Zeta variants, reducing COVID-19 pathological outcomes.
Collapse
Affiliation(s)
- Marcilio Jorge Fumagalli
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (L.A.C.-J.); (C.F.C.); (V.G.F.); (B.A.L.d.F.); (L.T.M.F.)
- Basic and Applied Immunology Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (V.L.D.B.); (R.T.G.)
| | - Luiza Antunes Castro-Jorge
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (L.A.C.-J.); (C.F.C.); (V.G.F.); (B.A.L.d.F.); (L.T.M.F.)
| | | | - Patrick Orestes de Azevedo
- Immunopathology Laboratory, René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte 30190-002, Minas Gerais, Brazil; (P.O.d.A.); (N.S.H.-S.); (J.T.d.C.)
| | - Carlos Fabiano Capato
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (L.A.C.-J.); (C.F.C.); (V.G.F.); (B.A.L.d.F.); (L.T.M.F.)
| | - Bruna Amanda Cruz Rattis
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (B.A.C.R.); (S.G.R.)
| | - Natália Satchiko Hojo-Souza
- Immunopathology Laboratory, René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte 30190-002, Minas Gerais, Brazil; (P.O.d.A.); (N.S.H.-S.); (J.T.d.C.)
| | - Vitor Gonçalves Floriano
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (L.A.C.-J.); (C.F.C.); (V.G.F.); (B.A.L.d.F.); (L.T.M.F.)
| | - Julia Teixeira de Castro
- Immunopathology Laboratory, René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte 30190-002, Minas Gerais, Brazil; (P.O.d.A.); (N.S.H.-S.); (J.T.d.C.)
| | - Simone Gusmão Ramos
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (B.A.C.R.); (S.G.R.)
| | - Benedito Antônio Lopes da Fonseca
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (L.A.C.-J.); (C.F.C.); (V.G.F.); (B.A.L.d.F.); (L.T.M.F.)
| | - Vânia Luiza Deperon Bonato
- Basic and Applied Immunology Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (V.L.D.B.); (R.T.G.)
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil;
| | - Ricardo Tostes Gazzinelli
- Basic and Applied Immunology Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (V.L.D.B.); (R.T.G.)
- Immunopathology Laboratory, René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte 30190-002, Minas Gerais, Brazil; (P.O.d.A.); (N.S.H.-S.); (J.T.d.C.)
- Platform of Translational Medicine, Fundação Oswaldo Cruz, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil
| | - Luiz Tadeu Moraes Figueiredo
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (L.A.C.-J.); (C.F.C.); (V.G.F.); (B.A.L.d.F.); (L.T.M.F.)
- Basic and Applied Immunology Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (V.L.D.B.); (R.T.G.)
| |
Collapse
|
337
|
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 vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 2021; 374:abm0829. [PMID: 34648302 PMCID: PMC9284784 DOI: 10.1126/science.abm0829] [Citation(s) in RCA: 566] [Impact Index Per Article: 188.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022]
Abstract
The durability of immune memory after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) messenger RNA (mRNA) vaccination remains unclear. In this study, we longitudinally profiled vaccine responses in SARS-CoV-2–naïve and –recovered individuals for 6 months after vaccination. Antibodies declined from peak levels but remained detectable in most subjects at 6 months. By contrast, mRNA vaccines generated functional memory B cells that increased from 3 to 6 months postvaccination, with the majority of these cells cross-binding the Alpha, Beta, and Delta variants. mRNA vaccination further induced antigen-specific CD4+ and CD8+ T cells, and early CD4+ T cell responses correlated with long-term humoral immunity. Recall responses to vaccination in individuals with preexisting immunity primarily increased antibody levels without substantially altering antibody decay rates. Together, these findings demonstrate robust cellular immune memory to SARS-CoV-2 and its variants for at least 6 months after mRNA vaccination.
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, USA
| | - 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, USA
| | - 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, USA
- 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, USA
| | - 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, NSW, Australia
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - 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, USA
| | - Harsh Sharma
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Herring
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott Korte
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - 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, USA
- 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, USA
| | - Sharon Adamski
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - The UPenn COVID Processing Unit‡
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, 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
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
- 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
- Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Disease, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- 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
| | - 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
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), 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, USA
| | - 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, 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
| |
Collapse
|
338
|
Does infection with or vaccination against SARS-CoV-2 lead to lasting immunity? THE LANCET. RESPIRATORY MEDICINE 2021; 9:1450-1466. [PMID: 34688434 PMCID: PMC8530467 DOI: 10.1016/s2213-2600(21)00407-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/26/2021] [Accepted: 08/21/2021] [Indexed: 12/17/2022]
Abstract
Many nations are pursuing the rollout of SARS-CoV-2 vaccines as an exit strategy from unprecedented COVID-19-related restrictions. However, the success of this strategy relies critically on the duration of protective immunity resulting from both natural infection and vaccination. SARS-CoV-2 infection elicits an adaptive immune response against a large breadth of viral epitopes, although the duration of the response varies with age and disease severity. Current evidence from case studies and large observational studies suggests that, consistent with research on other common respiratory viruses, a protective immunological response lasts for approximately 5-12 months from primary infection, with reinfection being more likely given an insufficiently robust primary humoral response. Markers of humoral and cell-mediated immune memory can persist over many months, and might help to mitigate against severe disease upon reinfection. Emerging data, including evidence of breakthrough infections, suggest that vaccine effectiveness might be reduced significantly against emerging variants of concern, and hence secondary vaccines will need to be developed to maintain population-level protective immunity. Nonetheless, other interventions will also be required, with further outbreaks likely to occur due to antigenic drift, selective pressures for novel variants, and global population mobility.
Collapse
|
339
|
Marfe G, Perna S, Shukla AK. Effectiveness of COVID-19 vaccines and their challenges (Review). Exp Ther Med 2021; 22:1407. [PMID: 34676000 PMCID: PMC8524740 DOI: 10.3892/etm.2021.10843] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
At the end of 2019, a new disease recognized such as severe acute respiratory syndrome (SARS), was reported in Wuhan, China. This disease was caused by an unknown SARS coronavirus 2 (SARS-CoV-2); a virus is characterized by high infectivity among humans. In some cases, this disease can be asymptomatic, while in other cases can induce flu-like symptoms or acute respiratory distress syndrome, pneumonia and death. For this reason, the World Health Organization and Public Health Emergency of International Concern declared a pandemic status in January 2020. Currently, numerous countries have been involved in the development of effective vaccines to protect humans against SARS-CoV-2 infection. The present review will discuss the four vaccines, AZD1222 (AstraZeneca or Vaxzevria), Janssen (Ad26.COV2.S), Moderna/mRNA-1273 and BioNTech/Fosun/Pfizer BNT162b1, that are currently in use worldwide to understand their efficacy, but also evaluate the difficulties and challenges of vaccine development. Although several questions should be addressed regarding these vaccines, the current review will examine the viral elements used in the coronavirus-19 vaccine that can play a crucial role in inducing a strong immune response, as well as the different adverse effects that they can cause to individuals.
Collapse
Affiliation(s)
- Gabriella Marfe
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Stefania Perna
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
- Inventra Medclin Biomedical Healthcare and Research Center, Katemanivli, Kalyan, Thane, Maharashtra 421306, India
| |
Collapse
|
340
|
Jaganathan S, Stieber F, Rao SN, Nikolayevskyy V, Manissero D, Allen N, Boyle J, Howard J. Preliminary Evaluation of QuantiFERON SARS-CoV-2 and QIAreach Anti-SARS-CoV-2 Total Test in Recently Vaccinated Individuals. Infect Dis Ther 2021; 10:2765-2776. [PMID: 34435336 PMCID: PMC8386336 DOI: 10.1007/s40121-021-00521-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/04/2021] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION There is an increasing body of evidence surrounding the importance of a T cell-mediated response to SARS-CoV-2 infection and after COVID-19 vaccination. In this internal feasibility study, we evaluated both the total antibody (IgA, IgM, and IgG) and T cell responses in a cohort of COVID-19 convalescents and vaccinated individuals. METHODS Whole blood specimens were collected weekly from 12 subjects at different time points within/after the COVID-19 mRNA vaccination regimen, and from 4 PCR-confirmed convalescent donors to measure durability of humoral and cell-mediated immune response. T cell and antibody responses were evaluated via the QuantiFERON SARS-CoV-2 research use only (QFN SARS-CoV-2) assay which is an interferon gamma release assay (IGRA) and QIAreach Anti-SARS-CoV-2 total (Anti-CoV-2) test, respectively. RESULTS In a cohort of recently vaccinated individuals, subjects demonstrated robust total antibody and CD4+/CD8+ T cell response to SARS-CoV-2 mRNA vaccines when followed for 2 months post-2nd dose. In most individuals, T cell response declined between the 1st and 2nd doses suggesting a need for a booster or the completion of the 2-dose vaccine series. In a group of convalescent donors tested with QFN SARS-CoV-2 and Anti-CoV-2 tests, all patients had an antibody and T cell response up to 1 year after natural infection. CONCLUSION This small feasibility study demonstrates that the QFN-SARS-CoV-2 test is able to identify CD4+ and CD8+ T cell-mediated responses in SARS-CoV-2-vaccinated subjects and those recovered from COVID-19, alongside a qualitative antibody response detectable via the QIAreach Anti-CoV2 test.
Collapse
Affiliation(s)
- Soumya Jaganathan
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Francis Stieber
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Sonia N Rao
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA.
| | | | | | - Nadia Allen
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Jeff Boyle
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Jenny Howard
- QIAGEN Sciences Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| |
Collapse
|
341
|
Baldwin J, Piplani S, Sakala IG, Honda-Okubo Y, Li L, Petrovsky N. Rapid development of analytical methods for evaluating pandemic vaccines: a COVID-19 perspective. Bioanalysis 2021; 13:1805-1826. [PMID: 34645288 PMCID: PMC8516068 DOI: 10.4155/bio-2021-0096] [Citation(s) in RCA: 6] [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: 05/06/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022] Open
Abstract
Vaccines are key in charting a path out of the COVID-19 pandemic. However, development of new vaccines is highly dependent on availability of analytical methods for their design and evaluation. This paper highlights the challenges presented in having to rapidly develop vaccine analytical tools during an ongoing pandemic, including the need to address progressive virus mutation and adaptation which can render initial assays unreliable or redundant. It also discusses the potential of new computational modeling techniques to model and analyze key viral proteins and their attributes to assist vaccine production and assay design. It then reviews the current range of analytical tools available for COVID-19 vaccine application, ranging from in vitro assays for immunogen characterization to assays to measure vaccine responses in vivo. Finally, it provides a future perspective for COVID-19 vaccine analytical tools and attempts to predict how the field might evolve over the next 5-10 years.
Collapse
Affiliation(s)
- Jeremy Baldwin
- Vaxine Pty Ltd, 11 Walkley Avenue, Adelaide, 5046, Australia
| | - Sakshi Piplani
- Vaxine Pty Ltd, 11 Walkley Avenue, Adelaide, 5046, Australia
- College of Medicine & Public Health, Flinders University, Adelaide, 5042, Australia
| | - Isaac G Sakala
- Vaxine Pty Ltd, 11 Walkley Avenue, Adelaide, 5046, Australia
- College of Medicine & Public Health, Flinders University, Adelaide, 5042, Australia
| | - Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, 11 Walkley Avenue, Adelaide, 5046, Australia
- College of Medicine & Public Health, Flinders University, Adelaide, 5042, Australia
| | - Lei Li
- Vaxine Pty Ltd, 11 Walkley Avenue, Adelaide, 5046, Australia
- College of Medicine & Public Health, Flinders University, Adelaide, 5042, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 11 Walkley Avenue, Adelaide, 5046, Australia
- College of Medicine & Public Health, Flinders University, Adelaide, 5042, Australia
| |
Collapse
|
342
|
Almendro-Vázquez P, Laguna-Goya R, Ruiz-Ruigomez M, Utrero-Rico A, Lalueza A, Maestro de la Calle G, Delgado P, Perez-Ordoño L, Muro E, Vila J, Zamarron I, Moreno-Batanero M, Chivite-Lacaba M, Gil-Etayo FJ, Martín-Higuera C, Meléndez-Carmona MÁ, Lumbreras C, Arellano I, Alarcon B, Allende LM, Aguado JM, Paz-Artal E. Longitudinal dynamics of SARS-CoV-2-specific cellular and humoral immunity after natural infection or BNT162b2 vaccination. PLoS Pathog 2021; 17:e1010211. [PMID: 34962970 PMCID: PMC8757952 DOI: 10.1371/journal.ppat.1010211] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/13/2022] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The timing of the development of specific adaptive immunity after natural SARS-CoV-2 infection, and its relevance in clinical outcome, has not been characterized in depth. Description of the long-term maintenance of both cellular and humoral responses elicited by real-world anti-SARS-CoV-2 vaccination is still scarce. Here we aimed to understand the development of optimal protective responses after SARS-CoV-2 infection and vaccination. We performed an early, longitudinal study of S1-, M- and N-specific IFN-γ and IL-2 T cell immunity and anti-S total and neutralizing antibodies in 88 mild, moderate or severe acute COVID-19 patients. Moreover, SARS-CoV-2-specific adaptive immunity was also analysed in 234 COVID-19 recovered subjects, 28 uninfected BNT162b2-vaccinees and 30 uninfected healthy controls. Upon natural infection, cellular and humoral responses were early and coordinated in mild patients, while weak and inconsistent in severe patients. The S1-specific cellular response measured at hospital arrival was an independent predictive factor against severity. In COVID-19 recovered patients, four to seven months post-infection, cellular immunity was maintained but antibodies and neutralization capacity declined. Finally, a robust Th1-driven immune response was developed in uninfected BNT162b2-vaccinees. Three months post-vaccination, the cellular response was comparable, while the humoral response was consistently stronger, to that measured in COVID-19 recovered patients. Thus, measurement of both humoral and cellular responses provides information on prognosis and protection from infection, which may add value for individual and public health recommendations.
Collapse
Affiliation(s)
| | - Rocio Laguna-Goya
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBER), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Ruiz-Ruigomez
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Utrero-Rico
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Antonio Lalueza
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBER), Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Guillermo Maestro de la Calle
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pilar Delgado
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas (CSIC), Universidad Autonoma de Madrid, Madrid, Spain
| | - Luis Perez-Ordoño
- Department of Emergency Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Eva Muro
- Department of Emergency Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Juan Vila
- Department of Emergency Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Isabel Zamarron
- Department of Emergency Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Marta Chivite-Lacaba
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Francisco Javier Gil-Etayo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Carmen Martín-Higuera
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - María Ángeles Meléndez-Carmona
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Carlos Lumbreras
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBER), Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Irene Arellano
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas (CSIC), Universidad Autonoma de Madrid, Madrid, Spain
| | - Balbino Alarcon
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas (CSIC), Universidad Autonoma de Madrid, Madrid, Spain
| | - Luis Miguel Allende
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense de Madrid, Madrid, Spain
| | - Jose Maria Aguado
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- Unit of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBER), Instituto de Salud Carlos III, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
343
|
Haveri A, Ekström N, Solastie A, Virta C, Österlund P, Isosaari E, Nohynek H, Palmu AA, Melin M. Persistence of neutralizing antibodies a year after SARS-CoV-2 infection in humans. Eur J Immunol 2021; 51:3202-3213. [PMID: 34580856 PMCID: PMC8646652 DOI: 10.1002/eji.202149535] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/02/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022]
Abstract
Most subjects develop antibodies to SARS-CoV-2 following infection. In order to estimate the duration of immunity induced by SARS-CoV-2 it is important to understand for how long antibodies persist after infection in humans. Here, we assessed the persistence of serum antibodies following WT SARS-CoV-2 infection at 8 and 13 months after diagnosis in 367 individuals. The SARS-CoV-2 spike IgG (S-IgG) and nucleoprotein IgG (N-IgG) concentrations and the proportion of subjects with neutralizing antibodies (NAb) were assessed. Moreover, the NAb titers among a smaller subset of participants (n = 78) against a WT virus (B) and variants of concern (VOCs): Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2) were determined. We found that NAb against the WT virus persisted in 89% and S-IgG in 97% of subjects for at least 13 months after infection. Only 36% had N-IgG by 13 months. The mean S-IgG concentrations declined from 8 to 13 months by less than one third; N-IgG concentrations declined by two-thirds. Subjects with severe infection had markedly higher IgG and NAb levels and are expected to remain seropositive for longer. Significantly lower NAb titers against the variants compared to the WT virus, especially after a mild disease, suggests reduced protection against VOCs.
Collapse
Affiliation(s)
- Anu Haveri
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Nina Ekström
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Anna Solastie
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Camilla Virta
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Pamela Österlund
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Elina Isosaari
- Department of Public Health and WelfareFinnish Institute for Health and WelfareHelsinkiFinland
| | - Hanna Nohynek
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Arto A Palmu
- Department of Public Health and WelfareFinnish Institute for Health and WelfareHelsinkiFinland
| | - Merit Melin
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| |
Collapse
|
344
|
Tran S, Truong TH, Narendran A. Evaluation of COVID-19 vaccine response in patients with cancer: An interim analysis. Eur J Cancer 2021; 159:259-274. [PMID: 34798454 PMCID: PMC8542448 DOI: 10.1016/j.ejca.2021.10.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/24/2021] [Accepted: 10/06/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Efficacy and safety data of COVID-19 vaccines among cancer populations have been limited; however, preliminary data from recent studies have emerged regarding their immunogenicity and safety in this population. In this review, we examined current peer-reviewed publications containing serological and safety data after COVID-19 vaccination of patients with cancer. METHODS This analysis examined 21 studies with a total of 5012 patients with cancer, of which 2676 (53%) had haematological malignancies, 2309 (46%) had solid cancers and 739 were healthy controls. Serological responses by anti-SARS-CoV-2 spike protein S1/S2 immunoglobulin G antibody levels and post-vaccination patient questionnaires regarding vaccine-related side-effects after the first and second dose were collected and analysed. RESULTS In general, a single dose of the COVID-19 vaccine yields weaker and heterogeneous serological responses in both patients with haematological and solid malignancies. On receiving a second dose, serological response rates indicate a substantial increase in seropositivity to the SARS-CoV-2 spike protein in all cancer cohorts, but antibody titres remain reduced in comparison with healthy controls. Furthermore, seroconversion in patients with haematological malignancies was significantly lower than that in patients with solid tumours. COVID-19 vaccines are safe and well-tolerated in patients with cancer based on current data of local and systemic effects. CONCLUSION Together, these data support the prioritisation of patients with cancer to receive their first and second doses to minimise the risk of COVID-19 infection and severe complications in this vulnerable population. Additional prophylactic measures must be considered for high-risk patients where current vaccination programs may not mount sufficient protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Son Tran
- Departments of Oncology and Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Tony H Truong
- Divisions of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Aru Narendran
- Departments of Oncology and Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Divisions of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| |
Collapse
|
345
|
Oosting SF, van der Veldt AAM, GeurtsvanKessel CH, Fehrmann RSN, van Binnendijk RS, Dingemans AMC, Smit EF, Hiltermann TJN, den Hartog G, Jalving M, Westphal TT, Bhattacharya A, van der Heiden M, Rimmelzwaan GF, Kvistborg P, Blank CU, Koopmans MPG, Huckriede ALW, van Els CACM, Rots NY, van Baarle D, Haanen JBAG, de Vries EGE. mRNA-1273 COVID-19 vaccination in patients receiving chemotherapy, immunotherapy, or chemoimmunotherapy for solid tumours: a prospective, multicentre, non-inferiority trial. Lancet Oncol 2021; 22:1681-1691. [PMID: 34767759 PMCID: PMC8577843 DOI: 10.1016/s1470-2045(21)00574-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Patients with cancer have an increased risk of complications from SARS-CoV-2 infection. Vaccination to prevent COVID-19 is recommended, but data on the immunogenicity and safety of COVID-19 vaccines for patients with solid tumours receiving systemic cancer treatment are scarce. Therefore, we aimed to assess the impact of immunotherapy, chemotherapy, and chemoimmunotherapy on the immunogenicity and safety of the mRNA-1273 (Moderna Biotech, Madrid, Spain) COVID-19 vaccine as part of the Vaccination Against COVID in Cancer (VOICE) trial. METHODS This prospective, multicentre, non-inferiority trial was done across three centres in the Netherlands. Individuals aged 18 years or older with a life expectancy of more than 12 months were enrolled into four cohorts: individuals without cancer (cohort A [control cohort]), and patients with solid tumours, regardless of stage and histology, treated with immunotherapy (cohort B), chemotherapy (cohort C), or chemoimmunotherapy (cohort D). Participants received two mRNA-1273 vaccinations of 100 μg in 0·5 mL intramuscularly, 28 days apart. The primary endpoint, analysed per protocol (excluding patients with a positive baseline sample [>10 binding antibody units (BAU)/mL], indicating previous SARS-CoV-2 infection), was defined as the SARS-CoV-2 spike S1-specific IgG serum antibody response (ie, SARS-CoV-2-binding antibody concentration of >10 BAU/mL) 28 days after the second vaccination. For the primary endpoint analysis, a non-inferiority design with a margin of 10% was used. We also assessed adverse events in all patients who received at least one vaccination, and recorded solicited adverse events in participants who received at least one vaccination but excluding those who already had seroconversion (>10 BAU/mL) at baseline. This study is ongoing and is registered with ClinicalTrials.gov, NCT04715438. FINDINGS Between Feb 17 and March 12, 2021, 791 participants were enrolled and followed up for a median of 122 days (IQR 118 to 128). A SARS-CoV-2-binding antibody response was found in 240 (100%; 95% CI 98 to 100) of 240 evaluable participants in cohort A, 130 (99%; 96 to >99) of 131 evaluable patients in cohort B, 223 (97%; 94 to 99) of 229 evaluable patients in cohort C, and 143 (100%; 97 to 100) of 143 evaluable patients in cohort D. The SARS-CoV-2-binding antibody response in each patient cohort was non-inferior compared with cohort A. No new safety signals were observed. Grade 3 or worse serious adverse events occurred in no participants in cohort A, three (2%) of 137 patients in cohort B, six (2%) of 244 patients in cohort C, and one (1%) of 163 patients in cohort D, with four events (two of fever, and one each of diarrhoea and febrile neutropenia) potentially related to the vaccination. There were no vaccine-related deaths. INTERPRETATION Most patients with cancer develop, while receiving chemotherapy, immunotherapy, or both for a solid tumour, an adequate antibody response to vaccination with the mRNA-1273 COVID-19 vaccine. The vaccine is also safe in these patients. The minority of patients with an inadequate response after two vaccinations might benefit from a third vaccination. FUNDING ZonMw, The Netherlands Organisation for Health Research and Development.
Collapse
Affiliation(s)
- Sjoukje F Oosting
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus Medical Centre, Rotterdam, Netherlands; Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, Netherlands.
| | | | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Rob S van Binnendijk
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | | | - Egbert F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - T Jeroen N Hiltermann
- Department of Pulmonary Diseases, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Gerco den Hartog
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Mathilda Jalving
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | | | - Arkajyoti Bhattacharya
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Guus F Rimmelzwaan
- Research Centre for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Pia Kvistborg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Anke L W Huckriede
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Cecile A C M van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands; Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Nynke Y Rots
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
346
|
Ferreira CS, Martins YC, Souza RC, Vasconcelos ATR. EpiCurator: an immunoinformatic workflow to predict and prioritize SARS-CoV-2 epitopes. PeerJ 2021; 9:e12548. [PMID: 34909278 PMCID: PMC8641484 DOI: 10.7717/peerj.12548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022] Open
Abstract
The ongoing coronavirus 2019 (COVID-19) pandemic, triggered by the emerging SARS-CoV-2 virus, represents a global public health challenge. Therefore, the development of effective vaccines is an urgent need to prevent and control virus spread. One of the vaccine production strategies uses the in silico epitope prediction from the virus genome by immunoinformatic approaches, which assist in selecting candidate epitopes for in vitro and clinical trials research. This study introduces the EpiCurator workflow to predict and prioritize epitopes from SARS-CoV-2 genomes by combining a series of computational filtering tools. To validate the workflow effectiveness, SARS-CoV-2 genomes retrieved from the GISAID database were analyzed. We identified 11 epitopes in the receptor-binding domain (RBD) of Spike glycoprotein, an important antigenic determinant, not previously described in the literature or published on the Immune Epitope Database (IEDB). Interestingly, these epitopes have a combination of important properties: recognized in sequences of the current variants of concern, present high antigenicity, conservancy, and broad population coverage. The RBD epitopes were the source for a multi-epitope design to in silico validation of their immunogenic potential. The multi-epitope overall quality was computationally validated, endorsing its efficiency to trigger an effective immune response since it has stability, high antigenicity and strong interactions with Toll-Like Receptors (TLR). Taken together, the findings in the current study demonstrated the efficacy of the workflow for epitopes discovery, providing target candidates for immunogen development.
Collapse
Affiliation(s)
- Cristina S. Ferreira
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| | - Yasmmin C. Martins
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| | - Rangel Celso Souza
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| | - Ana Tereza R. Vasconcelos
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| |
Collapse
|
347
|
Sariol CA, Serrano-Collazo C, Ortiz EJ, Pantoja P, Cruz L, Arana T, Atehortua D, Pabon-Carrero C, Espino AM. Limited Impact of Delta Variant's Mutations on the Effectiveness of Neutralization Conferred by Natural Infection or COVID-19 Vaccines in a Latino Population. Viruses 2021; 13:2405. [PMID: 34960674 PMCID: PMC8707683 DOI: 10.3390/v13122405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/23/2022] Open
Abstract
The SARS-CoV-2 pandemic has impacted public health systems all over the world. The Delta variant seems to possess enhanced transmissibility, but no clear evidence suggests it has increased virulence. Our data show that pre-exposed individuals had similar neutralizing activity against the authentic COVID-19 strain and the Delta and Epsilon variants. After only one vaccine dose, the neutralization capacity expanded to all tested variants in pre-exposed individuals. Healthy vaccinated individuals showed a limited breadth of neutralization. One vaccine dose did induce similar neutralizing antibodies against the Delta as against the authentic strain. However, even after two doses, this capacity only expanded to the Epsilon variant.
Collapse
Affiliation(s)
- Carlos A. Sariol
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (L.C.); (T.A.); (A.M.E.)
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (C.S.-C.); (E.J.O.); (P.P.)
- Department of Internal Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA
| | - Crisanta Serrano-Collazo
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (C.S.-C.); (E.J.O.); (P.P.)
| | - Edwin J. Ortiz
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (C.S.-C.); (E.J.O.); (P.P.)
- Puerto Rico Science, Technology and Research Trust, San Juan, PR 00927, USA; (D.A.); (C.P.-C.)
| | - Petraleigh Pantoja
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (C.S.-C.); (E.J.O.); (P.P.)
| | - Lorna Cruz
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (L.C.); (T.A.); (A.M.E.)
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (C.S.-C.); (E.J.O.); (P.P.)
| | - Teresa Arana
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (L.C.); (T.A.); (A.M.E.)
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (C.S.-C.); (E.J.O.); (P.P.)
| | - Dianne Atehortua
- Puerto Rico Science, Technology and Research Trust, San Juan, PR 00927, USA; (D.A.); (C.P.-C.)
| | - Christina Pabon-Carrero
- Puerto Rico Science, Technology and Research Trust, San Juan, PR 00927, USA; (D.A.); (C.P.-C.)
| | - Ana M. Espino
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936, USA; (L.C.); (T.A.); (A.M.E.)
| |
Collapse
|
348
|
Simon D, Tascilar K, Fagni F, Schmidt K, Krönke G, Kleyer A, Ramming A, Schoenau V, Bohr D, Knitza J, Harrer T, Manger K, Manger B, Schett G. Efficacy and safety of SARS-CoV-2 revaccination in non-responders with immune-mediated inflammatory disease. Ann Rheum Dis 2021; 81:1023-1027. [PMID: 34819271 DOI: 10.1136/annrheumdis-2021-221554] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To test whether patients with immune-mediated inflammatory disease (IMIDs), who did not respond to two doses of the SARS-CoV-2 vaccine, develop protective immunity, if a third vaccine dose is administered. METHODS Patients with IMID who failed to seroconvert after two doses of SARS-CoV-2 vaccine were subjected to a third vaccination with either mRNA or vector-based vaccines. Anti-SARS-CoV-2 IgG, neutralising activity and T cell responses were assessed at baseline and 3 weeks after revaccination and also evaluated seprarately in rituximab (RTX) and non-RTX exposed patients. RESULTS 66 non-responders were recruited, 33 treated with RTX, and 33 non-exposed to RTX. Overall, 49.2% patients seroconverted and 50.0% developed neutralising antibody activity. Seroconversion (78.8% vs 18.2%) and neutralising activity (80.0% vs 21.9%) was higher in non-RTX than RTX-treated patients with IMID, respectively. Humoral vaccination responses were not different among patients showing positive (59.3%) or negative (49.7%) T cell responses at baseline. Patients remaining on mRNA-based vaccines showed similar vaccination responses compared with those switching to vector-based vaccines. CONCLUSIONS Overall, these data strongly argue in favor of a third vaccination in patients with IMID lacking response to standard vaccination irrespective of their B cell status.
Collapse
Affiliation(s)
- David Simon
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Koray Tascilar
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Katja Schmidt
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Andreas Ramming
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Verena Schoenau
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Daniela Bohr
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Johannes Knitza
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Thomas Harrer
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Karin Manger
- Rheumatology Practice Bamberg, Erlangen, Germany
| | - Bernhard Manger
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany .,Deutsches Zentrum Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universiätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
349
|
Rosati E, Pogorelyy MV, Minervina AA, Scheffold A, Franke A, Bacher P, Thomas PG. Characterization of SARS-CoV-2 public CD4+ αβ T cell clonotypes through reverse epitope discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.11.19.469229. [PMID: 34845450 PMCID: PMC8629193 DOI: 10.1101/2021.11.19.469229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
UNLABELLED The amount of scientific data and level of public sharing produced as a consequence of the COVID-19 pandemic, as well as the speed at which these data were produced, far exceeds any previous effort against a specific disease condition. This unprecedented situation allows for development and application of new research approaches. One of the major technical hurdles in immunology is the characterization of HLA-antigen-T cell receptor (TCR) specificities. Most approaches aim to identify reactive T cells starting from known antigens using functional assays. However, the need for a reverse approach identifying the antigen specificity of orphan TCRs is increasing. Utilizing large public single-cell gene expression and TCR datasets, we identified highly public CD4 + T cell responses to SARS-CoV-2, covering >75% of the analysed population. We performed an integrative meta-analysis to deeply characterize these clonotypes by TCR sequence, gene expression, HLA-restriction, and antigen-specificity, identifying strong and public CD4 + immunodominant responses with confirmed specificity. CD4 + COVID-enriched clonotypes show T follicular helper functional features, while clonotypes depleted in SARS-CoV-2 individuals preferentially had a central memory phenotype. In total we identify more than 1200 highly public CD4+ T cell clonotypes reactive to SARS-CoV-2. TCR similarity analysis showed six prominent TCR clusters, for which we predicted both HLA-restriction and cognate SARS-CoV-2 immunodominant epitopes. To validate our predictions we used an independent cohort of TCR repertoires before and after vaccination with ChAdOx1 , a replication-deficient simian adenovirus-vectored vaccine, encoding the SARS-CoV-2 spike protein. We find statistically significant enrichment of the predicted spike-reactive TCRs after vaccination with ChAdOx1 , while the frequency of TCRs specific to other SARS-CoV-2 proteins remains stable. Thus, the CD4-associated TCR repertoire differentiates vaccination from natural infection. In conclusion, our study presents a novel reverse epitope discovery approach that can be used to infer HLA- and antigen-specificity of orphan TCRs in any context, such as viral infections, antitumor immune responses, or autoimmune disease. HIGHLIGHTS Identification of highly public CD4+ T cell responses to SARS-CoV-2Systematic prediction of exact immunogenic HLA class II epitopes for CD4+ T cell responseMethodological framework for reverse epitope discovery, which can be applied to other disease contexts and may provide essential insights for future studies and clinical applications.
Collapse
|
350
|
Wild K, Smits M, Killmer S, Strohmeier S, Neumann-Haefelin C, Bengsch B, Krammer F, Schwemmle M, Hofmann M, Thimme R, Zoldan K, Boettler T. Pre-existing immunity and vaccine history determine hemagglutinin-specific CD4 T cell and IgG response following seasonal influenza vaccination. Nat Commun 2021; 12:6720. [PMID: 34795301 PMCID: PMC8602312 DOI: 10.1038/s41467-021-27064-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Effectiveness of seasonal influenza vaccination varies between individuals and might be affected by vaccination history among other factors. Here we show, by monitoring frequencies of CD4 T cells specific to the conserved hemagglutinin epitope HA118-132 and titres of IgG against the corresponding recombinant hemagglutinin protein, that antigen-specific CD4 T cell and antibody responses are closely linked to pre-existing immunity and vaccine history. Upon immunization, a strong early reaction is observed in all vaccine naïve participants and also in vaccine experienced individuals who have not received the respective seasonal vaccine in the previous year. This response is characterized by HA118-132 specific CD4 T cells with a follicular helper T cell phenotype and by ascending titers of hemagglutinin-specific antibodies from baseline to day 28 following vaccination. This trend was observed in only a proportion of those participants who received the seasonal vaccine the year preceding the study. Regardless of history, levels of pre-existing antibodies and CD127 expression on CD4 T cells at baseline were the strongest predictors of robust early response. Thus, both pre-existing immunity and vaccine history contribute to the response to seasonal influenza vaccines.
Collapse
Affiliation(s)
- Katharina Wild
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Pharmacy, University of Freiburg, Freiburg, Germany
| | - Maike Smits
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Saskia Killmer
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christoph Neumann-Haefelin
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Schwemmle
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katharina Zoldan
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Boettler
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|