1201
|
Riou C, du Bruyn E, Stek C, Daroowala R, Goliath RT, Abrahams F, Said-Hartley Q, Allwood BW, Hsiao NY, Wilkinson KA, Arlehamn CSL, Sette A, Wasserman S, Wilkinson RJ, on behalf of the HIATUS consortium. Relationship of SARS-CoV-2-specific CD4 response to COVID-19 severity and impact of HIV-1 and tuberculosis coinfection. J Clin Invest 2021; 131:149125. [PMID: 33945513 PMCID: PMC8203446 DOI: 10.1172/jci149125] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
T cells are involved in control of coronavirus disease 2019 (COVID-19), but limited knowledge is available on the relationship between antigen-specific T cell response and disease severity. Here, we used flow cytometry to assess the magnitude, function, and phenotype of SARS coronavirus 2-specific (SARS-CoV-2-specific) CD4+ T cells in 95 hospitalized COVID-19 patients, 38 of them being HIV-1 and/or tuberculosis (TB) coinfected, and 38 non-COVID-19 patients. We showed that SARS-CoV-2-specific CD4+ T cell attributes, rather than magnitude, were associated with disease severity, with severe disease being characterized by poor polyfunctional potential, reduced proliferation capacity, and enhanced HLA-DR expression. Moreover, HIV-1 and TB coinfection skewed the SARS-CoV-2 T cell response. HIV-1-mediated CD4+ T cell depletion associated with suboptimal T cell and humoral immune responses to SARS-CoV-2, and a decrease in the polyfunctional capacity of SARS-CoV-2-specific CD4+ T cells was observed in COVID-19 patients with active TB. Our results also revealed that COVID-19 patients displayed reduced frequency of Mycobacterium tuberculosis-specific CD4+ T cells, with possible implications for TB disease progression. These results corroborate the important role of SARS-CoV-2-specific T cells in COVID-19 pathogenesis and support the concept of altered T cell functions in patients with severe disease.
Collapse
Affiliation(s)
- Catherine Riou
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
- Division of Medical Virology, Department of Pathology, and
| | - Elsa du Bruyn
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Cari Stek
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | - Remy Daroowala
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | - Rene T. Goliath
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
| | - Fatima Abrahams
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
| | | | - Brian W. Allwood
- Division of Pulmonology, Department of Medicine, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Nei-Yuan Hsiao
- Division of Medical Virology, Department of Pathology, and
- National Health Laboratory Service, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Katalin A. Wilkinson
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- The Francis Crick Institute, London, United Kingdom
| | | | - Alessandro Sette
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sean Wasserman
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Robert J. Wilkinson
- Wellcome Centre for Infectious Disease Research in Africa and Institute of Infectious Disease and Molecular Medicine
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | | |
Collapse
|
1202
|
Anand SP, Prévost J, Nayrac M, Beaudoin-Bussières G, Benlarbi M, Gasser R, Brassard N, Laumaea A, Gong SY, Bourassa C, Brunet-Ratnasingham E, Medjahed H, Gendron-Lepage G, Goyette G, Gokool L, Morrisseau C, Bégin P, Martel-Laferrière V, Tremblay C, Richard J, Bazin R, Duerr R, Kaufmann DE, Finzi A. Longitudinal analysis of humoral immunity against SARS-CoV-2 Spike in convalescent individuals up to 8 months post-symptom onset. Cell Rep Med 2021; 2:100290. [PMID: 33969322 PMCID: PMC8097665 DOI: 10.1016/j.xcrm.2021.100290] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/25/2021] [Accepted: 04/27/2021] [Indexed: 12/20/2022]
Abstract
With the recent approval of highly effective coronavirus disease 2019 (COVID-19) vaccines, functional and lasting immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently under investigation as antibody levels in plasma were shown to decline during convalescence. Since the absence of antibodies does not equate to absence of immune memory, we evaluate the presence of SARS-CoV-2-specific memory B cells in convalescent individuals. Here, we report a longitudinal assessment of humoral immune responses on 32 donors up to 8 months post-symptom onset. Our observations indicate that anti-Spike and anti-receptor binding domain (RBD) immunoglobulin M (IgM) in plasma decay rapidly, whereas the reduction of IgG is less prominent. Neutralizing activity also declines rapidly when compared to Fc-effector functions. Concomitantly, the frequencies of RBD-specific IgM+ B cells wane significantly when compared to RBD-specific IgG+ B cells, which remain stable. Our results add to the current understanding of immune memory following SARS-CoV-2 infection, which is critical for secondary infection prevention and vaccine efficacy.
Collapse
Affiliation(s)
- Sai Priya Anand
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
| | - Jérémie Prévost
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Manon Nayrac
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Mehdi Benlarbi
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
| | - Romain Gasser
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | | | - Annemarie Laumaea
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Shang Yu Gong
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
| | | | - Elsa Brunet-Ratnasingham
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | | | | | | | - Laurie Gokool
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
| | | | - Philippe Bégin
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- CHU Ste-Justine, Montreal, QC H3T 1C5, Canada
| | - Valérie Martel-Laferrière
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Cécile Tremblay
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Jonathan Richard
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Renée Bazin
- Héma-Québec, Affaires Médicales et Innovation, Quebec, QC G1V 5C3, Canada
| | - Ralf Duerr
- Departments of Pathology and Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Daniel E. Kaufmann
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Médecine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Andrés Finzi
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| |
Collapse
|
1203
|
Meyerowitz EA, Sen P, Schoenfeld SR, Neilan TG, Frigault MJ, Stone JH, Kim AY, Mansour MK. Immunomodulation as Treatment for Severe Coronavirus Disease 2019: A Systematic Review of Current Modalities and Future Directions. Clin Infect Dis 2021; 72:e1130-e1143. [PMID: 33216852 PMCID: PMC7717185 DOI: 10.1093/cid/ciaa1759] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
In severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, viral load peaks early and declines quickly after symptom onset. Severe coronavirus disease 2019 (COVID-19) is marked by aberrant innate and adaptive immune responses with an abnormal cytokine profile and multiorgan system dysfunction that persists well after viral clearance. A purely antiviral treatment strategy may therefore be insufficient, and antiviral agents have not shown a benefit later in the illness course. A number of immunomodulatory strategies are being tested, including corticosteroids, cytokine and anticytokine therapies, small molecule inhibitors, and cellular therapeutics. To date, the only drug to show a mortality benefit for COVID-19 in a randomized, controlled trial is dexamethasone. However, there remains uncertainty about which patients may benefit most and about longer-term complications, including secondary infections. Here, we review the immune dysregulation of severe COVID-19 and the existing data behind various immunomodulatory strategies, and we consider future directions of study.
Collapse
Affiliation(s)
- Eric A Meyerowitz
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Pritha Sen
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
| | - Sara R Schoenfeld
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tomas G Neilan
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew J Frigault
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John H Stone
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Arthur Y Kim
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
| | - Michael K Mansour
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
1204
|
Routhu NK, Cheedarla N, Bollimpelli VS, Gangadhara S, Edara VV, Lai L, Sahoo A, Shiferaw A, Styles TM, Floyd K, Fischinger S, Atyeo C, Shin SA, Gumber S, Kirejczyk S, Dinnon KH, Shi PY, Menachery VD, Tomai M, Fox CB, Alter G, Vanderford TH, Gralinski L, Suthar MS, Amara RR. SARS-CoV-2 RBD trimer protein adjuvanted with Alum-3M-052 protects from SARS-CoV-2 infection and immune pathology in the lung. Nat Commun 2021; 12:3587. [PMID: 34117252 PMCID: PMC8196016 DOI: 10.1038/s41467-021-23942-y] [Citation(s) in RCA: 73] [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: 02/23/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
There is a great need for the development of vaccines that induce potent and long-lasting protective immunity against SARS-CoV-2. Multimeric display of the antigen combined with potent adjuvant can enhance the potency and longevity of the antibody response. The receptor binding domain (RBD) of the spike protein is a primary target of neutralizing antibodies. Here, we developed a trimeric form of the RBD and show that it induces a potent neutralizing antibody response against live virus with diverse effector functions and provides protection against SARS-CoV-2 challenge in mice and rhesus macaques. The trimeric form induces higher neutralizing antibody titer compared to monomer with as low as 1μg antigen dose. In mice, adjuvanting the protein with a TLR7/8 agonist formulation alum-3M-052 induces 100-fold higher neutralizing antibody titer and superior protection from infection compared to alum. SARS-CoV-2 infection causes significant loss of innate cells and pathology in the lung, and vaccination protects from changes in innate cells and lung pathology. These results demonstrate RBD trimer protein as a suitable candidate for vaccine against SARS-CoV-2.
Collapse
Affiliation(s)
- Nanda Kishore Routhu
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Narayanaiah Cheedarla
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Venkata Satish Bollimpelli
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Sailaja Gangadhara
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Venkata Viswanadh Edara
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Lilin Lai
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Anusmita Sahoo
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ayalnesh Shiferaw
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Tiffany M Styles
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Katharine Floyd
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Caroline Atyeo
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sally A Shin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sanjeev Gumber
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Shannon Kirejczyk
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kenneth H Dinnon
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Vineet D Menachery
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Mark Tomai
- 3M Corporate Research Materials Laboratory, Saint Paul, MN, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Thomas H Vanderford
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Lisa Gralinski
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Rama Rao Amara
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
1205
|
Potter DA, Thomas A, Rugo HS. A Neoadjuvant Chemotherapy Trial for Early Breast Cancer is Impacted by COVID-19: Addressing Vaccination and Cancer Trials Through Education, Equity, and Outcomes. Clin Cancer Res 2021; 27:4486-4490. [PMID: 34108186 DOI: 10.1158/1078-0432.ccr-21-1133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/14/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022]
Abstract
While COVID-19 vaccine distribution has addressed vulnerabilities related to age and comorbidities, there is a need to ensure vaccination of patients with cancer receiving experimental and routine treatment, where interruption of treatment by infection is likely to result in inferior outcomes. Among patients with cancer, those undergoing neoadjuvant chemotherapy (NAC) or adjuvant chemotherapy (Adj chemo) for early breast cancer (EBC) are at particularly high risk for inferior outcomes, in part, because optimal timing of chemotherapy is essential for promoting distant disease-free survival. COVID-19 data from the ongoing multicenter I-SPY 2 trial of NAC for EBC provides a window into the magnitude of the problem of treatment interruption, not only for the trial itself but also for routine Adj chemo. In the I-SPY 2 trial, 4.5% of patients had disruption of therapy by COVID-19, prior to wide vaccine availability, suggesting that nationally up to 5,700 patients with EBC were at risk for adverse outcomes from COVID-19 infection in 2020. To address this problem, vaccine education and public engagement are essential to overcome hesitancy, while equity of distribution is needed to address access. To accomplish these goals, healthcare organizations (HCO) need to not only call out disinformation but also engage the public with vaccine education and find common ground for vaccine acceptance, while partnering with state/local governments to improve efficiency of vaccine distribution. These approaches are important to improve trial access and to reduce susceptibility to COVID-19, as the pandemic could continue to impact access to clinical trials and routine cancer treatment.
Collapse
Affiliation(s)
- David A Potter
- University of Minnesota Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, Minneapolis, Minnesota.
| | - Alexandra Thomas
- Wake Forest Baptist Health Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, California
| |
Collapse
|
1206
|
Vanshylla K, Di Cristanziano V, Kleipass F, Dewald F, Schommers P, Gieselmann L, Gruell H, Schlotz M, Ercanoglu MS, Stumpf R, Mayer P, Zehner M, Heger E, Johannis W, Horn C, Suárez I, Jung N, Salomon S, Eberhardt KA, Gathof B, Fätkenheuer G, Pfeifer N, Eggeling R, Augustin M, Lehmann C, Klein F. Kinetics and correlates of the neutralizing antibody response to SARS-CoV-2 infection in humans. Cell Host Microbe 2021; 29:917-929.e4. [PMID: 33984285 PMCID: PMC8090990 DOI: 10.1016/j.chom.2021.04.015] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/17/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022]
Abstract
Understanding antibody-based SARS-CoV-2 immunity is critical for overcoming the COVID-19 pandemic and informing vaccination strategies. We evaluated SARS-CoV-2 antibody dynamics over 10 months in 963 individuals who predominantly experienced mild COVID-19. Investigating 2,146 samples, we initially detected SARS-CoV-2 antibodies in 94.4% of individuals, with 82% and 79% exhibiting serum and IgG neutralization, respectively. Approximately 3% of individuals demonstrated exceptional SARS-CoV-2 neutralization, with these "elite neutralizers" also possessing SARS-CoV-1 cross-neutralizing IgG. Multivariate statistical modeling revealed age, symptomatic infection, disease severity, and gender as key factors predicting SARS-CoV-2-neutralizing activity. A loss of reactivity to the virus spike protein was observed in 13% of individuals 10 months after infection. Neutralizing activity had half-lives of 14.7 weeks in serum versus 31.4 weeks in purified IgG, indicating a rather long-term IgG antibody response. Our results demonstrate a broad spectrum in the initial SARS-CoV-2-neutralizing antibody response, with sustained antibodies in most individuals for 10 months after mild COVID-19.
Collapse
Affiliation(s)
- Kanika Vanshylla
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Veronica Di Cristanziano
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Franziska Kleipass
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Felix Dewald
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Philipp Schommers
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Lutz Gieselmann
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Henning Gruell
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Maike Schlotz
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Meryem S Ercanoglu
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Ricarda Stumpf
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Petra Mayer
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Matthias Zehner
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Eva Heger
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Wibke Johannis
- Institute for Clinical Chemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Carola Horn
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Isabelle Suárez
- German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Norma Jung
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Susanne Salomon
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Kirsten Alexandra Eberhardt
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and Department of Medicine, University Medical Center Hamburg-Eppendorf, 20359 Hamburg, Germany; Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Birgit Gathof
- Institute of Transfusion Medicine, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Gerd Fätkenheuer
- German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Nico Pfeifer
- Methods in Medical Informatics, Department of Computer Science, University of Tübingen, 72076 Tübingen, Germany; Faculty of Medicine, University of Tübingen, 72076 Tübingen, Germany; German Center for Infection Research, Partner Site Tübingen, 72076 Tübingen, Germany
| | - Ralf Eggeling
- Methods in Medical Informatics, Department of Computer Science, University of Tübingen, 72076 Tübingen, Germany
| | - Max Augustin
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Clara Lehmann
- German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
1207
|
Lyski ZL, Brunton AE, Strnad MI, Sullivan PE, Siegel SA, Tafesse FG, Slifka MK, Messer WB. SARS-CoV-2 specific memory B-cells from individuals with diverse disease severities recognize SARS-CoV-2 variants of concern. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.05.28.21258025. [PMID: 34100028 PMCID: PMC8183027 DOI: 10.1101/2021.05.28.21258025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this investigation we examined the magnitude, breadth, and durability of SARS-CoV-2 specific antibodies in two distinct B-cell compartments: long-lived plasma cell-derived antibodies in the plasma, and peripheral memory B-cells along with their associated antibody profiles elicited after in vitro stimulation. We found that magnitude varied amongst individuals, but was the highest in hospitalized subjects. Variants of concern (VoC) -RBD-reactive antibodies were found in the plasma of 72% of samples in this investigation, and VoC-RBD-reactive memory B-cells were found in all but 1 subject at a single time-point. This finding, that VoC-RBD-reactive MBCs are present in the peripheral blood of all subjects including those that experienced asymptomatic or mild disease, provides a reason for optimism regarding the capacity of vaccination, prior infection, and/or both, to limit disease severity and transmission of variants of concern as they continue to arise and circulate.
Collapse
Affiliation(s)
- Zoe L. Lyski
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | | | - Matt I. Strnad
- OHSU-PSU School of Public Health, Portland, OR 97239, USA
| | | | | | - Fikadu G. Tafesse
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Mark K. Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - William B. Messer
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- OHSU-PSU School of Public Health, Portland, OR 97239, USA
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| |
Collapse
|
1208
|
Vashishtha VM, Kumar P. Development of SARS-CoV-2 vaccines: challenges, risks, and the way forward. Hum Vaccin Immunother 2021; 17:1635-1649. [PMID: 33270478 PMCID: PMC7754925 DOI: 10.1080/21645515.2020.1845524] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/25/2020] [Accepted: 10/29/2020] [Indexed: 01/13/2023] Open
Abstract
The COVID-19 pandemic mandates the development of a safe and effective Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) vaccine. This review analyzes the complexities, challenges, and other vital issues associated with the development of the SARS-CoV-2 vaccine. A brief review of the immune responses (innate, antibody, and T-cell) to SARS-CoV-2, including immune targets, correlates of protection, and duration of immunity is presented. Approaches to vaccine development including different vaccine platforms, critical attributes of novel vaccine candidates, the status of the ongoing clinical trials, and the ways to speed up vaccine development are also reviewed. Despite a historical average success rate of only 6%, and a usual gestation period of 10-12 years for the development of a new vaccine, the world is on the verge of developing COVID-19 vaccines in an extraordinary short time span.
Collapse
Affiliation(s)
- Vipin M. Vashishtha
- Department of Pediatrics, Mangla Hospital & Research Center, Shakti Chowk, Bijnor, India
| | | |
Collapse
|
1209
|
Diefenbach C, Caro J, Koide A, Grossbard M, Goldberg JD, Raphael B, Hymes K, Moskovits T, Kreditor M, Kaminetzky D, Fleur-Lominy SS, Choi J, Thannickal SA, Stapleford KA, Koide S. Impaired Humoral Immunity to SARS-CoV-2 Vaccination in Non-Hodgkin Lymphoma and CLL Patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34100025 DOI: 10.1101/2021.06.02.21257804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Patients with hematologic malignancies are a high priority for SARS-CoV-2 vaccination, yet the benefit they will derive is uncertain. We investigated the humoral response to vaccination in 53 non-Hodgkin lymphoma (NHL), Hodgkin lymphoma (HL), or CLL patients. Peripheral blood was obtained 2 weeks after first vaccination and 6 weeks after second vaccination for antibody profiling using the multiplex bead-binding assay. Serum IgG, IgA, and IgM antibody levels to the spike specific receptor binding domain (RBD) were evaluated as a measure of response. Subsequently, antibody-positive serum were assayed for neutralization capacity against authentic SARS-CoV-2. Histology was 68% lymphoma and 32% CLL; groups were: patients receiving anti-CD20-based therapy (45%), monitored with disease (28%), receiving BTK inhibitors (19%), or chemotherapy (all HL) (8%). SARS-CoV-2 specific RBD IgG antibody response was decreased across all NHL and CLL groups: 25%, 73%, and 40%, respectively. Antibody IgG titers were significantly reduced (p < 0.001) for CD20 treated and targeted therapy patients, and (p = 0.003) for monitored patients. In 94% of patients evaluated after first and second vaccination, antibody titers did not significantly boost after second vaccination. Only 13% of CD20 treated and 13% of monitored patients generated neutralizing antibodies to SARS-CoV-2 with ICD50s 135 to 1767, and 445 and > 10240. This data has profound implications given the current guidance relaxing masking restrictions and for timing of vaccinations. Unless immunity is confirmed with laboratory testing, these patients should continue to mask, socially distance, and to avoid close contact with non-vaccinated individuals. Statement of Translational Relevance Non Hodgkin lymphoma (NHL) and Chronic Lymphocytic leukemia (CLL) patients who are treated with anti-CD20 antibody therapy, BTK inhibitor therapy, or who are monitored with active disease, have decreased antibody response to SARS-CoV-2 vaccination and decreased antibody titers compared to healthy controls. Antibody titers do not boost following second vaccination, and very few patients generate neutralizing antibodies against SARS-CoV-2. This data is of particular importance, given the recent guidance from the CDC that vaccinated patients no longer need to be masked indoors as well as outdoors. Patients with NHL or CLL who fall into these categories should not consider their immunity from vaccination to be assured. If infected with SARS-CoV-2, they should be a high priority for monoclonal antibody directed therapy. Unless immune response to vaccination is confirmed with laboratory testing, they should continue to mask, socially distance, and to avoid close contact with non-vaccinated individuals.
Collapse
|
1210
|
Scaglione A, Opp S, Hurtado A, Lin Z, Pampeno C, Noval MG, Thannickal SA, Stapleford KA, Meruelo D. Combination of a Sindbis-SARS-CoV-2 spike vaccine and αOX40 antibody elicits protective immunity against SARS-CoV-2 induced disease and potentiates long-term SARS-CoV-2-specific humoral and T-cell immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.28.446009. [PMID: 34075383 PMCID: PMC8168399 DOI: 10.1101/2021.05.28.446009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 is a major global public threat. Currently, a worldwide effort has been mounted to generate billions of effective SARS-CoV-2 vaccine doses to immunize the world's population at record speeds. However, there is still demand for alternative effective vaccines that rapidly confer long-term protection and rely upon cost-effective, easily scaled-up manufacturing. Here, we present a Sindbis alphavirus vector (SV), transiently expressing the SARS-CoV-2 spike protein (SV.Spike), combined with the OX40 immunostimulatory antibody (αOX40) as a novel, highly effective vaccine approach. We show that SV.Spike plus αOX40 elicits long-lasting neutralizing antibodies and a vigorous T-cell response in mice. Protein binding, immunohistochemical and cellular infection assays all show that vaccinated mice sera inhibits spike functions. Immunophenotyping, RNA Seq transcriptome profiles and metabolic analysis indicate a reprogramming of T-cells in vaccinated mice. Activated T-cells were found to mobilize to lung tissue. Most importantly, SV.Spike plus αOX40 provided robust immune protection against infection with authentic coronavirus in transgenic mice expressing the human ACE2 receptor (hACE2-Tg). Finally, our immunization strategy induced strong effector memory response, potentiating protective immunity against re-exposure to SARS-CoV-2 spike protein. Our results show the potential of a new Sindbis virus-based vaccine platform to counteract waning immune response that can be used as a new candidate to combat SARS-CoV-2. Given the strong T-cell responses elicited, our vaccine is likely to be effective against variants that are proving challenging, as well as, serve as a platform to develop a broader spectrum pancoronavirus vaccine. Similarly, the vaccine approach is likely to be applicable to other pathogens.
Collapse
Affiliation(s)
- Antonella Scaglione
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Silvana Opp
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alicia Hurtado
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ziyan Lin
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Christine Pampeno
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maria G Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sara A. Thannickal
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel Meruelo
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
1211
|
Mather MW, Jardine L, Talks B, Gardner L, Haniffa M. Complexity of immune responses in COVID-19. Semin Immunol 2021; 55:101545. [PMID: 34865933 PMCID: PMC8626289 DOI: 10.1016/j.smim.2021.101545] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022]
Abstract
The global COVID-19 pandemic has caused substantial morbidity and mortality to humanity. Remarkable progress has been made in understanding both the innate and adaptive mechanisms involved in the host response to the causative SARS-CoV-2 virus, but much remains to be discovered. Robust upper airway defenses are critical in restricting SARS-CoV-2 replication and propagation. Further, the nasal abundance of viral uptake receptor, ACE2, and the host epithelial transcriptional landscape, are associated with differential disease outcomes across different patient cohorts. The adaptive host response to systemic COVID-19 is heterogeneous and complex. Blunted responses to interferon and robust cytokine generation are hallmarks of the disease, particularly at the advanced stages. Excessive immune cell influx into tissues can lead to substantial collateral damage to the host akin to sepsis. This review offers a contemporary summary of these mechanisms of disease and highlights potential avenues for diagnostic and therapeutic development. These include improved disease stratification, targeting effectors of immune-mediated tissue damage, and blunting of immune cell-mediated tissue damage.
Collapse
Affiliation(s)
- Michael William Mather
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Otolaryngology, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Haematology Department, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Ben Talks
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Otolaryngology, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Louis Gardner
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE2 4LP, UK
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE2 4LP, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| |
Collapse
|
1212
|
Clarke CL, Prendecki M, Dhutia A, Gan J, Edwards C, Prout V, Lightstone L, Parker E, Marchesin F, Griffith M, Charif R, Pickard G, Cox A, McClure M, Tedder R, Randell P, Greathead L, Guckian M, McAdoo SP, Kelleher P, Willicombe M. Longevity of SARS-CoV-2 immune responses in hemodialysis patients and protection against reinfection. Kidney Int 2021; 99:1470-1477. [PMID: 33774082 PMCID: PMC7992297 DOI: 10.1016/j.kint.2021.03.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 01/19/2023]
Abstract
Patients with end stage kidney disease receiving in-center hemodialysis (ICHD) have had high rates of SARS-CoV-2 infection. Following infection, patients receiving ICHD frequently develop circulating antibodies to SARS-CoV-2, even with asymptomatic infection. Here, we investigated the durability and functionality of the immune responses to SARS-CoV-2 infection in patients receiving ICHD. Three hundred and fifty-six such patients were longitudinally screened for SARS-CoV-2 antibodies and underwent routine PCR-testing for symptomatic and asymptomatic infection. Patients were regularly screened for nucleocapsid protein (anti-NP) and receptor binding domain (anti-RBD) antibodies, and those who became seronegative at six months were screened for SARS-CoV-2 specific T-cell responses. One hundred and twenty-nine (36.2%) patients had detectable antibody to anti-NP at time zero, of whom 127 also had detectable anti-RBD. Significantly, at six months, 71/111 (64.0%) and 99/116 (85.3%) remained anti-NP and anti-RBD seropositive, respectively. For patients who retained antibody, both anti-NP and anti-RBD levels were reduced significantly after six months. Eleven patients who were anti-NP seropositive at time zero, had no detectable antibody at six months; of whom eight were found to have SARS-CoV-2 antigen specific T cell responses. Independent of antibody status at six months, patients with baseline positive SARS-CoV-2 serology were significantly less likely to have PCR confirmed infection over the following six months. Thus, patients receiving ICHD mount durable immune responses six months post SARS-CoV-2 infection, with fewer than 3% of patients showing no evidence of humoral or cellular immunity.
Collapse
Affiliation(s)
- Candice L Clarke
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK; Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK.
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK; Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Amrita Dhutia
- Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Jaslyn Gan
- Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Claire Edwards
- Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Virginia Prout
- Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Liz Lightstone
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK; Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Eleanor Parker
- Immunology of Infection Group, Department of Infectious Diseases, Imperial College London, London, UK
| | - Federica Marchesin
- Immunology of Infection Group, Department of Infectious Diseases, Imperial College London, London, UK
| | - Megan Griffith
- Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Rawya Charif
- Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Graham Pickard
- Department of Infection and Immunity, North West London Pathology NHS Trust, London, UK
| | - Alison Cox
- Department of Infection and Immunity, North West London Pathology NHS Trust, London, UK
| | - Myra McClure
- Immunology of Infection Group, Department of Infectious Diseases, Imperial College London, London, UK
| | - Richard Tedder
- Immunology of Infection Group, Department of Infectious Diseases, Imperial College London, London, UK
| | - Paul Randell
- Department of Infection and Immunity, North West London Pathology NHS Trust, London, UK
| | - Louise Greathead
- Department of Infection and Immunity, North West London Pathology NHS Trust, London, UK
| | - Mary Guckian
- Department of Infection and Immunity, North West London Pathology NHS Trust, London, UK
| | - Stephen P McAdoo
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK; Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Peter Kelleher
- Immunology of Infection Group, Department of Infectious Diseases, Imperial College London, London, UK; Department of Infection and Immunity, North West London Pathology NHS Trust, London, UK
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK; Imperial College Renal and Transplant Centre, Division of Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| |
Collapse
|
1213
|
Davenport BJ, Morrison TE, Kedl RM, Klarquist J. Conserved and Novel Mouse CD8 T Cell Epitopes within SARS-CoV-2 Spike Receptor Binding Domain Protein Identified following Subunit Vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2503-2507. [PMID: 33972373 PMCID: PMC8165008 DOI: 10.4049/jimmunol.2100195] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/31/2021] [Indexed: 01/04/2023]
Abstract
The prior existence of human ACE2 protein-expressing mice used to study SARS-CoV and the rapid development of mouse-adapted virus strains have allowed the study of SARS-CoV-2 in mice, even as we are still learning about its natural pathology in humans. With myriad genetically altered strains on the C57BL/6 background and the abundance of immunological reagents available to interrogate its immune responses, the C57BL/6 mice may provide useful insight into the immunology of SARS-CoV-2 infection and vaccination. To conduct more detailed studies on their T cell responses to vaccines and infection, the epitopes eliciting those responses must be characterized in further detail. In this study, we mapped CD8 T cell epitopes within the receptor binding domain of the SARS-CoV-2 spike protein in C57BL/6 mice. Our study identified five major CD8 T cell epitopes in immunized C57BL/6 mice, including one, VVLSFELL, presented by H-2Kb and common between SARS-CoV and SARS-CoV-2.
Collapse
Affiliation(s)
- Bennett J Davenport
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
1214
|
Cimolai N. Passive Immunity Should and Will Work for COVID-19 for Some Patients. Clin Hematol Int 2021; 3:47-68. [PMID: 34595467 PMCID: PMC8432400 DOI: 10.2991/chi.k.210328.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
In the absence of effective antiviral chemotherapy and still in the context of emerging vaccines for severe acute respiratory syndrome-CoV-2 infections, passive immunotherapy remains a key treatment and possible prevention strategy. What might initially be conceived as a simplified donor-recipient process, the intricacies of donor plasma, IV immunoglobulins, and monoclonal antibody modality applications are becoming more apparent. Key targets of such treatment have largely focused on virus neutralization and the specific viral components of the attachment Spike protein and its constituents (e.g., receptor binding domain, N-terminal domain). The cumulative laboratory and clinical experience suggests that beneficial protective and treatment outcomes are possible. Both a dose- and a time-dependency emerge. Lesser understood are the concepts of bioavailability and distribution. Apart from direct antigen binding from protective immunoglobulins, antibody effector functions have potential roles in outcome. In attempting to mimic the natural but variable response to infection or vaccination, a strong functional polyclonal approach attracts the potential benefits of attacking antigen diversity, high antibody avidity, antibody persistence, and protection against escape viral mutation. The availability and ease of administration for any passive immunotherapy product must be considered in the current climate of need. There is never a perfect product, but yet there is considerable room for improving patient outcomes. Given the variability of human genetics, immunity, and disease, and given the nuances of the virus and its potential for change, passive immunotherapy can be developed that will be effective for some but not all patients. An understanding of such patient variability and limitations is just as important as the understanding of the direct interactions between immunotherapy and virus.
Collapse
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, Children’s and Women’s Health Centre of British Columbia, 4480 Oak Street, Vancouver, BC, Canada V6H 3V4
| |
Collapse
|
1215
|
Cromer D, Juno JA, Khoury D, Reynaldi A, Wheatley AK, Kent SJ, Davenport MP. Prospects for durable immune control of SARS-CoV-2 and prevention of reinfection. Nat Rev Immunol 2021; 21:395-404. [PMID: 33927374 PMCID: PMC8082486 DOI: 10.1038/s41577-021-00550-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
Immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is central to long-term control of the current pandemic. Despite our rapidly advancing knowledge of immune memory to SARS-CoV-2, understanding how these responses translate into protection against reinfection at both the individual and population levels remains a major challenge. An ideal outcome following infection or after vaccination would be a highly protective and durable immunity that allows for the establishment of high levels of population immunity. However, current studies suggest a decay of neutralizing antibody responses in convalescent patients, and documented cases of SARS-CoV-2 reinfection are increasing. Understanding the dynamics of memory responses to SARS-CoV-2 and the mechanisms of immune control are crucial for the rational design and deployment of vaccines and for understanding the possible future trajectories of the pandemic. Here, we summarize our current understanding of immune responses to and immune control of SARS-CoV-2 and the implications for prevention of reinfection.
Collapse
Affiliation(s)
- Deborah Cromer
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| | | |
Collapse
|
1216
|
Radulescu A, Istrate A, Flonta M, Lupse M. Antibody and viral RNA kinetics in SARS-CoV2 infected patients admitted to a Romanian University Hospital of Infectious Diseases. Int J Infect Dis 2021; 107:205-211. [PMID: 33901653 PMCID: PMC8064896 DOI: 10.1016/j.ijid.2021.04.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES To assess the antibody and viral kinetics in asymptomatic/mild confirmed SARS-CoV-2 infections compared to more severe patients. MATERIAL AND METHODS Retrospective analysis of data obtained from adult patients with a confirmed SARS-CoV2 infection having at least one SARS-CoV-2 pair of specific IgM/IgG tests, admitted in The University Hospital of Infectious Diseases Cluj-Napoca, Romania (28 February to 31 August 2020). The database also included: demographic, clinical, chest X-ray and/or CT scan results, RT-PCR SARS-CoV-2, and dexamethasone treatment. A total of 469 patients were evaluated as "asymptomatic/mild" and "moderate/severe/critical" cases. RESULTS The median time since confirmation to SARS-CoV-2 PCR negativity was 15 days [95% CI: 13-18] in asymptomatic/mild cases and 17 days [95% CI: 16-21] in moderate/severe ones. The median time to seroconversion for both IgM and IgG was 13 days [95% CI: 13-14] in asymptomatic/mild cases and 11 days [95% CI: 10-13] in moderate/severe ones. For both antibody types, the highest reactivity was significantly associated with more severe presentation (IgM: OR = 10.30, IgG: OR = 7.97). CONCLUSION Asymptomatic/mild COVID-19 cases had a faster RT-PCR negativity rate compared to moderate/severe/critical patients. IgG and IgM dynamics were almost simultaneous, more robust for IgG in more severe cases, and at one month after confirmation, almost all patients had detectable antibody titers.
Collapse
Affiliation(s)
- Amanda Radulescu
- The "Iuliu Hatieganu" University of Medicine and Pharmacy, Epidemiology Department, Cluj-Napoca, Romania; The University Hospital of Infectious Diseases, Cluj-Napoca, Romania
| | - Alexandru Istrate
- The "Iuliu Hatieganu" University of Medicine and Pharmacy, Epidemiology Department, Cluj-Napoca, Romania
| | - Mirela Flonta
- The University Hospital of Infectious Diseases, Clinical Laboratory, Cluj-Napoca, Romania
| | - Mihaela Lupse
- The "Iuliu Hatieganu" University of Medicine and Pharmacy, Infectious Diseases Department, Cluj-Napoca, Romania; The University Hospital of Infectious Diseases, Cluj-Napoca, Romania.
| |
Collapse
|
1217
|
Hagin D, Freund T, Navon M, Halperin T, Adir D, Marom R, Levi I, Benor S, Alcalay Y, Freund NT. Immunogenicity of Pfizer-BioNTech COVID-19 vaccine in patients with inborn errors of immunity. J Allergy Clin Immunol 2021; 148:739-749. [PMID: 34087242 PMCID: PMC8168345 DOI: 10.1016/j.jaci.2021.05.029] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 01/10/2023]
Abstract
Background In mid-December 2020, Israel started a nationwide mass vaccination campaign against coronavirus disease 2019 (COVID-19). In the first few weeks, medical personnel, elderly citizens, and patients with chronic diseases were prioritized. As such, patients with primary and secondary immunodeficiencies were encouraged to receive the vaccine. Although the efficacy of RNA-based COVID-19 vaccines has been demonstrated in the general population, little is known about their efficacy and safety in patients with inborn errors of immunity (IEI). Objective Our aim was to evaluate the humoral and cellular immune response to COVID-19 vaccine in a cohort of patients with IEI. Methods A total of 26 adult patients were enrolled, and plasma and peripheral blood mononuclear cells were collected from them 2 weeks following the second dose of Pfizer-BioNTech COVID-19 vaccine. Humoral response was evaluated by testing anti–SARS-CoV-2 spike (S) receptor-binding domain and antinucleocapsid antibody titers and evaluating neutralizing ability by inhibition of receptor-binding domain–angiotensin-converting enzyme 2 binding. Cellular immune response was evaluated by using ELISpot, estimating IL-2 and IFN-γ secretion in response to pooled SARS-CoV-2 S- or M-peptides. Results Our cohort included 18 patients with a predominantly antibody deficiency, 2 with combined immunodeficiency, 3 with immune dysregulation, and 3 with other genetically defined diagnoses. Twenty-two of them were receiving immunoglobulin replacement therapy. Of the 26 patients, 18 developed specific antibody response, and 19 showed S-peptide–specific T-cell response. None of the patients reported significant adverse events. Conclusion Vaccinating patients with IEI is safe, and most patients were able to develop vaccine-specific antibody response, S-protein–specific cellular response, or both.
Collapse
Affiliation(s)
- David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Tal Freund
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Navon
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tami Halperin
- Laboratory for HIV Diagnosis, the AIDS Center, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dikla Adir
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rotem Marom
- Laboratory for HIV Diagnosis, the AIDS Center, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Levi
- Laboratory for HIV Diagnosis, the AIDS Center, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shira Benor
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yifat Alcalay
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Natalia T Freund
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
1218
|
Mazzoni A, Salvati L, Maggi L, Annunziato F, Cosmi L. Hallmarks of immune response in COVID-19: Exploring dysregulation and exhaustion. Semin Immunol 2021; 55:101508. [PMID: 34728121 PMCID: PMC8547971 DOI: 10.1016/j.smim.2021.101508] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023]
Abstract
One and half year following the occurrence of COVID-19 pandemic, significant efforts from laboratories all over the world generated a huge amount of data describing the prototypical features of immunity in the course of SARS-CoV-2 infection. In this Review, we rationalize and organize the main observations, trying to define a "core" signature of immunity in COVID-19. We identified six hallmarks describing the main alterations occurring in the early infection phase and in the course of the disease, which predispose to severe illness. The six hallmarks are dysregulated type I IFN activity, hyperinflammation, lymphopenia, lymphocyte impairment, dysregulated myeloid response, and heterogeneous adaptive immunity to SARS-CoV-2. Dysregulation and exhaustion came out as the trait d'union, connecting abnormalities affecting both innate and adaptive immunity, humoral and cellular responses.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, 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.
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
1219
|
Steffes LC, Cornfield DN. Coronavirus disease 2019 respiratory disease in children: clinical presentation and pathophysiology. Curr Opin Pediatr 2021; 33:302-310. [PMID: 33938476 DOI: 10.1097/mop.0000000000001013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Pediatric coronavirus disease 2019 (COVID-19) respiratory disease is a distinct entity from adult illness, most notable in its milder phenotype. This review summarizes the current knowledge of the clinical patterns, cellular pathophysiology, and epidemiology of COVID-19 respiratory disease in children with specific attention toward factors that account for the maturation-related differences in disease severity. RECENT FINDINGS Over the past 14 months, knowledge of the clinical presentation and pathophysiology of COVID-19 pneumonia has rapidly expanded. The decreased disease severity of COVID-19 pneumonia in children was an early observation. Differences in the efficiency of viral cell entry and timing of immune recognition and response between children and adults remain at the center of ongoing research. SUMMARY The clinical spectrum of COVID-19 respiratory disease in children is well defined. The age-related differences protecting children from severe disease and death remain incompletely understood.
Collapse
Affiliation(s)
- Lea C Steffes
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Center for Excellence in Pulmonary Biology, Stanford University Medical School, Stanford, California, USA
| | | |
Collapse
|
1220
|
Lehmann AA, Kirchenbaum GA, Zhang T, Reche PA, Lehmann PV. Deconvoluting the T Cell Response to SARS-CoV-2: Specificity Versus Chance and Cognate Cross-Reactivity. Front Immunol 2021; 12:635942. [PMID: 34127926 PMCID: PMC8196231 DOI: 10.3389/fimmu.2021.635942] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/11/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 infection takes a mild or clinically inapparent course in the majority of humans who contract this virus. After such individuals have cleared the virus, only the detection of SARS-CoV-2-specific immunological memory can reveal the exposure, and hopefully the establishment of immune protection. With most viral infections, the presence of specific serum antibodies has provided a reliable biomarker for the exposure to the virus of interest. SARS-CoV-2 infection, however, does not reliably induce a durable antibody response, especially in sub-clinically infected individuals. Consequently, it is plausible for a recently infected individual to yield a false negative result within only a few months after exposure. Immunodiagnostic attention has therefore shifted to studies of specific T cell memory to SARS-CoV-2. Most reports published so far agree that a T cell response is engaged during SARS-CoV-2 infection, but they also state that in 20-81% of SARS-CoV-2-unexposed individuals, T cells respond to SARS-CoV-2 antigens (mega peptide pools), allegedly due to T cell cross-reactivity with Common Cold coronaviruses (CCC), or other antigens. Here we show that, by introducing irrelevant mega peptide pools as negative controls to account for chance cross-reactivity, and by establishing the antigen dose-response characteristic of the T cells, one can clearly discern between cognate T cell memory induced by SARS-CoV-2 infection vs. cross-reactive T cell responses in individuals who have not been infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Alexander A Lehmann
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| | - Greg A Kirchenbaum
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| | - Ting Zhang
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| | - Pedro A Reche
- Laboratorio de Inmunomedicina & Inmunoinformatica, Departamento de Immunologia & O2, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Paul V Lehmann
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| |
Collapse
|
1221
|
Prior COVID-19 protects against reinfection, even in the absence of detectable antibodies. J Infect 2021; 83:237-279. [PMID: 34052242 DOI: 10.1016/j.jinf.2021.05.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 01/31/2023]
|
1222
|
Wang K, Qu M, Ding L, Shi X, Wang C, Cheng S, Hao X. Liver and Kidney Function Biomarkers, Blood Cell Traits and Risk of Severe COVID-19: A Mendelian Randomization Study. Front Genet 2021; 12:647303. [PMID: 34122505 PMCID: PMC8191502 DOI: 10.3389/fgene.2021.647303] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/29/2021] [Indexed: 01/14/2023] Open
Abstract
The pandemic of Coronavirus disease 2019 (COVID-19) has posed an enormous threat to human health. According to observational studies, abnormal liver and kidney functions and blood cell traits were associated with severe COVID-19, yet the causal risk factors for COVID-19 severity and the underlying mechanism remained elusive. We performed Mendelian randomization analyses to assess the potential causal role of eight liver function biomarkers, one kidney function biomarker, and 14 hematological traits on COVID-19 severity using genetic association summary statistics from Europeans. Our findings showed that albumin, direct bilirubin, white blood cell count, neutrophil count, lymphocyte count, and mean corpuscular hemoglobin are casually associated with the risk of severe COVID-19. Notably, lymphocyte count and mean corpuscular hemoglobin had an independent effect on severe COVID-19 risk. These causal evidences provide insights into directions for the risk stratification of individuals with abnormal liver function or blood cell indices and motivate more studies to unveil the roles of these abnormalities in COVID-19 pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Shanshan Cheng
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
1223
|
Yeo JG, Leong JY, Tay SH, Nadua KD, Anderson DE, Lim AJM, Ng XW, Poh SL, Guo D, Yaung KN, Kumar P, Wasser M, Hazirah SN, Sutamam N, Chua CJH, Qui M, Foo R, Gamage AM, Yeo KT, Ramakrishna L, Arkachaisri T, Young BE, Lye DC, Wang LF, Chong CY, Tan NWH, Li J, Kam KQ, Ginhoux F, Thoon KC, Chan JKY, Yung CF, Albani S. A Virus-Specific Immune Rheostat in the Immunome of Patients Recovering From Mild COVID-19. Front Immunol 2021; 12:674279. [PMID: 34113347 PMCID: PMC8185226 DOI: 10.3389/fimmu.2021.674279] [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: 03/01/2021] [Accepted: 05/05/2021] [Indexed: 01/10/2023] Open
Abstract
An accurate depiction of the convalescent COVID-19 immunome will help delineate the immunological milieu crucial for disease resolution and protection. Using mass cytometry, we characterized the immune architecture in patients recovering from mild COVID-19. We identified a virus-specific immune rheostat composed of an effector T (Teff) cell recall response that is balanced by the enrichment of a highly specialized regulatory T (Treg) cell subset. Both components were reactive against a peptide pool covering the receptor binding domain (RBD) of the SARS-CoV-2 spike glycoprotein. We also observed expansion of IFNγ+ memory CD4+ T cells and virus-specific follicular helper T (TFH) cells. Overall, these findings pinpoint critical immune effector and regulatory mechanisms essential for a potent, yet harmless resolution of COVID-19 infection.
Collapse
Affiliation(s)
- Joo Guan Yeo
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Rheumatology and Immunology Service, Department of Pediatric Subspecialities, KK Women's and Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Jing Yao Leong
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Shi Huan Tay
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Karen Donceras Nadua
- Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | | | - Amanda Jin Mei Lim
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Xiang Wen Ng
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Su Li Poh
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Dianyan Guo
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Katherine Nay Yaung
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Pavanish Kumar
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Martin Wasser
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Sharifah Nur Hazirah
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Nursyuhadah Sutamam
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Camillus Jian Hui Chua
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Martin Qui
- Duke-NUS Medical School, Singapore, Singapore
| | - Randy Foo
- Duke-NUS Medical School, Singapore, Singapore
| | | | - Kee Thai Yeo
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Department of Neonatology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Lakshmi Ramakrishna
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Thaschawee Arkachaisri
- Rheumatology and Immunology Service, Department of Pediatric Subspecialities, KK Women's and Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Barnaby E Young
- Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - David Chien Lye
- Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Lin-Fa Wang
- Duke-NUS Medical School, Singapore, Singapore
| | - Chia Yin Chong
- Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Natalie Woon Hui Tan
- Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Jiahui Li
- Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Kai-Qian Kam
- Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Florent Ginhoux
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Koh Cheng Thoon
- Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore.,Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Jerry Kok Yen Chan
- Duke-NUS Medical School, Singapore, Singapore.,Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Chee Fu Yung
- Duke-NUS Medical School, Singapore, Singapore.,Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Salvatore Albani
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Rheumatology and Immunology Service, Department of Pediatric Subspecialities, KK Women's and Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
1224
|
Hanley JP, Tu HA, Dragon JA, Dickson DM, Rio-Guerra RD, Tighe SW, Eckstrom KM, Selig N, Scarpino SV, Whitehead SS, Durbin AP, Pierce KK, Kirkpatrick BD, Rizzo DM, Frietze S, Diehl SA. Immunotranscriptomic profiling the acute and clearance phases of a human challenge dengue virus serotype 2 infection model. Nat Commun 2021; 12:3054. [PMID: 34031380 PMCID: PMC8144425 DOI: 10.1038/s41467-021-22930-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
About 20-25% of dengue virus (DENV) infections become symptomatic ranging from self-limiting fever to shock. Immune gene expression changes during progression to severe dengue have been documented in hospitalized patients; however, baseline or kinetic information is difficult to standardize in natural infection. Here we profile the host immunotranscriptome response in humans before, during, and after infection with a partially attenuated rDEN2Δ30 challenge virus (ClinicalTrials.gov NCT02021968). Inflammatory genes including type I interferon and viral restriction pathways are induced during DENV2 viremia and return to baseline after viral clearance, while others including myeloid, migratory, humoral, and growth factor immune regulation factors pathways are found at non-baseline levels post-viremia. Furthermore, pre-infection baseline gene expression is useful to predict rDEN2Δ30-induced immune responses and the development of rash. Our results suggest a distinct immunological profile for mild rDEN2Δ30 infection and offer new potential biomarkers for characterizing primary DENV infection.
Collapse
Affiliation(s)
- John P Hanley
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Huy A Tu
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cellular and Molecular Biomedical Sciences Program, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Julie A Dragon
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Integrated Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Dorothy M Dickson
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Roxana Del Rio-Guerra
- Flow Cytometry and Cell Sorting Facility, Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Scott W Tighe
- Vermont Integrated Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Korin M Eckstrom
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Integrated Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Nicholas Selig
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Stephen S Whitehead
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Anna P Durbin
- Center for Immunization Research, Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen K Pierce
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Beth D Kirkpatrick
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Donna M Rizzo
- Department of Civil and Environmental Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, USA
| | - Seth Frietze
- Cellular and Molecular Biomedical Sciences Program, University of Vermont, Burlington, VT, USA
- Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Sean A Diehl
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- Cellular and Molecular Biomedical Sciences Program, University of Vermont, Burlington, VT, USA.
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
1225
|
Maecker HT. Immune profiling of COVID-19: preliminary findings and implications for the pandemic. J Immunother Cancer 2021; 9:jitc-2021-002550. [PMID: 33963016 PMCID: PMC8108128 DOI: 10.1136/jitc-2021-002550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 infection can have widely diverse clinical outcomes, from asymptomatic infection to death, with many possible clinical symptoms and syndromes. It is thus essential to understand how the virus interacts with the host immune system to bring about these varied outcomes and to inform vaccine development. We now know that both antibody and T cell responses are induced in the majority of infected individuals, and that cross-reactive responses from other coronaviruses also exist in the uninfected population. Innate immune responses are a key focus of research and may influence the course of disease and the character of subsequent adaptive responses. Finally, baseline immune profiles and changes during early acute infection may be key to predicting the course of disease. Understanding all these aspects can help to create better immune monitoring tools for COVID-19, including tools for predicting disease severity or specific sequelae, perhaps even prior to infection.
Collapse
Affiliation(s)
- Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
1226
|
Repeated Exposure to Subinfectious Doses of SARS-CoV-2 May Promote T Cell Immunity and Protection against Severe COVID-19. Viruses 2021; 13:v13060961. [PMID: 34067349 PMCID: PMC8224680 DOI: 10.3390/v13060961] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022] Open
Abstract
Europe is experiencing a third wave of COVID-19 due to the spread of highly transmissible SARS-CoV-2 variants. A number of positive and negative factors constantly shape the rates of COVID-19 infections, hospitalization, and mortality. Among these factors, the rise in increasingly transmissible variants on one side and the effect of vaccinations on the other side create a picture deeply different from that of the first pandemic wave. Starting from the observation that in several European countries the number of COVID-19 infections in the second and third pandemic wave increased without a proportional rise in disease severity and mortality, we hypothesize the existence of an additional factor influencing SARS-CoV-2 dynamics. This factor consists of an immune defence against severe COVID-19, provided by SARS-CoV-2-specific T cells progressively developing upon natural exposure to low virus doses present in populated environments. As suggested by recent studies, low-dose viral particles entering the respiratory and intestinal tracts may be able to induce T cell memory in the absence of inflammation, potentially resulting in different degrees of immunization. In this scenario, non-pharmaceutical interventions would play a double role, one in the short term by reducing the detrimental spreading of SARS-CoV-2 particles, and one in the long term by allowing the development of a widespread (although heterogeneous and uncontrollable) form of immune protection.
Collapse
|
1227
|
Grau-Expósito J, Sánchez-Gaona N, Massana N, Suppi M, Astorga-Gamaza A, Perea D, Rosado J, Falcó A, Kirkegaard C, Torrella A, Planas B, Navarro J, Suanzes P, Álvarez-Sierra D, Ayora A, Sansano I, Esperalba J, Andrés C, Antón A, Ramón Y Cajal S, Almirante B, Pujol-Borrell R, Falcó V, Burgos J, Buzón MJ, Genescà M. Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat Commun 2021; 12:3010. [PMID: 34021148 PMCID: PMC8140108 DOI: 10.1038/s41467-021-23333-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/24/2021] [Indexed: 12/13/2022] Open
Abstract
Resident memory T cells (TRM) positioned within the respiratory tract are probably required to limit SARS-CoV-2 spread and COVID-19. Importantly, TRM are mostly non-recirculating, which reduces the window of opportunity to examine these cells in the blood as they move to the lung parenchyma. Here, we identify circulating virus-specific T cell responses during acute infection with functional, migratory and apoptotic patterns modulated by viral proteins and associated with clinical outcome. Disease severity is associated predominantly with IFNγ and IL-4 responses, increased responses against S peptides and apoptosis, whereas non-hospitalized patients have increased IL-12p70 levels, degranulation in response to N peptides and SARS-CoV-2-specific CCR7+ T cells secreting IL-10. In convalescent patients, lung-TRM are frequently detected even 10 months after initial infection, in which contemporaneous blood does not reflect tissue-resident profiles. Our study highlights a balanced anti-inflammatory antiviral response associated with a better outcome and persisting TRM cells as important for future protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Judith Grau-Expósito
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Nerea Sánchez-Gaona
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Núria Massana
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Marina Suppi
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Antonio Astorga-Gamaza
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - David Perea
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Joel Rosado
- Thoracic Surgery and Lung Transplantation Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Anna Falcó
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Kirkegaard
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ariadna Torrella
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Bibiana Planas
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Jordi Navarro
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Paula Suanzes
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Daniel Álvarez-Sierra
- Diagnostic Immunology Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Alfonso Ayora
- Occupational Risk Prevention Unit, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Irene Sansano
- Pathology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Departament de Ciències morfològiques, Universitat Autònoma de Barcelona, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Juliana Esperalba
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Andrés
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Andrés Antón
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Santiago Ramón Y Cajal
- Pathology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Departament de Ciències morfològiques, Universitat Autònoma de Barcelona, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Benito Almirante
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ricardo Pujol-Borrell
- Diagnostic Immunology Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,FOCIS Center of Excellence, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Vicenç Falcó
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Joaquín Burgos
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - María J Buzón
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
| |
Collapse
|
1228
|
Renner K, Schwittay T, Chaabane S, Gottschling J, Müller C, Tiefenböck C, Salewski JN, Winter F, Buchtler S, Balam S, Malfertheiner MV, Lubnow M, Lunz D, Graf B, Hitzenbichler F, Hanses F, Poeck H, Kreutz M, Orsó E, Burkhardt R, Niedermair T, Brochhausen C, Gessner A, Salzberger B, Mack M. Severe T cell hyporeactivity in ventilated COVID-19 patients correlates with prolonged virus persistence and poor outcomes. Nat Commun 2021; 12:3006. [PMID: 34021143 PMCID: PMC8140132 DOI: 10.1038/s41467-021-23334-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) can lead to pneumonia and hyperinflammation. Here we show a sensitive method to measure polyclonal T cell activation by downstream effects on responder cells like basophils, plasmacytoid dendritic cells, monocytes and neutrophils in whole blood. We report a clear T cell hyporeactivity in hospitalized COVID-19 patients that is pronounced in ventilated patients, associated with prolonged virus persistence and reversible with clinical recovery. COVID-19-induced T cell hyporeactivity is T cell extrinsic and caused by plasma components, independent of occasional immunosuppressive medication of the patients. Monocytes respond stronger in males than females and IL-2 partially restores T cell activation. Downstream markers of T cell hyporeactivity are also visible in fresh blood samples of ventilated patients. Based on our data we developed a score to predict fatal outcomes and identify patients that may benefit from strategies to overcome T cell hyporeactivity.
Collapse
Affiliation(s)
- Kerstin Renner
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Tobias Schwittay
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Sophia Chaabane
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Johanna Gottschling
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Christine Müller
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | | | - Jan-Niklas Salewski
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Frederike Winter
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Simone Buchtler
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Saidou Balam
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | | | - Matthias Lubnow
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Dirk Lunz
- Department of Anesthesiology, University Hospital Regensburg, Regensburg, Germany
| | - Bernhard Graf
- Department of Anesthesiology, University Hospital Regensburg, Regensburg, Germany
| | - Florian Hitzenbichler
- Department of Infection Prevention and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
| | - Frank Hanses
- Department of Infection Prevention and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
| | - Hendrik Poeck
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Evelyn Orsó
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Tanja Niedermair
- Institute of Pathology, University of Regensburg, Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital, Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital, Regensburg, Germany
| | - André Gessner
- Institute Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Bernd Salzberger
- Department of Infection Prevention and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany.
- Regensburg Center for Interventional Immunology, Regensburg, Germany.
| |
Collapse
|
1229
|
Martín MC, Jurado A, Abad-Molina C, Orduña A, Yarce O, Navas AM, Cunill V, Escobar D, Boix F, Burillo-Sanz S, Vegas-Sánchez MC, Jiménez-de Las Pozas Y, Melero J, Aguilar M, Sobieschi OI, López-Hoyos M, Ocejo-Vinyals G, San Segundo D, Almeida D, Medina S, Fernández L, Vergara E, Quirant B, Martínez-Cáceres E, Boiges M, Alonso M, Esparcia-Pinedo L, López-Sanz C, Muñoz-Vico J, López-Palmero S, Trujillo A, Álvarez P, Prada Á, Monzón D, Ontañón J, Marco FM, Mora S, Rojo R, González-Martínez G, Martínez-Saavedra MT, Gil-Herrera J, Cantenys-Molina S, Hernández M, Perurena-Prieto J, Rodríguez-Bayona B, Martínez A, Ocaña E, Molina J. The age again in the eye of the COVID-19 storm: evidence-based decision making. IMMUNITY & AGEING 2021; 18:24. [PMID: 34016150 PMCID: PMC8134808 DOI: 10.1186/s12979-021-00237-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND One hundred fifty million contagions, more than 3 million deaths and little more than 1 year of COVID-19 have changed our lives and our health management systems forever. Ageing is known to be one of the significant determinants for COVID-19 severity. Two main reasons underlie this: immunosenescence and age correlation with main COVID-19 comorbidities such as hypertension or dyslipidaemia. This study has two aims. The first is to obtain cut-off points for laboratory parameters that can help us in clinical decision-making. The second one is to analyse the effect of pandemic lockdown on epidemiological, clinical, and laboratory parameters concerning the severity of the COVID-19. For these purposes, 257 of SARSCoV2 inpatients during pandemic confinement were included in this study. Moreover, 584 case records from a previously analysed series, were compared with the present study data. RESULTS Concerning the characteristics of lockdown series, mild cases accounted for 14.4, 54.1% were moderate and 31.5%, severe. There were 32.5% of home contagions, 26.3% community transmissions, 22.5% nursing home contagions, and 8.8% corresponding to frontline worker contagions regarding epidemiological features. Age > 60 and male sex are hereby confirmed as severity determinants. Equally, higher severity was significantly associated with higher IL6, CRP, ferritin, LDH, and leukocyte counts, and a lower percentage of lymphocyte, CD4 and CD8 count. Comparing this cohort with a previous 584-cases series, mild cases were less than those analysed in the first moment of the pandemic and dyslipidaemia became more frequent than before. IL-6, CRP and LDH values above 69 pg/mL, 97 mg/L and 328 U/L respectively, as well as a CD4 T-cell count below 535 cells/μL, were the best cut-offs predicting severity since these parameters offered reliable areas under the curve. CONCLUSION Age and sex together with selected laboratory parameters on admission can help us predict COVID-19 severity and, therefore, make clinical and resource management decisions. Demographic features associated with lockdown might affect the homogeneity of the data and the robustness of the results.
Collapse
Affiliation(s)
- María C Martín
- Centro de Hemoterapia y Hemodonación de Castilla y León, Valladolid, Spain
| | - Aurora Jurado
- Department of Immunology and Allergology, Hospital Universitario Reina Sofía-Instituto de Investigación Biomédica de Córdoba (IMIBIC), Avd. Menéndez Pidal s/n, 14004, Córdoba, Spain.
| | - Cristina Abad-Molina
- Department of Microbiology and Immunology, Hospital Clínico Universitario, Valladolid, Spain
| | - Antonio Orduña
- Department of Microbiology and Immunology, Hospital Clínico Universitario, Valladolid, Spain
| | - Oscar Yarce
- Department of Immunology and Allergology, Hospital Universitario Reina Sofía-Instituto de Investigación Biomédica de Córdoba (IMIBIC), Avd. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Ana M Navas
- Department of Immunology and Allergology, Hospital Universitario Reina Sofía-Instituto de Investigación Biomédica de Córdoba (IMIBIC), Avd. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Vanesa Cunill
- Department of Immunology, Hospital Universitario Son Espases-Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Danilo Escobar
- Department of Immunology, Hospital Universitario Son Espases-Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Francisco Boix
- Department of Immunology, Hospital Clínico Universitario, Salamanca, Spain
| | | | | | | | - Josefa Melero
- Department of Immunology, Hospital Universitario de Badajoz, Badajoz, Spain
| | - Marta Aguilar
- Department of Immunology, Hospital Universitario de Badajoz, Badajoz, Spain
| | | | - Marcos López-Hoyos
- Department of Immunology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Gonzalo Ocejo-Vinyals
- Department of Immunology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - David San Segundo
- Department of Immunology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Delia Almeida
- Laboratory of Immunology, Complejo Hospitalario Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Medina
- Laboratory of Immunology, Complejo Hospitalario Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - Luis Fernández
- Laboratoy of Immunology and Genetics, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - Esther Vergara
- Laboratoy of Immunology and Genetics, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - Bibiana Quirant
- Department of Immunology, Hospital Germans Trias i Pujols, Barcelona, Spain
| | | | - Marc Boiges
- Department of Immunology, Hospital Germans Trias i Pujols, Barcelona, Spain
| | - Marta Alonso
- Department of Immunology, Hospital de Cruces, Baracaldo, Spain
| | | | - Celia López-Sanz
- Department of Immunology, Hospital Universitario La Princesa, Madrid, Spain
| | | | | | - Antonio Trujillo
- Department of Immunology and Allergology, Hospital Universitario Reina Sofía-Instituto de Investigación Biomédica de Córdoba (IMIBIC), Avd. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Paula Álvarez
- Department of Immunology and Allergology, Hospital Universitario Reina Sofía-Instituto de Investigación Biomédica de Córdoba (IMIBIC), Avd. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Álvaro Prada
- Department of Immunology, Hospital de Donostia, San Sebastián, Spain
| | - David Monzón
- Department of Immunology, Hospital de Donostia, San Sebastián, Spain
| | - Jesús Ontañón
- Unit of Immunology, Hospital General Universitario, Albacete, Spain
| | | | - Sergio Mora
- Laboratory Unit, Hospital General, Alicante, Spain
| | - Ricardo Rojo
- Department of Immunology, Complejo Hospitalario, La Coruña, Spain
| | - Gema González-Martínez
- Unit of Immunology, Hospital Universitario Insular-Materno Infantil, Las Palmas de Gran Canaria, Spain
| | - María T Martínez-Saavedra
- Unit of Immunology, Hospital Universitario Insular-Materno Infantil, Las Palmas de Gran Canaria, Spain
| | - Juana Gil-Herrera
- Department of Immunology, Hospital General Universitario e Instituto de Investigación Sanitaria, "Gregorio Marañón", Madrid, Spain
| | - Sergi Cantenys-Molina
- Department of Immunology, Hospital General Universitario e Instituto de Investigación Sanitaria, "Gregorio Marañón", Madrid, Spain
| | - Manuel Hernández
- Department of Immunology, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | | | | | | | - Esther Ocaña
- Laboratory Unit, Complejo Hospitalario, Jaén, Spain
| | - Juan Molina
- Department of Immunology and Allergology, Hospital Universitario Reina Sofía-Instituto de Investigación Biomédica de Córdoba (IMIBIC), Avd. Menéndez Pidal s/n, 14004, Córdoba, Spain
| |
Collapse
|
1230
|
Adaptive immune determinants of viral clearance and protection in mouse models of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34031656 DOI: 10.1101/2021.05.19.444825] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused more than 160 million infections and more than 3 million deaths worldwide. While effective vaccines are currently being deployed, the adaptive immune determinants which promote viral clearance and confer protection remain poorly defined. Using mouse models of SARS-CoV-2, we demonstrate that both humoral and cellular adaptive immunity contributes to viral clearance in the setting of primary infection. Furthermore, we find that either convalescent mice, or mice that receive mRNA vaccination are protected from both homologous infection and infection with a variant of concern, B.1.351. Additionally, we find this protection to be largely mediated by antibody response and not cellular immunity. These results highlight the in vivo protective capacity of antibodies generated to both vaccine and natural infection. One-Sentence Summary Defining the roles of humoral and cellular adaptive immunity in viral clearance and protection from SARS-CoV-2 and a variant of concern.
Collapse
|
1231
|
Filbin MR, Mehta A, Schneider AM, Kays KR, Guess JR, Gentili M, Fenyves BG, Charland NC, Gonye AL, Gushterova I, Khanna HK, LaSalle TJ, Lavin-Parsons KM, Lilley BM, Lodenstein CL, Manakongtreecheep K, Margolin JD, McKaig BN, Rojas-Lopez M, Russo BC, Sharma N, Tantivit J, Thomas MF, Gerszten RE, Heimberg GS, Hoover PJ, Lieb DJ, Lin B, Ngo D, Pelka K, Reyes M, Smillie CS, Waghray A, Wood TE, Zajac AS, Jennings LL, Grundberg I, Bhattacharyya RP, Parry BA, Villani AC, Sade-Feldman M, Hacohen N, Goldberg MB. Longitudinal proteomic analysis of severe COVID-19 reveals survival-associated signatures, tissue-specific cell death, and cell-cell interactions. Cell Rep Med 2021; 2:100287. [PMID: 33969320 PMCID: PMC8091031 DOI: 10.1016/j.xcrm.2021.100287] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/08/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Mechanisms underlying severe coronavirus disease 2019 (COVID-19) disease remain poorly understood. We analyze several thousand plasma proteins longitudinally in 306 COVID-19 patients and 78 symptomatic controls, uncovering immune and non-immune proteins linked to COVID-19. Deconvolution of our plasma proteome data using published scRNA-seq datasets reveals contributions from circulating immune and tissue cells. Sixteen percent of patients display reduced inflammation yet comparably poor outcomes. Comparison of patients who died to severely ill survivors identifies dynamic immune-cell-derived and tissue-associated proteins associated with survival, including exocrine pancreatic proteases. Using derived tissue-specific and cell-type-specific intracellular death signatures, cellular angiotensin-converting enzyme 2 (ACE2) expression, and our data, we infer whether organ damage resulted from direct or indirect effects of infection. We propose a model in which interactions among myeloid, epithelial, and T cells drive tissue damage. These datasets provide important insights and a rich resource for analysis of mechanisms of severe COVID-19 disease.
Collapse
Affiliation(s)
- Michael R. Filbin
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Emergency Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Arnav Mehta
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alexis M. Schneider
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kyle R. Kays
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Matteo Gentili
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Bánk G. Fenyves
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Emergency Medicine, Semmelweis University, Budapest, Hungary
| | - Nicole C. Charland
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anna L.K. Gonye
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Irena Gushterova
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hargun K. Khanna
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Thomas J. LaSalle
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Brendan M. Lilley
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Carl L. Lodenstein
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kasidet Manakongtreecheep
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Justin D. Margolin
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Brenna N. McKaig
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Maricarmen Rojas-Lopez
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Brian C. Russo
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Nihaarika Sharma
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica Tantivit
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Molly F. Thomas
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Robert E. Gerszten
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- CardioVascular Institute, Department of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Graham S. Heimberg
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Paul J. Hoover
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - David J. Lieb
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Brian Lin
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Regenerative Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Debby Ngo
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Karin Pelka
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Miguel Reyes
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christopher S. Smillie
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Avinash Waghray
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Regenerative Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Thomas E. Wood
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Amanda S. Zajac
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | | | | | - Roby P. Bhattacharyya
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Blair Alden Parry
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alexandra-Chloé Villani
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Moshe Sade-Feldman
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nir Hacohen
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marcia B. Goldberg
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
1232
|
Spencer AJ, McKay PF, Belij-Rammerstorfer S, Ulaszewska M, Bissett CD, Hu K, Samnuan K, Blakney AK, Wright D, Sharpe HR, Gilbride C, Truby A, Allen ER, Gilbert SC, Shattock RJ, Lambe T. Heterologous vaccination regimens with self-amplifying RNA and adenoviral COVID vaccines induce robust immune responses in mice. Nat Commun 2021; 12:2893. [PMID: 34001897 PMCID: PMC8129084 DOI: 10.1038/s41467-021-23173-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 01/08/2023] Open
Abstract
Several vaccines have demonstrated efficacy against SARS-CoV-2 mediated disease, yet there is limited data on the immune response induced by heterologous vaccination regimens using alternate vaccine modalities. Here, we present a detailed description of the immune response, in mice, following vaccination with a self-amplifying RNA (saRNA) vaccine and an adenoviral vectored vaccine (ChAdOx1 nCoV-19/AZD1222) against SARS-CoV-2. We demonstrate that antibody responses are higher in two-dose heterologous vaccination regimens than single-dose regimens. Neutralising titres after heterologous prime-boost were at least comparable or higher than the titres measured after homologous prime boost vaccination with viral vectors. Importantly, the cellular immune response after a heterologous regimen is dominated by cytotoxic T cells and Th1+ CD4 T cells, which is superior to the response induced in homologous vaccination regimens in mice. These results underpin the need for clinical trials to investigate the immunogenicity of heterologous regimens with alternate vaccine technologies.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- ChAdOx1 nCoV-19
- Immunization, Secondary
- Immunogenicity, Vaccine
- Mice
- RNA, Viral/administration & dosage
- RNA, Viral/genetics
- RNA, Viral/immunology
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Th1 Cells/immunology
- Vaccination/methods
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Alexandra J Spencer
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK.
| | - Paul F McKay
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Marta Ulaszewska
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Cameron D Bissett
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Kai Hu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Karnyart Samnuan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Anna K Blakney
- Department of Infectious Disease, Imperial College London, London, UK
| | - Daniel Wright
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Hannah R Sharpe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Ciaran Gilbride
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Adam Truby
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Elizabeth R Allen
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, London, UK
| | - Teresa Lambe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
1233
|
Jones JM, Faruqi AJ, Sullivan JK, Calabrese C, Calabrese LH. COVID-19 Outcomes in Patients Undergoing B Cell Depletion Therapy and Those with Humoral Immunodeficiency States: A Scoping Review. Pathog Immun 2021; 6:76-103. [PMID: 34056149 PMCID: PMC8150936 DOI: 10.20411/pai.v6i1.435] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The role of humoral immunity has been well established in reducing infection risk and facilitating viral clearance in patients with COVID-19. However, the relationship between specific antibody responses and severity of COVID-19 is less well understood. METHODS To address this question and identify gaps in knowledge, we utilized the methodology of a scoping review to interrogate risk of infection and clinical outcomes of COVID-19 in patients with iatrogenic and inborn humoral immunodeficiency states based on existing literature. RESULTS Among patients with iatrogenic B-cell depletion, particularly with agents targeting CD20, our analysis found increased risk of severe COVID-19 and death across a range of underlying disease states. Among patients with humoral inborn errors of immunity with COVID-19, our synthesis found that patients with dysregulated humoral immunity, predominantly common variable immunodeficiency (CVID), may be more susceptible to severe COVID-19 than patients with humoral immunodeficiency states due to X-linked agammaglobulinemia and other miscellaneous forms of humoral immunodeficiency. There were insufficient data to appraise the risk of COVID-19 infection in both populations of patients. CONCLUSIONS Our work identifies potentially significant predictors of COVID-19 severity in patients with humoral immunodeficiency states and highlights the need for larger studies to control for clinical and biologic confounders of disease severity.
Collapse
Affiliation(s)
- Jessica M. Jones
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Aiman J. Faruqi
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - James K. Sullivan
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Cassandra Calabrese
- Cleveland Clinic, Department of Rheumatic and Immunologic Diseases, Cleveland, Ohio
| | - Leonard H. Calabrese
- Cleveland Clinic, Department of Rheumatic and Immunologic Diseases, Cleveland, Ohio
| |
Collapse
|
1234
|
Blumental S, Debré P. Challenges and Issues of Anti-SARS-CoV-2 Vaccines. Front Med (Lausanne) 2021; 8:664179. [PMID: 34055838 PMCID: PMC8163222 DOI: 10.3389/fmed.2021.664179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
At the beginning of 2021, anti-SARS-CoV-2 vaccination campaigns had been launched in almost 60 countries with more than 500 million doses having been distributed. In addition to the few vaccines already in use, many other candidates are in preclinical phases or experimental stages in humans. Despite the fact that the availability of anti-SARS-CoV-2 vaccine constitutes a major advance and appear to be the only way to control the pandemic, some investigation remains to be carried out, and this is notably concerning the impact on transmissibility, the duration of the conferred protection in the mid- and long term, the effectiveness against present and future viral mutants, or the ideal schedule that should be applied. In this paper, we review the circumstances that facilitated such a rapid development of anti-SARS-CoV-2 vaccines and summarize the different vaccine platforms under investigation as well as their present results and perspectives in different settings. We also discuss the indications of vaccination under special conditions, such as a history of previous COVID-19 infection or belonging to extreme age categories like children and elderly. Overall, this review highlights the multiple challenges to face if aiming to find a global solution to the pandemic through high vaccination coverage all over the world.
Collapse
Affiliation(s)
- Sophie Blumental
- Pediatric Infectious Disease Unit, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Patrice Debré
- Immunology Department, APHP, Sorbonne Université CIMI (Inserm U1135), Hôpital Pitie Salpêtrière, Paris, France
| |
Collapse
|
1235
|
Palacios-Pedrero MÁ, Osterhaus ADME, Becker T, Elbahesh H, Rimmelzwaan GF, Saletti G. Aging and Options to Halt Declining Immunity to Virus Infections. Front Immunol 2021; 12:681449. [PMID: 34054872 PMCID: PMC8149791 DOI: 10.3389/fimmu.2021.681449] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Immunosenescence is a process associated with aging that leads to dysregulation of cells of innate and adaptive immunity, which may become dysfunctional. Consequently, older adults show increased severity of viral and bacterial infections and impaired responses to vaccinations. A better understanding of the process of immunosenescence will aid the development of novel strategies to boost the immune system in older adults. In this review, we focus on major alterations of the immune system triggered by aging, and address the effect of chronic viral infections, effectiveness of vaccination of older adults and strategies to improve immune function in this vulnerable age group.
Collapse
Affiliation(s)
| | - Albert D M E Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Tanja Becker
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Husni Elbahesh
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Giulietta Saletti
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
1236
|
Old and new coronaviruses in the elderly. Aging (Albany NY) 2021; 13:12295-12296. [PMID: 33982672 PMCID: PMC8148481 DOI: 10.18632/aging.203065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 11/25/2022]
|
1237
|
Barreiro P, Candel FJ, Sanz JC, San Román J, del Mar Carretero M, Pérez-Abeledo M, Ramos B, Viñuela-Prieto JM, Canora J, Martínez-Peromingo FJ, Zapatero A. Virological Correlates of IgM-IgG Patterns of Response to SARS-CoV-2 Infection According to Targeted Antigens. Viruses 2021; 13:874. [PMID: 34068703 PMCID: PMC8151912 DOI: 10.3390/v13050874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/17/2022] Open
Abstract
The virological meaning of the different patterns of serology in COVID-19 has been little examined in clinical settings. Asymptomatic subjects with IgM-spike (S) and IgG-nucleocapsid (N) determinations by chemiluminescence were studied for SARS-CoV-2 shedding in respiratory secretions by transcription-mediated amplification (TMA). In subjects showing IgM-S positive and IgG-N negative, IgG-S was determined by lateral flow assay. A total of 712 individuals were tested: 30.0% presented IgM-S(+)/IgG-N(-), 25.8% had IgM-S(+)/IgG-N(+) and 44.2% had IgM-S(-)/IgG-N(+); the proportion with TMA(+) were comparable in these three groups: 12.1, 8.7 and 10.5%, respectively. In individuals with IgM-S(+)/IgG-N(-), IgG-S(+) was detected in 66.5%. The frequency of IgM-S(+)/IgG-S(-) in the total population was 10.0%, of whom 24.1% had TMA(+); the chances for TMA(+) in subjects with an IgM-S(+) alone pattern were 2.4%. Targeting of the same SARS-CoV-2 antigen seems to be better for the characterization of IgM/IgG patterns of response. IgM-S(+) alone reactivity is rare, and a small proportion is associated with viral shedding.
Collapse
Affiliation(s)
- Pablo Barreiro
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
- Council of Public Health, Community of Madrid, Calle O’Donnell, 50, 28009 Madrid, Spain; (J.C.); (F.J.M.-P.); (A.Z.)
| | - Francisco Javier Candel
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
- Council of Public Health, Community of Madrid, Calle O’Donnell, 50, 28009 Madrid, Spain; (J.C.); (F.J.M.-P.); (A.Z.)
| | - Juan Carlos Sanz
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
| | - Jesús San Román
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
- Council of Public Health, Community of Madrid, Calle O’Donnell, 50, 28009 Madrid, Spain; (J.C.); (F.J.M.-P.); (A.Z.)
| | - María del Mar Carretero
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
- Council of Public Health, Community of Madrid, Calle O’Donnell, 50, 28009 Madrid, Spain; (J.C.); (F.J.M.-P.); (A.Z.)
| | - Marta Pérez-Abeledo
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
| | - Belén Ramos
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
| | - José Manuel Viñuela-Prieto
- Public Health Regional Laboratory, Hospital Isabel Zendal, Av. Manuel Fraga Iribarne, 2, 28055 Madrid, Spain; (F.J.C.); (J.C.S.); (J.S.R.); (M.d.M.C.); (M.P.-A.); (B.R.); (J.M.V.-P.)
| | - Jesús Canora
- Council of Public Health, Community of Madrid, Calle O’Donnell, 50, 28009 Madrid, Spain; (J.C.); (F.J.M.-P.); (A.Z.)
| | | | - Antonio Zapatero
- Council of Public Health, Community of Madrid, Calle O’Donnell, 50, 28009 Madrid, Spain; (J.C.); (F.J.M.-P.); (A.Z.)
| |
Collapse
|
1238
|
Verbeke R, Lentacker I, De Smedt SC, Dewitte H. The dawn of mRNA vaccines: The COVID-19 case. J Control Release 2021; 333:511-520. [PMID: 33798667 PMCID: PMC8008785 DOI: 10.1016/j.jconrel.2021.03.043] [Citation(s) in RCA: 282] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
In less than one year since the outbreak of the COVID-19 pandemic, two mRNA-based vaccines, BNT162b2 and mRNA-1273, were granted the first historic authorization for emergency use, while another mRNA vaccine, CVnCoV, progressed to phase 3 clinical testing. The COVID-19 mRNA vaccines represent a new class of vaccine products, which consist of synthetic mRNA strands encoding the SARS-CoV-2 Spike glycoprotein, packaged in lipid nanoparticles to deliver mRNA to cells. This review digs deeper into the scientific breakthroughs of the last decades that laid the foundations for the rapid rise of mRNA vaccines during the COVID-19 pandemic. As well as providing momentum for mRNA vaccines, SARS-CoV-2 represents an ideal case study allowing to compare design-activity differences between the different mRNA vaccine candidates. Therefore, a detailed overview of the composition and (pre)clinical performance of the three most advanced mRNA vaccines is provided and the influence of choices in their structural design on to their immunogenicity and reactogenicity profile is discussed in depth. In addition to the new fundamental insights in the mRNA vaccines' mode of action highlighted here, we also point out which unknowns remain that require further investigation and possibly, optimization in future mRNA vaccine development.
Collapse
Affiliation(s)
- Rein Verbeke
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium.
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
1239
|
Gangaev A, Ketelaars SLC, Isaeva OI, Patiwael S, Dopler A, Hoefakker K, De Biasi S, Gibellini L, Mussini C, Guaraldi G, Girardis M, Ormeno CMPT, Hekking PJM, Lardy NM, Toebes M, Balderas R, Schumacher TN, Ovaa H, Cossarizza A, Kvistborg P. Identification and characterization of a SARS-CoV-2 specific CD8 + T cell response with immunodominant features. Nat Commun 2021; 12:2593. [PMID: 33972535 PMCID: PMC8110804 DOI: 10.1038/s41467-021-22811-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/26/2021] [Indexed: 02/03/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 is a continuous challenge worldwide, and there is an urgent need to map the landscape of immunogenic and immunodominant epitopes recognized by CD8+ T cells. Here, we analyze samples from 31 patients with COVID-19 for CD8+ T cell recognition of 500 peptide-HLA class I complexes, restricted by 10 common HLA alleles. We identify 18 CD8+ T cell recognized SARS-CoV-2 epitopes, including an epitope with immunodominant features derived from ORF1ab and restricted by HLA-A*01:01. In-depth characterization of SARS-CoV-2-specific CD8+ T cell responses of patients with acute critical and severe disease reveals high expression of NKG2A, lack of cytokine production and a gene expression profile inhibiting T cell re-activation and migration while sustaining survival. SARS-CoV-2-specific CD8+ T cell responses are detectable up to 5 months after recovery from critical and severe disease, and these responses convert from dysfunctional effector to functional memory CD8+ T cells during convalescence.
Collapse
Affiliation(s)
- Anastasia Gangaev
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Steven L. C. Ketelaars
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Olga I. Isaeva
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Sanne Patiwael
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Anna Dopler
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Kelly Hoefakker
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Sara De Biasi
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Lara Gibellini
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Cristina Mussini
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Giovanni Guaraldi
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Massimo Girardis
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Cami M. P. Talavera Ormeno
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, South Holland The Netherlands
| | - Paul J. M. Hekking
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, South Holland The Netherlands
| | - Neubury M. Lardy
- grid.417732.40000 0001 2234 6887Department of Immunogenetics, Sanquin Diagnostics B.V., Amsterdam, North Holland The Netherlands
| | - Mireille Toebes
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Robert Balderas
- grid.420052.10000 0004 0543 6807Department of Biological Sciences, BD Biosciences, San Jose, CA USA
| | - Ton N. Schumacher
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Huib Ovaa
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, South Holland The Netherlands
| | - Andrea Cossarizza
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Pia Kvistborg
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| |
Collapse
|
1240
|
de Mattos Barbosa MG, Liu H, Huynh D, Shelley G, Keller ET, Emmer BT, Sherman E, Ginsburg D, Kennedy AA, Tai AW, Wobus C, Mirabeli C, Lanigan TM, Samaniego M, Meng W, Rosenfeld AM, Prak ETL, Platt JL, Cascalho M. IgV somatic mutation of human anti-SARS-CoV-2 monoclonal antibodies governs neutralization and breadth of reactivity. JCI Insight 2021; 6:147386. [PMID: 33769311 PMCID: PMC8262290 DOI: 10.1172/jci.insight.147386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
Abs that neutralize SARS-CoV-2 are thought to provide the most immediate and effective treatment for those severely afflicted by this virus. Because coronavirus potentially diversifies by mutation, broadly neutralizing Abs are especially sought. Here, we report a possibly novel approach to rapid generation of potent broadly neutralizing human anti-SARS-CoV-2 Abs. We isolated SARS-CoV-2 spike protein-specific memory B cells by panning from the blood of convalescent subjects after infection with SARS-CoV-2 and sequenced and expressed Ig genes from individual B cells as human mAbs. All of 43 human mAbs generated in this way neutralized SARS-CoV-2. Eighteen of the forty-three human mAbs exhibited half-maximal inhibitory concentrations (IC50) of 6.7 × 10-12 M to 6.7 × 10-15 M for spike-pseudotyped virus. Seven of the human mAbs also neutralized (with IC50 < 6.7 × 10-12 M) viruses pseudotyped with mutant spike proteins (including receptor-binding domain mutants and the S1 C-terminal D614G mutant). Neutralization of the Wuhan Hu-1 founder strain and of some variants decreased when coding sequences were reverted to germline, suggesting that potency of neutralization was acquired by somatic hypermutation and selection of B cells. These results indicate that infection with SARS-CoV-2 evokes high-affinity B cell responses, some products of which are broadly neutralizing and others highly strain specific. We also identify variants that would potentially resist immunity evoked by infection with the Wuhan Hu-1 founder strain or by vaccines developed with products of that strain, suggesting evolutionary courses that SARS-CoV-2 could take.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Emily Sherman
- Department of Internal Medicine
- Life Sciences Institute
| | - David Ginsburg
- Department of Internal Medicine
- Life Sciences Institute
- Departments of Human Genetics and Pediatrics and Howard Hughes Medical Institute
| | | | | | | | | | - Thomas M. Lanigan
- Department of Internal Medicine
- Vector Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, Michigan, USA
| | - Milagros Samaniego
- Department of Medicine, Henry Ford Health Systems, Detroit, Michigan, USA
| | - Wenzhao Meng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Aaron M. Rosenfeld
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eline T. Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeffrey L. Platt
- Department of Surgery
- Department of Microbiology and Immunology, and
| | - Marilia Cascalho
- Department of Surgery
- Department of Microbiology and Immunology, and
| |
Collapse
|
1241
|
Townsend L, Dyer AH, Naughton A, Kiersey R, Holden D, Gardiner M, Dowds J, O’Brien K, Bannan C, Nadarajan P, Dunne J, Martin-Loeches I, Fallon PG, Bergin C, O’Farrelly C, Cheallaigh CN, Bourke NM, Conlon N. Longitudinal Analysis of COVID-19 Patients Shows Age-Associated T Cell Changes Independent of Ongoing Ill-Health. Front Immunol 2021; 12:676932. [PMID: 34025675 PMCID: PMC8138306 DOI: 10.3389/fimmu.2021.676932] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives The immunological and inflammatory changes following acute COVID-19 are hugely variable. Persistent clinical symptoms following resolution of initial infection, termed long COVID, are also hugely variable, but association with immunological changes has not been described. We investigate changing immunological parameters in convalescent COVID-19 and interrogate their potential relationships with persistent symptoms. Methods We performed paired immunophenotyping at initial SARS-CoV-2 infection and convalescence (n=40, median 68 days) and validated findings in 71 further patients at median 101 days convalescence. Results were compared to 40 pre-pandemic controls. Fatigue and exercise tolerance were assessed as cardinal features of long COVID using the Chalder Fatigue Scale and 6-minute-walk test. The relationships between these clinical outcomes and convalescent immunological results were investigated. Results We identify persistent expansion of intermediate monocytes, effector CD8+, activated CD4+ and CD8+ T cells, and reduced naïve CD4+ and CD8+ T cells at 68 days, with activated CD8+ T cells remaining increased at 101 days. Patients >60 years also demonstrate reduced naïve CD4+ and CD8+ T cells and expanded activated CD4+ T cells at 101 days. Ill-health, fatigue, and reduced exercise tolerance were common in this cohort. These symptoms were not associated with immune cell populations or circulating inflammatory cytokines. Conclusion We demonstrate myeloid recovery but persistent T cell abnormalities in convalescent COVID-19 patients more than three months after initial infection. These changes are more marked with age and are independent of ongoing subjective ill-health, fatigue and reduced exercise tolerance.
Collapse
Affiliation(s)
- Liam Townsend
- Department of Infectious Diseases, St James’s Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Adam H. Dyer
- Department of Medical Gerontology, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Aifric Naughton
- Department of Immunology, St James’s Hospital, Dublin, Ireland
| | - Rachel Kiersey
- Department of Immunology, St James’s Hospital, Dublin, Ireland
| | - Dean Holden
- Department of Immunology, St James’s Hospital, Dublin, Ireland
| | - Mary Gardiner
- Department of Immunology, St James’s Hospital, Dublin, Ireland
| | - Joanne Dowds
- Department of Physiotherapy, St James’s Hospital, Dublin, Ireland
| | - Kate O’Brien
- Department of Physiotherapy, St James’s Hospital, Dublin, Ireland
| | - Ciaran Bannan
- Department of Infectious Diseases, St James’s Hospital, Dublin, Ireland
| | | | - Jean Dunne
- Department of Immunology, St James’s Hospital, Dublin, Ireland
| | | | - Padraic G. Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Colm Bergin
- Department of Infectious Diseases, St James’s Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Cliona O’Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Cliona Ni Cheallaigh
- Department of Infectious Diseases, St James’s Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Nollaig M. Bourke
- Department of Medical Gerontology, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Niall Conlon
- Department of Immunology, St James’s Hospital, Dublin, Ireland
- Department of Immunology, School of Medicine, Trinity College, Dublin, Ireland
| |
Collapse
|
1242
|
Mazzoni A, Maggi L, Capone M, Vanni A, Spinicci M, Salvati L, Tekle Kiros S, Semeraro R, Pengue L, Colao MG, Magi A, Rossolini GM, Liotta F, Cosmi L, Bartoloni A, Annunziato F. Heterogeneous magnitude of immunological memory to SARS-CoV-2 in recovered individuals. Clin Transl Immunology 2021; 10:e1281. [PMID: 33976879 PMCID: PMC8101693 DOI: 10.1002/cti2.1281] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 12/26/2022] Open
Abstract
Objective Although the adaptive immune response to SARS‐CoV‐2 has been characterised in the acute and early convalescent phase of the disease, few studies explore whether natural infection elicits long‐lasting immunological memory in recovered individuals. In this work, we aimed to assess the maintenance of immunological memory to SARS‐CoV‐2. Methods We evaluated the long‐term virus‐specific cellular and humoral immune response in the members of an Italian Serie A football team, who experienced a cluster of COVID‐19 in March 2020, which was strictly evaluated in the following months. Results Our results highlight a heterogeneous magnitude of immunological memory at 5 months after infection. Indeed, 20% of the subjects displayed a weak cellular and humoral memory to SARS‐CoV‐2, suggesting that they may be at higher risk of reinfection. In addition, a history of symptomatic COVID‐19 was associated with higher levels of SARS‐CoV‐2‐reactive CD4+ T cells and specific antibody levels than in asymptomatic individuals. Conclusion Collectively, these data demonstrate that immunity to SARS‐CoV‐2 is maintained five months postinfection even if the magnitude of response is heterogeneous among individuals. This finding suggests that some COVID‐19‐recovered subjects may benefit from vaccination.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Manuela Capone
- 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 Diseases Unit Careggi University Hospital Florence Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Seble Tekle Kiros
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine University of Florence Florence Italy
| | - Luca Pengue
- Sports Medicine Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Maria Grazia Colao
- Microbiology and Virology Unit Careggi University Hospital Florence Italy
| | - Alberto Magi
- Department of Information Engineering University of Florence 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
| | - 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 (CDCI) 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
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Infectious and Tropical Diseases Unit Careggi University Hospital Florence Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine University of Florence Florence Italy.,Flow Cytometry Diagnostic Center and Immunotherapy (CDCI) Careggi University Hospital Florence Italy
| |
Collapse
|
1243
|
Mysore V, Cullere X, Settles ML, Ji X, Kattan MW, Desjardins M, Durbin-Johnson B, Gilboa T, Baden LR, Walt DR, Lichtman A, Jehi L, Mayadas TN. Protective heterologous T cell immunity in COVID-19 induced by MMR and Tdap vaccine antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.03.441323. [PMID: 33972940 PMCID: PMC8109200 DOI: 10.1101/2021.05.03.441323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
T cells are critical for control of viral infection and effective vaccination. We investigated whether prior Measles-Mumps-Rubella (MMR) or Tetanus-Diphtheria-pertussis (Tdap) vaccination elicit cross-reactive T cells that mitigate COVID-19. Using co-cultures of antigen presenting cells (APC) loaded with antigens and autologous T cells, we found a high correlation between responses to SARS-CoV-2 (Spike-S1 and Nucleocapsid) and MMR and Tdap vaccine proteins in both SARS-CoV-2 infected individuals and individuals immunized with mRNA-based SARS-CoV-2 vaccines. The overlapping T cell population contained effector memory T cells (TEMRA) previously implicated in anti-viral immunity and their activation required APC-derived IL-15. TCR- and scRNA-sequencing detected cross-reactive clones with TEMRA features among the cells recognizing SARS-CoV-2, MMR and Tdap epitopes. A propensity-weighted analysis of 73,582 COVID-19 patients revealed that severe disease outcomes (hospitalization and transfer to intensive care unit or death) were reduced in MMR or Tdap vaccinated individuals by 38-32% and 23-20% respectively. In summary, SARS-CoV-2 re-activates memory T cells generated by Tdap and MMR vaccines, which may reduce disease severity.
Collapse
|
1244
|
Gallagher KM, Leick MB, Larson RC, Berger TR, Katsis K, Yam JY, Brini G, Grauwet K, COVID-19 Collection & Processing Team, Maus MV. SARS -CoV-2 T-cell immunity to variants of concern following vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.03.442455. [PMID: 33972942 PMCID: PMC8109204 DOI: 10.1101/2021.05.03.442455] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, two mRNA vaccines to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have become available, but there is also an emergence of SARS-CoV-2 variants with increased transmissibility and virulence1-6. A major concern is whether the available vaccines will be equally effective against these variants. The vaccines are designed to induce an immune response against the SARS-CoV-2 spike protein7,8, which is required for viral entry to host cells9. Immunity to SARS-CoV-2 is often evaluated by antibody production, while less is known about the T-cell response. Here we developed, characterized, and implemented two standardized, functional assays to measure T-cell immunity to SARS-CoV-2 in uninfected, convalescent, and vaccinated individuals. We found that vaccinated individuals had robust T-cell responses to the wild type spike and nucleocapsid proteins, even more so than convalescent patients. We also found detectable but diminished T-cell responses to spike variants (B.1.1.7, B.1.351, and B.1.1.248) among vaccinated but otherwise healthy donors. Since decreases in antibody neutralization have also been observed with some variants10-12, investigation into the T-cell response to these variants as an alternative means of viral control is imperative. Standardized measurements of T-cell responses to SARS-CoV-2 are feasible and can be easily adjusted to determine changes in response to variants.
Collapse
Affiliation(s)
- Kathleen M.E. Gallagher
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark B. Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca C. Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Trisha R. Berger
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Katelin Katsis
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jennifer Y. Yam
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gabrielle Brini
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Korneel Grauwet
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Marcela V. Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
1245
|
Jangra S, Landers JJ, Rathnasinghe R, O'Konek JJ, Janczak KW, Cascalho M, Kennedy AA, Tai AW, Baker JR, Schotsaert M, Wong PT. A Combination Adjuvant for the Induction of Potent Antiviral Immune Responses for a Recombinant SARS-CoV-2 Protein Vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.18.431484. [PMID: 33619480 PMCID: PMC7899444 DOI: 10.1101/2021.02.18.431484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Several SARS-CoV-2 vaccines have received EUAs, but many issues remain unresolved, including duration of conferred immunity and breadth of cross-protection. Adjuvants that enhance and shape adaptive immune responses that confer broad protection against SARS-CoV-2 variants will be pivotal for long-term protection. We developed an intranasal, rationally designed adjuvant integrating a nanoemulsion (NE) that activates TLRs and NLRP3 with an RNA agonist of RIG-I (IVT DI). The combination adjuvant with spike protein antigen elicited robust responses to SARS-CoV-2 in mice, with markedly enhanced T H 1-biased cellular responses and high virus-neutralizing antibody titers towards both homologous SARS-CoV-2 and a variant harboring the N501Y mutation shared by B1.1.7, B.1.351 and P.1 variants. Furthermore, passive transfer of vaccination-induced antibodies protected naive mice against heterologous viral challenge. NE/IVT DI enables mucosal vaccination, and has the potential to improve the immune profile of a variety of SARS-CoV-2 vaccine candidates to provide effective cross-protection against future drift variants.
Collapse
|
1246
|
Croce L, Gangemi D, Ancona G, Liboà F, Bendotti G, Minelli L, Chiovato L. The cytokine storm and thyroid hormone changes in COVID-19. J Endocrinol Invest 2021; 44:891-904. [PMID: 33559848 PMCID: PMC7871522 DOI: 10.1007/s40618-021-01506-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/09/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND COVID-19 is now a worldwide pandemic. Among the many extra-pulmonary manifestations of COVID-19, recent evidence suggested a possible occurrence of thyroid dysfunction. PURPOSE The Aim of the present review is to summarize available studies regarding thyroid function alterations in patients with COVID-19 and to overview the possible physio-pathological explanations. CONCLUSIONS The repercussions of the thyroid of COVID-19 seem to be related, in part, with the occurrence of a "cytokine storm" that would, in turn, induce a "non-thyroidal illness". Some specific cytokines and chemokines appear to have a direct role on the hypothalamus-pituitary-thyroid axis. On the other hand, some authors have observed an increased incidence of a destructive thyroiditis, either subacute or painless, in patients with COVID-19. The hypothesis of a direct infection of the thyroid by SARS-Cov-2 stems from the observation that its receptor, ACE2, is strongly expressed in thyroid tissue. Lastly, it is highly probable that some pharmaceutical agents largely used for the treatment of COVID-19 can act as confounding factors in the laboratory evaluation of thyroid function parameters.
Collapse
Affiliation(s)
- L Croce
- Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
- PHD Course in Experimental Medicine, University of Pavia, 27100, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
| | - D Gangemi
- Postgraduate School in Endocrinology and Metabolism, University of Pavia, 27100, Pavia, Italy
| | - G Ancona
- Postgraduate School in Endocrinology and Metabolism, University of Pavia, 27100, Pavia, Italy
| | - F Liboà
- Postgraduate School in Endocrinology and Metabolism, University of Pavia, 27100, Pavia, Italy
| | - G Bendotti
- Postgraduate School in Endocrinology and Metabolism, University of Pavia, 27100, Pavia, Italy
| | - L Minelli
- Postgraduate School in Endocrinology and Metabolism, University of Pavia, 27100, Pavia, Italy
| | - L Chiovato
- Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy.
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy.
| |
Collapse
|
1247
|
Fournier PE, Colson P, Levasseur A, Devaux CA, Gautret P, Bedotto M, Delerce J, Brechard L, Pinault L, Lagier JC, Fenollar F, Raoult D. Emergence and outcomes of the SARS-CoV-2 'Marseille-4' variant. Int J Infect Dis 2021; 106:228-236. [PMID: 33785459 PMCID: PMC7997945 DOI: 10.1016/j.ijid.2021.03.068] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND In Marseille, France, following a first severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak in March-May 2020, a second epidemic phase occurred from June, involving 10 new variants. The Marseille-4 variant caused an epidemic that started in August and is still ongoing. METHODS The 1038 SARS-CoV-2 whole genome sequences obtained in our laboratory by next-generation sequencing with Illumina technology were analysed using Nextclade and nextstrain/ncov pipelines and IQ-TREE. A Marseille-4-specific qPCR assay was implemented. Demographic and clinical features were compared between patients with the Marseille-4 variant and those with earlier strains. RESULTS Marseille-4 harbours 13 hallmark mutations. One leads to an S477N substitution in the receptor binding domain of the spike protein targeted by current vaccines. Using a specific qPCR, it was observed that Marseille-4 caused 12-100% of SARS-CoV-2 infections in Marseille from September 2020, being involved in 2106 diagnoses. This variant was more frequently associated with hypoxemia than were clade 20A strains before May 2020. It caused a re-infection in 11 patients diagnosed with different SARS-CoV-2 strains before June 2020, suggesting either short-term protective immunity or a lack of cross-immunity. CONCLUSIONS Marseille-4 should be considered as a major SARS-CoV-2 variant. Its sudden appearance points towards an animal reservoir, possibly mink. The protective role of past exposure and current vaccines against this variant should be evaluated.
Collapse
Affiliation(s)
- Pierre-Edouard Fournier
- IHU Méditerranée Infection, Marseille, France; Vecteurs - Infections Tropicales et Méditerranéennes (VITROME), Marseille, France.
| | - Philippe Colson
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Aix-Marseille Université, Marseille, France
| | - Anthony Levasseur
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Aix-Marseille Université, Marseille, France
| | - Christian A Devaux
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Aix-Marseille Université, Marseille, France
| | - Philippe Gautret
- IHU Méditerranée Infection, Marseille, France; Vecteurs - Infections Tropicales et Méditerranéennes (VITROME), Marseille, France
| | | | | | | | | | - Jean-Christophe Lagier
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Aix-Marseille Université, Marseille, France
| | - Florence Fenollar
- IHU Méditerranée Infection, Marseille, France; Vecteurs - Infections Tropicales et Méditerranéennes (VITROME), Marseille, France
| | - Didier Raoult
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Aix-Marseille Université, Marseille, France.
| |
Collapse
|
1248
|
Ambrosioni J, Blanco JL, Reyes-Urueña JM, Davies MA, Sued O, Marcos MA, Martínez E, Bertagnolio S, Alcamí J, Miro JM. Overview of SARS-CoV-2 infection in adults living with HIV. Lancet HIV 2021; 8:e294-e305. [PMID: 33915101 PMCID: PMC8075775 DOI: 10.1016/s2352-3018(21)00070-9] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022]
Abstract
Around 2·5 million deaths and more than 110 million COVID-19 cases have been reported globally. Although it initially appeared that HIV infection was not a risk factor for COVID-19 or more severe disease, more recent large studies suggest that people living with HIV (particularly with low CD4 cell counts or untreated HIV infection) might have a more severe clinical course than those who are HIV-negative. Moreover, the COVID-19 pandemic has disrupted HIV prevention and treatment services worldwide, creating huge challenges to the continuity of essential activities. We have reviewed the most relevant features of COVID-19 in people living with HIV and highlighted topics where further research is required.
Collapse
Affiliation(s)
- Juan Ambrosioni
- Infectious Diseases Service, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain.
| | - José Luis Blanco
- Infectious Diseases Service, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Juliana M Reyes-Urueña
- Centre d'Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), Agència de Salut Pública de Catalunya, Barcelona, Spain; CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
| | - Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina
| | - Maria Angeles Marcos
- Microbiology Service, Hospital Clinic-ISGlobal, University of Barcelona, Barcelona, Spain
| | - Esteban Martínez
- Infectious Diseases Service, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Silvia Bertagnolio
- Department of Global HIV, Hepatitis, and STI Programmes, World Health Organization, Geneva, Switzerland; Department of Infection and Population Health, University College London, London, UK
| | - Jose Alcamí
- Infectious Diseases Service, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain; AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose M Miro
- Infectious Diseases Service, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
1249
|
Zhao C, Fang X, Feng Y, Fang X, He J, Pan H. Emerging role of air pollution and meteorological parameters in COVID-19. J Evid Based Med 2021; 14:123-138. [PMID: 34003571 PMCID: PMC8207011 DOI: 10.1111/jebm.12430] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 01/09/2023]
Abstract
Exposure to air pollutants has been associated with respiratory viral infections. Epidemiological studies have shown that air pollution exposure is related to increased cases of SARS-COV-2 infection and COVID-19-associated mortality. In addition, the changes of meteorological parameters have also been implicated in the occurrence and development of COVID-19. However, the molecular mechanisms by which pollutant exposure and changes of meteorological parameters affects COVID-19 remains unknown. This review summarizes the biology of COVID-19 and the route of viral transmission, and elaborates on the relationship between air pollution and climate indicators and COVID-19. Finally, we envisaged the potential roles of air pollution and meteorological parameters in COVID-19.
Collapse
Affiliation(s)
- Channa Zhao
- Anhui Provincial Tuberculosis InstituteHefeiAnhuiChina
| | - Xinyu Fang
- Department of Epidemiology and Biostatistics, School of Public HealthAnhui Medical UniversityHefeiAnhuiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceHefeiAnhuiChina
| | - Yating Feng
- Department of Epidemiology and Biostatistics, School of Public HealthAnhui Medical UniversityHefeiAnhuiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceHefeiAnhuiChina
| | - Xuehui Fang
- Anhui Provincial Tuberculosis InstituteHefeiAnhuiChina
| | - Jun He
- Anhui Provincial Center for Disease Control and PreventionHefeiChina
- Key Laboratory for Medical and Health of the 13th Five‐Year PlanHefeiAnhuiChina
| | - Haifeng Pan
- Department of Epidemiology and Biostatistics, School of Public HealthAnhui Medical UniversityHefeiAnhuiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceHefeiAnhuiChina
| |
Collapse
|
1250
|
Mazereel V, Van Assche K, Detraux J, De Hert M. COVID-19 vaccination for people with severe mental illness: why, what, and how? Lancet Psychiatry 2021; 8:444-450. [PMID: 33548184 PMCID: PMC7906686 DOI: 10.1016/s2215-0366(20)30564-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
Psychiatric disorders, and especially severe mental illness, are associated with an increased risk of severe acute respiratory syndrome coronavirus 2 infection and COVID-19-related morbidity and mortality. People with severe mental illness should therefore be prioritised in vaccine allocation strategies. Here, we discuss the risk for worse COVID-19 outcomes in this vulnerable group, the effect of severe mental illness and psychotropic medications on vaccination response, the attitudes of people with severe mental illness towards vaccination, and, the potential barriers to, and possible solutions for, an efficient vaccination programme in this population.
Collapse
Affiliation(s)
- Victor Mazereel
- Center for Clinical Psychiatry, KU Leuven, Leuven, Belgium; Department of Neurosciences, and University Psychiatric Center, KU Leuven, Leuven, Belgium
| | - Kristof Van Assche
- Research Group Personal Rights and Property Rights, Faculty of Law, University of Antwerp, Antwerp, Belgium
| | - Johan Detraux
- Public Health Psychiatry, KU Leuven, Leuven, Belgium; Department of Neurosciences, and University Psychiatric Center, KU Leuven, Leuven, Belgium
| | - Marc De Hert
- Center for Clinical Psychiatry, KU Leuven, Leuven, Belgium; Department of Neurosciences, and University Psychiatric Center, KU Leuven, Leuven, Belgium; Antwerp Health Law and Ethics Chair, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|