1301
|
Koch T, Mellinghoff SC, Shamsrizi P, Addo MM, Dahlke C. Correlates of Vaccine-Induced Protection against SARS-CoV-2. Vaccines (Basel) 2021; 9:238. [PMID: 33801831 PMCID: PMC8035658 DOI: 10.3390/vaccines9030238] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
We are in the midst of a pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes the coronavirus disease 2019 (COVID-19). SARS-CoV-2 has caused more than two million deaths after one year of the pandemic. The world is experiencing a deep economic recession. Safe and effective vaccines are needed to prevent further morbidity and mortality. Vaccine candidates against COVID-19 have been developed at an unprecedented speed, with more than 200 vaccine candidates currently under investigation. Among those, 20 candidates have entered the clinical Phase 3 to evaluate efficacy, and three have been approved by the European Medicines Agency. The aim of immunization is to act against infection, disease and/or transmission. However, the measurement of vaccine efficacy is challenging, as efficacy trials need to include large cohorts with verum and placebo cohorts. In the future, this will be even more challenging as further vaccine candidates will receive approval, an increasing number of humans will receive vaccinations and incidence might decrease. To evaluate novel and second-generation vaccine candidates, randomized placebo-controlled trials might not be appropriate anymore. Correlates of protection (CoP) could be an important tool to evaluate novel vaccine candidates, but vaccine-induced CoP have not been clearly defined for SARS-CoV-2 vaccines. In this review, we report on immunogenicity against natural SARS-CoV-2 infection, vaccine-induced immune responses and discuss immunological markers that can be linked to protection. By discussing the immunogenicity and efficacy of forerunner vaccines, we aim to give a comprehensive overview of possible efficacy measures and CoP.
Collapse
Affiliation(s)
- Till Koch
- Division of Infectious Diseases, 1st Department of Medicine, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany; (T.K.); (S.C.M.); (P.S.); (M.M.A.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Sibylle C. Mellinghoff
- Division of Infectious Diseases, 1st Department of Medicine, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany; (T.K.); (S.C.M.); (P.S.); (M.M.A.)
- Excellence Centre for Medical Mycology (ECMM), 1st Department of Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Translational Research, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, 50937 Cologne, Germany
| | - Parichehr Shamsrizi
- Division of Infectious Diseases, 1st Department of Medicine, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany; (T.K.); (S.C.M.); (P.S.); (M.M.A.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Marylyn M. Addo
- Division of Infectious Diseases, 1st Department of Medicine, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany; (T.K.); (S.C.M.); (P.S.); (M.M.A.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Christine Dahlke
- Division of Infectious Diseases, 1st Department of Medicine, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany; (T.K.); (S.C.M.); (P.S.); (M.M.A.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| |
Collapse
|
1302
|
Park J, Foox J, Hether T, Danko D, Warren S, Kim Y, Reeves J, Butler DJ, Mozsary C, Rosiene J, Shaiber A, Afshinnekoo E, MacKay M, Bram Y, Chandar V, Geiger H, Craney A, Velu P, Melnick AM, Hajirasouliha I, Beheshti A, Taylor D, Saravia-Butler A, Singh U, Wurtele ES, Schisler J, Fennessey S, Corvelo A, Zody MC, Germer S, Salvatore S, Levy S, Wu S, Tatonetti N, Shapira S, Salvatore M, Loda M, Westblade LF, Cushing M, Rennert H, Kriegel AJ, Elemento O, Imielinski M, Borczuk AC, Meydan C, Schwartz RE, Mason CE. Systemic Tissue and Cellular Disruption from SARS-CoV-2 Infection revealed in COVID-19 Autopsies and Spatial Omics Tissue Maps. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33758858 DOI: 10.1101/2021.03.08.434433] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) virus has infected over 115 million people and caused over 2.5 million deaths worldwide. Yet, the molecular mechanisms underlying the clinical manifestations of COVID-19, as well as what distinguishes them from common seasonal influenza virus and other lung injury states such as Acute Respiratory Distress Syndrome (ARDS), remains poorly understood. To address these challenges, we combined transcriptional profiling of 646 clinical nasopharyngeal swabs and 39 patient autopsy tissues, matched with spatial protein and expression profiling (GeoMx) across 357 tissue sections. These results define both body-wide and tissue-specific (heart, liver, lung, kidney, and lymph nodes) damage wrought by the SARS-CoV-2 infection, evident as a function of varying viral load (high vs. low) during the course of infection and specific, transcriptional dysregulation in splicing isoforms, T cell receptor expression, and cellular expression states. In particular, cardiac and lung tissues revealed the largest degree of splicing isoform switching and cell expression state loss. Overall, these findings reveal a systemic disruption of cellular and transcriptional pathways from COVID-19 across all tissues, which can inform subsequent studies to combat the mortality of COVID-19, as well to better understand the molecular dynamics of lethal SARS-CoV-2 infection and other viruses.
Collapse
|
1303
|
Le Bert N, Clapham HE, Tan AT, Chia WN, Tham CYL, Lim JM, Kunasegaran K, Tan LWL, Dutertre CA, Shankar N, Lim JME, Sun LJ, Zahari M, Tun ZM, Kumar V, Lim BL, Lim SH, Chia A, Tan YJ, Tambyah PA, Kalimuddin S, Lye D, Low JGH, Wang LF, Wan WY, Hsu LY, Bertoletti A, Tam CC. Highly functional virus-specific cellular immune response in asymptomatic SARS-CoV-2 infection. J Exp Med 2021; 218:211835. [PMID: 33646265 PMCID: PMC7927662 DOI: 10.1084/jem.20202617] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/21/2021] [Accepted: 02/11/2021] [Indexed: 12/23/2022] Open
Abstract
The efficacy of virus-specific T cells in clearing pathogens involves a fine balance between antiviral and inflammatory features. SARS-CoV-2–specific T cells in individuals who clear SARS-CoV-2 without symptoms could reveal nonpathological yet protective characteristics. We longitudinally studied SARS-CoV-2–specific T cells in a cohort of asymptomatic (n = 85) and symptomatic (n = 75) COVID-19 patients after seroconversion. We quantified T cells reactive to structural proteins (M, NP, and Spike) using ELISpot and cytokine secretion in whole blood. Frequencies of SARS-CoV-2–specific T cells were similar between asymptomatic and symptomatic individuals, but the former showed an increased IFN-γ and IL-2 production. This was associated with a proportional secretion of IL-10 and proinflammatory cytokines (IL-6, TNF-α, and IL-1β) only in asymptomatic infection, while a disproportionate secretion of inflammatory cytokines was triggered by SARS-CoV-2–specific T cell activation in symptomatic individuals. Thus, asymptomatic SARS-CoV-2–infected individuals are not characterized by weak antiviral immunity; on the contrary, they mount a highly functional virus-specific cellular immune response.
Collapse
Affiliation(s)
- Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Hannah E Clapham
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Anthony T Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Wan Ni Chia
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Christine Y L Tham
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Jane M Lim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Kamini Kunasegaran
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Linda Wei Lin Tan
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | | | - Nivedita Shankar
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Joey M E Lim
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Louisa Jin Sun
- Infectious Diseases, Alexandra Hospital, National University Health System, Singapore
| | - Marina Zahari
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Zaw Myo Tun
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Vishakha Kumar
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Beng Lee Lim
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Siew Hoon Lim
- Department of Microbiology, Singapore General Hospital, Singapore
| | - Adeline Chia
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Yee-Joo Tan
- Infectious Diseases Translational Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | - Shirin Kalimuddin
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - David Lye
- Infectious Diseases Translational Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,National Center of Infectious Diseases, Singapore.,Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Singapore
| | - Jenny G H Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Wei Yee Wan
- Department of Microbiology, Singapore General Hospital, Singapore
| | - Li Yang Hsu
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Singapore Immunology Network, A*STAR, Singapore
| | - Clarence C Tam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore.,London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
1304
|
Gao L, Zhou J, Yang S, Wang L, Chen X, Yang Y, Li R, Pan Z, Zhao J, Li Z, Huang Q, Tang J, Hu L, Liu P, Zhang G, Chen Y, Ye L. The dichotomous and incomplete adaptive immunity in COVID-19 patients with different disease severity. Signal Transduct Target Ther 2021; 6:113. [PMID: 33686064 PMCID: PMC7938043 DOI: 10.1038/s41392-021-00525-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/27/2021] [Accepted: 02/04/2021] [Indexed: 12/28/2022] Open
Abstract
The adaptive immunity that protects patients from coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is not well characterized. In particular, the asymptomatic patients have been found to induce weak and transient SARS-CoV-2 antibody responses, but the underlying mechanisms remain unknown; meanwhile, the protective immunity that guide the recovery of these asymptomatic patients is elusive. Here, we characterized SARS-CoV-2-specific B-cell and T-cell responses in 10 asymptomatic patients and 64 patients with other disease severity (mild, n = 10, moderate, n = 32, severe, n = 12) and found that asymptomatic or mild symptomatic patients failed to mount virus-specific germinal center (GC) B cell responses that result in robust and prolonged humoral immunity, assessed by GC response indicators including follicular helper T (TFH) cell and memory B cell responses as well as serum CXCL13 levels. Alternatively, these patients mounted potent virus-specific TH1 and CD8+ T cell responses. In sharp contrast, patients of moderate or severe disease induced vigorous virus-specific GC B cell responses and associated TFH responses; however, the virus-specific TH1 and CD8+ T cells were minimally induced in these patients. These results, therefore, uncovered the protective immunity in asymptomatic patients and also revealed the strikingly dichotomous and incomplete humoral and cellular immune responses in COVID-19 patients with different disease severity, providing important insights into rational design of effective COVID-19 vaccines.
Collapse
Affiliation(s)
- Leiqiong Gao
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jing Zhou
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Sen Yang
- Chongqing Public Health Medical Center, Chongqing, China
| | - Lisha Wang
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Xiangyu Chen
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Yang Yang
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Ren Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Zhiwei Pan
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jing Zhao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhirong Li
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Qizhao Huang
- Cancer Center, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Jianfang Tang
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Li Hu
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Pinghuang Liu
- Comparative Immunology Research Center, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Guozhong Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Yaokai Chen
- Chongqing Public Health Medical Center, Chongqing, China.
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, China.
| |
Collapse
|
1305
|
Winchester N, Calabrese C, Calabrese L. The Intersection of COVID-19 and Autoimmunity: What is Our Current Understanding? Pathog Immun 2021; 6:31-54. [PMID: 33969248 PMCID: PMC8097827 DOI: 10.20411/pai.v6i1.417] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/16/2021] [Indexed: 12/16/2022] Open
Abstract
Viral infections have historically had a complex relationship with autoimmune diseases. For patients with preexisting autoimmune disorders, often complicated by immunosuppressive therapies, there are numerous potential effects of COVID-19, a disease of complex immunobiology, including the potential for an altered natural history of COVID-19 when infected. In addition, individuals without recognized autoimmune disease may be vulnerable to virally induced autoimmunity in the forms of autoantibody formation, as well as the development of clinical immune-mediated inflammatory diseases. Until quite recently in the pandemic, this relationship between COVID-19 and autoimmune diseases has been relatively underexplored; yet such investigation offers potential insights into immunopathogenesis as well as for the development of new immune-based therapeutics. Our review examines this relationship through exploration of a series of questions with relevance to both immunopathogenic mechanisms as well as some clinical implications.
Collapse
Affiliation(s)
- N. Winchester
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - C. Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH
| | - L.H. Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
1306
|
Perelson AS, Ke R. Mechanistic Modeling of SARS-CoV-2 and Other Infectious Diseases and the Effects of Therapeutics. Clin Pharmacol Ther 2021; 109:829-840. [PMID: 33410134 DOI: 10.1002/cpt.2160] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022]
Abstract
Modern viral kinetic modeling and its application to therapeutics is a field that attracted the attention of the medical, pharmaceutical, and modeling communities during the early days of the AIDS epidemic. Its successes led to applications of modeling methods not only to HIV but a plethora of other viruses, such as hepatitis C virus (HCV), hepatitis B virus and cytomegalovirus, which along with HIV cause chronic diseases, and viruses such as influenza, respiratory syncytial virus, West Nile virus, Zika virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which generally cause acute infections. Here we first review the historical development of mathematical models to understand HIV and HCV infections and the effects of treatment by fitting the models to clinical data. We then focus on recent efforts and contributions of applying these models towards understanding SARS-CoV-2 infection and highlight outstanding questions where modeling can provide crucial insights and help to optimize nonpharmaceutical and pharmaceutical interventions of the coronavirus disease 2019 (COVID-19) pandemic. The review is written from our personal perspective emphasizing the power of simple target cell limited models that provided important insights and then their evolution into more complex models that captured more of the virology and immunology. To quote Albert Einstein, "Everything should be made as simple as possible, but not simpler," and this idea underlies the modeling we describe below.
Collapse
Affiliation(s)
- Alan S Perelson
- Los Alamos National Laboratory, Theoretical Biology and Biophysics Group, Los Alamos, New Mexico, USA.,New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Ruian Ke
- Los Alamos National Laboratory, Theoretical Biology and Biophysics Group, Los Alamos, New Mexico, USA.,New Mexico Consortium, Los Alamos, New Mexico, USA
| |
Collapse
|
1307
|
Della Valle P, Fabbri M, Madotto F, Ferrara P, Cozzolino P, Calabretto E, D'Orso MI, Longhi E, Polosa R, Riva MA, Mazzaglia G, Sommese C, Mantovani LG, The Mustang-Occupation-Covid-Study Group. Occupational Exposure in the Lombardy Region (Italy) to SARS-CoV-2 Infection: Results from the MUSTANG-OCCUPATION-COVID-19 Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2567. [PMID: 33806578 PMCID: PMC7967539 DOI: 10.3390/ijerph18052567] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 01/12/2023]
Abstract
Sero-epidemiological surveys are valuable attempts to estimate the circulation of SARS-CoV-2 in general or selected populations. Within this context, a prospective observational study was conducted to estimate the prevalence and persistence of SARS-CoV-2 antibodies in different categories of workers and factors associated with positivity, through the detection of virus-specific immunoglobulin G and M (IgG/IgM) in serum samples. Enrollees were divided in low exposure and medium-high groups on the basis of their work activity. Antibody responders were re-contacted after 3 months for the follow-up. Of 2255 sampled workers, 4.8% tested positive for SARS-CoV-2 IgG/IgM antibodies, with 81.7% to IgG only. Workers who continued to go to their place of work, were healthcare workers, or experienced at least one COVID-19-related symptom were more likely to test positive for SARS-CoV-2 antibodies. SARS-CoV-2 antibodies prevalence was significantly higher in the medium-high risk vs. low-risk group (7.2% vs. 3.0%, p < 0.0001). At 3-month follow-up, 81.3% of subjects still had antibody response. This study provided important information of SARS-CoV-2 infection prevalence among workers in northern Italy, where the impact of COVID-19 was particularly intense. The presented surveillance data give a contribution to refine current estimates of the disease burden expected from the SARS-CoV-2.
Collapse
Affiliation(s)
- Paola Della Valle
- Center for Public Health Research, University of Milan Bicocca, 20900 Monza, Italy
- IRCCS MultiMedica, 20099 Sesto San Giovanni, Italy
| | - Marco Fabbri
- Center for Public Health Research, University of Milan Bicocca, 20900 Monza, Italy
- IRCCS MultiMedica, 20099 Sesto San Giovanni, Italy
| | | | - Pietro Ferrara
- Center for Public Health Research, University of Milan Bicocca, 20900 Monza, Italy
- IRCCS MultiMedica, 20099 Sesto San Giovanni, Italy
| | | | | | - Marco Italo D'Orso
- Consortium for Occupational and Environmental Medicine, Department of Medicine and Surgery, University of Milan-Bicocca, 20900 Monza, Italy
| | | | - Riccardo Polosa
- Center of Excellence for the Acceleration of HArm Reduction (CoEHAR), University of Catania, 95131 Catania, Italy
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy
| | - Michele Augusto Riva
- Center for Public Health Research, University of Milan Bicocca, 20900 Monza, Italy
| | - Giampiero Mazzaglia
- Center for Public Health Research, University of Milan Bicocca, 20900 Monza, Italy
| | | | - Lorenzo Giovanni Mantovani
- Center for Public Health Research, University of Milan Bicocca, 20900 Monza, Italy
- IRCCS MultiMedica, 20099 Sesto San Giovanni, Italy
| | | |
Collapse
|
1308
|
Yang J, Zhong M, Hong K, Yang Q, Zhang E, Zhou D, Xia J, Chen Y, Sun M, Zhao B, Xiang J, Liu Y, Han Y, Xu M, Zhou X, Huang C, Shang Y, Yan H. Characteristics of T-cell responses in COVID-19 patients with prolonged SARS-CoV-2 positivity - a cohort study. Clin Transl Immunology 2021; 10:e1259. [PMID: 33728049 PMCID: PMC7932004 DOI: 10.1002/cti2.1259] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/22/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE SARS-CoV-2 has caused a worldwide pandemic of COVID-19. The existence of prolonged SARS-CoV-2 positivity (PP) has further increased the burden on the health system. Since T cells are vital for viral control, we aimed to evaluate the characteristics of T-cell responses associated with PP. METHODS We established a PP cohort and two age- and sex-matched control cohorts: a regular clinical recovery (CR) cohort and a healthy donor (HD) cohort. The mean time for RNA negativity conversion in the PP cohort was markedly longer than that in the CR cohort (66.2 vs 25.3 days), while the time from illness onset to sampling was not significantly different. T-cell responses in the PP cohort were assayed, analysed and compared with those in the CR and HD cohorts by flow cytometry and ELISpot analysis of peripheral blood mononuclear cells. RESULTS Compared with the CR cohort, the proliferation, activation and functional potential of CD8+ and CD4+ T cells in the PP cohort were not significantly different. However, the frequencies and counts of Teff and Tem in CD8+ but not in CD4+ T cells of the PP cohort were prominently lower. Moreover, a weaker SARS-CoV-2 N protein-specific IFN-γ+ T-cell response and a higher frequency of Tregs were detected in the PP cohort. CONCLUSION Suppressed CD8+ T-cell differentiation is associated with PP and may be an indicator for the prediction of prolonged SARS-CoV-2 positivity in COVID-19 patients. The association between suppressed CD8+ T-cell differentiation and elevated Tregs warrants studies in the future.
Collapse
|
1309
|
Joag V, Wijeyesinghe S, Stolley JM, Quarnstrom CF, Dileepan T, Soerens AG, Sangala JA, O'Flanagan SD, Gavil NV, Hong SW, Bhela S, Gangadhara S, Weyu E, Matchett WE, Thiede J, Krishna V, Cheeran MCJ, Bold TD, Amara R, Southern P, Hart GT, Schifanella L, Vezys V, Jenkins MK, Langlois RA, Masopust D. Cutting Edge: Mouse SARS-CoV-2 Epitope Reveals Infection and Vaccine-Elicited CD8 T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:931-935. [PMID: 33441437 PMCID: PMC8136468 DOI: 10.4049/jimmunol.2001400] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/23/2020] [Indexed: 01/27/2023]
Abstract
The magnitude of SARS-CoV-2-specific T cell responses correlates inversely with human disease severity, suggesting T cell involvement in primary control. Whereas many COVID-19 vaccines focus on establishing humoral immunity to viral spike protein, vaccine-elicited T cell immunity may bolster durable protection or cross-reactivity with viral variants. To better enable mechanistic and vaccination studies in mice, we identified a dominant CD8 T cell SARS-CoV-2 nucleoprotein epitope. Infection of human ACE2 transgenic mice with SARS-CoV-2 elicited robust responses to H2-Db/N219-227, and 40% of HLA-A*02+ COVID-19 PBMC samples isolated from hospitalized patients responded to this peptide in culture. In mice, i.m. prime-boost nucleoprotein vaccination with heterologous vectors favored systemic CD8 T cell responses, whereas intranasal boosting favored respiratory immunity. In contrast, a single i.v. immunization with recombinant adenovirus established robust CD8 T cell memory both systemically and in the respiratory mucosa.
Collapse
Affiliation(s)
- Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Clare F Quarnstrom
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Thamotharampillai Dileepan
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Andrew G Soerens
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Jules A Sangala
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Division of Infectious Disease and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| | - Stephen D O'Flanagan
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Noah V Gavil
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Sung-Wook Hong
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Siddheshvar Bhela
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Sailaja Gangadhara
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Eyob Weyu
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - William E Matchett
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Joshua Thiede
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Division of Infectious Disease and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| | - Venkatramana Krishna
- Veterinary Population Medicine Department, University of Minnesota, St. Paul, MN 55108; and
| | - Maxim C-J Cheeran
- Veterinary Population Medicine Department, University of Minnesota, St. Paul, MN 55108; and
| | - Tyler D Bold
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Division of Infectious Disease and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| | - Rama Amara
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Peter Southern
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Geoffrey T Hart
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Division of Infectious Disease and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| | - Luca Schifanella
- Surgical Outcomes and Precision Medicine Research Division, Department of Surgery, University of Minnesota, Minneapolis, MN 55455
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Marc K Jenkins
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455;
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455;
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455;
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
1310
|
Peluso MJ, Deitchman AN, Torres L, Iyer NS, Nixon CC, Munter SE, Donatelli J, Thanh C, Takahashi S, Hakim J, Turcios K, Janson O, Hoh R, Tai V, Hernandez Y, Fehrman E, Spinelli MA, Gandhi M, Trinh L, Wrin T, Petropoulos CJ, Aweeka FT, Rodriguez-Barraquer I, Kelly JD, Martin JN, Deeks SG, Greenhouse B, Rutishauser RL, Henrich TJ. Long-Term SARS-CoV-2-Specific Immune and Inflammatory Responses Across a Clinically Diverse Cohort of Individuals Recovering from COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 33688685 PMCID: PMC7941662 DOI: 10.1101/2021.02.26.21252308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A detailed understanding of long-term SARS-CoV-2-specific T cell responses and their relationship to humoral immunity and markers of inflammation in diverse groups of individuals representing the spectrum of COVID-19 illness and recovery is urgently needed. Data are also lacking as to whether and how adaptive immune and inflammatory responses differ in individuals that experience persistent symptomatic sequelae months following acute infection compared to those with complete, rapid recovery. We measured SARS-CoV-2-specific T cell responses, soluble markers of inflammation, and antibody levels and neutralization capacity longitudinally up to 9 months following infection in a diverse group of 70 individuals with PCR-confirmed SARS-CoV-2 infection. The participants had varying degrees of initial disease severity and were enrolled in the northern California Long-term Impact of Infection with Novel Coronavirus (LIINC) cohort. Adaptive T cell responses remained remarkably stable in all participants across disease severity during the entire study interval. Whereas the magnitude of the early CD4+ T cell immune response is determined by the severity of initial infection (participants requiring hospitalization or intensive care), pre-existing lung disease was significantly associated with higher long-term SARS-CoV2-specific CD8+ T cell responses, independent of initial disease severity or age. Neutralizing antibody levels were strongly correlated with SARS-CoV-2-specific CD4+ T but not CD8+ T cell responses. Importantly, we did not identify substantial differences in long-term virus-specific T cell or antibody responses between participants with and without COVID-19-related symptoms that persist months after initial infection.
Collapse
Affiliation(s)
- Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Amelia N Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, USA
| | - Leonel Torres
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA.,Division of Experimental Medicine, University of California, San Francisco, USA
| | - Nikita S Iyer
- Division of Experimental Medicine, University of California, San Francisco, USA
| | - Christopher C Nixon
- Division of Experimental Medicine, University of California, San Francisco, USA
| | - Sadie E Munter
- Division of Experimental Medicine, University of California, San Francisco, USA
| | - Joanna Donatelli
- Division of Experimental Medicine, University of California, San Francisco, USA
| | - Cassandra Thanh
- Division of Experimental Medicine, University of California, San Francisco, USA
| | - Saki Takahashi
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Jill Hakim
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Keirstinne Turcios
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Owen Janson
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Viva Tai
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Yanel Hernandez
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Emily Fehrman
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Matthew A Spinelli
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Monica Gandhi
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Lan Trinh
- Monogram Biosciences, Inc., South San Francisco, USA
| | - Terri Wrin
- Monogram Biosciences, Inc., South San Francisco, USA
| | | | - Francesca T Aweeka
- Department of Clinical Pharmacy, University of California, San Francisco, USA
| | - Isabel Rodriguez-Barraquer
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA.,Division of Experimental Medicine, University of California, San Francisco, USA
| | - J Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - Jeffrey N Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA
| | - Bryan Greenhouse
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, USA.,Division of Experimental Medicine, University of California, San Francisco, USA
| | | | | |
Collapse
|
1311
|
Cantenys‐Molina S, Fernández‐Cruz E, Francos P, Lopez Bernaldo de Quirós JC, Muñoz P, Gil‐Herrera J. Lymphocyte subsets early predict mortality in a large series of hospitalized COVID-19 patients in Spain. Clin Exp Immunol 2021; 203:424-432. [PMID: 33187018 PMCID: PMC7753314 DOI: 10.1111/cei.13547] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/27/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
The role of lymphocytes and their main subsets as prognostic factors of death in SARS-CoV-2-infected patients remains unclear, with no information obtained from patients outside China. We aimed to assess whether measuring lymphocyte subpopulations added clinical value to the total lymphocyte counting regarding mortality when they were simultaneously tested at hospital admission. Peripheral blood was analysed in 701 polymerase chain reaction (PCR)-confirmed consecutive patients by lysed-no washed flow cytometry. Demographic and clinical features were registered in electronic medical records. Statistical analysis was performed after a 3-month follow-up. The 112 patients who died were older and had significantly higher frequencies of known co-morbidities than survivor COVID-19 patients. A significant reduction in total lymphocytes, CD3+ , CD4+ , CD8+ and CD19+ counts and CD3+ percentage was found in the group of deceased patients (P < 0·001), while the percentage of CD56+ /CD16+ natural killer (NK) cells was significantly higher (P < 0·001). Multivariate logistic regression analysis showed a significantly increased risk of in-hospital death associated to age [odds ratio (OR) = 2·36, 95% confidence interval (CI) = 1·9-3·0 P < 0·001]; CD4+ T counts ≤ 500 cells/μl, (OR = 2·79, 95% CI = 1·1-6·7, P = 0·021); CD8+ T counts ≤ 100 cells/μl, (OR = 1·98, 95% CI = 1·2-3·3) P = 0·009) and CD56+ /CD16+ NK ≥ 30%, (OR = 1·97, 95% CI = 1·1-3·1, P = 0·002) at admission, independent of total lymphocyte numbers and co-morbidities, with area under the curve 0·85 (95% CI = 0·81-0·88). Reduced counts of CD4+ and CD8+ T cells with proportional expansion of NK lymphocytes at admission were prognostic factors of death in this Spanish series. In COVID-19 patients with normal levels of lymphocytes or mild lymphopenia, imbalanced lymphocyte subpopulations were early markers of in-hospital mortality.
Collapse
Affiliation(s)
- S. Cantenys‐Molina
- Division of ImmunologyHospital General Universitario ‘Gregorio Marañón’MadridSpain
| | - E. Fernández‐Cruz
- Division of ImmunologyHospital General Universitario ‘Gregorio Marañón’MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)MadridSpain
| | - P. Francos
- Division of ImmunologyHospital General Universitario ‘Gregorio Marañón’MadridSpain
| | - J. C. Lopez Bernaldo de Quirós
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)MadridSpain
- Department of Clinical Microbiology and Infectious DiseasesHospital General Universitario ‘Gregorio Marañón’MadridSpain
| | - P. Muñoz
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)MadridSpain
- Department of Clinical Microbiology and Infectious DiseasesHospital General Universitario ‘Gregorio Marañón’MadridSpain
- Medicine DepartmentSchool of MedicineUniversidad Complutense de Madrid (UCM)MadridSpain
- CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058)MadridSpain
| | - J. Gil‐Herrera
- Division of ImmunologyHospital General Universitario ‘Gregorio Marañón’MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)MadridSpain
| |
Collapse
|
1312
|
Tarke A, Sidney J, Methot N, Zhang Y, Dan JM, Goodwin B, Rubiro P, Sutherland A, da Silva Antunes R, Frazier A, Rawlings SA, Smith DM, Peters B, Scheuermann RH, Weiskopf D, Crotty S, Grifoni A, Sette A. Negligible impact of SARS-CoV-2 variants on CD4 + and CD8 + T cell reactivity in COVID-19 exposed donors and vaccinees. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.27.433180. [PMID: 33688655 PMCID: PMC7941626 DOI: 10.1101/2021.02.27.433180] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The emergence of SARS-CoV-2 variants highlighted the need to better understand adaptive immune responses to this virus. It is important to address whether also CD4+ and CD8+ T cell responses are affected, because of the role they play in disease resolution and modulation of COVID-19 disease severity. Here we performed a comprehensive analysis of SARS-CoV-2-specific CD4+ and CD8+ T cell responses from COVID-19 convalescent subjects recognizing the ancestral strain, compared to variant lineages B.1.1.7, B.1.351, P.1, and CAL.20C as well as recipients of the Moderna (mRNA-1273) or Pfizer/BioNTech (BNT162b2) COVID-19 vaccines. Similarly, we demonstrate that the sequences of the vast majority of SARS-CoV-2 T cell epitopes are not affected by the mutations found in the variants analyzed. Overall, the results demonstrate that CD4+ and CD8+ T cell responses in convalescent COVID-19 subjects or COVID-19 mRNA vaccinees are not substantially affected by mutations found in the SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Internal Medicine and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, 16132, Italy
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Nils Methot
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA 92037, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Benjamin Goodwin
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Paul Rubiro
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Aaron Sutherland
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Stephen A Rawlings
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Davey M Smith
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Richard H Scheuermann
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- J. Craig Venter Institute, La Jolla, CA 92037, USA
- Department of Pathology, University of California, San Diego, CA 92093
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
1313
|
Gallo O, Locatello LG, Mazzoni A, Novelli L, Annunziato F. The central role of the nasal microenvironment in the transmission, modulation, and clinical progression of SARS-CoV-2 infection. Mucosal Immunol 2021; 14:305-316. [PMID: 33244161 PMCID: PMC7690066 DOI: 10.1038/s41385-020-00359-2] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023]
Abstract
The novel coronavirus SARS-CoV-2 enters into the human body mainly through the ACE2 + TMPRSS2+ nasal epithelial cells. The initial host response to this pathogen occurs in a peculiar immune microenvironment that, starting from the Nasopharynx-Associated Lymphoid Tissue (NALT) system, is the product of a long evolutionary process that is aimed to first recognize exogenous airborne agents. In the present work, we want to critically review the latest molecular and cellular findings on the mucosal response to SARS-CoV-2 in the nasal cavity and in NALT, and to analyze its impact in the subsequent course of COVID-19. Finally, we want to explore the possibility that the regulation of the systemic inflammatory network against the virus can be modulated starting from the initial phases of the nasal and nasopharyngeal response and this may have several clinical and epidemiological implications starting from a mucosal vaccine development.
Collapse
Affiliation(s)
- Oreste Gallo
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| | - Luca Giovanni Locatello
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luca Novelli
- Department of Pathology, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy,Flow Cytometry and Immunotherapy Diagnostic Center, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| |
Collapse
|
1314
|
Abstract
PURPOSE OF REVIEW The aim of this study was to evaluate the relationship between infection with SARS-CoV-2 and autoimmunity. RECENT FINDINGS Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome (SARS) associated coronavirus 2 (SARS-CoV-2). Although most of the infected individuals are asymptomatic, a proportion of patients with COVID-19 develop severe disease with multiple organ injuries. Evidence suggests that some medications used to treat autoimmune rheumatologic diseases might have therapeutic effect in patients with severe COVID-19 infections, drawing attention to the relationship between COVID-19 and autoimmune diseases. COVID-19 shares similarities with autoimmune diseases in clinical manifestations, immune responses and pathogenic mechanisms. Robust immune reactions participate in the pathogenesis of both disease conditions. Autoantibodies as a hallmark of autoimmune diseases can also be detected in COVID-19 patients. Moreover, some patients have been reported to develop autoimmune diseases, such as Guillain--Barré syndrome or systemic lupus erythematosus, after COVID-19 infection. It is speculated that SARS-CoV-2 can disturb self-tolerance and trigger autoimmune responses through cross-reactivity with host cells. The infection risk and prognosis of COVID-19 in patients with autoimmune diseases remains controversial, but patient adherence to medication regimens to prevent autoimmune disease flares is strongly recommended. SUMMARY We present a review of the association between COVID-19 and autoimmune diseases, focusing on similarities in immune responses, cross-reactivity of SARS-CoV-2, the development of autoimmune diseases in COVID-19 patients and the risk of COVID-19 infection in patients with preexisting autoimmune conditions.
Collapse
Affiliation(s)
- Yu Liu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenetics, Changsha, Hunan, PR China
| | - Amr H. Sawalha
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenetics, Changsha, Hunan, PR China
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
1315
|
Affiliation(s)
- Jennifer L Hope
- Tumor Microenvironment and Cancer Immunology Program, NCI-Designated Cancer Center, and Immunity and Pathogenesis Program, Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Linda M Bradley
- Tumor Microenvironment and Cancer Immunology Program, NCI-Designated Cancer Center, and Immunity and Pathogenesis Program, Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
1316
|
Triggle CR, Bansal D, Ding H, Islam MM, Farag EABA, Hadi HA, Sultan AA. A Comprehensive Review of Viral Characteristics, Transmission, Pathophysiology, Immune Response, and Management of SARS-CoV-2 and COVID-19 as a Basis for Controlling the Pandemic. Front Immunol 2021; 12:631139. [PMID: 33717166 PMCID: PMC7952616 DOI: 10.3389/fimmu.2021.631139] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 emerged from China in December 2019 and during 2020 spread to every continent including Antarctica. The coronavirus, SARS-CoV-2, has been identified as the causative pathogen, and its spread has stretched the capacities of healthcare systems and negatively affected the global economy. This review provides an update on the virus, including the genome, the risks associated with the emergence of variants, mode of transmission, immune response, COVID-19 in children and the elderly, and advances made to contain, prevent and manage the disease. Although our knowledge of the mechanics of virus transmission and the immune response has been substantially demystified, concerns over reinfection, susceptibility of the elderly and whether asymptomatic children promote transmission remain unanswered. There are also uncertainties about the pathophysiology of COVID-19 and why there are variations in clinical presentations and why some patients suffer from long lasting symptoms-"the long haulers." To date, there are no significantly effective curative drugs for COVID-19, especially after failure of hydroxychloroquine trials to produce positive results. The RNA polymerase inhibitor, remdesivir, facilitates recovery of severely infected cases but, unlike the anti-inflammatory drug, dexamethasone, does not reduce mortality. However, vaccine development witnessed substantial progress with several being approved in countries around the globe.
Collapse
Affiliation(s)
- Chris R. Triggle
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Devendra Bansal
- Department of Health Protection & Communicable Diseases Control, Ministry of Public Health, Doha, Qatar
| | - Hong Ding
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Md Mazharul Islam
- Department of Animal Resources, Ministry of Municipality and Environment, Doha, Qatar
| | | | - Hamad Abdel Hadi
- Communicable Diseases Centre, Hamad Medical Corporations, Doha, Qatar
| | - Ali A. Sultan
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Doha, Qatar
| |
Collapse
|
1317
|
Lee E, Sandgren K, Duette G, Stylianou VV, Khanna R, Eden JS, Blyth E, Gottlieb D, Cunningham AL, Palmer S. Identification of SARS-CoV-2 Nucleocapsid and Spike T-Cell Epitopes for Assessing T-Cell Immunity. J Virol 2021; 95:e02002-20. [PMID: 33443088 PMCID: PMC8579755 DOI: 10.1128/jvi.02002-20] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
Developing optimal T-cell response assays to severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is critical for measuring the duration of immunity to this disease and assessing the efficacy of vaccine candidates. These assays need to target conserved regions of SARS-CoV-2 global variants and avoid cross-reactivity to seasonal human coronaviruses. To contribute to this effort, we employed an in silico immunoinformatics analysis pipeline to identify immunogenic peptides resulting from conserved and highly networked regions with topological importance from the SARS-CoV-2 nucleocapsid and spike proteins. A total of 57 highly networked T-cell epitopes that are conserved across geographic viral variants were identified from these viral proteins, with a binding potential to diverse HLA alleles and 80 to 100% global population coverage. Importantly, 18 of these T-cell epitope derived peptides had limited homology to seasonal human coronaviruses making them promising candidates for SARS-CoV-2-specific T-cell immunity assays. Moreover, two of the NC-derived peptides elicited effector/polyfunctional responses of CD8+ T cells derived from SARS-CoV-2 convalescent patients.IMPORTANCE The development of specific and validated immunologic tools is critical for understanding the level and duration of the cellular response induced by SARS-CoV-2 infection and/or vaccines against this novel coronavirus disease. To contribute to this effort, we employed an immunoinformatics analysis pipeline to define 57 SARS-CoV-2 immunogenic peptides within topologically important regions of the nucleocapsid (NC) and spike (S) proteins that will be effective for detecting cellular immune responses in 80 to 100% of the global population. Our immunoinformatics analysis revealed that 18 of these peptides had limited homology to circulating seasonal human coronaviruses and therefore are promising candidates for distinguishing SARS-CoV-2-specific immune responses from pre-existing coronavirus immunity. Importantly, CD8+ T cells derived from SARS-CoV-2 survivors exhibited polyfunctional effector responses to two novel NC-derived peptides identified as HLA-binders. These studies provide a proof of concept that our immunoinformatics analysis pipeline identifies novel immunogens which can elicit polyfunctional SARS-CoV-2-specific T-cell responses.
Collapse
Affiliation(s)
- Eunok Lee
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Kerrie Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Gabriel Duette
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Vicki V Stylianou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Rajiv Khanna
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John-Sebastian Eden
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Emily Blyth
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- BMT and Cell Therapies Program, Westmead Hospital, Westmead, New South Wales, Australia
| | - David Gottlieb
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- BMT and Cell Therapies Program, Westmead Hospital, Westmead, New South Wales, Australia
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Sarah Palmer
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
1318
|
Sarmiento-Monroy JC, Parra-Medina R, Garavito E, Rojas-Villarraga A. T Helper 17 Response to Severe Acute Respiratory Syndrome Coronavirus 2: A Type of Immune Response with Possible Therapeutic Implications. Viral Immunol 2021; 34:190-200. [PMID: 33625297 DOI: 10.1089/vim.2020.0177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The initial immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) includes an interferon-dependent antiviral response. A late and uncontrolled inflammatory response characterized by high activity of proinflammatory cytokines and the recruitment of neutrophils and macrophages develops in predisposed individuals and is potentially harmful in some cases. Interleukin (IL)-17 is one of the many cytokines released during coronavirus disease 2019 (COVID-19). IL-17 is crucial in recruiting and activating neutrophils, cells that can migrate to the lung, and are heavily involved in the pathogenesis of COVID-19. During the infection T helper 17 (Th17) cells and IL-17-related pathways are associated with a worse outcome of the disease. All these have practical consequences considering that some drugs with therapeutic targets related to the Th17 response may have a beneficial effect on patients with SARS-CoV-2 infection. Herein, we present the arguments underlying our assumption that blocking the IL-23/IL-17 axis using targeted biological therapies as well as drugs that act indirectly on this pathway such as convalescent plasma therapy and colchicine may be good therapeutic options.
Collapse
Affiliation(s)
| | - Rafael Parra-Medina
- Research Department, Fundación Universitaria de Ciencias de la Salud-FUCS, Bogotá, Colombia.,Pathology Department, Fundación Universitaria de Ciencias de la Salud-FUCS, Bogotá, Colombia
| | - Edgar Garavito
- Pathology Department, Fundación Universitaria de Ciencias de la Salud-FUCS, Bogotá, Colombia.,Basic Sciences Department, Fundación Universitaria Sanitas, Bogotá, Colombia.,Department of Morphology, Universidad Nacional de Colombia, Bogotá, Colombia
| | | |
Collapse
|
1319
|
Bertoletti A, Tan AT, Le Bert N. The T-cell response to SARS-CoV-2: kinetic and quantitative aspects and the case for their protective role. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab006. [PMID: 38626271 PMCID: PMC7928654 DOI: 10.1093/oxfimm/iqab006] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/23/2022] Open
Abstract
Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2), the etiological agent of Coronavirus Diseases 2019 (COVID-19), triggers an adaptive immunity in the infected host that results in the production of virus-specific antibodies and T cells. Although kinetic and quantitative aspects of antibodies have been analyzed in large patient cohorts, similar information about SARS-CoV-2-specific T cells are scarce. We summarize the available knowledge of quantitative and temporal features of the SARS-CoV-2 T-cell response in this review. Currently, most of the data are derived only from the analysis of the circulatory compartment. Despite this limitation, early appearance, multi-specificity and functionality of SARS-CoV-2-specific T cells are associated with accelerated viral clearance and with protection from severe COVID-19.
Collapse
Affiliation(s)
- Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Singapore Immunology Network, A*STAR, Singapore
| | - Anthony T Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| |
Collapse
|
1320
|
Richmond P, Hatchuel L, Dong M, Ma B, Hu B, Smolenov I, Li P, Liang P, Han HH, Liang J, Clemens R. Safety and immunogenicity of S-Trimer (SCB-2019), a protein subunit vaccine candidate for COVID-19 in healthy adults: a phase 1, randomised, double-blind, placebo-controlled trial. Lancet 2021; 397:682-694. [PMID: 33524311 PMCID: PMC7906655 DOI: 10.1016/s0140-6736(21)00241-5] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND As part of the accelerated development of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we report a dose-finding and adjuvant justification study of SCB-2019, a protein subunit vaccine candidate containing a stabilised trimeric form of the spike (S)-protein (S-Trimer) combined with two different adjuvants. METHODS Our study is a phase 1, randomised, double-blind placebo-controlled trial at a specialised clinical trials centre in Australia. We enrolled healthy adult volunteers in two age groups: younger adults (aged 18-54 years) and older adults (aged 55-75 years). Participants were randomly allocated either vaccine or placebo using a list prepared by the study funder. Participants were to receive two doses of SCB-2019 (either 3 μg, 9 μg, or 30 μg) or a placebo (0·9% NaCl) 21 days apart. SCB-2019 either had no adjuvant (S-Trimer protein alone) or was adjuvanted with AS03 or CpG/Alum. The assigned treatment was administered in opaque syringes to maintain masking of assignments. Reactogenicity was assessed for 7 days after each vaccination. Humoral responses were measured as SCB-2019 binding IgG antibodies and ACE2-competitive blocking IgG antibodies by ELISA and as neutralising antibodies by wild-type SARS-CoV-2 microneutralisation assay. Cellular responses to pooled S-protein peptides were measured by flow-cytometric intracellular cytokine staining. This trial is registered with ClinicalTrials.gov, NCT04405908; this is an interim analysis and the study is continuing. FINDINGS Between June 19 and Sept 23, 2020, 151 volunteers were enrolled; three people withdrew, two for personal reasons and one with an unrelated serious adverse event (pituitary adenoma). 148 participants had at least 4 weeks of follow-up after dose two and were included in this analysis (database lock, Oct 23, 2020). Vaccination was well tolerated, with two grade 3 solicited adverse events (pain in 9 μg AS03-adjuvanted and 9 μg CpG/Alum-adjuvanted groups). Most local adverse events were mild injection-site pain, and local events were more frequent with SCB-2019 formulations containing AS03 adjuvant (44-69%) than with those containing CpG/Alum adjuvant (6-44%) or no adjuvant (3-13%). Systemic adverse events were more frequent in younger adults (38%) than in older adults (17%) after the first dose but increased to similar levels in both age groups after the second dose (30% in older and 34% in younger adults). SCB-2019 with no adjuvant elicited minimal immune responses (three seroconversions by day 50), but SCB-2019 with fixed doses of either AS03 or CpG/Alum adjuvants induced high titres and seroconversion rates of binding and neutralising antibodies in both younger and older adults (anti-SCB-2019 IgG antibody geometric mean titres at day 36 were 1567-4452 with AS03 and 174-2440 with CpG/Alum). Titres in all AS03 dose groups and the CpG/Alum 30 μg group were higher than were those recorded in a panel of convalescent serum samples from patients with COVID-19. Both adjuvanted SCB-2019 formulations elicited T-helper-1-biased CD4+ T-cell responses. INTERPRETATION The SCB-2019 vaccine, comprising S-Trimer protein formulated with either AS03 or CpG/Alum adjuvants, elicited robust humoral and cellular immune responses against SARS-CoV-2, with high viral neutralising activity. Both adjuvanted vaccine formulations were well tolerated and are suitable for further clinical development. FUNDING Clover Biopharmaceuticals and the Coalition for Epidemic Preparedness Innovations.
Collapse
Affiliation(s)
- Peter Richmond
- Division of Paediatrics, University of Western Australia, Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute and Perth Children's Hospital, Perth, WA, Australia
| | | | - Min Dong
- Clover Biopharmaceuticals, Chengdu, China
| | - Brenda Ma
- Clover Biopharmaceuticals, Chengdu, China
| | - Branda Hu
- Clover Biopharmaceuticals, Chengdu, China
| | | | - Ping Li
- Clover Biopharmaceuticals, Chengdu, China
| | - Peng Liang
- Clover Biopharmaceuticals, Chengdu, China
| | | | | | - Ralf Clemens
- Global Research in Infectious Diseases, Rio de Janeiro, Brazil.
| |
Collapse
|
1321
|
Sette A, Crotty S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021; 184:861-880. [PMID: 33497610 PMCID: PMC7803150 DOI: 10.1016/j.cell.2021.01.007] [Citation(s) in RCA: 1279] [Impact Index Per Article: 319.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022]
Abstract
The adaptive immune system is important for control of most viral infections. The three fundamental components of the adaptive immune system are B cells (the source of antibodies), CD4+ T cells, and CD8+ T cells. The armamentarium of B cells, CD4+ T cells, and CD8+ T cells has differing roles in different viral infections and in vaccines, and thus it is critical to directly study adaptive immunity to SARS-CoV-2 to understand COVID-19. Knowledge is now available on relationships between antigen-specific immune responses and SARS-CoV-2 infection. Although more studies are needed, a picture has begun to emerge that reveals that CD4+ T cells, CD8+ T cells, and neutralizing antibodies all contribute to control of SARS-CoV-2 in both non-hospitalized and hospitalized cases of COVID-19. The specific functions and kinetics of these adaptive immune responses are discussed, as well as their interplay with innate immunity and implications for COVID-19 vaccines and immune memory against re-infection.
Collapse
Affiliation(s)
- Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
1322
|
Rha MS, Kim AR, Shin EC. SARS-CoV-2-Specific T Cell Responses in Patients with COVID-19 and Unexposed Individuals. Immune Netw 2021; 21:e2. [PMID: 33728095 PMCID: PMC7937509 DOI: 10.4110/in.2021.21.e2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes coronavirus disease 2019 (COVID-19), an ongoing pandemic disease. In the current review, we describe SARS-CoV-2-specific CD4+ and CD8+ T-cell responses in acute and convalescent COVID-19 patients. We also discuss the relationships between COVID-19 severity and SARS-CoV-2-specific T-cell responses and summarize recent reports regarding SARS-CoV-2-reactive T cells in SARS-CoV-2-unexposed individuals. These T cells may be cross-reactive cells primed by previous infection with human common-cold coronaviruses. Finally, we outline SARS-CoV-2-specific T-cell responses in the context of vaccination. A better understanding of SARS-CoV-2-specific T-cell responses is needed to develop effective vaccines and therapeutics.
Collapse
Affiliation(s)
- Min-Seok Rha
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - A Reum Kim
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.,The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
1323
|
Alrubayyi A, Gea-Mallorquí E, Touizer E, Hameiri-Bowen D, Kopycinski J, Charlton B, Fisher-Pearson N, Muir L, Rosa A, Roustan C, Earl C, Cherepanov P, Pellegrino P, Waters L, Burns F, Kinloch S, Dong T, Dorrell L, Rowland-Jones S, McCoy LE, Peppa D. Characterization of humoral and SARS-CoV-2 specific T cell responses in people living with HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.15.431215. [PMID: 33619489 PMCID: PMC7899453 DOI: 10.1101/2021.02.15.431215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
There is an urgent need to understand the nature of immune responses generated against SARS-CoV-2, to better inform risk-mitigation strategies for people living with HIV (PLWH). Although not all PLWH are considered immunosuppressed, residual cellular immune deficiency and ongoing inflammation could influence COVID-19 disease severity, the evolution and durability of protective memory responses. Here, we performed an integrated analysis, characterizing the nature, breadth and magnitude of SARS-CoV-2-specific immune responses in PLWH, controlled on ART, and HIV negative subjects. Both groups were in the convalescent phase of predominately mild COVID-19 disease. The majority of PLWH mounted SARS-CoV-2 Spike- and Nucleoprotein-specific antibodies with neutralizing activity and SARS-CoV-2-specific T cell responses, as measured by ELISpot, at levels comparable to HIV negative subjects. T cell responses against Spike, Membrane and Nucleocapsid were the most prominent, with SARS-CoV-2-specific CD4 T cells outnumbering CD8 T cells. Notably, the overall magnitude of SARS-CoV-2-specific T cell responses related to the size of the naive CD4 T cell pool and the CD4:CD8 ratio in PLWH, in whom disparate antibody and T cell responses were observed. Both humoral and cellular responses to SARS-CoV-2 were detected at 5-7 months post-infection, providing evidence of medium-term durability of responses irrespective of HIV serostatus. Incomplete immune reconstitution on ART and a low CD4:CD8 ratio could, however, hamper the development of immunity to SARS-CoV-2 and serve as a useful tool for risk stratification of PLWH. These findings have implications for the individual management and potential effectiveness of vaccination against SARS-CoV-2 in PLWH.
Collapse
Affiliation(s)
| | | | - Emma Touizer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dan Hameiri-Bowen
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | - Jakub Kopycinski
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | - Bethany Charlton
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | | | - Luke Muir
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Annachiara Rosa
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Chloe Roustan
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Christopher Earl
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Pierre Pellegrino
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, United Kingdom
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, United Kingdom
| | - Fiona Burns
- Institute for Global Health UCL, London, United Kingdom
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sabine Kinloch
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Tao Dong
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | - Lucy Dorrell
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
| | | | - Laura E. McCoy
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dimitra Peppa
- Nuffield Dept of Clinical Medicine, University of Oxford, United Kingdom
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, United Kingdom
| |
Collapse
|
1324
|
Liu H, Wei P, Zhang Q, Chen Z, Aviszus K, Downing W, Peterson S, Reynoso L, Downey GP, Frankel SK, Kappler J, Marrack P, Zhang G. 501Y.V2 and 501Y.V3 variants of SARS-CoV-2 lose binding to Bamlanivimab in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33619479 DOI: 10.1101/2021.02.16.431305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We generated several versions of the receptor binding domain (RBD) of the Spike protein with mutations existing within newly emerging variants from South Africa and Brazil. We found that the mutant RBD with K417N, E484K, and N501Y exchanges has higher binding affinity to the human receptor compared to the wildtype RBD. This mutated version of RBD also completely abolishes the binding to a therapeutic antibody, Bamlanivimab, in vitro .
Collapse
|
1325
|
Laing ED, Sterling SL, Richard SA, Epsi NJ, Coggins S, Samuels EC, Phogat S, Yan L, Moreno N, Coles CL, Drew M, Mehalko J, English CE, Merritt S, Mende K, Munster VJ, de Wit E, Chung KK, Millar EV, Tribble DR, Simons MP, Pollett SD, Agan BK, Esposito D, Lanteri C, Clifton GT, Mitre E, Burgess TH, Broder CC. Antigen-based multiplex strategies to discriminate SARS-CoV-2 natural and vaccine induced immunity from seasonal human coronavirus humoral responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.10.21251518. [PMID: 33594376 PMCID: PMC7885935 DOI: 10.1101/2021.02.10.21251518] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sensitive and specific SARS-CoV-2 antibody assays remain critical for community and hospital-based SARS-CoV-2 sero-surveillance. With the rollout of SARS-CoV-2 vaccines, such assays must be able to distinguish vaccine from natural immunity to SARS-CoV-2 and related human coronaviruses. Here, we developed and implemented multiplex microsphere-based immunoassay strategies for COVD-19 antibody studies that incorporates spike protein trimers of SARS-CoV-2 and the endemic seasonal human coronaviruses (HCoV), enabling high throughout measurement of pre-existing cross-reactive antibodies. We varied SARS-CoV-2 antigen compositions within the multiplex assay, allowing direct comparisons of the effects of spike protein, receptor-binding domain protein (RBD) and nucleocapsid protein (NP) based SARS-CoV-2 antibody detection. Multiplex immunoassay performance characteristics are antigen-dependent, and sensitivities and specificities range 92-99% and 94-100%, respectively, for human subject samples collected as early as 7-10 days from symptom onset. SARS-CoV-2 spike and RBD had a strong correlative relationship for the detection of IgG. Correlation between detectable IgG reactive with spike and NP also had strong relationship, however, several PCR-positive and spike IgG-positive serum samples were NP IgG-negative. This spike and NP multiplex immunoassay has the potential to be useful for differentiation between vaccination and natural infection induced antibody responses. We also assessed the induction of de novo SARS-CoV-2 IgG cross reactions with SARS-CoV and MERS-CoV spike proteins. Furthermore, multiplex immunoassays that incorporate spike proteins of SARS-CoV-2 and HCoVs will permit investigations into the influence of HCoV antibodies on COVID-19 clinical outcomes and SARS-CoV-2 antibody durability.
Collapse
Affiliation(s)
- Eric D. Laing
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Spencer L. Sterling
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Stephanie A. Richard
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nusrat J. Epsi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Si’Ana Coggins
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Emily C. Samuels
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Shreshta Phogat
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Nicole Moreno
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Christian L. Coles
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Matthew Drew
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jennifer Mehalko
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Caroline E. English
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Scott Merritt
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Brooke Army Medical Center, JBSA Fort Sam Houston, TX, USA
| | - Katrin Mende
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Brooke Army Medical Center, JBSA Fort Sam Houston, TX, USA
| | - Vincent J. Munster
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Emmie de Wit
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kevin K. Chung
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Eugene V. Millar
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - David R. Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Mark P. Simons
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Simon D. Pollett
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Brian K. Agan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Dominic Esposito
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Charlotte Lanteri
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | | | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Timothy H. Burgess
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
1326
|
The impact of immuno-aging on SARS-CoV-2 vaccine development. GeroScience 2021; 43:31-51. [PMID: 33569701 PMCID: PMC7875765 DOI: 10.1007/s11357-021-00323-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
The SARS-CoV-2 pandemic has almost 56 million confirmed cases resulting in over 1.3 million deaths as of November 2020. This infection has proved more deadly to older adults (those >65 years of age) and those with immunocompromising conditions. The worldwide population aged 65 years and older is increasing, and the total number of aged individuals will outnumber those younger than 65 years by the year 2050. Aging is associated with a decline in immune function and chronic activation of inflammation that contributes to enhanced viral susceptibility and reduced responses to vaccination. Here we briefly review the pathogenicity of the virus, epidemiology and clinical response, and the underlying mechanisms of human aging in improving vaccination. We review current methods to improve vaccination in the older adults using novel vaccine platforms and adjuvant systems. We conclude by summarizing the existing clinical trials for a SARS-CoV-2 vaccine and discussing how to address the unique challenges for vaccine development presented with an aging immune system.
Collapse
|
1327
|
Abstract
Understanding the precise nature and durability of protective immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is essential in order to gain insight into the pathophysiology of coronavirus disease 2019 (COVID-19) and to develop novel treatment strategies to this disease. Here I succinctly summarize what is currently known and unknown about the immune response during COVID-19 and discuss whether natural infections can lead to herd immunity.
Collapse
Affiliation(s)
- Masayuki Miyasaka
- Immunology Frontier Research Center (IFReC), Osaka University, Japan
| |
Collapse
|
1328
|
Demirdag YY, Gupta S. Update on Infections in Primary Antibody Deficiencies. Front Immunol 2021; 12:634181. [PMID: 33643318 PMCID: PMC7905085 DOI: 10.3389/fimmu.2021.634181] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/07/2021] [Indexed: 11/14/2022] Open
Abstract
Bacterial respiratory tract infections are the hallmark of primary antibody deficiencies (PADs). Because they are also among the most common infections in healthy individuals, PADs are usually overlooked in these patients. Careful evaluation of the history, including frequency, chronicity, and presence of other infections, would help suspect PADs. This review will focus on infections in relatively common PADs, discussing diagnostic challenges, and some management strategies to prevent infections.
Collapse
Affiliation(s)
- Yesim Yilmaz Demirdag
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | | |
Collapse
|
1329
|
Castro A, Ozturk K, Zanetti M, Carter H. In silico analysis suggests less effective MHC-II presentation of SARS-CoV-2 RBM peptides: Implication for neutralizing antibody responses. PLoS One 2021; 16:e0246731. [PMID: 33571241 PMCID: PMC7877779 DOI: 10.1371/journal.pone.0246731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/25/2021] [Indexed: 12/27/2022] Open
Abstract
SARS-CoV-2 antibodies develop within two weeks of infection, but wane relatively rapidly post-infection, raising concerns about whether antibody responses will provide protection upon re-exposure. Here we revisit T-B cooperation as a prerequisite for effective and durable neutralizing antibody responses centered on a mutationally constrained RBM B cell epitope. T-B cooperation requires co-processing of B and T cell epitopes by the same B cell and is subject to MHC-II restriction. We evaluated MHC-II constraints relevant to the neutralizing antibody response to a mutationally-constrained B cell epitope in the receptor binding motif (RBM) of the spike protein. Examining common MHC-II alleles, we found that peptides surrounding this key B cell epitope are predicted to bind poorly, suggesting a lack MHC-II support in T-B cooperation, impacting generation of high-potency neutralizing antibodies in the general population. Additionally, we found that multiple microbial peptides had potential for RBM cross-reactivity, supporting previous exposures as a possible source of T cell memory.
Collapse
Affiliation(s)
- Andrea Castro
- Biomedical Informatics Program, University of California San Diego, La Jolla, CA, United States of America
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Kivilcim Ozturk
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine, University of California San Diego, La Jolla, CA, United States of America
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States of America
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, United States of America
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States of America
| |
Collapse
|
1330
|
Breton G, Mendoza P, Hägglöf T, Oliveira TY, Schaefer-Babajew D, Gaebler C, Turroja M, Hurley A, Caskey M, Nussenzweig MC. Persistent cellular immunity to SARS-CoV-2 infection. J Exp Med 2021; 218:211727. [PMID: 33533915 PMCID: PMC7845919 DOI: 10.1084/jem.20202515] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 is responsible for an ongoing pandemic that has affected millions of individuals around the globe. To gain further understanding of the immune response in recovered individuals, we measured T cell responses in paired samples obtained an average of 1.3 and 6.1 mo after infection from 41 individuals. The data indicate that recovered individuals show persistent polyfunctional SARS-CoV-2 antigen–specific memory that could contribute to rapid recall responses. Recovered individuals also show enduring alterations in relative overall numbers of CD4+ and CD8+ memory T cells, including expression of activation/exhaustion markers, and cell division.
Collapse
Affiliation(s)
- Gaëlle Breton
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Pilar Mendoza
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Thomas Hägglöf
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | | | - Christian Gaebler
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Martina Turroja
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Arlene Hurley
- Hospital Program Direction, The Rockefeller University, New York, NY
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY.,Howard Hughes Medical Institute, Baltimore, MD
| |
Collapse
|
1331
|
Gevertz JL, Greene JM, Sanchez-Tapia CH, Sontag ED. A novel COVID-19 epidemiological model with explicit susceptible and asymptomatic isolation compartments reveals unexpected consequences of timing social distancing. J Theor Biol 2021; 510:110539. [PMID: 33242489 PMCID: PMC7840295 DOI: 10.1016/j.jtbi.2020.110539] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/07/2020] [Accepted: 11/06/2020] [Indexed: 02/08/2023]
Abstract
Motivated by the current COVID-19 epidemic, this work introduces an epidemiological model in which separate compartments are used for susceptible and asymptomatic "socially distant" populations. Distancing directives are represented by rates of flow into these compartments, as well as by a reduction in contacts that lessens disease transmission. The dynamical behavior of this system is analyzed, under various different rate control strategies, and the sensitivity of the basic reproduction number to various parameters is studied. One of the striking features of this model is the existence of a critical implementation delay (CID) in issuing distancing mandates: while a delay of about two weeks does not have an appreciable effect on the peak number of infections, issuing mandates even slightly after this critical time results in a far greater incidence of infection. Thus, there is a nontrivial but tight "window of opportunity" for commencing social distancing in order to meet the capacity of healthcare resources. However, if one wants to also delay the timing of peak infections - so as to take advantage of potential new therapies and vaccines - action must be taken much faster than the CID. Different relaxation strategies are also simulated, with surprising results. Periodic relaxation policies suggest a schedule which may significantly inhibit peak infective load, but that this schedule is very sensitive to parameter values and the schedule's frequency. Furthermore, we considered the impact of steadily reducing social distancing measures over time. We find that a too-sudden reopening of society may negate the progress achieved under initial distancing guidelines, but the negative effects can be mitigated if the relaxation strategy is carefully designed.
Collapse
Affiliation(s)
- Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, United States
| | - James M Greene
- Department of Mathematics, Clarkson University, Potsdam, NY, United States
| | - Cynthia H Sanchez-Tapia
- Department of Mathematics, College of Natural and Behavioral Sciences, California State University Dominguez Hills, Carson, CA, United States
| | - Eduardo D Sontag
- Department of Electrical and Computer Engineering and Department of Bioengineering, Northeastern University, Boston, MA, United States; Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
1332
|
Gevertz JL, Greene JM, Sanchez-Tapia CH, Sontag ED. A novel COVID-19 epidemiological model with explicit susceptible and asymptomatic isolation compartments reveals unexpected consequences of timing social distancing. J Theor Biol 2021. [PMID: 33242489 DOI: 10.1101/2020.05.11.20098335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Motivated by the current COVID-19 epidemic, this work introduces an epidemiological model in which separate compartments are used for susceptible and asymptomatic "socially distant" populations. Distancing directives are represented by rates of flow into these compartments, as well as by a reduction in contacts that lessens disease transmission. The dynamical behavior of this system is analyzed, under various different rate control strategies, and the sensitivity of the basic reproduction number to various parameters is studied. One of the striking features of this model is the existence of a critical implementation delay (CID) in issuing distancing mandates: while a delay of about two weeks does not have an appreciable effect on the peak number of infections, issuing mandates even slightly after this critical time results in a far greater incidence of infection. Thus, there is a nontrivial but tight "window of opportunity" for commencing social distancing in order to meet the capacity of healthcare resources. However, if one wants to also delay the timing of peak infections - so as to take advantage of potential new therapies and vaccines - action must be taken much faster than the CID. Different relaxation strategies are also simulated, with surprising results. Periodic relaxation policies suggest a schedule which may significantly inhibit peak infective load, but that this schedule is very sensitive to parameter values and the schedule's frequency. Furthermore, we considered the impact of steadily reducing social distancing measures over time. We find that a too-sudden reopening of society may negate the progress achieved under initial distancing guidelines, but the negative effects can be mitigated if the relaxation strategy is carefully designed.
Collapse
Affiliation(s)
- Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, United States
| | - James M Greene
- Department of Mathematics, Clarkson University, Potsdam, NY, United States
| | - Cynthia H Sanchez-Tapia
- Department of Mathematics, College of Natural and Behavioral Sciences, California State University Dominguez Hills, Carson, CA, United States
| | - Eduardo D Sontag
- Department of Electrical and Computer Engineering and Department of Bioengineering, Northeastern University, Boston, MA, United States; Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
1333
|
Dan JM, Mateus J, Kato Y, Hastie KM, Yu ED, Faliti CE, Grifoni A, Ramirez SI, Haupt S, Frazier A, Nakao C, Rayaprolu V, Rawlings SA, Peters B, Krammer F, Simon V, Saphire EO, Smith DM, Weiskopf D, Sette A, Crotty S. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 2021; 371:eabf4063. [PMID: 33408181 PMCID: PMC7919858 DOI: 10.1126/science.abf4063] [Citation(s) in RCA: 1915] [Impact Index Per Article: 478.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Understanding immune memory to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical for improving diagnostics and vaccines and for assessing the likely future course of the COVID-19 pandemic. We analyzed multiple compartments of circulating immune memory to SARS-CoV-2 in 254 samples from 188 COVID-19 cases, including 43 samples at ≥6 months after infection. Immunoglobulin G (IgG) to the spike protein was relatively stable over 6+ months. Spike-specific memory B cells were more abundant at 6 months than at 1 month after symptom onset. SARS-CoV-2-specific CD4+ T cells and CD8+ T cells declined with a half-life of 3 to 5 months. By studying antibody, memory B cell, CD4+ T cell, and CD8+ T cell memory to SARS-CoV-2 in an integrated manner, we observed that each component of SARS-CoV-2 immune memory exhibited distinct kinetics.
Collapse
Affiliation(s)
- Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Yu Kato
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Kathryn M Hastie
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Caterina E Faliti
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Sydney I Ramirez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Sonya Haupt
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Catherine Nakao
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Vamseedhar Rayaprolu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Stephen A Rawlings
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Davey M Smith
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
1334
|
Neidleman J, Luo X, George AF, McGregor M, Yang J, Yun C, Murray V, Gill G, Greene WC, Vasquez J, Lee S, Ghosn E, Lynch K, Roan NR. Distinctive features of SARS-CoV-2-specific T cells predict recovery from severe COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.22.21250054. [PMID: 33532792 PMCID: PMC7852243 DOI: 10.1101/2021.01.22.21250054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although T cells are likely players in SARS-CoV-2 immunity, little is known about the phenotypic features of SARS-CoV-2-specific T cells associated with recovery from severe COVID-19. We analyzed T cells from longitudinal specimens of 34 COVID-19 patients with severities ranging from mild (outpatient) to critical culminating in death. Relative to patients that succumbed, individuals that recovered from severe COVID-19 harbored elevated and increasing numbers of SARS-CoV-2-specific T cells capable of homeostatic proliferation. In contrast, fatal COVID-19 displayed elevated numbers of SARS-CoV-2-specific regulatory T cells and a time-dependent escalation in activated bystander CXCR4+ T cells. Together with the demonstration of increased proportions of inflammatory CXCR4+ T cells in the lungs of severe COVID-19 patients, these results support a model whereby lung-homing T cells activated through bystander effects contribute to immunopathology, while a robust, non-suppressive SARS-CoV-2-specific T cell response limits pathogenesis and promotes recovery from severe COVID-19.
Collapse
Affiliation(s)
- Jason Neidleman
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Xiaoyu Luo
- Gladstone Institutes, San Francisco, CA, USA
| | - Ashley F. George
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Matthew McGregor
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Junkai Yang
- Department of Medicine, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Cassandra Yun
- Department of Laboratory Medicine, University of California, San Francisco, CA USA
| | - Victoria Murray
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Gurjot Gill
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Warner C. Greene
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Joshua Vasquez
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Sulggi Lee
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Eliver Ghosn
- Department of Medicine, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Kara Lynch
- Department of Laboratory Medicine, University of California, San Francisco, CA USA
| | - Nadia R. Roan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| |
Collapse
|
1335
|
Cohen CA, Li APY, Hachim A, Hui DSC, Kwan MYW, Tsang OTY, Chiu SS, Chan WH, Yau YS, Kavian N, Ma FNL, Lau EHY, Cheng SMS, Poon LLM, Peiris JSM, Valkenburg SA. SARS-CoV-2 specific T cell responses are lower in children and increase with age and time after infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.02.21250988. [PMID: 33564773 PMCID: PMC7872365 DOI: 10.1101/2021.02.02.21250988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
SARS-CoV-2 infection of children leads to a mild illness and the immunological differences with adults remains unclear. We quantified the SARS-CoV-2 specific T cell responses in adults and children (<13 years of age) with RT-PCR confirmed asymptomatic and symptomatic infection for long-term memory, phenotype and polyfunctional cytokines. Acute and memory CD4+ T cell responses to structural SARS-CoV-2 proteins significantly increased with age, whilst CD8+ T cell responses increased with time post infection. Infected children had significantly lower CD4+ and CD8+ T cell responses to SARS-CoV-2 structural and ORF1ab proteins compared to infected adults. SARS-CoV-2-specific CD8+ T cell responses were comparable in magnitude to uninfected negative adult controls. In infected adults CD4+ T cell specificity was skewed towards structural peptides, whilst children had increased contribution of ORF1ab responses. This may reflect differing T cell compartmentalisation for antigen processing during antigen exposure or lower recruitment of memory populations. T cell polyfunctional cytokine production was comparable between children and adults, but children had a lower proportion of SARS-CoV-2 CD4+ T cell effector memory. Compared to adults, children had significantly lower levels of antibodies to β-coronaviruses, indicating differing baseline immunity. Total T follicular helper responses was increased in children during acute infection indicating rapid co-ordination of the T and B cell responses. However total monocyte responses were reduced in children which may be reflective of differing levels of inflammation between children and adults. Therefore, reduced prior β-coronavirus immunity and reduced activation and recruitment of de novo responses in children may drive milder COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Carolyn A Cohen
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Athena PY Li
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David SC Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mike YW Kwan
- Department of Paediatric and Adolescent Medicine, Hong Kong Hospital Authority Infectious Disease Center, Princess Margaret Hospital, Special Administrative Region of Hong Kong, China
| | - Owen TY Tsang
- Infectious Diseases Centre, Princess Margaret Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Susan S Chiu
- Department of Paediatric and Adolescent Medicine, The University of Hong Kong and Queen Mary Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Wai Hung Chan
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Yat Sun Yau
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Niloufar Kavian
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Fionn NL Ma
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Eric HY Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Samuel MS Cheng
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Leo LM Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - JS Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
1336
|
Roberts J, Pritchard AL, Treweeke AT, Rossi AG, Brace N, Cahill P, MacRury SM, Wei J, Megson IL. Why Is COVID-19 More Severe in Patients With Diabetes? The Role of Angiotensin-Converting Enzyme 2, Endothelial Dysfunction and the Immunoinflammatory System. Front Cardiovasc Med 2021; 7:629933. [PMID: 33614744 PMCID: PMC7886785 DOI: 10.3389/fcvm.2020.629933] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/15/2020] [Indexed: 01/08/2023] Open
Abstract
Meta-analyses have indicated that individuals with type 1 or type 2 diabetes are at increased risk of suffering a severe form of COVID-19 and have a higher mortality rate than the non-diabetic population. Patients with diabetes have chronic, low-level systemic inflammation, which results in global cellular dysfunction underlying the wide variety of symptoms associated with the disease, including an increased risk of respiratory infection. While the increased severity of COVID-19 amongst patients with diabetes is not yet fully understood, the common features associated with both diseases are dysregulated immune and inflammatory responses. An additional key player in COVID-19 is the enzyme, angiotensin-converting enzyme 2 (ACE2), which is essential for adhesion and uptake of virus into cells prior to replication. Changes to the expression of ACE2 in diabetes have been documented, but they vary across different organs and the importance of such changes on COVID-19 severity are still under investigation. This review will examine and summarise existing data on how immune and inflammatory processes interplay with the pathogenesis of COVID-19, with a particular focus on the impacts that diabetes, endothelial dysfunction and the expression dynamics of ACE2 have on the disease severity.
Collapse
Affiliation(s)
- Jacob Roberts
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Antonia L. Pritchard
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Andrew T. Treweeke
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Adriano G. Rossi
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicole Brace
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Paul Cahill
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Sandra M. MacRury
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Jun Wei
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Ian L. Megson
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| |
Collapse
|
1337
|
Ni L, Cheng ML, Feng Y, Zhao H, Liu J, Ye F, Ye Q, Zhu G, Li X, Wang P, Shao J, Deng YQ, Wei P, Chen F, Qin CF, Wang G, Li F, Zeng H, Dong C. Impaired Cellular Immunity to SARS-CoV-2 in Severe COVID-19 Patients. Front Immunol 2021; 12:603563. [PMID: 33603759 PMCID: PMC7884325 DOI: 10.3389/fimmu.2021.603563] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
The high infection rate and rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) make it a world-wide pandemic. Individuals infected by the virus exhibited different degrees of symptoms, and most convalescent individuals have been shown to develop both cellular and humoral immune responses. However, virus-specific adaptive immune responses in severe patients during acute phase have not been thoroughly studied. Here, we found that in a group of COVID-19 patients with acute respiratory distress syndrome (ARDS) during hospitalization, most of them mounted SARS-CoV-2-specific antibody responses, including neutralizing antibodies. However, compared to healthy controls, the percentages and absolute numbers of both NK cells and CD8+ T cells were significantly reduced, with decreased IFNγ expression in CD4+ T cells in peripheral blood from severe patients. Most notably, their peripheral blood lymphocytes failed in producing IFNγ against viral proteins. Thus, severe COVID-19 patients at acute infection stage developed SARS-CoV-2-specific antibody responses but were impaired in cellular immunity, which emphasizes on the role of cellular immunity in COVID-19.
Collapse
Affiliation(s)
- Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Center for Human Disease Immuno-monitoring, Beijing Friendship Hospital, Beijing, China
| | - Meng-Li Cheng
- College of Basic Medical Science, Jilin University, Changchun, China
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yu Feng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Hui Zhao
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Jingyuan Liu
- Beijing Ditan Hospital, Capital Medical University, and Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Fang Ye
- Department of Hematology, Chui Yang Liu Hospital Affiliated to Tsinghua University, Beijing, China
| | - Qing Ye
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Gengzhen Zhu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xiaoli Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Pengzhi Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Jing Shao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Yong-Qiang Deng
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Peng Wei
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Fang Chen
- Department of Cardiology, Chui Yang Liu Hospital Affiliated to Tsinghua University, Beijing, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Guoqing Wang
- College of Basic Medical Science, Jilin University, Changchun, China
| | - Fan Li
- College of Basic Medical Science, Jilin University, Changchun, China
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Hui Zeng
- Beijing Ditan Hospital, Capital Medical University, and Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Center for Human Disease Immuno-monitoring, Beijing Friendship Hospital, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
1338
|
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CA, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella L, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, The UPenn COVID Processing Unit, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer N, DeMichele AM, Vardhana SA, Mamtani R, Huang AC. CD8 T cells compensate for impaired humoral immunity in COVID-19 patients with hematologic cancer. RESEARCH SQUARE 2021:rs.3.rs-162289. [PMID: 33564756 PMCID: PMC7872363 DOI: 10.21203/rs.3.rs-162289/v1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer patients have increased morbidity and mortality from Coronavirus Disease 2019 (COVID-19), but the underlying immune mechanisms are unknown. In a cohort of 100 cancer patients hospitalized for COVID-19 at the University of Pennsylvania Health System, we found that patients with hematologic cancers had a significantly higher mortality relative to patients with solid cancers after accounting for confounders including ECOG performance status and active cancer status. We performed flow cytometric and serologic analyses of 106 cancer patients and 113 non-cancer controls from two additional cohorts at Penn and Memorial Sloan Kettering Cancer Center. Patients with solid cancers exhibited an immune phenotype similar to non-cancer patients during acute COVID-19 whereas patients with hematologic cancers had significant impairment of B cells and SARS-CoV-2-specific antibody responses. High dimensional analysis of flow cytometric data revealed 5 distinct immune phenotypes. An immune phenotype characterized by CD8 T cell depletion was associated with a high viral load and the highest mortality of 71%, among all cancer patients. In contrast, despite impaired B cell responses, patients with hematologic cancers and preserved CD8 T cells had a lower viral load and mortality. These data highlight the importance of CD8 T cells in acute COVID-19, particularly in the setting of impaired humoral immunity. Further, depletion of B cells with anti-CD20 therapy resulted in almost complete abrogation of SARS-CoV-2-specific IgG and IgM antibodies, but was not associated with increased mortality compared to other hematologic cancers, when adequate CD8 T cells were present. Finally, higher CD8 T cell counts were associated with improved overall survival in patients with hematologic cancers. Thus, CD8 T cells likely compensate for deficient humoral immunity and influence clinical recovery of COVID-19. These observations have important implications for cancer and COVID-19-directed treatments, immunosuppressive therapies, and for understanding the role of B and T cells in acute COVID-19.
Collapse
Affiliation(s)
- Erin M. Bange
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Nicholas A. Han
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Paul Wileyto
- Abramson Cancer Center, University of Pennsylvania
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania
| | - Justin Y. Kim
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - Allison R. Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Florence Porterfield
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Olutosin Owoyemi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Karan Naik
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Cathy Zheng
- Abramson Cancer Center, University of Pennsylvania
| | | | - Ariel R. Weisman
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Caroline A.G. Ittner
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Emily M. Kugler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Amy E. Baxter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Olutwatosin Oniyide
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Roseline S. Agyekum
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Thomas G. Dunn
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Tiffanie K. Jones
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Heather M. Giannini
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Madison E. Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - N. Esther Babady
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Adam J Widman
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | | | | | | | - Carla Wright
- Abramson Cancer Center, University of Pennsylvania
| | - Tara Perloff
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Josephine R. Giles
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Derek A. Oldridge
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Jennifer E. Wu
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Cécile Alanio
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Sharon Adamski
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Alfred L. Garfall
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Laura Vella
- Department of Pediatrics, Perelman School of Medicine, Children’s Hospital of Philadelphia
| | - Samuel J. Kerr
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Justine V. Cohen
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Randall A. Oyer
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Ryan Massa
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital
| | - Ivan P. Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | | | - Kara N. Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - John P. Reilly
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Peter G. Maslak
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Robert H. Vonderheide
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Jedd D. Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Scott E. Hensley
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Nuala Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Angela M. DeMichele
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Santosha A. Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Parker Institute for Cancer Immunotherapy
| | - Ronac Mamtani
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Alexander C. Huang
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| |
Collapse
|
1339
|
Beltrán-Pavez C, Riquelme-Barrios S, Oyarzún-Arrau A, Gaete-Argel A, González-Stegmaier R, Cereceda-Solis K, Aguirre A, Travisany D, Palma-Vejares R, Barriga GP, Gaggero A, Martínez-Valdebenito C, Corre NL, Ferrés M, Balcells ME, Fernandez J, Ramírez E, Villarroel F, Valiente-Echeverría F, Soto-Rifo R. Insights into neutralizing antibody responses in individuals exposed to SARS-CoV-2 in Chile. SCIENCE ADVANCES 2021; 7:eabe6855. [PMID: 33579701 PMCID: PMC7880587 DOI: 10.1126/sciadv.abe6855] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/23/2020] [Indexed: 05/08/2023]
Abstract
Chile has one of the worst numbers worldwide in terms of SARS-CoV-2 positive cases and COVID-19-related deaths per million inhabitants; thus, characterization of neutralizing antibody (NAb) responses in the general population is critical to understanding of immunity at the local level. Given our inability to perform massive classical neutralization assays due to the scarce availability of BSL-3 facilities in the country, we developed and fully characterized an HIV-based SARS-CoV-2 pseudotype, which was used in a 96-well plate format to investigate NAb responses in samples from individuals exposed to SARS-CoV-2 or treated with convalescent plasma. We also identified samples with decreased or enhanced neutralization activity against the D614G spike variant compared with the wild type, indicating the relevance of this variant in host immunity. The data presented here represent the first insights into NAb responses in individuals from Chile, serving as a guide for future studies in the country.
Collapse
Affiliation(s)
- Carolina Beltrán-Pavez
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Sebastián Riquelme-Barrios
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Aarón Oyarzún-Arrau
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Aracelly Gaete-Argel
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Karina Cereceda-Solis
- Translational Medicine Laboratory, Fundación Arturo López Pérez Cancer Center, Santiago, Chile
| | - Adam Aguirre
- Translational Medicine Laboratory, Fundación Arturo López Pérez Cancer Center, Santiago, Chile
| | - Dante Travisany
- Centro de Modelamiento Matemático UMI-CNRS 2807, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile and Fondap Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- INRIA Chile Research Center, Santiago, Chile
| | - Ricardo Palma-Vejares
- Centro de Modelamiento Matemático UMI-CNRS 2807, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile and Fondap Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gonzalo P Barriga
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Emerging Viruses, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Aldo Gaggero
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Environmental Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Constanza Martínez-Valdebenito
- Departamento de Enfermedades Infecciosas e Inmunología Pediátricas, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Infectología y Virología Molecular, Laboratorio de Bioseguridad Nivel 3, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicole Le Corre
- Departamento de Enfermedades Infecciosas e Inmunología Pediátricas, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Infectología y Virología Molecular, Laboratorio de Bioseguridad Nivel 3, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcela Ferrés
- Departamento de Enfermedades Infecciosas e Inmunología Pediátricas, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Infectología y Virología Molecular, Laboratorio de Bioseguridad Nivel 3, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Elvira Balcells
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge Fernandez
- Subdepartamento de Genética Molecular, Instituto de Salud Pública, Santiago, Chile
| | - Eugenio Ramírez
- Sección Virus Oncogénicos, Subdepartamento de Enfermedades Virales, Instituto de Salud Pública, Santiago, Chile
| | - Franz Villarroel
- Translational Medicine Laboratory, Fundación Arturo López Pérez Cancer Center, Santiago, Chile
| | - Fernando Valiente-Echeverría
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ricardo Soto-Rifo
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
1340
|
Barrett JR, Belij-Rammerstorfer S, Dold C, Ewer KJ, Folegatti PM, Gilbride C, Halkerston R, Hill J, Jenkin D, Stockdale L, Verheul MK, Aley PK, Angus B, Bellamy D, Berrie E, Bibi S, Bittaye M, Carroll MW, Cavell B, Clutterbuck EA, Edwards N, Flaxman A, Fuskova M, Gorringe A, Hallis B, Kerridge S, Lawrie AM, Linder A, Liu X, Madhavan M, Makinson R, Mellors J, Minassian A, Moore M, Mujadidi Y, Plested E, Poulton I, Ramasamy MN, Robinson H, Rollier CS, Song R, Snape MD, Tarrant R, Taylor S, Thomas KM, Voysey M, Watson MEE, Wright D, Douglas AD, Green CM, Hill AVS, Lambe T, Gilbert S, Pollard AJ. Phase 1/2 trial of SARS-CoV-2 vaccine ChAdOx1 nCoV-19 with a booster dose induces multifunctional antibody responses. Nat Med 2021; 27:279-288. [PMID: 33335322 DOI: 10.1038/s41591-020-01179-4] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022]
Abstract
More than 190 vaccines are currently in development to prevent infection by the novel severe acute respiratory syndrome coronavirus 2. Animal studies suggest that while neutralizing antibodies against the viral spike protein may correlate with protection, additional antibody functions may also be important in preventing infection. Previously, we reported early immunogenicity and safety outcomes of a viral vector coronavirus vaccine, ChAdOx1 nCoV-19 (AZD1222), in a single-blinded phase 1/2 randomized controlled trial of healthy adults aged 18-55 years ( NCT04324606 ). Now we describe safety and exploratory humoral and cellular immunogenicity of the vaccine, from subgroups of volunteers in that trial, who were subsequently allocated to receive a homologous full-dose (SD/SD D56; n = 20) or half-dose (SD/LD D56; n = 32) ChAdOx1 booster vaccine 56 d following prime vaccination. Previously reported immunogenicity data from the open-label 28-d interval prime-boost group (SD/SD D28; n = 10) are also presented to facilitate comparison. Additionally, we describe volunteers boosted with the comparator vaccine (MenACWY; n = 10). In this interim report, we demonstrate that a booster dose of ChAdOx1 nCoV-19 is safe and better tolerated than priming doses. Using a systems serology approach we also demonstrate that anti-spike neutralizing antibody titers, as well as Fc-mediated functional antibody responses, including antibody-dependent neutrophil/monocyte phagocytosis, complement activation and natural killer cell activation, are substantially enhanced by a booster dose of vaccine. A booster dose of vaccine induced stronger antibody responses than a dose-sparing half-dose boost, although the magnitude of T cell responses did not increase with either boost dose. These data support the two-dose vaccine regime that is now being evaluated in phase 3 clinical trials.
Collapse
Affiliation(s)
- Jordan R Barrett
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Katie J Ewer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pedro M Folegatti
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ciaran Gilbride
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Daniel Jenkin
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Lisa Stockdale
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Marije K Verheul
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Duncan Bellamy
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eleanor Berrie
- Clinical BioManufacturing Facility, The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Mustapha Bittaye
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Miles W Carroll
- Clinical BioManufacturing Facility, The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Nick Edwards
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Amy Flaxman
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Michelle Fuskova
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Simon Kerridge
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alison M Lawrie
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aline Linder
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Xinxue Liu
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Meera Madhavan
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rebecca Makinson
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Angela Minassian
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Maria Moore
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Yama Mujadidi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Ian Poulton
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Maheshi N Ramasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Hannah Robinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Rinn Song
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Richard Tarrant
- Clinical BioManufacturing Facility, The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Marion E E Watson
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel Wright
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexander D Douglas
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Catherine M Green
- Clinical BioManufacturing Facility, The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | | |
Collapse
|
1341
|
Batty CJ, Heise MT, Bachelder EM, Ainslie KM. Vaccine formulations in clinical development for the prevention of severe acute respiratory syndrome coronavirus 2 infection. Adv Drug Deliv Rev 2021; 169:168-189. [PMID: 33316346 PMCID: PMC7733686 DOI: 10.1016/j.addr.2020.12.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an unprecedented effort toward the development of an effective and safe vaccine. Aided by extensive research efforts into characterizing and developing countermeasures towards prior coronavirus epidemics, as well as recent developments of diverse vaccine platform technologies, hundreds of vaccine candidates using dozens of delivery vehicles and routes have been proposed and evaluated preclinically. A high demand coupled with massive effort from researchers has led to the advancement of at least 31 candidate vaccines in clinical trials, many using platforms that have never before been approved for use in humans. This review will address the approach and requirements for a successful vaccine against SARS-CoV-2, the background of the myriad of vaccine platforms currently in clinical trials for COVID-19 prevention, and a summary of the present results of those trials. It concludes with a perspective on formulation problems which remain to be addressed in COVID-19 vaccine development and antigens or adjuvants which may be worth further investigation.
Collapse
Affiliation(s)
- Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Mark T Heise
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
1342
|
Byazrova M, Yusubalieva G, Spiridonova A, Efimov G, Mazurov D, Baranov K, Baklaushev V, Filatov A. Pattern of circulating SARS-CoV-2-specific antibody-secreting and memory B-cell generation in patients with acute COVID-19. Clin Transl Immunology 2021; 10:e1245. [PMID: 33552508 PMCID: PMC7848539 DOI: 10.1002/cti2.1245] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES To predict the spread of coronavirus disease (COVID-19), information regarding the immunological memory for disease-specific antigens is necessary. The possibility of reinfection, as well as the efficacy of vaccines for COVID-19 that are currently under development, will largely depend on the quality and longevity of immunological memory in patients. To elucidate the process of humoral immunity development, we analysed the generation of plasmablasts and virus receptor-binding domain (RBD)-specific memory B (Bmem) cells in patients during the acute phase of COVID-19. METHODS The frequencies of RBD-binding plasmablasts and RBD-specific antibody-secreting cells (ASCs) in the peripheral blood samples collected from patients with COVID-19 were measured using flow cytometry and the ELISpot assay. RESULTS The acute phase of COVID-19 was characterised by the transient appearance of total as well as RBD-binding plasmablasts. ELISpot analysis indicated that most patients exhibited a spontaneous secretion of RBD-specific ASCs in the circulation with good correlation between the IgG and IgM subsets. IL-21/CD40L stimulation of purified B cells induced the activation and proliferation of Bmem cells, which led to the generation of plasmablast phenotypic cells as well as RBD-specific ASCs. No correlation was observed between the frequency of Bmem cell-derived and spontaneous ASCs, suggesting that the two types of ASCs were weakly associated with each other. CONCLUSION Our findings reveal that SARS-CoV-2-specific Bmem cells are generated during the acute phase of COVID-19. These findings can serve as a basis for further studies on the longevity of SARS-CoV-2-specific B-cell memory.
Collapse
Affiliation(s)
- Maria Byazrova
- National Research Center Institute of Immunology of Federal Medical Biological Agency of RussiaMoscowRussia
- Department of ImmunologyFaculty of BiologyLomonosov Moscow State UniversityMoscowRussia
| | - Gaukhar Yusubalieva
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of RussiaMoscowRussia
| | - Anna Spiridonova
- National Research Center Institute of Immunology of Federal Medical Biological Agency of RussiaMoscowRussia
| | | | - Dmitriy Mazurov
- Institute of Gene Biology Russian Academy of SciencesCenter for Precision Genome Editing and Genetic Technologies for BiomedicineMoscowRussia
| | - Konstantin Baranov
- Institute of Molecular and Cellular Biology SB RASLomonosov Moscow State UniversityNovosibirskRussia
| | - Vladimir Baklaushev
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of RussiaMoscowRussia
| | - Alexander Filatov
- National Research Center Institute of Immunology of Federal Medical Biological Agency of RussiaMoscowRussia
- Department of ImmunologyFaculty of BiologyLomonosov Moscow State UniversityMoscowRussia
| |
Collapse
|
1343
|
Sui Y, Bekele Y, Berzofsky JA. Potential SARS-CoV-2 Immune Correlates of Protection in Infection and Vaccine Immunization. Pathogens 2021; 10:pathogens10020138. [PMID: 33573221 PMCID: PMC7912691 DOI: 10.3390/pathogens10020138] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 01/08/2023] Open
Abstract
Both SARS-CoV-2 infections and vaccines induce robust immune responses. Current data suggested that high neutralizing antibody titers with sustained Th1 responses might correlate with protection against viral transmission and disease development and severity. In addition, genetic and innate immune factors, including higher levels of type I interferons, as well as the induction of trained immunity and local mucosal immunity also contribute to lower risk of infection and amelioration of disease severity. The identification of immune correlates of protection will facilitate the development of effective vaccines and therapeutics strategies.
Collapse
|
1344
|
DiPiazza AT, Graham BS, Ruckwardt TJ. T cell immunity to SARS-CoV-2 following natural infection and vaccination. Biochem Biophys Res Commun 2021; 538:211-217. [PMID: 33190827 PMCID: PMC7584424 DOI: 10.1016/j.bbrc.2020.10.060] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022]
Abstract
SARS-CoV-2 first emerged in the human population in late 2019 in Wuhan, China, and in a matter of months, spread across the globe resulting in the Coronavirus Disease 19 (COVID-19) pandemic and substantial economic fallout. SARS-CoV-2 is transmitted between humans via respiratory particles, with infection presenting a spectrum of clinical manifestations ranging from asymptomatic to respiratory failure with multiorgan dysfunction and death in severe cases. Prior experiences with human pathogenic coronaviruses and respiratory virus diseases in general have revealed an important role for cellular immunity in limiting disease severity. Here, we review some of the key mechanisms underlying cell-mediated immunity to respiratory viruses and summarize our current understanding of the functional capacity and role of SARS-CoV-2-specific T cells following natural infection and vaccination.
Collapse
Affiliation(s)
- Anthony T DiPiazza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, USA.
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
1345
|
Kikuchi T. Valuable Insights into the Immune Responses against Coronavirus Disease 2019. JMA J 2021; 4:61-62. [PMID: 33575504 PMCID: PMC7872788 DOI: 10.31662/jmaj.2020-0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 11/09/2022] Open
Affiliation(s)
- Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
1346
|
Carrillo J, Izquierdo-Useros N, Ávila-Nieto C, Pradenas E, Clotet B, Blanco J. Humoral immune responses and neutralizing antibodies against SARS-CoV-2; implications in pathogenesis and protective immunity. Biochem Biophys Res Commun 2021; 538:187-191. [PMID: 33187644 PMCID: PMC7648524 DOI: 10.1016/j.bbrc.2020.10.108] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
The magnitude and the quality of humoral responses against SARS-CoV-2 have been associated with clinical outcome. Although the elicitation of humoral responses against different viral proteins is rapid and occurs in most infected individuals, its magnitude is highly variable among them and positively correlates with COVID-19 disease severity. This rapid response is characterized by the almost concomitant appearance of virus-specific IgG, IgA and IgM antibodies that contain neutralizing antibodies directed against different epitopes of the Spike glycoprotein. Of particularly interest, the antibodies against domain of the Spike that interacts with the cellular receptor ACE2, known as the receptor binding domain (RBD), are present in most infected individuals and are block viral entry and infectivity. Such neutralizing antibodies protect different animal species when administered before virus exposure; therefore, its elicitation is the main target of current vaccine approaches and their clinical use as recombinant monoclonal antibodies (mAbs) is being explored. Yet, little information exists on the duration of humoral responses during natural infection. This is a key issue that will impact the management of the pandemic and determine the utility of seroconversion studies and the level of herd immunity. Certainly, several cases of reinfection have been reported, suggesting that immunity could be transient, as reported for other coronaviruses. In summary, although the kinetics of the generation of antibodies against SASR-CoV-2 and their protective activity have been clearly defined, their role in COVID-19 pathogenesis and the length of these responses are still open questions.
Collapse
Affiliation(s)
- Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Carlos Ávila-Nieto
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Edwards Pradenas
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Infectious Diseases Department, Germans Trias i Pujol Hospital, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| |
Collapse
|
1347
|
Scourfield DO, Reed SG, Quastel M, Alderson J, Bart VMT, Teijeira Crespo A, Jones R, Pring E, Richter FC, Burnell SEA. The role and uses of antibodies in COVID-19 infections: a living review. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab003. [PMID: 34192270 PMCID: PMC7928637 DOI: 10.1093/oxfimm/iqab003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
Coronavirus disease 2019 has generated a rapidly evolving field of research, with the global scientific community striving for solutions to the current pandemic. Characterizing humoral responses towards SARS-CoV-2, as well as closely related strains, will help determine whether antibodies are central to infection control, and aid the design of therapeutics and vaccine candidates. This review outlines the major aspects of SARS-CoV-2-specific antibody research to date, with a focus on the various prophylactic and therapeutic uses of antibodies to alleviate disease in addition to the potential of cross-reactive therapies and the implications of long-term immunity.
Collapse
Affiliation(s)
- D Oliver Scourfield
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sophie G Reed
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Max Quastel
- Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Jennifer Alderson
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, OX3 FTY, UK
| | - Valentina M T Bart
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Alicia Teijeira Crespo
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN UK
| | - Ruth Jones
- Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Ellie Pring
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Felix Clemens Richter
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, OX3 FTY, UK
| | - Stephanie E A Burnell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
1348
|
Sellner J, Rommer PS. Multiple Sclerosis and SARS-CoV-2 Vaccination: Considerations for Immune-Depleting Therapies. Vaccines (Basel) 2021; 9:vaccines9020099. [PMID: 33525459 PMCID: PMC7911298 DOI: 10.3390/vaccines9020099] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 12/19/2022] Open
Abstract
Several concerns have been raised about the use of immunodepleting agents including alemtuzumab, cladribine and CD20-depleting antibodies in people with multiple sclerosis (pwMS) during the coronavirus disease (COVID) 2019 pandemic. As the end of the pandemic is not yet in sight, vaccination against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) may be an elegant strategy to overcome the potential hazards associated with initiating and continuing treatment with immune-depleting agents. In this review, we summarize the immunological effects of immune-depleting therapy and underlying considerations for the hitherto existing recommendations that suggest a restricted use of immune-deleting therapies during the pandemic. Moreover, we critically discuss open questions regarding vaccination in general and against SARS-CoV-2 in pwMS.
Collapse
Affiliation(s)
- Johann Sellner
- Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Correspondence: ; Tel.: +43-2572-9004-12850; Fax: +43-2572-9004-49281
| | - Paulus S. Rommer
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
1349
|
Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases. CELL REPORTS MEDICINE 2021; 2:100204. [PMID: 33521695 PMCID: PMC7837622 DOI: 10.1016/j.xcrm.2021.100204] [Citation(s) in RCA: 367] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/17/2020] [Accepted: 01/20/2021] [Indexed: 12/19/2022]
Abstract
T cells are involved in control of SARS-CoV-2 infection. To establish the patterns of immunodominance of different SARS-CoV-2 antigens and precisely measure virus-specific CD4+ and CD8+ T cells, we study epitope-specific T cell responses of 99 convalescent coronavirus disease 2019 (COVID-19) cases. The SARS-CoV-2 proteome is probed using 1,925 peptides spanning the entire genome, ensuring an unbiased coverage of human leukocyte antigen (HLA) alleles for class II responses. For HLA class I, we study an additional 5,600 predicted binding epitopes for 28 prominent HLA class I alleles, accounting for wide global coverage. We identify several hundred HLA-restricted SARS-CoV-2-derived epitopes. Distinct patterns of immunodominance are observed, which differ for CD4+ T cells, CD8+ T cells, and antibodies. The class I and class II epitopes are combined into epitope megapools to facilitate identification and quantification of SARS-CoV-2-specific CD4+ and CD8+ T cells.
Collapse
|
1350
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33532774 DOI: 10.1101/2021.01.25.428097] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Functional and lasting immune responses to the novel coronavirus (SARS-CoV-2) are currently under intense investigation as antibody titers in plasma have been shown to decline during convalescence. Since the absence of antibodies does not equate to absence of immune memory, we sought to determine the presence of SARS-CoV-2-specific memory B cells in COVID-19 convalescent patients. In this study, we report on the evolution of the overall humoral immune responses on 101 blood samples obtained from 32 COVID-19 convalescent patients between 16 and 233 days post-symptom onset. Our observations indicate that anti-Spike and anti-RBD IgM in plasma decay rapidly, whereas the reduction of IgG is less prominent. Neutralizing activity in convalescent plasma declines rapidly 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 increase over time, and the number of IgG+ memory B cells which remain stable thereafter for up to 8 months after symptoms onset. With the recent approval of highly effective vaccines for COVID-19, data on the persistence of immune responses are of central importance. Even though overall circulating SARS-CoV-2 Spike-specific antibodies contract over time during convalescence, we demonstrate that RBD-specific B cells increase and persist up to 8 months post symptom onset. We also observe modest increases in RBD-specific IgG+ memory B cells and importantly, detectable IgG and sustained Fc-effector activity in plasma over the 8-month period. Our results add to the current understanding of immune memory following SARS-CoV-2 infection, which is critical for the prevention of secondary infections, vaccine efficacy and herd immunity against COVID-19.
Collapse
|