1
|
Leekha A, Saeedi A, Sefat KMSR, Kumar M, Martinez-Paniagua M, Damian A, Kulkarni R, Reichel K, Rezvan A, Masoumi S, Liu X, Cooper LJN, Sebastian M, Sands CM, Das VE, Patel NB, Hurst B, Varadarajan N. Multi-antigen intranasal vaccine protects against challenge with sarbecoviruses and prevents transmission in hamsters. Nat Commun 2024; 15:6193. [PMID: 39043645 PMCID: PMC11266618 DOI: 10.1038/s41467-024-50133-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
Immunization programs against SARS-CoV-2 with commercial intramuscular vaccines prevent disease but are less efficient in preventing infections. Mucosal vaccines can provide improved protection against transmission, ideally for different variants of concern (VOCs) and related sarbecoviruses. Here, we report a multi-antigen, intranasal vaccine, NanoSTING-SN (NanoSTING-Spike-Nucleocapsid), eliminates virus replication in both the lungs and the nostrils upon challenge with the pathogenic SARS-CoV-2 Delta VOC. We further demonstrate that NanoSTING-SN prevents transmission of the SARS-CoV-2 Omicron VOC (BA.5) to vaccine-naïve hamsters. To evaluate protection against other sarbecoviruses, we immunized mice with NanoSTING-SN. We showed that immunization affords protection against SARS-CoV, leading to protection from weight loss and 100% survival in mice. In non-human primates, animals immunized with NanoSTING-SN show durable serum IgG responses (6 months) and nasal wash IgA responses cross-reactive to SARS-CoV-2 (XBB1.5), SARS-CoV and MERS-CoV antigens. These observations have two implications: (1) mucosal multi-antigen vaccines present a pathway to reducing transmission of respiratory viruses, and (2) eliciting immunity against multiple antigens can be advantageous in engineering pan-sarbecovirus vaccines.
Collapse
Affiliation(s)
- Ankita Leekha
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Arash Saeedi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - K M Samiur Rahman Sefat
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Monish Kumar
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Melisa Martinez-Paniagua
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Adrian Damian
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Rohan Kulkarni
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Kate Reichel
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Ali Rezvan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Shalaleh Masoumi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | | | | | | | - Vallabh E Das
- College of Optometry, University of Houston, Houston, TX, USA
| | - Nimesh B Patel
- College of Optometry, University of Houston, Houston, TX, USA
| | - Brett Hurst
- Institute of Antiviral Research, Utah State University, UT, Logan, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
2
|
Richards KA, Changrob S, Thomas PG, Wilson PC, Sant AJ. Lack of memory recall in human CD4 T cells elicited by the first encounter with SARS-CoV-2. iScience 2024; 27:109992. [PMID: 38868209 PMCID: PMC11166706 DOI: 10.1016/j.isci.2024.109992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/11/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
The studies reported here focus on the impact of pre-existing CD4 T cell immunity on the first encounter with SARS-CoV-2. They leverage PBMC samples from plasma donors collected after a first SARS-CoV-2 infection, prior to vaccine availability and compared to samples collected prior to the emergence of SARS-CoV-2. Analysis of CD4 T cell specificity across the entire SARS-CoV-2 proteome revealed that the recognition of SARS-CoV-2-derived epitopes by CD4 memory cells prior to the pandemic are enriched for reactivity toward non-structural proteins conserved across endemic CoV strains. However, CD4 T cells after primary infection with SARS-CoV-2 focus on epitopes from structural proteins. We observed little evidence for preferential recall to epitopes conserved between SARS-CoV-2 and seasonal CoV, a finding confirmed through use of selectively curated conserved and SARS-unique peptides. Our data suggest that SARS-CoV-2 CD4 T cells elicited by the first infection are primarily established from the naive CD4 T cell pool.
Collapse
Affiliation(s)
- Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Siriruk Changrob
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Patrick C. Wilson
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
3
|
Maličev E, Žiberna K, Jazbec K, Kolenc A, Mali P, Potokar UR, Rožman P. Cytokine, Anti-SARS-CoV-2 Antibody, and Neutralizing Antibody Levels in Conventional Blood Donors Who Have Recovered from COVID-19. Transfus Med Hemother 2024; 51:175-184. [PMID: 38867805 PMCID: PMC11166906 DOI: 10.1159/000531942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/02/2023] [Indexed: 06/14/2024] Open
Abstract
Background At the beginning of the pandemic, COVID-19 convalescent plasma (CCP) containing anti-SARS-CoV-2 antibodies was suggested as a source of therapy. In the last 3 years, many trials have demonstrated the limited usefulness of CCP therapy. This led us to the hypothesis that CCP could contain other elements, along with the desired neutralizing antibodies, which could potentially prevent it from having a therapeutic effect, among them cytokines, chemokines, growth factors, clotting factors, and autoantibodies. Methods In total, 39 cytokines were analyzed in the plasma of 190 blood donors, and further research focused on the levels of 23 different cytokines in CCP (sCD40L, eotaxin, FGF-2, FLT-3L, ractalkine, GRO-α, IFNα2, IL-1β, IL-1RA, IL-5, IL-6, IL-8, IL-12, IL-13, IL-15, IL-17E, IP-10, MCP-1, MIP-1b, PDGF-AA, TGFα, TNFα, and TRAIL). Anti-SARS-CoV-2 antibodies and neutralizing antibodies were detected in CCP. Results We found no significant differences between CCP taken within a maximum of 180 days from the onset of the first COVID-19 symptoms and the controls. We also made a comparison of the cytokine levels between the low neutralizing antibodies (<160) group and the high neutralizing antibodies (≥160) group and found there were no differences between the groups. Our research also showed no correlation either to levels of anti-SARS-CoV-2 IgG Ab or to the levels of neutralizing antibodies. There were also no significant changes in cytokine levels based on the period after the start of COVID-19 symptoms. Conclusions No elements which could potentially be responsible for preventing CCP from having a therapeutic effect were found.
Collapse
Affiliation(s)
- Elvira Maličev
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Klemen Žiberna
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | | | - Ana Kolenc
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Polonca Mali
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | | | - Primož Rožman
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
4
|
Tripodi D, Dominici R, Sacco D, Pozzobon C, Spiti S, Falbo R, Brambilla P, Mascagni P, Leoni V. Antibody Response against SARS-CoV-2 after mRNA Vaccine in a Cohort of Hospital Healthy Workers Followed for 17 Months. Vaccines (Basel) 2024; 12:506. [PMID: 38793757 PMCID: PMC11125999 DOI: 10.3390/vaccines12050506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
The assessment of antibody response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is of critical importance to verify the protective efficacy of available vaccines. Hospital healthcare workers play an essential role in the care and treatment of patients and were particularly at risk of contracting the SARS-CoV-2 infection during the pandemic. The vaccination protocol introduced in our hospital protected the workers and contributed to the containment of the infection' s spread and transmission, although a reduction in vaccine efficacy against symptomatic and breakthrough infections in vaccinated individuals was observed over time. Here, we present the results of a longitudinal and prospective analysis of the anti-SARS-CoV-2 antibodies at multiple time points over a 17-month period to determine how circulating antibody levels change over time following natural infection and vaccination for SARS-CoV-2 before (T0-T4) and after the spread of the omicron variant (T5-T6), analyzing the antibody response of 232 healthy workers at the Pio XI hospital in Desio. A General Estimating Equation model indicated a significant association of the antibody response with time intervals and hospital area, independent of age and sex. Specifically, a similar pattern of antibody response was observed between the surgery and administrative departments, and a different pattern with higher peaks of average antibody response was observed in the emergency and medical departments. Furthermore, using a logistic model, we found no differences in contracting SARS-CoV-2 after the third dose based on the hospital department. Finally, analysis of antibody distribution following the spread of the omicron variant, subdividing the cohort of positive individuals into centiles, highlighted a cut-off of 550 BAU/mL and showed that subjects with antibodies below this are more susceptible to infection than those with a concentration above the established cut-off value.
Collapse
Affiliation(s)
- Domenico Tripodi
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberto Dominici
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Davide Sacco
- Department of Brain and Behavioural Sciences, Università degli Studi di Pavia, 27100 Pavia, Italy;
- Laboratory of Medical Genetics, Centro Diagnostico Italiano, 20100 Milan, Italy
| | - Claudia Pozzobon
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Simona Spiti
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Rosanna Falbo
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Paolo Brambilla
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Paolo Mascagni
- Clinical Unit of Occupational Health, Desio Hospital, ASST Brianza, 20832 Desio, Italy
| | - Valerio Leoni
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
5
|
Moy JN, Amin AM, Chalmers-Watson C, Chowdhury R, Forssten C, Fu J, Ghosh S, Harris JD, Kordowich S, Li Y, Lin W, Mackay-Thomas S, Mickiewicz M, Patel N, Resino S, Sevenoaks T, Tugetman MA, Valencia J, Vijesurier R, White N, Woods CW, Kennedy PT, Ryan P. Evaluation of the Panbio™ COVID-19 IgG rapid test device performance. Heliyon 2023; 9:e22612. [PMID: 38125420 PMCID: PMC10730567 DOI: 10.1016/j.heliyon.2023.e22612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Background The Panbio™ COVID-19 IgG Rapid Test Device ("Panbio™") detects IgG antibodies against the SARS-CoV-2 spike protein from viral infection or vaccination. Objectives To determine the diagnostic sensitivity and specificity of the Panbio™ professional use test, using fingerstick whole blood and venous plasma. Study design Fingerstick whole blood and venous plasma from each participant were tested with Panbio™ and compared against the SARS-CoV-2 IgG II assay on the Abbott Architect™ platform (Europe) or the equivalent AdviseDx SARS-CoV-2 IgG II Abbott Alinity i™ platform (US). 447 evaluable participants were enrolled across 6 US and 9 European clinical centers. Results For unvaccinated participants with PCR-confirmed infection ≥21 days post-symptom onset, the Panbio™ sensitivity with fingerstick whole blood was 92.6 % (95 % CI: 85.9, 96.7), and the specificity was 97.0 % (95 % CI: 93.1, 99.0). For venous plasma, the sensitivity was 90.0 % (95 % CI: 79.5, 96.2) for participants with PCR-confirmed infection and symptom onset 22-180 days ago; the specificity was 96.3 % (92.2, 98.6). For vaccinated participants, the sensitivity was 98.4 % (95 % CI: 91.2, 100.0) for fingerstick whole blood and 96.7 % (95 % CI: 88.7, 99.6) for venous plasma. Conclusion The Panbio™ test had high sensitivity and specificity for detecting IgG against the SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
- James N. Moy
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | - Jun Fu
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | | | - Jeffrey D. Harris
- Urgent Care Clinical Trials at AFC Urgent Care, Easley and Powdersville, SC, USA
| | | | - Yin Li
- Abbott Rapid and Molecular Diagnostics, Carlsbad, CA, USA
| | - Wenchi Lin
- Abbott Rapid and Molecular Diagnostics, Galway, Ireland
| | | | - Marc Mickiewicz
- Urgent Care Clinical Trials at Complete Urgent Care, Nashville, TN, USA
| | | | - Salvador Resino
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain
| | | | | | - Jorge Valencia
- Servicio de Medicina Interna, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Roy Vijesurier
- Abbott Rapid and Molecular Diagnostics, Abbott Park, IL, USA
| | - Nikki White
- The Princess Alexandra Hospital NHS Trust, Essex, UK
| | - Christopher W. Woods
- Center for Infectious Disease Diagnostic and Innovation, Duke University School of Medicine, Durham, NC, USA
| | | | - Pablo Ryan
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Medicina Interna, Hospital Universitario Infanta Leonor, Madrid, Spain
- Universidad complutense de Madrid, Madrid, Spain
| |
Collapse
|
6
|
Wang Y, Zhao B, Zhang X, Zhang X, Gao F, Yuan X, Ren X, Li M, Liu D. How immune breakthroughs could slow disease progression and improve prognosis in COVID-19 patients: a retrospective study. Front Immunol 2023; 14:1246751. [PMID: 37936709 PMCID: PMC10627193 DOI: 10.3389/fimmu.2023.1246751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/04/2023] [Indexed: 11/09/2023] Open
Abstract
Background Previous infections and vaccinations have produced preexisting immunity, which differs from primary infection in the organism immune response and may lead to different disease severities and prognoses when reinfected. Objectives The purpose of this retrospective cohort study was to investigate the impact of immune breakthroughs on disease progression and prognosis in patients with COVID-19. Methods A retrospective cohort study was conducted on 1513 COVID-19 patients in Chengdu Public Health Clinical Medical Center from January 2020 to November 2022. All patients were divided into the no immunity group (primary infection and unvaccinated, n=1102) and the immune breakthrough group (previous infection or vaccination, n=411). The immune breakthrough group was further divided into the natural immunity subgroup (n=73), the acquired immunity subgroup (n=322) and the mixed immunity subgroup (n=16). The differences in clinical and outcome data and T lymphocyte subsets and antibody levels between two groups or between three subgroups were compared by ANOVA, t test and chi-square test, and the relationship between T lymphocyte subsets and antibody levels and the disease progression and prognosis of COVID-19 patients was assessed by univariate analysis and logistic regression analysis. Results The total critical rate and the total mortality rate were 2.11% and 0.53%, respectively. The immune breakthrough rate was 27.16%. In the no immunity group, the critical rate and the mortality rate were all higher, and the coronavirus negative conversion time was longer than those in the immune breakthrough group. The differences in the critical rate and the coronavirus negative conversion time between the two groups were all statistically significant (3.72% vs. 0.24%, 14.17 vs. 11.90 days, all p<0.001). In addition, in the no immunity group, although lymphocyte counts and T subsets at admission were higher, all of them decreased consistently and significantly and were significantly lower than those in the immune breakthrough group at the same time from the first week to the fourth week after admission (all p<0.01). The total antibody levels and specific Immunoglobulin G (IgG) levels increased gradually and were always significantly lower than those in the immune breakthrough group at the same time from admission to the fourth week after admission (all p<0.001). Moreover, in the natural immunity subgroup, lymphocyte counts and T subsets at admission were the highest, and total antibody levels and specific IgG levels at admission were the lowest. Then, all of them decreased significantly and were the lowest among the three subgroups at the same time from admission to one month after admission (total antibody: from 546.07 to 158.89, IgG: from 6.00 to 3.95) (all p<0.001). Those in the mixed immunity subgroup were followed by those in the acquired immunity subgroup. While lymphocyte counts and T subsets in these two subgroups and total antibody levels (from 830.84 to 1008.21) and specific IgG levels (from 6.23 to 7.51) in the acquired immunity subgroup increased gradually, total antibody levels (from 1100.82 to 908.58) and specific IgG levels (from 7.14 to 6.58) in the mixed immunity subgroup decreased gradually. Furthermore, T lymphocyte subsets and antibody levels were negatively related to disease severity, prognosis and coronavirus negative conversion time. The total antibody, specific IgM and IgG levels showed good utility for predicting critical COVID-19 patients and dead COVID-19 patients. Conclusion Among patients with COVID-19 patients, immune breakthroughs resulting from previous infection or vaccination, could decelerate disease progression and enhance prognosis by expediting host cellular and humoral immunity to accelerate virus clearance, especially in individuals who have been vaccinated and previously infected. Clinical trial registry Chinese Clinical Trial Register ChiCTR2000034563.
Collapse
Affiliation(s)
- Yiting Wang
- School of Public Health, Chengdu Medical College, Chengdu, China
| | - Bennan Zhao
- The First Ward of Internal Medicine, Public Health Clinic Centre of Chengdu, Chengdu, China
| | - Xinyi Zhang
- Department of Endocrinology & Metabolism, Sichuan University West China Hospital, Chengdu, China
| | - Xia Zhang
- The First Ward of Internal Medicine, Public Health Clinic Centre of Chengdu, Chengdu, China
| | - Fengjiao Gao
- The First Ward of Internal Medicine, Public Health Clinic Centre of Chengdu, Chengdu, China
| | - Xiaoyan Yuan
- The First Ward of Internal Medicine, Public Health Clinic Centre of Chengdu, Chengdu, China
| | - Xiaoxia Ren
- The First Ward of Internal Medicine, Public Health Clinic Centre of Chengdu, Chengdu, China
| | - Maoquan Li
- School of Public Health, Chengdu Medical College, Chengdu, China
| | - Dafeng Liu
- The First Ward of Internal Medicine, Public Health Clinic Centre of Chengdu, Chengdu, China
| |
Collapse
|
7
|
Curtis NC, Shin S, Hederman AP, Connor RI, Wieland-Alter WF, Ionov S, Boylston J, Rose J, Sakharkar M, Dorman DB, Dessaint JA, Gwilt LL, Crowley AR, Feldman J, Hauser BM, Schmidt AG, Ashare A, Walker LM, Wright PF, Ackerman ME, Lee J. Characterization of SARS-CoV-2 Convalescent Patients' Serological Repertoire Reveals High Prevalence of Iso-RBD Antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556349. [PMID: 37745524 PMCID: PMC10515772 DOI: 10.1101/2023.09.08.556349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
While our understanding of SARS-CoV-2 pathogenesis and antibody responses following infection and vaccination has improved tremendously since the outbreak in 2019, the sequence identities and relative abundances of the individual constituent antibody molecules in circulation remain understudied. Using Ig-Seq, we proteomically profiled the serological repertoire specific to the whole ectodomain of SARS-CoV-2 prefusion-stabilized spike (S) as well as to the receptor binding domain (RBD) over a 6-month period in four subjects following SARS-CoV-2 infection before SARS-CoV-2 vaccines were available. In each individual, we identified between 59 and 167 unique IgG clonotypes in serum. To our surprise, we discovered that ∼50% of serum IgG specific for RBD did not recognize prefusion-stabilized S (referred to as iso-RBD antibodies), suggesting that a significant fraction of serum IgG targets epitopes on RBD inaccessible on the prefusion-stabilized conformation of S. On the other hand, the abundance of iso-RBD antibodies in nine individuals who received mRNA-based COVID-19 vaccines encoding prefusion-stabilized S was significantly lower (∼8%). We expressed a panel of 12 monoclonal antibodies (mAbs) that were abundantly present in serum from two SARS-CoV-2 infected individuals, and their binding specificities to prefusion-stabilized S and RBD were all in agreement with the binding specificities assigned based on the proteomics data, including 1 iso-RBD mAb which bound to RBD but not to prefusion-stabilized S. 2 of 12 mAbs demonstrated neutralizing activity, while other mAbs were non-neutralizing. 11 of 12 mAbs also bound to S (B.1.351), but only 1 maintained binding to S (B.1.1.529). This particular mAb binding to S (B.1.1.529) 1) represented an antibody lineage that comprised 43% of the individual's total S-reactive serum IgG binding titer 6 months post-infection, 2) bound to the S from a related human coronavirus, HKU1, and 3) had a high somatic hypermutation level (10.9%), suggesting that this antibody lineage likely had been elicited previously by pre-pandemic coronavirus and was re-activated following the SARS-CoV-2 infection. All 12 mAbs demonstrated their ability to engage in Fc-mediated effector function activities. Collectively, our study provides a quantitative overview of the serological repertoire following SARS-CoV-2 infection and the significant contribution of iso-RBD antibodies, demonstrating how vaccination strategies involving prefusion-stabilized S may have reduced the elicitation of iso-RBD serum antibodies which are unlikely to contribute to protection.
Collapse
|
8
|
Seekircher L, Bánki Z, Kimpel J, Rössler A, Schäfer H, Falkensammer B, Bante D, Forer L, Schönherr S, Harthaller T, Sacher M, Ower C, Tschiderer L, Ulmer H, Krammer F, von Laer D, Borena W, Willeit P. Immune response after two doses of the BNT162b2 COVID-19 vaccine and risk of SARS-CoV-2 breakthrough infection in Tyrol, Austria: an open-label, observational phase 4 trial. THE LANCET. MICROBE 2023; 4:e612-e621. [PMID: 37354911 PMCID: PMC10284585 DOI: 10.1016/s2666-5247(23)00107-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Correlates of protection could help to assess the extent to which a person is protected from SARS-CoV-2 infection after vaccination (so-called breakthrough infection). We aimed to clarify associations of antibody and T-cell responses after vaccination against COVID-19 with risk of a SARS-CoV-2 breakthrough infection and whether measurement of these responses enhances risk prediction. METHODS We did an open-label, phase 4 trial in two community centres in the Schwaz district of the Federal State of Tyrol, Austria, before the emergence of the omicron (B.1.1.529) variant of SARS-CoV-2. We included individuals (aged ≥16 years) a mean of 35 days (range 27-43) after they had received a second dose of the BNT162b2 (Pfizer-BioNTech) COVID-19 vaccine. We quantified associations between immunological parameters and breakthrough infection and assessed whether information on these parameters improves risk discrimination. The study is registered with the European Union Drug Regulating Authorities Clinical Trials Database, 2021-002030-16. FINDINGS 2760 individuals (1682 [60·9%] female, 1078 [39·1%] male, mean age 47·4 years [SD 14·5]) were enrolled into this study between May 15 and May 21, 2021, 712 (25·8%) of whom had a previous SARS-CoV-2 infection. Over a median follow-up of 5·9 months, 68 (2·5%) participants had a breakthrough infection. In models adjusted for age, sex, and previous infection, hazard ratios for breakthrough infection for having twice the immunological parameter level at baseline were 0·72 (95% CI 0·60-0·86) for anti-spike IgG, 0·80 (0·70-0·92) for neutralising antibodies in a surrogate virus neutralisation assay, 0·84 (0·58-1·21) for T-cell response after stimulation with a CD4 peptide pool, and 0·77 (0·54-1·08) for T-cell response after stimulation with a combined CD4 and CD8 peptide pool. For neutralising antibodies measured in a nested case-control sample using a pseudotyped virus neutralisation assay, the corresponding odds ratio was 0·78 (0·62-1·00). Among participants with previous infection, the corresponding hazard ratio was 0·73 (0·61-0·88) for anti-nucleocapsid Ig. Addition of anti-spike IgG information to a model containing information on age and sex improved the C-index by 0·085 (0·027-0·143). INTERPRETATION In contrast to T-cell response, higher levels of binding and neutralising antibodies were associated with a reduced risk of breakthrough SARS-CoV-2 infection. The assessment of anti-spike IgG enhances the prediction of incident breakthrough SARS-CoV-2 infection and could therefore be a suitable correlate of protection in practice. Our phase 4 trial measured both humoral and cellular immunity and had a 6-month follow-up period; however, the longer-term protection against emerging variants of SARS-CoV-2 remains unclear. FUNDING None.
Collapse
Affiliation(s)
- Lisa Seekircher
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltán Bánki
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Kimpel
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Annika Rössler
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helena Schäfer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - David Bante
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Teresa Harthaller
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Sacher
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Ower
- Department of Surgery, University Hospital of Trauma Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Lena Tschiderer
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hanno Ulmer
- Institute of Medical Statistics and Informatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krammer
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dorothee von Laer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wegene Borena
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Willeit
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Vienna, Austria; Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| |
Collapse
|
9
|
Li Z, Xiang T, Liang B, Liu J, Deng H, Yang X, Wang H, Feng X, Zelinskyy G, Trilling M, Sutter K, Lu M, Dittmer U, Wang B, Yang D, Zheng X, Liu J. SARS-CoV-2-specific T cell responses wane profoundly in convalescent individuals 10 months after primary infection. Virol Sin 2023; 38:606-619. [PMID: 37414153 PMCID: PMC10436107 DOI: 10.1016/j.virs.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 06/28/2023] [Indexed: 07/08/2023] Open
Abstract
A key question in the coronavirus disease 2019 (COVID-19) pandemic is the duration of specific T cell responses against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) post primary infection, which is difficult to address due to the large-scale COVID-19 vaccination and re-exposure to the virus. Here, we conducted an analysis of the long-term SARS-CoV-2-specific T cell responses in a unique cohort of convalescent individuals (CIs) that were among the first to be infected worldwide and without any possible antigen re-exposure since then. The magnitude and breadth of SARS-CoV-2-specific T cell responses correlated inversely with the time that had elapsed from disease onset and the age of those CIs. The mean magnitude of SARS-CoV-2-specific CD4 and CD8 T cell responses decreased about 82% and 76%, respectively, over the time period of ten months after infection. Accordingly, the longitudinal analysis also demonstrated that SARS-CoV-2-specific T cell responses waned significantly in 75% of CIs during the follow-up. Collectively, we provide a comprehensive characterization of the long-term memory T cell response in CIs, suggesting that robust SARS-CoV-2-specific T cell immunity post primary infection may be less durable than previously expected.
Collapse
Affiliation(s)
- Ziwei Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tiandan Xiang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Boyun Liang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Deng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuecheng Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuemei Feng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mirko Trilling
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kathrin Sutter
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengji Lu
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ulf Dittmer
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Baoju Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jia Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
10
|
Augustin M, Heyn F, Ullrich S, Sandaradura de Silva U, Albert MC, Linne V, Schlotz M, Schommers P, Pracht E, Horn C, Suarez I, Simonis A, Picard LK, Zoufaly A, Wenisch C, Fätkenheuer G, Gruell H, Klein F, Hallek M, Walczak H, Rybniker J, Theobald SJ, Lehmann C. Immunological fingerprint in coronavirus disease-19 convalescents with and without post-COVID syndrome. Front Med (Lausanne) 2023; 10:1129288. [PMID: 37168268 PMCID: PMC10165999 DOI: 10.3389/fmed.2023.1129288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Background Symptoms lasting longer than 12 weeks after severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection are called post-coronavirus disease (COVID) syndrome (PCS). The identification of new biomarkers that predict the occurrence or course of PCS in terms of a post-viral syndrome is vital. T-cell dysfunction, cytokine imbalance, and impaired autoimmunity have been reported in PCS. Nevertheless, there is still a lack of conclusive information on the underlying mechanisms due to, among other things, a lack of controlled study designs. Methods Here, we conducted a prospective, controlled study to characterize the humoral and cellular immune response in unvaccinated patients with and without PCS following SARS-CoV-2 infection over 7 months and unexposed donors. Results Patients with PCS showed as early as 6 weeks and 7 months after symptom onset significantly increased frequencies of SARS-CoV-2-specific CD4+ and CD8+ T-cells secreting IFNγ, TNF, and expressing CD40L, as well as plasmacytoid dendritic cells (pDC) with an activated phenotype. Remarkably, the immunosuppressive counterparts type 1 regulatory T-cells (TR1: CD49b/LAG-3+) and IL-4 were more abundant in PCS+. Conclusion This work describes immunological alterations between inflammation and immunosuppression in COVID-19 convalescents with and without PCS, which may provide potential directions for future epidemiological investigations and targeted treatments.
Collapse
Affiliation(s)
- Max Augustin
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Ferdinand Heyn
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Stella Ullrich
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Ute Sandaradura de Silva
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Marie-Christine Albert
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Viktoria Linne
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Maike Schlotz
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Schommers
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Elisabeth Pracht
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carola Horn
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Isabelle Suarez
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Alexander Simonis
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Lea Katharina Picard
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexander Zoufaly
- Department IV of Internal Medicine, Klinik Favoriten, Vienna Healthcare Group, Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Christoph Wenisch
- Department IV of Internal Medicine, Klinik Favoriten, Vienna Healthcare Group, Vienna, Austria
| | - Gerd Fätkenheuer
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Henning Gruell
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Klein
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Henning Walczak
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Jan Rybniker
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Sebastian J. Theobald
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| | - Clara Lehmann
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Bonn-Cologne, Germany
| |
Collapse
|
11
|
Jazbec K, Jež M, Žiberna K, Mali P, Ramšak Ž, Potokar UR, Kvrzić Z, Černilec M, Gracar M, Šprohar M, Jovanovič P, Vuletić S, Rožman P. Simple prediction of COVID-19 convalescent plasma units with high levels of neutralization antibodies. Virol J 2023; 20:53. [PMID: 36973781 PMCID: PMC10042109 DOI: 10.1186/s12985-023-02007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Hyperimmune convalescent COVID-19 plasma (CCP) containing anti-SARS-CoV-2 neutralizing antibodies (NAbs) was proposed as a therapeutic option for patients early in the new coronavirus disease pandemic. The efficacy of this therapy depends on the quantity of neutralizing antibodies (NAbs) in the CCP units, with titers ≥ 1:160 being recommended. The standard neutralizing tests (NTs) used for determining appropriate CCP donors are technically demanding and expensive and take several days. We explored whether they could be replaced by high-throughput serology tests and a set of available clinical data. METHODS Our study included 1302 CCP donors after PCR-confirmed COVID-19 infection. To predict donors with high NAb titers, we built four (4) multiple logistic regression models evaluating the relationships of demographic data, COVID-19 symptoms, results of various serological testing, the period between disease and donation, and COVID-19 vaccination status. RESULTS The analysis of the four models showed that the chemiluminescent microparticle assay (CMIA) for the quantitative determination of IgG Abs to the RBD of the S1 subunit of the SARS-CoV-2 spike protein was enough to predict the CCP units with a high NAb titer. CCP donors with respective results > 850 BAU/ml SARS-CoV-2 IgG had a high probability of attaining sufficient NAb titers. Including additional variables such as donor demographics, clinical symptoms, or time of donation into a particular predictive model did not significantly increase its sensitivity and specificity. CONCLUSION A simple quantitative serological determination of anti-SARS-CoV-2 antibodies alone is satisfactory for recruiting CCP donors with high titer NAbs.
Collapse
Affiliation(s)
- Katerina Jazbec
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia.
| | - Mojca Jež
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Klemen Žiberna
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Polonca Mali
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Živa Ramšak
- NIB-National Institute of Biology, Ljubljana, Slovenia
| | - Urška Rahne Potokar
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Zdravko Kvrzić
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Maja Černilec
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Melita Gracar
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Marjana Šprohar
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Petra Jovanovič
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Sonja Vuletić
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| | - Primož Rožman
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, Ljubljana, 1000, Slovenia
| |
Collapse
|
12
|
Immune Dynamics Involved in Acute and Convalescent COVID-19 Patients. IMMUNO 2023. [DOI: 10.3390/immuno3010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
COVID-19 is a viral disease that has caused millions of deaths around the world since 2020. Many strategies have been developed to manage patients in critical conditions; however, comprehension of the immune system is a key factor in viral clearance, tissue repairment, and adaptive immunity stimulus. Participation of immunity has been identified as a major factor, along with biomarkers, prediction of clinical outcomes, and antibody production after infection. Immune cells have been proposed not only as a hallmark of severity, but also as a predictor of clinical outcomes, while dynamics of inflammatory molecules can also induce worse consequences for acute patients. For convalescent patients, mild disease was related to higher antibody production, although the factors related to the specific antibodies based on a diversity of antigens were not clear. COVID-19 was explored over time; however, the study of immunological predictors of outcomes is still lacking discussion, especially in convalescent patients. Here, we propose a review using previously published studies to identify immunological markers of COVID-19 outcomes and their relation to antibody production to further contribute to the clinical and laboratorial management of patients.
Collapse
|
13
|
Tanaka S, Umezawa J, Yamaji T, Abe SK, Hamada A, Kobayashi O, Ushijima T, Inoue M. SARS-CoV-2 Antibody Response to Symptoms Indicative of COVID-19 in a Non-Infected Population in Japan: a Cross-Sectional Study. Jpn J Infect Dis 2023; 76:46-54. [PMID: 36184396 DOI: 10.7883/yoken.jjid.2022.093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This study was aimed at investigating differences in antibody titers indicative of the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) between those who had experienced symptoms of coronavirus disease 2019 (COVID-19) and those who had not. We used data from a cross-sectional survey conducted at the National Cancer Center, Japan, of 434 individuals with no previous COVID-19 infection. The participants self-reported symptoms from the start of 2020. A generalized linear model was used to compare the mean SARS-CoV-2-specific IgG nucleocapsid protein (N-IgG) titer with estimated confidence intervals according to the onset of symptoms indicative of COVID-19. We observed a tendency toward an association between higher mean N-IgG titers and occurrence of high fever within the past 8 months (P = 0.053). The mean N-IgG titer was higher in those with prior symptoms (P = 0.03) and those with over three symptoms (P < 0.01) than in those without symptoms. The mean N-IgG titer was higher in those with symptoms and those with more symptoms that might indicate COVID-19 relative to those without symptoms, suggesting that transient but contained SARS-CoV-2 infection had occurred.
Collapse
Affiliation(s)
- Shiori Tanaka
- Division of Prevention, National Cancer Center, Japan
| | - Jun Umezawa
- Division of Cohort Research, National Cancer Center, Japan
| | - Taiki Yamaji
- Division of Epidemiology, Institute for Cancer Control, National Cancer Center, Japan
| | - Sarah K Abe
- Division of Prevention, National Cancer Center, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center, Japan
| | - Osamu Kobayashi
- Department of Infectious Diseases, National Cancer Center, Japan
| | | | - Manami Inoue
- Division of Prevention, National Cancer Center, Japan
| |
Collapse
|
14
|
Bekbossynova M, Akhmaltdinova L, Dossybayeva K, Tauekelova A, Smagulova Z, Tsechoeva T, Turebayeva G, Sailybayeva A, Kalila Z, Mirashirova T, Muratov T, Poddighe D. Central and effector memory T cells in peripheral blood of patients with interstitial pneumonia: preliminary clues from a COVID-19 study. Respir Res 2022; 23:278. [PMID: 36217141 PMCID: PMC9549841 DOI: 10.1186/s12931-022-02190-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND SARS-CoV-2 pre-existing T-cell immune reactivity can be present in some people. A general perturbation of the main peripheral lymphocyte subsets has been described in severe COVID-19 patients, but very few studies assessed the general memory T-cell homeostasis in the acute phase of COVID-19. Here, we performed a general analysis of the main memory T cell populations in the peripheral blood of patients admitted to the hospital for a confirmed or probable COVID-19 diagnosis. METHODS In this cross-sectional study, adult patients (aged ≥ 18 years) needing hospital admission for respiratory disease due to confirmed or probable COVID-19, were recruited before starting the therapeutic protocol for this disease. In addition to the assessment of the general lymphocyte subpopulations in the early phase of COVID-19, central memory T cells (Tmcentr cells: CD45RO+CCR7+) and effector memory T cells (Tmeff cells: CD45RO+CCR7-) were assessed by multi-color flow cytometry, in comparison to a control group. RESULTS During the study period, 148 study participants were recruited. Among them, 58 patients turned out positive for SARS-CoV-2 PCR (including both patients with interstitial pneumonia [PCR+Pn+] and without this complication [PCR+Pn-]), whereas the remaining 90 patients resulted to be SARS-CoV-2 PCR negative, even though all were affected with interstitial pneumonia [PCR-Pn+]. Additionally, 28 control patients without any ongoing respiratory disease were recruited. A clear unbalance in the T memory compartment emerged from this analysis on the whole pool of T cells (CD3+ cells), showing a significant increase in Tmcentr cells and, conversely, a significant decrease in Tmeff cells in both pneumonia groups (PCR+Pn+ and PCR-Pn+) compared to the controls; PCR+Pn- group showed trends comprised between patients with pneumonia (from one side) and the control group (from the other side). This perturbation inside the memory T cell compartment was also observed in the individual analysis of the four main T cell subpopulations, based upon the differential expression of CD4 and/or CD8 markers. CONCLUSION Overall, we observed both absolute and relative increases of Tmcentr cells and decrease of Tmeff cells in patients affected with interstitial pneumonia (regardless of the positive or negative results of SARS-CoV-2 PCR), compared to controls. These results need confirmation from additional research, in order to consider this finding as a potential biological marker of interstitial lung involvement in patients affected with viral respiratory infections.
Collapse
Affiliation(s)
| | | | - Kuanysh Dossybayeva
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan.,Nazarbayev University School of Medicine (NUSOM), Kerei-Zhanibek Str. 5/1, 010000, Nur-Sultan, Kazakhstan
| | - Ainur Tauekelova
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan
| | - Zauresh Smagulova
- City Infectious Disease Center at Multidisciplinary Medical Center, 010000, Nur-Sultan, Kazakhstan
| | - Tatyana Tsechoeva
- City Infectious Disease Center at Multidisciplinary Medical Center, 010000, Nur-Sultan, Kazakhstan
| | - Gulsimzhan Turebayeva
- City Infectious Disease Center at Multidisciplinary Medical Center, 010000, Nur-Sultan, Kazakhstan
| | - Aliya Sailybayeva
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan
| | - Zhanar Kalila
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan
| | | | - Timur Muratov
- Department of Public Health of Nur‑Sultan City, 010000, Nur-Sultan, Kazakhstan
| | - Dimitri Poddighe
- Nazarbayev University School of Medicine (NUSOM), Kerei-Zhanibek Str. 5/1, 010000, Nur-Sultan, Kazakhstan. .,University Medical Center (UMC), 010000, Nur-Sultan, Kazakhstan.
| |
Collapse
|
15
|
Palacios-Pedrero MÁ, Jansen JM, Blume C, Stanislawski N, Jonczyk R, Molle A, Hernandez MG, Kaiser FK, Jung K, Osterhaus ADME, Rimmelzwaan GF, Saletti G. Signs of immunosenescence correlate with poor outcome of mRNA COVID-19 vaccination in older adults. NATURE AGING 2022; 2:896-905. [PMID: 37118289 PMCID: PMC10154205 DOI: 10.1038/s43587-022-00292-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/02/2022] [Indexed: 04/30/2023]
Abstract
Vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is effective in preventing COVID-19 hospitalization and fatal outcome. However, several studies indicated that there is reduced vaccine effectiveness among older individuals, which is correlated with their general health status1,2. How and to what extent age-related immunological defects are responsible for the suboptimal vaccine responses observed in older individuals receiving SARS-CoV-2 messenger RNA vaccine, is unclear and not fully investigated1,3-5. In this observational study, we investigated adaptive immune responses in adults of various ages (22-99 years old) receiving 2 doses of the BNT162b2 mRNA vaccine. Vaccine-induced Spike-specific antibody, and T and memory B cell responses decreased with increasing age. These responses positively correlated with the percentages of peripheral naïve CD4+ and CD8+ T cells and negatively with CD8+ T cells expressing signs of immunosenescence. Older adults displayed a preferred T cell response to the S2 region of the Spike protein, which is relatively conserved and a target for cross-reactive T cells induced by human 'common cold' coronaviruses. Memory T cell responses to influenza virus were not affected by age-related changes, nor the SARS-CoV-2-specific response induced by infection. Collectively, we identified signs of immunosenescence correlating with the outcome of vaccination against a new viral antigen to which older adults are immunologically naïve. This knowledge is important for the management of COVID-19 infections in older adults.
Collapse
Affiliation(s)
| | - Janina M Jansen
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Cornelia Blume
- Institute of Technical Chemistry, Leibniz University, Hanover, Germany
| | - Nils Stanislawski
- Institute of Microelectronic Systems, Leibniz University, Hanover, Germany
| | - Rebecca Jonczyk
- Institute of Technical Chemistry, Leibniz University, Hanover, Germany
| | - Antonia Molle
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Mariana Gonzalez Hernandez
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Franziska K Kaiser
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, Genomics and Bioinformatics of Infectious Diseases, University of Veterinary Medicine, Hanover, Germany
| | - Albert D M E Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
- Global Virus Network, Center of Excellence, Buffalo, NY, USA
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany.
| | - Giulietta Saletti
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany.
| |
Collapse
|
16
|
Xu W, Song D, Liu J, Han X, Xu J, Zhu A, Long F. Development of chemiluminescent lab-on-fiber immunosensors for rapid point-of-care testing of anti-SARS-CoV-2 antibodies and evaluation of longitudinal immune response kinetics following three-dose inactivation virus vaccination. J Med Virol 2022; 95:e28190. [PMID: 36180404 PMCID: PMC9539144 DOI: 10.1002/jmv.28190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 01/11/2023]
Abstract
Developing reliable, rapid, and quantitative point-of-care testing (POCT) technology of SARS-CoV-2-specific antibodies and understanding longitudinal vaccination response kinetics are highly required to restrain the ongoing coronavirus disease 2019 (COVID-19) pandemic. We demonstrate a novel portable, sensitive, and rapid chemiluminescent lab-on-fiber detection platform for detection of anti-SARS-CoV-2 antibodies: the chemiluminescent lab-on-fiber immunosensor (c-LOFI). Using SARS-CoV-2 Spike S1 RBD protein functionalized fiber bio-probe, the c-LOFI can detect anti-SARS-CoV-2 immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies with high sensitivity based on their respective horseradish peroxidase-labeled secondary antibodies. The limits of detection of anti-SARS-CoV-2 IgG and IgM antibodies were 0.6 and 0.3 ng/ml, respectively. The c-LOFI was successfully applied for direct detection of anti-SARS-CoV-2 antibodies in whole blood samples with simple dilution, which can serve as a finger prick test to rapidly detect antibodies. Furthermore, the longitudinal immune response (>12 months) kinetics following three-dose inactivated virus vaccines was evaluated based on anti-SARS-CoV-2 IgG detection results, which can provide important significance for understanding the immune mechanism against COVID-19 and identify individuals who may benefit from the vaccination and booster vaccination. The c-LOFI has great potential to become a sensitive, low-cost, rapid, high-frequency POCT tool for the detection of both SARS-CoV-2-specific antibodies and other biomarkers.
Collapse
Affiliation(s)
- Wenjuan Xu
- School of Environment and Natural ResourcesRenmin University of ChinaBeijing100872China
| | - Dan Song
- School of Environment and Natural ResourcesRenmin University of ChinaBeijing100872China
| | - Jiayao Liu
- School of Environment and Natural ResourcesRenmin University of ChinaBeijing100872China
| | - Xiangzhi Han
- School of Environment and Natural ResourcesRenmin University of ChinaBeijing100872China
| | - Jiaxin Xu
- School of Environment and Natural ResourcesRenmin University of ChinaBeijing100872China
| | - Anna Zhu
- State Key Laboratory of NBC Protection for CivilianBeijing102205China
| | - Feng Long
- School of Environment and Natural ResourcesRenmin University of ChinaBeijing100872China,Department of ChemistryRenmin University of ChinaBeijing100872China
| |
Collapse
|
17
|
Duan LJ, Jiang WG, Wang ZY, Yao L, Zhu KL, Meng QC, Wang BS, Li LB, Wang GL, Ma MJ. Neutralizing immunity against SARS-CoV-2 Omicron BA.1 by infection and vaccination. iScience 2022; 25:104886. [PMID: 35966041 PMCID: PMC9359924 DOI: 10.1016/j.isci.2022.104886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 12/03/2022] Open
Abstract
The emergence of the SARS-CoV-2 Omicron BA.1 (B.1.1.529) variant has raised questions regarding resistance to neutralizing antibodies elicited by natural infection or immunization. We examined the neutralization activity of sera collected from previously SARS-CoV-2-infected individuals and SARS-CoV-2 naive individuals who received BBIBP-CorV or CoronaVac to BA.1 and the earlier variants Alpha, Beta, and Delta. Both sera from convalescent patients over three months after infection and two-dose BBIBP-CorV or CoronaVac vaccine recipients barely inhibited BA.1, less effectively neutralized Beta and Delta, and moderately neutralized Alpha. However, administering a single dose of BBIBP-CorV or CoronaVac in previously infected individuals or a third dose booster vaccination of BBIBP-CorV or CoronaVac in previously vaccinated individuals enhances neutralizing activity against BA.1 and other variants, albeit with a lower antibody titer for BA.1. Our data suggest that a booster vaccination is important to broaden neutralizing antibody responses against the variants. Limited duration of antibody response against BA.1 in convalescent individuals Infection before BBIBP-CorV or CoronaVac vaccination boosts neutralization Two doses of BBIBP-CorV or CoronaVac elicit limited neutralizing activity against VOCs Neutralization breadth for BA.1 is boosted by a third dose of BBIBP-CorV or CoronaVac
Collapse
Affiliation(s)
- Li-Jun Duan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Wen-Guo Jiang
- Jining Center for Disease Control and Prevention, Jining, China
| | - Zhuang-Ye Wang
- Dezhou Center for Disease Control and Prevention, Dezhou, China
| | - Lin Yao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Ka-Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Qing-Chuan Meng
- Ningjin County Community Health Service Center, Dezhou, China
| | - Bao-Shan Wang
- Decheng District Center for Disease Control and Prevention, Dezhou, China
| | - Li-Bo Li
- Jining Center for Disease Control and Prevention, Jining, China
| | - Guo-Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Mai-Juan Ma
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
18
|
Abstract
The COVID-19 pandemic has caused an unprecedented health crisis and economic burden worldwide. Its etiological agent SARS-CoV-2, a new virus in the coronavirus family, has infected hundreds of millions of people worldwide. SARS-CoV-2 has evolved over the past 2 years to increase its transmissibility as well as to evade the immunity established by previous infection and vaccination. Nevertheless, strong immune responses can be elicited by viral infection and vaccination, which have proved to be protective against the emergence of variants, particularly with respect to hospitalization or severe disease. Here, we review our current understanding of how the virus enters the host cell and how our immune system is able to defend against cell entry and infection. Neutralizing antibodies are a major component of our immune defense and have been extensively studied for SARS-CoV-2 and its variants. Structures of these neutralizing antibodies have provided valuable insights into epitopes that are protective against the original ancestral virus and the variants that have emerged. The molecular characterization of neutralizing epitopes as well as epitope conservation and resistance are important for design of next-generation vaccines and antibody therapeutics.
Collapse
Affiliation(s)
- Hejun Liu
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- The Skaggs Institute for Chemical BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| |
Collapse
|
19
|
Safont G, Latorre I, Villar-Hernández R, Stojanovic Z, Marín A, Pérez-Cano C, Lacoma A, Molina-Moya B, Solis AJ, Arméstar F, Matllo J, Díaz-Fernández S, Cendón A, Sokalchuk L, Tolosa G, Casas I, Rosell A, Domínguez J. Measuring T-Cell Responses against SARS-CoV-2 Is of Utility for Disease and Vaccination Management. J Clin Med 2022; 11:5103. [PMID: 36079033 PMCID: PMC9457376 DOI: 10.3390/jcm11175103] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
The measurement of specific T-cell responses can be a useful tool for COVID-19 diagnostics and clinical management. In this study, we evaluated the IFN-γ T-cell response against the main SARS-CoV-2 antigens (spike, nucleocapsid and membrane) in acute and convalescent individuals classified according to severity, and in vaccinated and unvaccinated controls. IgG against spike and nucleocapsid were also measured. Spike antigen triggered the highest number of T-cell responses. Acute patients showed a low percentage of positive responses when compared to convalescent (71.6% vs. 91.7%, respectively), but increased during hospitalization and with severity. Some convalescent patients showed an IFN-γ T-cell response more than 200 days after diagnosis. Only half of the vaccinated individuals displayed an IFN-γ T-cell response after the second dose. IgG response was found in a higher percentage of individuals compared to IFN-γ T-cell responses, and moderate correlations between both responses were seen. However, in some acute COVID-19 patients specific T-cell response was detected, but not IgG production. We found that the chances of an IFN-γ T-cell response against SARS-CoV-2 is low during acute phase, but may increase over time, and that only half of the vaccinated individuals had an IFN-γ T-cell response after the second dose.
Collapse
Affiliation(s)
- Guillem Safont
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Irene Latorre
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Raquel Villar-Hernández
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Zoran Stojanovic
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Alicia Marín
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Cristina Pérez-Cano
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Alicia Lacoma
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Bárbara Molina-Moya
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Alan Jhunior Solis
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Fernando Arméstar
- Intensive Care Medicine Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Joan Matllo
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Sergio Díaz-Fernández
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Arnau Cendón
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Liliya Sokalchuk
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Guillermo Tolosa
- Diagnostic and Research in Immunodeficiencies Jeffrey Modell Center, Cytometry and Cellular Culture Area, La Frontera University, Temuco 01145, Chile
| | - Irma Casas
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Preventive Medicine Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Antoni Rosell
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - José Domínguez
- Institut d’Investigació Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
20
|
High Seroprevalence of SARS-CoV-2 IgG and RNA among Asymptomatic Blood Donors in Makkah Region, Saudi Arabia. Vaccines (Basel) 2022; 10:vaccines10081279. [PMID: 36016167 PMCID: PMC9414004 DOI: 10.3390/vaccines10081279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/23/2022] [Accepted: 08/04/2022] [Indexed: 01/08/2023] Open
Abstract
The gold-standard approach for diagnosing and confirming Severe Acute Respiratory Syndrome Corona Virus-2 (SARS-CoV-2) infection is reverse transcription-polymerase chain reaction (RT-PCR). This method, however, is inefficient in detecting previous or dormant viral infections. The presence of antigen-specific antibodies is the fingerprint and cardinal sign for diagnosis and determination of exposure to infectious agents including Corona virus disease-2019 (COVID-19). This cross-sectional study examined the presence of SARS-CoV-2 spike-specific immunoglobulin G (IgG) among asymptomatic blood donors in Makkah region. A total of 4368 asymptomatic blood donors were enrolled. They were screened for spike-specific IgG using ELISA and COVID-19 RNA by real-time PCR. COVID-19 IgG was detected among 2248 subjects (51.5%) while COVID-19-RNA was detected among 473 (10.8%) subjects. The IgG frequency was significantly higher among males and non-Saudi residents (p < 0.001 each) with no significant variation in IgG positivity among blood donors with different blood groups. In addition, COVID-19 RNA frequency was significantly higher among donors below 40-years old (p = 0.047, χ2 = 3.95), and non-Saudi residents (p = 0.001, χ2 = 304.5). The COVID-19 IgG levels were significantly higher among the RNA-positive donors (p = 001), and non-Saudi residents (p = 0.041), with no variations with age or blood group (p > 0.05). This study reveals a very high prevalence of COVID-19 IgG and RNA among asymptomatic blood donors in Makkah, Saudi Arabia indicating a high exposure rate of the general population to COVID-19; particularly foreign residents. It sheds light on the spread on COVID-19 among apparently healthy individuals at the beginning of the pandemic and could help in designing various control measures to minimize viral spread.
Collapse
|
21
|
Li E, Wang S, He W, He J, Liu L, Zhang X, Yang S, Yan F, Gao Y, Liu B, Xia X. Clinical Characteristics of Immune Response in Asymptomatic Carriers and Symptomatic Patients With COVID-19. Front Microbiol 2022; 13:896965. [PMID: 35685940 PMCID: PMC9171238 DOI: 10.3389/fmicb.2022.896965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/12/2022] [Indexed: 01/08/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) has emerged as a major public health challenge worldwide. A comprehensive understanding of clinical characteristics and immune responses in asymptomatic carriers and symptomatic patients with COVID-19 is of great significance to the countermeasures of patients with COVID-19. Herein, we described the clinical information and laboratory findings of 43 individuals from Hunan Province, China, including 13 asymptomatic carriers and 10 symptomatic patients with COVID-19, as well as 20 healthy controls in the period from 25 January to 18 May 2020. The serum samples of these individuals were analyzed to measure the cytokine responses, receptor-binding domain (RBD), and nucleocapsid (N) protein-specific antibody titers, as well as SARS-CoV-2 neutralizing antibodies (nAbs). For cytokines, significantly higher Th1 cytokines including IL-2, IL-8, IL-12p70, IFN-γ, and TNF-α, as well as Th2 cytokines including IL-10 and IL-13 were observed in symptomatic patients compared with asymptomatic carriers. Compared with symptomatic patients, higher N-specific IgG4/IgG1 ratio and RBD-specific/N-specific IgG1 ratio were observed in asymptomatic carriers. Comparable nAbs were detected in both asymptomatic carriers and symptomatic patients with COVID-19. In the symptomatic group, nAbs in patients with underlying diseases were weaker than those of patients without underlying diseases. Our retrospective study will enrich and verify the clinical characteristics and serology diversities in asymptomatic carriers and symptomatic patients with COVID-19.
Collapse
Affiliation(s)
- Entao Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Wenwen He
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Department of Laboratory Medicine, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Jun He
- Department of Laboratory Medicine, Nanhua Hospital, University of South China, Hengyang, China
| | - Luogeng Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Xiaotuan Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bin Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
22
|
Štěpánek L, Janošíková M, Štěpánek L, Nakládalová M, Boriková A. The kinetics and predictors of anti-SARS-CoV-2 antibodies up to eight months after symptomatic COVID-19: a Czech cross-sectional study. J Med Virol 2022; 94:3731-3738. [PMID: 35419860 PMCID: PMC9088611 DOI: 10.1002/jmv.27784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 11/08/2022]
Abstract
The presence of neutralizing SARS‐CoV‐2‐specific antibodies indicates protection against (re)infection, however, the knowledge of their long‐term kinetics is limited. This study analyzed the presence of COVID‐19‐induced antibodies in unvaccinated healthcare workers (HCWs) over the period of 1–8 months post symptom onset (SO) and explored the determinants of persisting immunoglobulin (Ig) seropositivity. Six hundred sixty‐two HCWs were interviewed for anamnestic data and tested for IgG targeting the spike protein (S1 and S2) and IgM targeting the receptor‐binding domain. A Cox regression model was used to explore potential predictors of seropositivity with respect to the time lapse between SO and serology testing. 82.9% and 44.7% of HCWs demonstrated IgG and IgM seropositivity, respectively, with a mean interval of 83 days between SARS‐CoV‐2 detection and serology testing. On average, HCWs reported seven symptoms in the acute phase lasting 20 days. IgG seropositivity rates among HCWs decreased gradually to 80%, 50%, and 35% at 3, 6, and 8 months after SO, while IgM seropositivity fell rapidly to 60%, 15%, and 0% over the same time intervals. The number of symptoms was the only predictor of persisting IgG seropositivity (odds ratio [OR] 1.096, 95% confidence interval [CI] 1.003–1.199, p = 0.043) and symptom duration a predictor of IgM seropositivity (OR 1.011, 95% CI 1.004–1.017, p = 0.002). Infection‐induced anti‐SARS‐CoV‐2 IgG rates drop to a third in seropositive participants over the course of 8 months. Symptom count and duration in the acute phase of COVID‐19 are both relevant to the subsequent kinetics of antibody responses. 60% and 35% of subjects maintain IgG seropositivity 6‐ and 8‐month post COVID‐19. Characteristics of the acute phase of COVID‐19 are relevant for antibody responses. The number of symptoms of acute COVID‐19 predicts persisting IgG seropositivity. Symptom duration predicts persisting IgM seropositivity. Anamnestic data may serve as simple predictors of seropositivity post COVID‐19.
Collapse
Affiliation(s)
- Ladislav Štěpánek
- Department of Occupational Medicine, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, I. P. Pavlova 185/6, 779 00, Olomouc, Czech Republic
| | - Magdaléna Janošíková
- Department of Occupational Medicine, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, I. P. Pavlova 185/6, 779 00, Olomouc, Czech Republic
| | - Lubomír Štěpánek
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovská 1, 120 00 Praha 2, Czech Republic
| | - Marie Nakládalová
- Department of Occupational Medicine, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, I. P. Pavlova 185/6, 779 00, Olomouc, Czech Republic
| | - Alena Boriková
- Department of Occupational Medicine, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, I. P. Pavlova 185/6, 779 00, Olomouc, Czech Republic
| |
Collapse
|
23
|
Rajamanickam A, Pavan Kumar N, Pandiaraj AN, Selvaraj N, Munisankar S, Renji RM, Venkataramani V, Murhekar M, Thangaraj JWV, Muthusamy SK, Chethrapilly Purushothaman GK, Bhatnagar T, Ponnaiah M, Ramasamy S, Velusamy S, Babu S. Characterization of memory T cell subsets and common γ-chain cytokines in convalescent COVID-19 individuals. J Leukoc Biol 2022; 112:201-212. [PMID: 35258122 PMCID: PMC9088480 DOI: 10.1002/jlb.5cova0721-392rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/30/2021] [Accepted: 01/21/2022] [Indexed: 01/08/2023] Open
Abstract
T cells are thought to be an important correlates of protection against SARS‐CoV2 infection. However, the composition of T cell subsets in convalescent individuals of SARS‐CoV2 infection has not been well studied. The authors determined the lymphocyte absolute counts, the frequency of memory T cell subsets, and the plasma levels of common γ−chain in 7 groups of COVID‐19 individuals, based on days since RT‐PCR confirmation of SARS‐CoV‐2 infection. The data show that both absolute counts and frequencies of lymphocytes as well as, the frequencies of CD4+ central and effector memory cells increased, and the frequencies of CD4+ naïve T cells, transitional memory, stem cell memory T cells, and regulatory cells decreased from Days 15–30 to Days 61–90 and plateaued thereafter. In addition, the frequencies of CD8+ central memory, effector, and terminal effector memory T cells increased, and the frequencies of CD8+ naïve cells, transitional memory, and stem cell memory T cells decreased from Days 15–30 to Days 61–90 and plateaued thereafter. The plasma levels of IL‐2, IL‐7, IL‐15, and IL‐21—common γc cytokines started decreasing from Days 15–30 till Days 151–180. Severe COVID‐19 patients exhibit decreased levels of lymphocyte counts and frequencies, higher frequencies of naïve cells, regulatory T cells, lower frequencies of central memory, effector memory, and stem cell memory, and elevated plasma levels of IL‐2, IL‐7, IL‐15, and IL‐21. Finally, there was a significant correlation between memory T cell subsets and common γc cytokines. Thus, the study provides evidence of alterations in lymphocyte counts, memory T cell subset frequencies, and common γ−chain cytokines in convalescent COVID‐19 individuals.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | | | - Arul Nancy Pandiaraj
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Nandhini Selvaraj
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Saravanan Munisankar
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Rachel Mariam Renji
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Vijayalakshmi Venkataramani
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Manoj Murhekar
- National Institute of Epidemiology (ICMR), Near Ambattur, Ayapakkam, Chennai, India
| | | | | | | | - Tarun Bhatnagar
- National Institute of Epidemiology (ICMR), Near Ambattur, Ayapakkam, Chennai, India
| | - Manickam Ponnaiah
- National Institute of Epidemiology (ICMR), Near Ambattur, Ayapakkam, Chennai, India
| | | | | | - Subash Babu
- ICMR-NIRT-International Center for Excellence in Research, Chennai, India
| |
Collapse
|
24
|
Peluso MJ, Donatelli J, Henrich TJ. Long-term immunologic effects of SARS-CoV-2 infection: leveraging translational research methodology to address emerging questions. Transl Res 2022; 241:1-12. [PMID: 34780969 PMCID: PMC8588584 DOI: 10.1016/j.trsl.2021.11.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 11/01/2022]
Abstract
The current era of COVID-19 is characterized by emerging variants of concern, waning vaccine- and natural infection-induced immunity, debate over the timing and necessity of vaccine boosting, and the emergence of post-acute sequelae of SARS-CoV-2 infection. As a result, there is an ongoing need for research to promote understanding of the immunology of both natural infection and prevention, especially as SARS-CoV-2 immunology is a rapidly changing field, with new questions arising as the pandemic continues to grow in complexity. The next phase of COVID-19 immunology research will need focus on clearer characterization of the immune processes defining acute illness, development of a better understanding of the immunologic processes driving protracted symptoms and prolonged recovery (ie, post-acute sequelae of SARS-CoV-2 infection), and a growing focus on the impact of therapeutic and prophylactic interventions on the long-term consequences of SARS-CoV-2 infection. In this review, we address what is known about the long-term immune consequences of SARS-CoV-2 infection and propose how experience studying the translational immunology of other infections might inform the approach to some of the key questions that remain.
Collapse
Affiliation(s)
- Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, California
| | - Joanna Donatelli
- Division Experimental Medicine, University of California, San Francisco, California
| | - Timothy J Henrich
- Division Experimental Medicine, University of California, San Francisco, California.
| |
Collapse
|
25
|
Zhang Y, Guo X, Li C, Kou Z, Lin L, Yao M, Pang B, Zhang X, Duan Q, Tian X, Xing Y, Jiang X. Transcriptome Analysis of Peripheral Blood Mononuclear Cells in SARS-CoV-2 Naïve and Recovered Individuals Vaccinated With Inactivated Vaccine. Front Cell Infect Microbiol 2022; 11:821828. [PMID: 35186784 PMCID: PMC8851474 DOI: 10.3389/fcimb.2021.821828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/30/2021] [Indexed: 12/13/2022] Open
Abstract
The urgent approval of the use of the inactivated COVID-19 vaccine is essential to reduce the threat and burden of the epidemic on global public health, however, our current understanding of the host immune response to inactivated vaccine remains limited. Herein, we performed serum IgG antibody detection and transcriptomics analysis on 20 SARS-CoV-2 naïve individuals who received multiple doses of inactivated vaccine and 5 SARS-CoV-2 recovered individuals who received single dose of inactivated vaccine. Our research revealed the important role of many innate immune pathways after vaccination, identified a significant correlation with the third dose of booster vaccine and proteasome-related genes, and found that SARS-CoV-2 recovered individuals can produces a strong immune response to a single dose of inactivated vaccine. These results help us understand the reaction mechanism of the host's molecular immune system to the inactivated vaccine, and provide a basis for the choice of vaccination strategy.
Collapse
Affiliation(s)
- Yuwei Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Xingyu Guo
- Infectious Disease Prevention and Control Section, School of Public Health and Health Management, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Cunbao Li
- Infectious Disease Prevention and Control Section, Lanshan Center for Disease Control and Prevention, Linyi, China
| | - Zengqiang Kou
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Lanfang Lin
- Infectious Disease Prevention and Control Section, Lanshan Center for Disease Control and Prevention, Linyi, China
| | - Mingxiao Yao
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Bo Pang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Xiaomei Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Qing Duan
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Xueying Tian
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Yufang Xing
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Xiaolin Jiang
- Ministry of Research and Education, Shandong Center for Disease Control and Prevention, Jinan, China
| |
Collapse
|
26
|
Favà A, Donadeu L, Jouve T, Gonzalez-Costello J, Lladó L, Santana C, Toapanta N, Lopez M, Pernin V, Facundo C, Cabañas NS, Thaunat O, Crespo M, Llinàs-Mallol L, Revuelta I, Sabé N, Rombauts A, Calatayud L, Ardanuy C, Esperalba J, Fernandez C, Lozano JJ, Preyer R, Strecker K, Couceiro C, García-Romero E, Cachero A, Meneghini M, Torija A, Le Quintrec M, Melilli E, Cruzado JM, Polo C, Moreso F, Crespo E, Bestard O. A comprehensive assessment of long-term SARS-CoV-2-specific adaptive immune memory in convalescent COVID-19 Solid Organ Transplant recipients. Kidney Int 2022; 101:1027-1038. [PMID: 35124011 PMCID: PMC8813192 DOI: 10.1016/j.kint.2021.12.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/25/2021] [Accepted: 12/09/2021] [Indexed: 01/08/2023]
Affiliation(s)
- Alexandre Favà
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Laura Donadeu
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Thomas Jouve
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Laura Lladó
- Liver Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Carolina Santana
- Primary Care Baix Llobregat Centre, Direcció d'Atenció Primària Costa Ponent, Equip d'Atenció Primària Gavarra, Cornellà de Llobregat, Barcelona, Spain
| | - Néstor Toapanta
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Lopez
- Lung Transplant Unit, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vincent Pernin
- Kidney Transplant Unit, Hospital de Montpellier, Montpellier, France
| | - Carme Facundo
- Kidney Transplant Unit, Nephrology Department, Fundació PuigVert, Barcelona, Spain
| | - Nuria Serra Cabañas
- Kidney Transplant Unit, Nephrology Department, Fundació PuigVert, Barcelona, Spain
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital Lyon, Hospices Civils de Lyon, Lyon, France
| | - Marta Crespo
- Kidney Transplant Unit, Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - Laura Llinàs-Mallol
- Kidney Transplant Unit, Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - Ignacio Revuelta
- Kidney Transplant Unit, Nephrology Department, Hospital Clínic, Barcelona, Spain
| | - Nuria Sabé
- Infectious Disease Department, Bellvitge University Hospital, Barcelona, Spain
| | - Alexander Rombauts
- Infectious Disease Department, Bellvitge University Hospital, Barcelona, Spain
| | - Laura Calatayud
- Microbiology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Carmen Ardanuy
- Microbiology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Juliana Esperalba
- Microbiology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Candela Fernandez
- Microbiology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan J Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Catalonia, Spain
| | | | | | - Carlos Couceiro
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | | | - Alba Cachero
- Liver Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Maria Meneghini
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Torija
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Edoardo Melilli
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Josep Maria Cruzado
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Carolina Polo
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Francesc Moreso
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Crespo
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Bestard
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
27
|
Mak WA, Koeleman JG, van der Vliet M, Keuren F, Ong DS. SARS-CoV-2 antibody and T cell responses one year after COVID-19 and the booster effect of vaccination: A prospective cohort study. J Infect 2022; 84:171-178. [PMID: 34896516 PMCID: PMC8656179 DOI: 10.1016/j.jinf.2021.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/04/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES First, to describe SARS-CoV-2 T cell and antibody responses in a prospective cohort of healthcare workers that suffered from mild to moderate COVID-19 approximately one year ago. Second, to assess COVID-19 vaccine-induced immune responses in these prior-infected individuals. METHODS SARS-CoV-2-specific T cell and anti-SARS-CoV-2-Spike-RBD immunoglobulin G (IgG) responses in blood were determined before COVID-19 vaccination with mRNA-1273, BNT162b2, Ad26.CoV2-S or ChAdOx1-S, two weeks after first vaccination, and after second vaccination. RESULTS 55 prior SARS-CoV-2 infected and seroconverted individuals were included. S1-specific T cell responses and anti-RBD IgG were detectable one year post SARS-CoV-2 infection: 24 spot-forming cells per 106 peripheral blood mononuclear cells (SFCs/106 PBMCs) after S1 stimulation and anti-RBD IgG concentration of 74 (IQR 36-158) IU/mL. Responses after the first and second vaccination were comparable with S1-specfic T cell responses of 198 (IQR 137-359) and 180 (IQR 103-347) SFCs/106 PBMCs, and IgG concentrations of 6792 (IQR 3386-15,180) and 6326 (IQR 2336-13,440) IU/mL, respectively. These responses retained up to four months after vaccination. CONCLUSIONS Both T cell and IgG responses against SARS-CoV-2 persist for up to one year after COVID-19. A second COVID-19 vaccination in prior-infected individuals did not further increase immune responses in comparison to one vaccination.
Collapse
Affiliation(s)
- Willem A. Mak
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - Johannes G.M. Koeleman
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - Marijke van der Vliet
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - Frans Keuren
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| | - David S.Y. Ong
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Universiteitsweg 100, Utrecht 3584 GC, the Netherlands,Corresponding author at: Department of Medical Microbiology and Infection Control, Franciscus Gasthuis and Vlietland, Kleiweg 500, Rotterdam 3045 PM, the Netherlands
| |
Collapse
|
28
|
Alcaide ML, Nogueira NF, Salazar AS, Montgomerie EK, Rodriguez VJ, Raccamarich PD, Barreto IT, McGaugh A, Sharkey ME, Mantero AM, Rodriguez AE, Beauchamps L, Jones DL. A Longitudinal Analysis of SARS-CoV-2 Antibody Responses Among People With HIV. Front Med (Lausanne) 2022; 9:768138. [PMID: 35330585 PMCID: PMC8940197 DOI: 10.3389/fmed.2022.768138] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background The concentration and duration of antibodies (Ab) to SARS-CoV-2 infection predicts the severity of the disease and the clinical outcomes. Older people and those with HIV have impaired immune responses, worse outcomes after SARS-CoV-2 infection, and lower antibody responses after viral infection and vaccination. This study evaluated an Ab response to SARS-CoV-2 in people with HIV (PWH) and without HIV (HIV-) and its association with age. Methods A total of 23 COVID+PWH and 21 COVID+HIV- participants were followed longitudinally for 6 months post-mild COVID-19. Immunoglobin G (IgG) and immunoglobin M (IgM) Ab responses were measured by an in-house developed ELISA. Time points and HIV status interaction were analyzed using Poisson generalized estimating equations, and correlations were analyzed using non-parametric tests. Results Median age in PWH was 55 years with 28.6% women, while in the HIV- group was 36 years with 60.9% women. The mean time from COVID-19 diagnosis to study enrollment was 16 days for PWH and 11 days for HIV-. The mean CD4+ T-cell count/μl for PWH was 772.10 (±365.21). SARS-CoV-2 IgM and IgG were detected at all time points and Ab response levels did not differ by HIV status (p > 0.05). At entry, age showed a weak direct association with IgG responses (ρ = 0.44, p < 0.05) in HIV- but did not show any association in PWH. Similar associations between age, IgG, and HIV status emerged at day 14 (T1; ρ = 0.50, p < 0.05), 3 months (T3; ρ = 0.50, p < 0.05), and 6 months visit (T4; ρ = 0.78, p < 0.05) in the HIV- group. Conclusion The Ab responses in the 6-month post-SARS-CoV-2 infection did not differ by HIV status, though a positive association was found between age and Ab response in older PWH. Results suggest that immune protection and vaccine responses are similar for PWH than for those without HIV infection.
Collapse
Affiliation(s)
- Maria L Alcaide
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nicholas F Nogueira
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ana S Salazar
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emily K Montgomerie
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Violeta J Rodriguez
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Patricia D Raccamarich
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Irma T Barreto
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Angela McGaugh
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mark E Sharkey
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alejandro M Mantero
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Allan E Rodriguez
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Laura Beauchamps
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Deborah L Jones
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
29
|
Wong LYR, Perlman S. Immune dysregulation and immunopathology induced by SARS-CoV-2 and related coronaviruses - are we our own worst enemy? Nat Rev Immunol 2022; 22:47-56. [PMID: 34837062 PMCID: PMC8617551 DOI: 10.1038/s41577-021-00656-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 02/04/2023]
Abstract
Human coronaviruses cause a wide spectrum of disease, ranging from mild common colds to acute respiratory distress syndrome and death. Three highly pathogenic human coronaviruses - severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus and SARS-CoV-2 - have illustrated the epidemic and pandemic potential of human coronaviruses, and a better understanding of their disease-causing mechanisms is urgently needed for the rational design of therapeutics. Analyses of patients have revealed marked dysregulation of the immune system in severe cases of human coronavirus infection, and there is ample evidence that aberrant immune responses to human coronaviruses are typified by impaired induction of interferons, exuberant inflammatory responses and delayed adaptive immune responses. In addition, various viral proteins have been shown to impair interferon induction and signalling and to induce inflammasome activation. This suggests that severe disease associated with human coronaviruses is mediated by both dysregulated host immune responses and active viral interference. Here we discuss our current understanding of the mechanisms involved in each of these scenarios.
Collapse
Affiliation(s)
- Lok-Yin Roy Wong
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA.
- Department of Paediatrics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
30
|
Zhao Z, Yang HS. Robust antibody response in children to acute COVID-19 infection and lasts for months. J Pediatr 2022; 240:310-313. [PMID: 34952665 PMCID: PMC8688797 DOI: 10.1016/j.jpeds.2021.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
31
|
Zhang J, Lu D, Li M, Liu M, Yao S, Zhan J, Liu WJ, Gao GF. A COVID-19 T-Cell Response Detection Method Based on a Newly Identified Human CD8 + T Cell Epitope from SARS-CoV-2 — Hubei Province, China, 2021. China CDC Wkly 2022; 4:83-87. [PMID: 35186375 PMCID: PMC8837460 DOI: 10.46234/ccdcw2021.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/01/2021] [Indexed: 12/01/2022] Open
Abstract
Introduction Similar to antibody detection, severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2)-specific T-cell response evaluation is also pivotal among the coronavirus disease 2019 (COVID-19) convalescents and the vaccinated populations. Nucleocapsid (N) protein is one of the main structural proteins of SARS-CoV-2 and can trigger T-cell responses in humans. Methods An overlapping peptide pool covering the full length of the N protein was designed, peptides with positive T-cell activating potency in COVID-19 convalescents were screened, and CD8+ T cell epitopes were further identified. The epitope was used to detect the SARS-CoV-2-specific CD8+ T cell responses in COVID-19 convalescents based in intracellular cytokine staining and tetramer staining in flow cytometry.
Results A human leukocyte antigen A (HLA-A)*1101-restricted CD8+ T cell epitope, which could stimulate the production of IFN-γ via peripheral blood mononuclear cells (PBMCs) of the convalescents was defined, and the tetramer generated with this epitope could detect SARS-CoV-2-specific T cells in the PBMCs of the convalescents. The structural investigation eliminated that the epitope was a typical HLA-A*1101-restricted T-cell epitope which was conserved among all the sarbecoviruses.
Discussion The newly identified SARS-CoV-2-derived T-cell epitope was helpful to detect the cellular immunity against different sarbecoviruses including SARS-CoV and SARS-CoV-2. This study provided an evaluation method and also a peptide candidate for the research and development of T-cell based vaccine for the virus.
Collapse
Affiliation(s)
- Jie Zhang
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dan Lu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Min Li
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Maoshun Liu
- College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sijia Yao
- College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianbo Zhan
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei, China
| | - William J. Liu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- William J. Liu,
| | - George F. Gao
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- George F. Gao,
| |
Collapse
|
32
|
Mouton W, Compagnon C, Saker K, Daniel S, Djebali S, Lacoux X, Pozzetto B, Oriol G, Laubreton D, Prieux M, Fassier J, Guibert N, Massardier‐Pilonchéry A, Alfaiate D, Berthier F, Walzer T, Marvel J, Brengel‐Pesce K, Trouiller‐Assant S. Specific detection of memory T-cells in COVID-19 patients using standardized whole-blood Interferon gammarelease assay. Eur J Immunol 2021; 51:3239-3242. [PMID: 34387859 PMCID: PMC8420580 DOI: 10.1002/eji.202149296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/30/2021] [Accepted: 08/04/2021] [Indexed: 12/25/2022]
Abstract
Antigen-specific T-cells are essential for protective immunity against SARS-CoV-2. We set up a semi-automated whole-blood Interferon-gamma release assay (WB IGRA) to monitor the T-cell response after stimulation with SARS-CoV-2 peptide pools. We report that the WB IGRA is complementary to serological assays to assess SARS-CoV-2 immunity.
Collapse
Affiliation(s)
- William Mouton
- Joint Research Unit Hospices Civils de Lyon‐bioMérieuxCivils Hospices of LyonPierre‐BéniteLyon Sud HospitalLyonFrance
- CIRI‐ International Centre for Research in Infectiology (CIRI)INSERM U1111CNRS UMR5308ENS de LyonClaude Bernard Lyon 1 UniversityLyonFrance
- Open Innovation & Partnerships (OIP)bioMérieux S.A.Marcy l'EtoileFrance
| | - Christelle Compagnon
- Joint Research Unit Hospices Civils de Lyon‐bioMérieuxCivils Hospices of LyonPierre‐BéniteLyon Sud HospitalLyonFrance
- Open Innovation & Partnerships (OIP)bioMérieux S.A.Marcy l'EtoileFrance
| | - Kahina Saker
- Joint Research Unit Hospices Civils de Lyon‐bioMérieuxCivils Hospices of LyonPierre‐BéniteLyon Sud HospitalLyonFrance
| | - Soizic Daniel
- R&D – ImmunoassaybioMérieux S.A.Marcy l'EtoileFrance
| | - Sophia Djebali
- CIRI‐ International Centre for Research in Infectiology (CIRI)INSERM U1111CNRS UMR5308ENS de LyonClaude Bernard Lyon 1 UniversityLyonFrance
| | - Xavier Lacoux
- R&D – ImmunoassaybioMérieux S.A.Marcy l'EtoileFrance
| | - Bruno Pozzetto
- GIMAP EA 3064 (Groupe Immunité des Muqueuses et Agents Pathogènes)Lyon UniversityJean Monnet UniversitySaint‐EtienneFrance
- Laboratoire des Agents Infectieux et HygièneSaint‐Etienne Univesity hospitalSaint‐EtienneFrance
| | - Guy Oriol
- Joint Research Unit Hospices Civils de Lyon‐bioMérieuxCivils Hospices of LyonPierre‐BéniteLyon Sud HospitalLyonFrance
- Open Innovation & Partnerships (OIP)bioMérieux S.A.Marcy l'EtoileFrance
| | - Daphné Laubreton
- CIRI‐ International Centre for Research in Infectiology (CIRI)INSERM U1111CNRS UMR5308ENS de LyonClaude Bernard Lyon 1 UniversityLyonFrance
| | - Margaux Prieux
- CIRI‐ International Centre for Research in Infectiology (CIRI)INSERM U1111CNRS UMR5308ENS de LyonClaude Bernard Lyon 1 UniversityLyonFrance
| | - Jean‐Baptiste Fassier
- Lyon UniversityIfsttarUMRESTTEUMR T_9405Claude Bernard Lyon 1 UniversityLyonFrance
- Occupational Health and Medicine DepartmentCivils Hospices of LyonLyonFrance
| | - Nicolas Guibert
- Lyon UniversityIfsttarUMRESTTEUMR T_9405Claude Bernard Lyon 1 UniversityLyonFrance
- Occupational Health and Medicine DepartmentCivils Hospices of LyonLyonFrance
| | - Amélie Massardier‐Pilonchéry
- Lyon UniversityIfsttarUMRESTTEUMR T_9405Claude Bernard Lyon 1 UniversityLyonFrance
- Occupational Health and Medicine DepartmentCivils Hospices of LyonLyonFrance
| | - Dulce Alfaiate
- Lyon UniversityIfsttarUMRESTTEUMR T_9405Claude Bernard Lyon 1 UniversityLyonFrance
- Occupational Health and Medicine DepartmentCivils Hospices of LyonLyonFrance
| | | | - Thierry Walzer
- CIRI‐ International Centre for Research in Infectiology (CIRI)INSERM U1111CNRS UMR5308ENS de LyonClaude Bernard Lyon 1 UniversityLyonFrance
| | - Jacqueline Marvel
- CIRI‐ International Centre for Research in Infectiology (CIRI)INSERM U1111CNRS UMR5308ENS de LyonClaude Bernard Lyon 1 UniversityLyonFrance
| | - Karen Brengel‐Pesce
- Joint Research Unit Hospices Civils de Lyon‐bioMérieuxCivils Hospices of LyonPierre‐BéniteLyon Sud HospitalLyonFrance
- Open Innovation & Partnerships (OIP)bioMérieux S.A.Marcy l'EtoileFrance
| | - Sophie Trouiller‐Assant
- Joint Research Unit Hospices Civils de Lyon‐bioMérieuxCivils Hospices of LyonPierre‐BéniteLyon Sud HospitalLyonFrance
- CIRI‐ International Centre for Research in Infectiology (CIRI)INSERM U1111CNRS UMR5308ENS de LyonClaude Bernard Lyon 1 UniversityLyonFrance
| |
Collapse
|
33
|
Persistence of SARS-CoV-2-Specific Antibodies for 13 Months after Infection. Viruses 2021; 13:v13112313. [PMID: 34835119 PMCID: PMC8622371 DOI: 10.3390/v13112313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Dynamics of antibody responses were investigated after a SARS-CoV-2 outbreak in a private company during the first wave of the pandemic. METHODS Workers of a sewing company (Lithuania) with known SARS-CoV-2 RT-PCR result during the outbreak (April 2020) were invited to participate in the study. Virus-specific IgG and IgM were monitored 2, 6 and 13 months after the outbreak via rapid IgG/IgM serological test and SARS-CoV-2 S protein-specific IgG ELISA. RESULTS Six months after the outbreak, 95% (CI 86-99%) of 59 previously infected individuals had virus-specific antibodies irrespective of the severity of infection. One-third of seropositive individuals had virus-specific IgM along with IgG indicating that IgM may persist for 6 months. Serological testing 13 months after the outbreak included 47 recovered individuals that remained non-vaccinated despite a wide accessibility of COVID-19 vaccines. The seropositivity rate was 83% (CI 69-91%) excluding one case of confirmed asymptomatic reinfection in this group. Between months 6 and 13, IgG levels either declined or remained stable in 31 individual and increased in 7 individuals possibly indicating an exposure to SARS-CoV-2 during the second wave of the pandemic. CONCLUSIONS Detectable levels of SARS-CoV-2-specific antibodies persist up to 13 months after infection for the majority of the cases.
Collapse
|
34
|
Abela IA, Pasin C, Schwarzmüller M, Epp S, Sickmann ME, Schanz MM, Rusert P, Weber J, Schmutz S, Audigé A, Maliqi L, Hunziker A, Hesselman MC, Niklaus CR, Gottschalk J, Schindler E, Wepf A, Karrer U, Wolfensberger A, Rampini SK, Meyer Sauteur PM, Berger C, Huber M, Böni J, Braun DL, Marconato M, Manz MG, Frey BM, Günthard HF, Kouyos RD, Trkola A. Multifactorial seroprofiling dissects the contribution of pre-existing human coronaviruses responses to SARS-CoV-2 immunity. Nat Commun 2021; 12:6703. [PMID: 34795285 PMCID: PMC8602384 DOI: 10.1038/s41467-021-27040-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Determination of SARS-CoV-2 antibody responses in the context of pre-existing immunity to circulating human coronavirus (HCoV) is critical for understanding protective immunity. Here we perform a multifactorial analysis of SARS-CoV-2 and HCoV antibody responses in pre-pandemic (N = 825) and SARS-CoV-2-infected donors (N = 389) using a custom-designed multiplex ABCORA assay. ABCORA seroprofiling, when combined with computational modeling, enables accurate definition of SARS-CoV-2 seroconversion and prediction of neutralization activity, and reveals intriguing interrelations with HCoV immunity. Specifically, higher HCoV antibody levels in SARS-CoV-2-negative donors suggest that pre-existing HCoV immunity may provide protection against SARS-CoV-2 acquisition. In those infected, higher HCoV activity is associated with elevated SARS-CoV-2 responses, indicating cross-stimulation. Most importantly, HCoV immunity may impact disease severity, as patients with high HCoV reactivity are less likely to require hospitalization. Collectively, our results suggest that HCoV immunity may promote rapid development of SARS-CoV-2-specific immunity, thereby underscoring the importance of exploring cross-protective responses for comprehensive coronavirus prevention.
Collapse
Affiliation(s)
- Irene A Abela
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Chloé Pasin
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Selina Epp
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Michèle E Sickmann
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Merle M Schanz
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Peter Rusert
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Jacqueline Weber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Stefan Schmutz
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Annette Audigé
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Liridona Maliqi
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Annika Hunziker
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Maria C Hesselman
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Cyrille R Niklaus
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | | | | | - Alexander Wepf
- Institute of Laboratory Medicine, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Urs Karrer
- Department of Medicine, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Aline Wolfensberger
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Silvana K Rampini
- Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Patrick M Meyer Sauteur
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Christoph Berger
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Jürg Böni
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Dominique L Braun
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Maddalena Marconato
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Beat M Frey
- Blood Transfusion Service Zurich, Zurich, Switzerland
| | - Huldrych F Günthard
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland.
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.
| | - Roger D Kouyos
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland.
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
Marklund E, Leach S, Nyström K, Lundgren A, Liljeqvist JÅ, Nilsson S, Yilmaz A, Andersson LM, Bemark M, Gisslén M. Longitudinal Follow Up of Immune Responses to SARS-CoV-2 in Health Care Workers in Sweden With Several Different Commercial IgG-Assays, Measurement of Neutralizing Antibodies and CD4 + T-Cell Responses. Front Immunol 2021; 12:750448. [PMID: 34795668 PMCID: PMC8593002 DOI: 10.3389/fimmu.2021.750448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/11/2021] [Indexed: 01/21/2023] Open
Abstract
Background The risk of SARS-CoV-2 infection among health care workers (HCWs) is a concern, but studies that conclusively determine whether HCWs are over-represented remain limited. Furthermore, methods used to confirm past infection vary and the immunological response after mild COVID-19 is still not well defined. Method 314 HCWs were recruited from a Swedish Infectious Diseases clinic caring for COVID-19 patients. IgG antibodies were measured using two commercial assays (Abbot Architect nucleocapsid (N)-assay and YHLO iFlash-1800 N and spike (S)-assays) at five time-points, from March 2020 to January 2021, covering two pandemic waves. Seroprevalence was assessed in matched blood donors at three time-points. More extensive analyses were performed in 190 HCWs in September/October 2020, including two additional IgG-assays (DiaSorin LiaisonXL S1/S2 and Abbot Architect receptor-binding domain (RBD)-assays), neutralizing antibodies (NAbs), and CD4+ T-cell reactivity using an in-house developed in vitro whole-blood assay based on flow cytometric detection of activated cells after stimulation with Spike S1-subunit or Spike, Membrane and Nucleocapsid (SMN) overlapping peptide pools. Findings Seroprevalence was higher among HCWs compared to sex and age-matched blood donors at all time-points. Seropositivity increased from 6.4% to 16.3% among HCWs between May 2020 and January 2021, compared to 3.6% to 11.9% among blood donors. We found significant correlations and high levels of agreement between NAbs and all four commercial IgG-assays. At 200-300 days post PCR-verified infection, there was a wide variation in sensitivity between the commercial IgG-assays, ranging from <30% in the N-assay to >90% in the RBD-assay. There was only moderate agreement between NAbs and CD4+ T-cell reactivity to S1 or SMN. Pre-existing CD4+ T-cell reactivity was present in similar proportions among HCW who subsequently became infected and those that did not. Conclusions HCWs in COVID-19 patient care in Sweden have been infected with SARS-CoV-2 at a higher rate compared to blood donors. We demonstrate substantial variation between different IgG-assays and propose that multiple serological targets should be used to verify past infection. Our data suggest that CD4+ T-cell reactivity is not a suitable measure of past infection and does not reliably indicate protection from infection in naive individuals.
Collapse
Affiliation(s)
- Emelie Marklund
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Susannah Leach
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Pharmacology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kristina Nyström
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Lundgren
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan-Åke Liljeqvist
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Aylin Yilmaz
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars-Magnus Andersson
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
36
|
Ha B, Jadhao S, Hussaini L, Gibson T, Stephens K, Salazar L, Ciric C, Taylor M, Rouphael N, Edupuganti S, Rostad CA, Tompkins SM, Anderson EJ, Anderson LJ. Evaluation of a SARS-CoV-2 Capture IgM Antibody Assay in Convalescent Sera. Microbiol Spectr 2021; 9:e0045821. [PMID: 34494855 PMCID: PMC8557898 DOI: 10.1128/spectrum.00458-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/08/2021] [Indexed: 01/19/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for a global pandemic with over 152 million cases and 3.19 million deaths reported by early May 2021. Understanding the serological response to SARS-CoV-2 is critical to determining the burden of infection and disease (coronavirus disease 2019 [COVID-19]) and transmission dynamics. We developed a capture IgM assay because it should have better sensitivity and specificity than the commonly used indirect assay. Here, we report the development and performance of a capture IgM enzyme-linked immunosorbent assay (ELISA) and a companion indirect IgG ELISA for the spike (S) and nucleocapsid (N) proteins and the receptor-binding domain (RBD) of S. We found that among the IgM ELISAs, the S ELISA was positive in 76% of 55 serum samples from SARS-CoV-2 PCR-positive patients, the RBD ELISA was positive in 55% of samples, and the N ELISA was positive in 15% of samples. The companion indirect IgG ELISAs were positive for S in 89% of the 55 serum samples, RBD in 78%, and N in 85%. While the specificities for IgM RBD, S, and N ELISAs and IgG S and RBD ELISAs were 97% to 100%, the specificity of the N IgG ELISA was lower (89%). RBD-specific IgM antibodies became undetectable by 3 to 6 months, and S IgM reached low levels at 6 months. The corresponding IgG S, RBD, and N antibodies persisted with some decreases in levels over this time period. These capture IgM ELISAs and the companion indirect IgG ELISAs should enhance serologic studies of SARS-CoV-2 infections. IMPORTANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has inflicted tremendous loss of lives, overwhelmed health care systems, and disrupted all aspects of life worldwide since its emergence in Wuhan, China, in December 2019. Detecting current and past infection by PCR or serology is important to understanding and controlling SARS-CoV-2. With increasing prevalence of past infection or vaccination, IgG antibodies are less helpful in diagnosing a current infection. IgM antibodies indicate a more recent infection and can supplement PCR diagnosis. We report an alternative method, capture IgM, to detect serum IgM antibodies, which should be more sensitive and specific than most currently used methods. We describe this capture IgM assay and a companion indirect IgG assay for the SARS-CoV-2 spike (S), nucleocapsid (N), and receptor-binding domain (RBD) proteins. These assays can add value to diagnostic and serologic studies of coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Binh Ha
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Samadhan Jadhao
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Laila Hussaini
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Theda Gibson
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Kathy Stephens
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Luis Salazar
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Caroline Ciric
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Meg Taylor
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - Nadine Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Srilatha Edupuganti
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christina A. Rostad
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| | - S. Mark Tompkins
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Emory-UGA Centers of Excellence for Influenza Research and Surveillance (CEIRS), Athens, Georgia, USA
| | - Evan J. Anderson
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Larry J. Anderson
- Division of Pediatric Infectious Diseases, Emory University School of Medicine and Children’s Health Care of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
37
|
Mild SARS-CoV-2 Illness Is Not Associated with Reinfections and Provides Persistent Spike, Nucleocapsid, and Virus-Neutralizing Antibodies. Microbiol Spectr 2021; 9:e0008721. [PMID: 34468184 PMCID: PMC8557889 DOI: 10.1128/spectrum.00087-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Uncertainty exists whether mild COVID-19 confers immunity to reinfection. Questions also remain regarding the persistence of antibodies against SARS-CoV-2 after mild infection. We prospectively followed at-risk individuals with and without SARS-CoV-2 for reinfection and monitored the spike and nucleocapsid antibodies. This prospective cohort study was conducted over two visits, 3 to 6 months apart, between May 2020 and February 2021. Adults with and without COVID-19, verified by FDA EUA-approved SARS-CoV-2 RT-PCR assays, were screened for spike and nucleocapsid antibody responses using FDA EUA-approved immunoassays and for pseudoviral neutralization activity. The subjects were monitored for symptoms, exposure to COVID-19, COVID-19 testing, seroconversion, reinfection, and vaccination. A total of 653 subjects enrolled; 129 (20%) had a history of COVID-19 verified by RT-PCR at enrollment. Most had mild disease, with only three requiring hospitalization. No initially seropositive subjects experienced a subsequent COVID-19 infection during the follow-up versus 15 infections among initially seronegative subjects (infection rates of 0.00 versus 2.05 per 10,000 days at risk [P = 0.0485]). In all, 90% of SARS-CoV-2-positive subjects produced spike and nucleocapsid responses, and all but one of these had persistent antibody levels at follow-up. Pseudoviral neutralization activity was widespread among participants, did not decrease over time, and correlated with clinical antibody assays. Reinfection with SARS-CoV-2 was not observed among individuals with mild clinical COVID-19, while infections continued in a group without known prior infection. Spike and nucleocapsid COVID-19 antibodies were associated with almost all infections and persisted at stable levels for the study duration. IMPORTANCE This article demonstrates that people who have mild COVID-19 illnesses and produce antibodies are protected from reinfection for up to 6 months afterward. The antibodies that people produce in this situation are stable for up to 6 months as well. Clinical antibody assays correlate well with evidence of antibody-related viral neutralization activity.
Collapse
|
38
|
Abstract
Cellular immunity may be involved in organ damage and rehabilitation in patients with coronavirus disease 2019 (COVID-19). We aimed to delineate immunological features of COVID-19 patients with pulmonary sequelae (PS) 1 year after discharge. Fifty COVID-19 survivors were recruited and classified according to radiological characteristics, including 24 patients with PS and 26 patients without PS. Phenotypic and functional characteristics of immune cells were evaluated by multiparametric flow cytometry. Patients with PS had an increased proportion of natural killer (NK) cells and a lower percentage of B cells than patients without PS. Phenotypic and functional features of T cells in patients with PS were predominated by the accumulation of CD4-positive (CD4+) T cells secreting interleukin 17A (IL-17A), short-lived effector-like CD8+ T cells (CD27-negative [CD27−] CD62L−), and senescent T cells with excessive secretion of granzyme B/perforin/interferon gamma (IFN-γ). NK cells were characterized by the excessive secretion of granzyme B and perforin and the downregulation of NKP30 and NKP46; highly activated NKT and γδ T cells exhibited NKP30 and TIM-3 upregulation and NKB1 downregulation in patients with PS. However, immunosuppressive cells were comparable between the two groups. The interrelationship of immune cells in COVID-19 was intrinsically identified, whereby T cells secreting IL-2, IL-4, and IL-17A were enriched among CD28+ and CD57− cells and cells secreting perforin/granzyme B/IFN-γ/tumor necrosis factor alpha (TNF-α)-expressed markers of terminal differentiation. CD57+ NK cells, CD4+Perforin+ T cells, and CD8+ CD27+ CD62L+ T cells were identified as the independent predictors for residual lesions. Overall, our findings unveil the profound imbalance of immune landscape that may correlate with organ damage and rehabilitation in COVID-19.
Collapse
|
39
|
Wang B, Goh YS, Prince T, Ngoh EZX, Salleh SNM, Hor PX, Loh CY, Fong SW, Hartley C, Tan SY, Young BE, Leo YS, Lye DC, Maurer-Stroh S, Ng LFP, Hiscox JA, Renia L, Wang CI. Resistance of SARS-CoV-2 variants to neutralization by convalescent plasma from early COVID-19 outbreak in Singapore. NPJ Vaccines 2021; 6:125. [PMID: 34697298 PMCID: PMC8546091 DOI: 10.1038/s41541-021-00389-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
The rapid spreading of SARS-CoV-2 variants B.1.1.7 originated from the United Kingdom and B.1.351 from South Africa has contributed to the second wave of COVID-19 cases in the respective countries and also around the world. In this study, we employed advanced biochemical and virological methodologies to evaluate the impact of Spike mutations of these strains on the degree of protection afforded by humoral immune responses following natural infection of the ancestral SARS-CoV-2 strain during the early stages of the outbreak. We found that antibody-mediated neutralization activity was partially reduced for B.1.1.7 variant and significantly attenuated for the B.1.351 strain. We also found that mutations outside the receptor-binding domain (RBD) can strongly influence antibody binding and neutralization, cautioning the use of solely RBD mutations in evaluating vaccine efficacy. These findings highlight an urgent need to develop new SARS-CoV-2 vaccines that are not based exclusively on the ancestral SARS-CoV-2 Spike gene sequence.
Collapse
Affiliation(s)
- Bei Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yun Shan Goh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Tessa Prince
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
| | - Eve Zi Xian Ngoh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siti Nazihah Mohd Salleh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei Xiang Hor
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chiew Yee Loh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siew Wai Fong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Catherine Hartley
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Seow-Yen Tan
- Department of Infectious Diseases, Changi General Hospital, Singapore, Singapore
| | - Barnaby Edward Young
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yee-Sin Leo
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - David C Lye
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Sebastian Maurer-Stroh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Bioinformatics Institute, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Julian A Hiscox
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
40
|
Luo H, Camilleri D, Garitaonandia I, Djumanov D, Chen T, Lorch U, Täubel J, Wang D. Kinetics of anti-SARS-CoV-2 IgG antibody levels and potential influential factors in subjects with COVID-19: A 11-month follow-up study. Diagn Microbiol Infect Dis 2021; 101:115537. [PMID: 34619569 PMCID: PMC8428032 DOI: 10.1016/j.diagmicrobio.2021.115537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022]
Abstract
We aim to study kinetics of anti-SARS-CoV-2 IgG antibody levels in subjects with COVID-19 for up to 11 months and the potential influential factors. The study was a prospective longitudinal study. The analyses were based on 77 serum/plasma samples with a mean of 4 samples per participant (range 1 – 18) in 20 participants with at least one positive Polymerase Chain Reaction testing result from 19 March 2020 up to 10 February 2021. Among the subjects (median age 34.5 years, 65% male), IgG level declined with the follow-up time (per month; geometric mean ratio [GMR] 0.73; 95% CI, 0.72 – 0.74). In a small sample of subjects from the general population with COVID-19, IgG levels declined non-linearly from month 2 to 11 with individual heterogeneity in quantity and changing speed and may be associated with gender, race and the loss of smell and taste.
Collapse
Affiliation(s)
- Huanyuan Luo
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Ibon Garitaonandia
- Richmond Research Institute, St George's University of London, London, UK
| | - Dilshat Djumanov
- Department of Data Science, Richmond Pharmacology Ltd, London, UK
| | - Tao Chen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Ulrike Lorch
- Department of Data Science, Richmond Pharmacology Ltd, London, UK
| | - Jörg Täubel
- Department of Data Science, Richmond Pharmacology Ltd, London, UK; Richmond Research Institute, St George's University of London, London, UK.
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
41
|
Rha MS, Shin EC. Activation or exhaustion of CD8 + T cells in patients with COVID-19. Cell Mol Immunol 2021; 18:2325-2333. [PMID: 34413488 PMCID: PMC8374113 DOI: 10.1038/s41423-021-00750-4] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
In addition to CD4+ T cells and neutralizing antibodies, CD8+ T cells contribute to protective immune responses against SARS-CoV-2 in patients with coronavirus disease 2019 (COVID-19), an ongoing pandemic disease. In patients with COVID-19, CD8+ T cells exhibiting activated phenotypes are commonly observed, although the absolute number of CD8+ T cells is decreased. In addition, several studies have reported an upregulation of inhibitory immune checkpoint receptors, such as PD-1, and the expression of exhaustion-associated gene signatures in CD8+ T cells from patients with COVID-19. However, whether CD8+ T cells are truly exhausted during COVID-19 has been a controversial issue. In the present review, we summarize the current understanding of CD8+ T-cell exhaustion and describe the available knowledge on the phenotypes and functions of CD8+ T cells in the context of activation and exhaustion. We also summarize recent reports regarding phenotypical and functional analyses of SARS-CoV-2-specific CD8+ T cells and discuss long-term SARS-CoV-2-specific CD8+ T-cell memory.
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, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of 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, Republic of Korea.
- The Center for Epidemic Preparedness, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
42
|
A bioluminescent and homogeneous SARS-CoV-2 spike RBD and hACE2 interaction assay for antiviral screening and monitoring patient neutralizing antibody levels. Sci Rep 2021; 11:18428. [PMID: 34531417 PMCID: PMC8445915 DOI: 10.1038/s41598-021-97330-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/24/2021] [Indexed: 01/07/2023] Open
Abstract
Here we describe a homogeneous bioluminescent immunoassay based on the interaction between Fc-tagged SARS-CoV-2 Spike RBD and human ACE2, and its detection by secondary antibodies labeled with NanoLuc luciferase fragments LgBit and SmBit. The assay utility for the discovery of novel inhibitors was demonstrated with a panel of anti-RBD antibodies, ACE2-derived miniproteins and soluble ACE2. Studying the effect of RBD mutations on ACE2 binding showed that the N501Y mutation increased RBD apparent affinity toward ACE2 tenfold that resulted in escaping inhibition by some anti-RBD antibodies. In contrast, while E484K mutation did not highly change the binding affinity, it still escaped antibody inhibition likely due to changes in the epitope recognized by the antibody. Also, neutralizing antibodies (NAbs) from COVID-19 positive samples from two distinct regions (USA and Brazil) were successfully detected and the results further suggest the persistence of NAbs for at least 6 months post symptom onset. Finally, sera from vaccinated individuals were tested for NAbs and showed varying neutralizing activity after first and second doses, suggesting the assay can be used to assess immunity of vaccinated populations. Our results demonstrate the broad utility and ease of use of this methodology both for drug discovery and clinical research applications.
Collapse
|
43
|
Zhao Y, Yang C, An X, Xiong Y, Shang Y, He J, Qiu Y, Zhang N, Huang L, Jia J, Xu Q, Zhang L, Zhao J, Pei G, Luo H, Wang J, Li Q, Gao Y, Xu A. Follow-up study on COVID-19 survivors one year after discharge from hospital. Int J Infect Dis 2021; 112:173-182. [PMID: 34520845 PMCID: PMC8434916 DOI: 10.1016/j.ijid.2021.09.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 01/25/2023] Open
Abstract
Objective To evaluate the long-term consequences of COVID-19 survivors one year after recovery, and to identify the risk factors associated with abnormal patterns in chest imaging manifestations or impaired lung function. Methods COVID-19 patients were recruited and prospectively followed up with symptoms, health-related quality of life, psychological questionnaires, 6-minute walking test, chest computed tomography (CT), pulmonary function tests, and blood tests. Multivariable logistic regression models were used to evaluate the association between the clinical characteristics and chest CT abnormalities or pulmonary function. Results Ninety-four patients with COVID-19 were recruited between January 16 and February 6, 2021. Muscle fatigue and insomnia were the most common symptoms. Chest CT scans were abnormal in 71.28% of participants. The results of multivariable regression showed an increased odds in age. Ten patients had diffusing capacity of the lung for carbon monoxide (DLCO) impairment. Urea nitrogen concentration on admission was significantly associated with impaired DLCO. IgG levels and neutralizing activity were significantly lower compared with those in the early phase. Conclusions One year after hospitalization for COVID-19, a cohort of survivors were mainly troubled with muscle fatigue and insomnia. Pulmonary structural abnormalities and pulmonary diffusion capacities were highly prevalent in surviving COVID-19 patients. It is necessary to intervene in the main target population for long-term recovery.
Collapse
Affiliation(s)
- Yumiao Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Chunxia Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Xiaocai An
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Yajun Xiong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Yaomin Shang
- Department of Respiration, Henan Provincial Chest Hospital, Zhengzhou 450003, China
| | - Jiarong He
- Department of Respiration, The Fifth People's Hospital of Xinyang, Xinyang 464000, China
| | - Yan Qiu
- Department of Respiration, The Fifth People's Hospital of Xinyang, Xinyang 464000, China
| | - Ning Zhang
- Clinical Laboratory, The Fifth People's Hospital of Xinyang, Xinyang 464000, China
| | - Lisha Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Junli Jia
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Qinfu Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Long Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Junjie Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China
| | - Guangzhong Pei
- Department of Neurology, Guangshan People's Hospital. Xinyang 465400, China
| | - Hong Luo
- Department of Respiratory and Critical Care Medicine, Guangshan People's Hospital, Xinyang 465400, China
| | - Jun Wang
- Department of Infectious Diseases, Luoshan People's Hospital, Xinyang 465400, China
| | - Qingquan Li
- Department of Respiratory and Critical Care Medicine, Xixian People's Hospital, Xinyang 464200, China.
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Aiguo Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450051, China.
| |
Collapse
|
44
|
Comelli A, Consonni D, Lombardi A, Viero G, Oggioni M, Bono P, Uceda Renteria SC, Ceriotti F, Mangioni D, Muscatello A, Piatti A, Pesatori AC, Castaldi S, Riboldi L, Bandera A, Gori A. Nasopharyngeal Testing among Healthcare Workers (HCWs) of a Large University Hospital in Milan, Italy during Two Epidemic Waves of COVID-19. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:8748. [PMID: 34444497 PMCID: PMC8392643 DOI: 10.3390/ijerph18168748] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND since October 2020, a second SARS-CoV-2 epidemic wave has hit Italy. We investigate the frequency of positive nasopharyngeal swabs among HCWs during the two waves and the association with occupation and demographic characteristics. METHODS this is a retrospective, observational study conducted in a large university hospital in Milan, Northern Italy. We defined two epidemic waves: 1st (February 2020-July 2020) and 2nd (August 2020-January 2021). Occupational and demographic characteristics of HCWs who underwent nasopharyngeal swabs for SARS-CoV-2 were collected. RESULTS in the 1st wave, 242 positive subjects (7.2%) were found among 3378 HCWs, whereas in the 2nd wave, the positive subjects were 545 out of 4465 (12.2%). In both epidemic waves positive NPSs were more frequent among HCWs with health-related tasks and lower among students (p < 0.001). However, in the 2nd wave, workers engaged in non-health-related tasks had a peak of 20.7% positivity. Among 160 positive HCWs in the 1st wave who were tested again in the 2nd wave, the rate of reinfection based on SARS-CoV2 RNA cycle quantification value was 0.6%. CONCLUSIONS during the 2nd epidemic wave, we confirmed a significant impact of COVID-19 among HCWs. The rise of infection rate among HCWs seems to reflect the increasing spread of SARS-CoV-2 among the overall population.
Collapse
Affiliation(s)
- Agnese Comelli
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.L.); (G.V.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Dario Consonni
- Epidemiology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (D.C.); (A.C.P.)
| | - Andrea Lombardi
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.L.); (G.V.); (D.M.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Giulia Viero
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.L.); (G.V.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Massimo Oggioni
- Clinical Laboratory, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.O.); (P.B.); (S.C.U.R.); (F.C.)
| | - Patrizia Bono
- Clinical Laboratory, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.O.); (P.B.); (S.C.U.R.); (F.C.)
| | - Sara Colonia Uceda Renteria
- Clinical Laboratory, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.O.); (P.B.); (S.C.U.R.); (F.C.)
| | - Ferruccio Ceriotti
- Clinical Laboratory, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.O.); (P.B.); (S.C.U.R.); (F.C.)
| | - Davide Mangioni
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.L.); (G.V.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Antonio Muscatello
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.L.); (G.V.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Alessandra Piatti
- Medical Direction, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Angela Cecilia Pesatori
- Epidemiology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (D.C.); (A.C.P.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Silvana Castaldi
- Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy;
- Quality Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Luciano Riboldi
- Occupational Health Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.L.); (G.V.); (D.M.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, 20122 Milan, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.L.); (G.V.); (D.M.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, 20122 Milan, Italy
| |
Collapse
|
45
|
Borena W, Bánki Z, Bates K, Winner H, Riepler L, Rössler A, Pipperger L, Theurl I, Falkensammer B, Ulmer H, Walser A, Pichler D, Baumgartner M, Schönherr S, Forer L, Knabl L, Würzner R, von Laer D, Paetzold J, Kimpel J. Persistence of immunity to SARS-CoV-2 over time in the ski resort Ischgl. EBioMedicine 2021; 70:103534. [PMID: 34392147 PMCID: PMC8358264 DOI: 10.1016/j.ebiom.2021.103534] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 11/22/2022] Open
Abstract
Background In early March 2020, a SARS-CoV-2 outbreak in the ski resort Ischgl in Austria triggered the spread of SARS-CoV-2 throughout Austria and Northern Europe. In a previous study, we found that the seroprevalence in the adult population of Ischgl had reached 45% by the end of April, representing an exceptionally high level of local seropositivity in Europe. We performed a follow-up study in Ischgl, which is the first to show persistence of immunity and protection against SARS-CoV-2 and some of its variants at a community level. Methods Of the 1259 adults that participated in the baseline study, 801 have been included in the follow-up in November 2020. The study involved the analysis of binding and neutralizing antibodies and T cell responses. In addition, the incidence of SARS-CoV-2 and its variants in Ischgl was compared to the incidence in similar municipalities in Tyrol until April 2021. Findings For the 801 individuals that participated in both studies, the seroprevalence declined from 51.4% (95% confidence interval (CI) 47.9-54.9) to 45.4% (95% CI 42.0-49.0). Median antibody concentrations dropped considerably (5.345, 95% CI 4.833 - 6.123 to 2.298, 95% CI 2.141 - 2.527) but antibody avidity increased (17.02, 95% CI 16.49 - 17.94 to 42.46, 95% CI 41.06 - 46.26). Only one person had lost detectable antibodies and T cell responses. In parallel to this persistent immunity, we observed that Ischgl was relatively spared, compared to similar municipalities, from the prominent second COVID-19 wave that hit Austria in November 2020. In addition, we used sequencing data to show that the local immunity acquired from wild-type infections also helped to curb infections from variants of SARS-CoV-2 which spread in Austria since January 2021. Interpretation The relatively high level of seroprevalence (40-45%) in Ischgl persisted and might have been associated with the observed protection of Ischgl residents against virus infection during the second COVID-19 wave as well as against variant spread in 2021. Funding Funding was provided by the government of Tyrol and the FWF Austrian Science Fund.
Collapse
Affiliation(s)
- Wegene Borena
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Zoltán Bánki
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Katie Bates
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Austria
| | - Hannes Winner
- University of Salzburg, Department of Economics, Residenzplatz 9, Salzburg A-5010, Austria
| | - Lydia Riepler
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Annika Rössler
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Lisa Pipperger
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Igor Theurl
- Labor Dr. Theurl, Franz-Fischerstr.7b, Innsbruck, Austria
| | - Barbara Falkensammer
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Hanno Ulmer
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Austria
| | | | - Daniel Pichler
- Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Matthias Baumgartner
- Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Innsbruck 6020, Austria
| | | | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Austria
| | - Ludwig Knabl
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Reinhard Würzner
- Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Dorothee von Laer
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria
| | - Jörg Paetzold
- University of Salzburg, Department of Economics, Residenzplatz 9, Salzburg A-5010, Austria.
| | - Janine Kimpel
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Peter-Mayr-Str. 4b, Innsbruck 6020, Austria.
| |
Collapse
|
46
|
Ibarrondo FJ, Hofmann C, Fulcher JA, Goodman-Meza D, Mu W, Hausner MA, Ali A, Balamurugan A, Taus E, Elliott J, Krogstad P, Tobin NH, Ferbas KG, Kitchen SG, Aldrovandi GM, Rimoin AW, Yang OO. Primary, Recall, and Decay Kinetics of SARS-CoV-2 Vaccine Antibody Responses. ACS NANO 2021; 15:11180-11191. [PMID: 34159781 DOI: 10.1021/acsnano.1c03972] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Studies of two SARS-CoV-2 mRNA vaccines suggested that they yield ∼95% protection from symptomatic infection at least short-term, but important clinical questions remain. It is unclear how vaccine-induced antibody levels quantitatively compare to the wide spectrum induced by natural SARS-CoV-2 infection. Vaccine response kinetics and magnitudes in persons with prior COVID-19 compared to virus-naı̈ve persons are not well-defined. The relative stability of vaccine-induced versus infection-induced antibody levels is unclear. We addressed these issues with longitudinal assessments of vaccinees with and without prior SARS-CoV-2 infection using quantitative enzyme-linked immunosorbent assay (ELISA) of anti-RBD antibodies. SARS-CoV-2-naı̈ve individuals achieved levels similar to mild natural infection after the first vaccination; a second dose generated levels approaching severe natural infection. In persons with prior COVID-19, one dose boosted levels to the high end of severe natural infection even in those who never had robust responses from infection, increasing no further after the second dose. Antiviral neutralizing assessments using a spike-pseudovirus assay revealed that virus-naı̈ve vaccinees did not develop physiologic neutralizing potency until the second dose, while previously infected persons exhibited maximal neutralization after one dose. Finally, antibodies from vaccination waned similarly to natural infection, resulting in an average of ∼90% loss within 90 days. In summary, our findings suggest that two doses are important for quantity and quality of humoral immunity in SARS-CoV-2-naı̈ve persons, while a single dose has maximal effects in those with past infection. Antibodies from vaccination wane with kinetics very similar to that seen after mild natural infection; booster vaccinations will likely be required.
Collapse
Affiliation(s)
- F Javier Ibarrondo
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Christian Hofmann
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Jennifer A Fulcher
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - David Goodman-Meza
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - William Mu
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Mary Ann Hausner
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Ayub Ali
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Arumugam Balamurugan
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Ellie Taus
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Julie Elliott
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Paul Krogstad
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Nicole H Tobin
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Kathie G Ferbas
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Scott G Kitchen
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Grace M Aldrovandi
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Anne W Rimoin
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Otto O Yang
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
47
|
Unique autoantibody prevalence in long-term recovered SARS-CoV-2-infected individuals. J Autoimmun 2021; 122:102682. [PMID: 34214763 PMCID: PMC8214939 DOI: 10.1016/j.jaut.2021.102682] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/24/2022]
Abstract
The variability in resolution of SARS-CoV-2-infections between individuals neither is comprehended, nor are the long-term immunological consequences. To assess the long-term impact of a SARS-CoV-2-infection on the immune system, we conducted a prospective study of 80 acute and former SARS-CoV-2 infected individuals and 39 unexposed donors to evaluate autoantibody responses and immune composition. Autoantibody levels against cyclic citrullinated peptide (CCP), a specific predictor for rheumatoid arthritis (RA), were significantly (p = 0.035) elevated in convalescents only, whereas both acute COVID-19 patients and long-term convalescents showed critically increased levels of anti-tissue transglutaminase (TG), a specific predictor of celiac disease (CD) (p = 0.002). Both, anti-CCP and anti-TG antibody levels were still detectable after 4–8 months post infection. Anti-TG antibodies occurred predominantly in aged patients in a context of a post-SARS-CoV-2-specific immune composition (R2 = 0.31; p = 0.044). This study shows that increased anti-CCP and anti-TG autoantibody levels can remain long-term after recovering even from mildly experienced COVID-19. The inter-relationship of the lung as viral entry side and RA- and CD-associated autoimmunity indicates that a SARS-CoV-2-infection could be a relevant environmental factor in their pathogenesis.
Collapse
|
48
|
De Giorgi V, West KA, Henning AN, Chen LN, Holbrook MR, Gross R, Liang J, Postnikova E, Trenbeath J, Pogue S, Scinto T, Alter HJ, Cantilena CC. Naturally acquired SARS-CoV-2 immunity persists for up to 11 months following infection. J Infect Dis 2021; 224:1294-1304. [PMID: 34089610 PMCID: PMC8195007 DOI: 10.1093/infdis/jiab295] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/04/2021] [Indexed: 11/23/2022] Open
Abstract
Background Characterizing the kinetics of the antibody response to severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) is of critical importance to developing strategies that may mitigate the public health burden of coronavirus disease 2019 (COVID-19). We conducted a prospective, longitudinal analysis of COVID-19 convalescent plasma donors at multiple time points over an 11-month period to determine how circulating antibody levels change over time following natural infection. Methods From April 2020 to February 2021, we enrolled 228 donors. At each study visit, subjects either donated plasma or had study samples drawn only. Anti–SARS-CoV-2 donor testing was performed using the VITROS Anti–SARS-CoV-2 Total and IgG assays and an in-house fluorescence reduction neutralization assay. Results Anti–SARS-CoV-2 antibodies were identified in 97% of COVID-19 convalescent donors at initial presentation. In follow-up analyses, of 116 donors presenting at repeat time points, 91.4% had detectable IgG levels up to 11 months after symptom recovery, while 63% had detectable neutralizing titers; however, 25% of donors had neutralizing levels that dropped to an undetectable titer over time. Conclusions Our data suggest that immunological memory is acquired in most individuals infected with SARS-CoV-2 and is sustained in a majority of patients for up to 11 months after recovery. Clinical Trials Registration. NCT04360278.
Collapse
Affiliation(s)
- Valeria De Giorgi
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Kamille A West
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Amanda N Henning
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Leonard N Chen
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Michael R Holbrook
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Robin Gross
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Janie Liang
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Elena Postnikova
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Joni Trenbeath
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Sarah Pogue
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Tania Scinto
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Harvey J Alter
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| | - Cathy Conry Cantilena
- National Institutes of Health (NIH), Clinical Center, Department of Transfusion Medicine, Bethesda, MD 20892, USA
| |
Collapse
|
49
|
Cromer D, Juno JA, Khoury D, Reynaldi A, Wheatley AK, Kent SJ, Davenport MP. Prospects for durable immune control of SARS-CoV-2 and prevention of reinfection. Nat Rev Immunol 2021; 21:395-404. [PMID: 33927374 PMCID: PMC8082486 DOI: 10.1038/s41577-021-00550-x] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
Immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is central to long-term control of the current pandemic. Despite our rapidly advancing knowledge of immune memory to SARS-CoV-2, understanding how these responses translate into protection against reinfection at both the individual and population levels remains a major challenge. An ideal outcome following infection or after vaccination would be a highly protective and durable immunity that allows for the establishment of high levels of population immunity. However, current studies suggest a decay of neutralizing antibody responses in convalescent patients, and documented cases of SARS-CoV-2 reinfection are increasing. Understanding the dynamics of memory responses to SARS-CoV-2 and the mechanisms of immune control are crucial for the rational design and deployment of vaccines and for understanding the possible future trajectories of the pandemic. Here, we summarize our current understanding of immune responses to and immune control of SARS-CoV-2 and the implications for prevention of reinfection.
Collapse
Affiliation(s)
- Deborah Cromer
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| | | |
Collapse
|
50
|
Vigón L, Fuertes D, García-Pérez J, Torres M, Rodríguez-Mora S, Mateos E, Corona M, Saez-Marín AJ, Malo R, Navarro C, Murciano-Antón MA, Cervero M, Alcamí J, García-Gutiérrez V, Planelles V, López-Huertas MR, Coiras M. Impaired Cytotoxic Response in PBMCs From Patients With COVID-19 Admitted to the ICU: Biomarkers to Predict Disease Severity. Front Immunol 2021; 12:665329. [PMID: 34122423 PMCID: PMC8187764 DOI: 10.3389/fimmu.2021.665329] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/05/2021] [Indexed: 01/08/2023] Open
Abstract
Infection by novel coronavirus SARS-CoV-2 causes different presentations of COVID-19 and some patients may progress to a critical, fatal form of the disease that requires their admission to ICU and invasive mechanical ventilation. In order to predict in advance which patients could be more susceptible to develop a critical form of COVID-19, it is essential to define the most adequate biomarkers. In this study, we analyzed several parameters related to the cellular immune response in blood samples from 109 patients with different presentations of COVID-19 who were recruited in Hospitals and Primary Healthcare Centers in Madrid, Spain, during the first pandemic peak between April and June 2020. Hospitalized patients with the most severe forms of COVID-19 showed a potent inflammatory response that was not translated into an efficient immune response. Despite the high levels of effector cytotoxic cell populations such as NK, NKT and CD8+ T cells, they displayed immune exhaustion markers and poor cytotoxic functionality against target cells infected with pseudotyped SARS-CoV-2 or cells lacking MHC class I molecules. Moreover, patients with critical COVID-19 showed low levels of the highly cytotoxic TCRγδ+ CD8+ T cell subpopulation. Conversely, CD4 count was greatly reduced in association to high levels of Tregs, low plasma IL-2 and impaired Th1 differentiation. The relative importance of these immunological parameters to predict COVID-19 severity was analyzed by Random Forest algorithm and we concluded that the most important features were related to an efficient cytotoxic response. Therefore, efforts to fight against SARS-CoV-2 infection should be focused not only to decrease the disproportionate inflammatory response, but also to elicit an efficient cytotoxic response against the infected cells and to reduce viral replication.
Collapse
Affiliation(s)
- Lorena Vigón
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Fuertes
- School of Telecommunications Engineering, Universidad Politécnica de Madrid, Madrid, Spain
| | - Javier García-Pérez
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Montserrat Torres
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Rodríguez-Mora
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Mateos
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Magdalena Corona
- Hematology Service, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Rosa Malo
- Neumology Service, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | | | | | - Miguel Cervero
- Internal Medicine Service, Hospital Universitario Severo Ochoa, Leganés, Spain
| | - José Alcamí
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Vicente Planelles
- Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - María Rosa López-Huertas
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Mayte Coiras
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|