Seroprevalence of anti-SARS-CoV-2 antibodies in COVID-19 patients and healthy volunteers up to 6 months post disease onset

Patrícia Figueiredo-Campos, Birte Blankenhaus, Catarina Mota, Andreia Gomes, Marta Serrano, Silvia Ariotti, Catarina Costa, Helena Nunes-Cabaço, António M Mendes, Pedro Gaspar, M Conceição Pereira-Santos, Fabiana Rodrigues, Jorge Condeço, M Antonia Escoval, Matilde Santos, Mario Ramirez, José Melo-Cristino, J Pedro Simas, Eugenia Vasconcelos, Ângela Afonso, Marc Veldhoen, Patrícia Figueiredo-Campos, Birte Blankenhaus, Catarina Mota, Andreia Gomes, Marta Serrano, Silvia Ariotti, Catarina Costa, Helena Nunes-Cabaço, António M Mendes, Pedro Gaspar, M Conceição Pereira-Santos, Fabiana Rodrigues, Jorge Condeço, M Antonia Escoval, Matilde Santos, Mario Ramirez, José Melo-Cristino, J Pedro Simas, Eugenia Vasconcelos, Ângela Afonso, Marc Veldhoen

Abstract

SARS-CoV-2 has emerged as a human pathogen, causing clinical signs, from fever to pneumonia-COVID-19-but may remain mild or asymptomatic. To understand the continuing spread of the virus, to detect those who are and were infected, and to follow the immune response longitudinally, reliable and robust assays for SARS-CoV-2 detection and immunological monitoring are needed. We quantified IgM, IgG, and IgA antibodies recognizing the SARS-CoV-2 receptor-binding domain (RBD) or the Spike (S) protein over a period of 6 months following COVID-19 onset. We report the detailed setup to monitor the humoral immune response from over 300 COVID-19 hospital patients and healthcare workers, 2500 University staff, and 198 post-COVID-19 volunteers. Anti-SARS-CoV-2 antibody responses follow a classic pattern with a rapid increase within the first three weeks after symptoms. Although titres reduce subsequently, the ability to detect anti-SARS-CoV-2 IgG antibodies remained robust with confirmed neutralization activity for up to 6 months in a large proportion of previously virus-positive screened subjects. Our work provides detailed information for the assays used, facilitating further and longitudinal analysis of protective immunity to SARS-CoV-2. Importantly, it highlights a continued level of circulating neutralising antibodies in most people with confirmed SARS-CoV-2.

Keywords: COVID-19; SARS-CoV-2; Seroprevalence; neutralizing antibodies.

Conflict of interest statement

The authors have no commercial or financial conflict of interest.

© 2020 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.

Figures

Figure 1
Figure 1
SARS‐CoV‐2 ELISA setup. SARS‐CoV‐2 IgG antibody detection in serum samples from SARS‐CoV‐2 PCR‐positive subjects or pre‐COVID‐19 controls using Immulon 4HBX 96‐well plates coated with RBD protein for ELISA. Absorbance (optical density, OD) was evaluated at 450 nm. (A) Three serum samples, pre‐COVID‐19 control, medium, and high titer were treated by the indicated methods to inactivate virus particles. (B) Isolated RBD monomer, RBD dimer, pooled RBD and Spike protein were used for coating at the same conditions, and four COVID‐19 serum samples (colored) were tested versus four pre‐COVID‐19 (open symbols). (C and D) 96‐well plate was coated with (C) RBD or (D) Spike protein at indicated concentrations and three COVID‐19 (colored) and pre‐COVID‐19 (black) sera were tested. (E) Secondary antibody dilution titration anti‐IgG, at indicated dilution on 96‐well plate coated with 2 μg/ml RBD protein for three sera. (F) Quality control (QC) and sample serum serial dilution. QC sample is a mix of the serum samples tested from four healthcare workers (S1–S4). Dashed line indicates blank values. Data show individual sample values.
Figure 2
Figure 2
SARS‐CoV‐2 ELISA testing. SARS‐CoV‐2 IgG antibody detection in serum samples from SARS‐CoV‐2 PCR‐positive subjects (colored) or pre‐COVID‐19 controls (open) using Immulon 4HBX 96‐well plates coated with RBD (circles) or Spike (squared) proteins for ELISA. Absorbance was evaluated at 450 nm. (A) Serum at 1/50 dilution from 19 SARS‐CoV‐2 PCR‐positive healthcare workers were assessed for anti‐SARS‐CoV‐2 RBD and Spike IgG and compared with 100 pre‐COVID‐19 sera. Bars indicate mean ± SD. (B and C) ROC analysis, plotting sensitivity against the specificity of (B) RBD or (C) Spike samples as shown in (A). D) Serum at 1/50 dilution from pre‐COVID‐19 cohorts, healthy (100 donors, open symbols), food allergies (61 donors, dark grey symbols), and bee/wasp allergies (20 donors, light grey symbols), were tested for RBD and Spike protein reactivity. (E) Example of cross‐reactivity on RBD or Spike protein from pre‐COVID‐19 serum as used in (D). Statistical analysis was performed using Kruskal–Wallis test with Graphpad Prism software. *p < 0.05, ***p < 0.001. Dashed lines indicate Black, blank values; Red, RBD cutoff; Green, Spike cutoff. Data show individual sample values.
Figure 3
Figure 3
Seroconversion in hospitalized patients. SARS‐CoV‐2 IgG antibody detection in serum samples from SARS‐CoV‐2 PCR‐positive hospital patients (colored) or pre‐COVID‐19 controls (open) using Immulon 4HBX 96‐well plates coated with RBD (circles) or Spike (squared) proteins for ELISA. Absorbance was evaluated at 450 nm. (A) Overview of all over 300 SARS‐CoV‐2 PCR‐positive tested subjects from Hospital Santa Maria and accumulated 181 pre‐COVID‐19 controls. (B) Selected samples presented in (A) post‐day 14 of the initial reported COVID‐19 symptoms (n = 73 COVID‐19 donors, n = 100 pre‐COVID‐19 healthy controls). (C) Selected samples presented in (A) pre‐day 7 of the initial reported COVID‐19 symptoms (n = 78 COVID‐19 donors, n = 100 pre‐COVID‐19 healthy controls). (D) Longitudinal follow‐up of patients sampled at the indicated week of COVID‐19 symptom onset and re‐sampling 7 days later (n = 76 COVID‐19 donors). Blue indicates continued high signal, Green those that seroconverted at the second sampling in week 2, Orange those that seroconverted at the second sampling past week 2, Red those in which no seroconversion was detected in first and second sampling. (E) Selected samples presented in (A) without reported COVID‐19 symptoms (n = 40 COVID‐19 donors, n = 100 pre‐COVID‐19 healthy controls). (F) Longitudinal follow‐up of asymptomatic patients sampled in the first week of COVID‐19 symptom onset and re‐sampling 7 days later, colors as used as in (D) (n = 10 donors). Dashed lines indicate Black, blank values; Red, RBD cutoff; Green, Spike cutoff. Data show individual sample values, bars indicate mean ± SD.
Figure 4
Figure 4
Seroconversion in subgroups and time. SARS‐CoV‐2 IgG antibody detection in serum samples from SARS‐CoV‐2 PCR‐positive hospital patients or pre‐COVID‐19 controls using Immulon 4HBX 96‐well plates coated with RBD (circles) or Spike (squared) proteins for ELISA. Absorbance was evaluated at 450 nm. (A and B) IgG OD signals were plotted of (A) female (n = 42) and male (n = 28) or (B) by age at the time of blood sampling, of those subjects 14‐days post first COVID‐19 symptoms. Red line marks the mean. (C and D) IgG OD signals of all subjects were plotted by (C) severity of symptoms or (D) over time since the day of first symptoms. (E and F) Hospital patients receiving (E) immunomodulatory medication (orange, n = 31) or (F) antiviral medication (purple, n = 12) who tested SARS‐CoV‐2 PCR‐positive were assessed for IgG antibodies and compared with pre‐COVID‐19 controls (open symbols, n = 100). Statistical analysis was performed using Kruskal–Wallis test using Graphpad Prism software. *p < 0.05, **p < 0.01, ***p < 0.001. Data show individual sample values, bars indicate mean ± SD.
Figure 5
Figure 5
Largescale seroconversion testing of Lisbon University staff. SARS‐CoV‐2 IgG antibody detection in 1/50 diluted serum samples from University of Lisbon staff using Immulon 4HBX 96‐well plates for ELISA. Absorbance was evaluated at 450 nm. (A) Schedule of screening plates, coated with 2 μg/ml RBD, accommodating 90 samples/plate, and includes two blanks, two quality control at high concentration (QChi), and two at low concentration (QClo). B) Overview of tested staff from Lisbon University (n = 2571 donors). Red symbols indicate negative scores, purple symbols indicate ODs above the cutoff. (C) Schedule of re‐screening plates, coated with 2 μg/ml RBD (left) or Spike (right), accommodating 21 samples/plate, and includes two dilutions per sample (1/50 and 1/150), two blanks, two QChi, and two QClo per protein used. (D) Screening results from the re‐screen (n = 68 donors), showing samples tested on Spike protein. Green depicts those samples above the cut off for RBD and Spike at both 1/50 and 1/150 dilution (n = 38); Red indicates those samples below the cut off on the second screen for either RBD or Spike (n = 30). (E–G) Showing RBD and Spike protein signals for the re‐assessed samples and an additional 10 negative samples, (E) all samples, (F) samples assessed negative (open to red symbols) and an additional 10 that were originally negative (red to red symbols), (G) samples assessed as positive (open to green symbols). (H) Quality control signals for 12 sequential plates, showing QChi (circles) and QClo (squares). Dotted lines indicate average signal ± SD for QChi (red, 200ng/ml) and QClo (blue (10 ng/ml)) of human anti‐SARS‐CoV‐2. Data show individual sample values.
Figure 6
Figure 6
Longitudinal SARS‐CoV‐2 antibody titers. SARS‐CoV‐2 antibodies were assessed in 1/50 diluted serum samples from donors in Portugal using Immulon 4HBX 96‐well plates for ELISA. Absorbance was evaluated at 450 nm. (A–C) Anti‐SARS‐CoV‐2 RBD antibody titers plotted over time for (A) IgM, (B) IgG, and (C) IgA (n = 356 total donors). Red line marks the geometric mean. (D–I) Anti‐SARS‐CoV‐2 antibody titers for males and females during (D, F, and H) early, days 7–40 (females n = 32, males n = 29) or (E, G, and H) late response, days 40–150 (females n = 60, males n = 114) for (D and E) IgM, (F and G) IgG or (H and I) IgA. Red line marks the mean. (J–L) Anti‐SARS‐CoV‐2 RBD antibody titers plotted by severity of COVID‐19 symptoms experienced for (J) IgM, (K) IgG, and (L) IgA. Red line indicates the mean (Asymptomatic = 13, Mild n = 28, moderate n = 54, severe n = 8). (M and N) SARS‐CoV‐2 neutralizing activity was determined in sera (n = 84 total donors) and plotted against (M) time since SARS‐CoV‐2 PCR+ or N) IgG titer. Red lines indicate geographic mean. Statistical analysis was performed using Mann–Whitney U‐test (d–i) or Kruskal–Wallis test using Graphpad Prism software. *p < 0.05, **p < 0.01, ***p < 0.001. Data show individual sample values.

References

    1. Buchholz, U. J. , Bukreyev, A. , Yang, L. , Lamirande, E. W. , Murphy, B. R. , Subbarao, K. and Collins, P. L. . Contributions of the structural proteins of severe acute respiratory syndrome coronavirus to protective immunity. PNAS 2004. 101: 9804–9809.
    1. Deming, D. , Sheahan, T. , Heise, M. , Yount, B. , Davis, N. , Sims, A. , Suthar, M. et al., Vaccine efficacy in senescent mice challenged with recombinant SARS‐CoV bearing epidemic and zoonotic spike variants. PLoS Med. 2006. 3: e525.
    1. Suthar, M. S. , Zimmerman, M. , Kauffman, R. , Mantus, G. , Linderman, S. , Vanderheiden, A. , Nyhoff, L. et al., Rapid generation of neutralizing antibody responses in COVID‐19 patients. medRxiv 2020.
    1. Robbiani, D. F. , Gaebler, C. , Muecksch, F. , Lorenzi, J. C. C. , Wang, Z. , Cho, A. , Agudelo, M. et al., Convergent antibody responses to SARS‐CoV‐2 in convalescent individuals. Nature 2020.
    1. Bosch, B. J. , van der Zee, R. , de Haan, C. A. and Rottier, P. J. . The coronavirus spike protein is a class I virus fusion protein: structural and functional characterization of the fusion core complex. J. Virol. 2003. 77: 8801–8811.
    1. Hoffmann, M. , Kleine‐Weber, H. , Schroeder, S. , Kruger, N. , Herrler, T. , Erichsen, S. , Schiergens, T. S. et al., SARS‐CoV‐2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020. 181: 271–280 e278.
    1. Wang, Q. , Zhang, Y. , Wu, L. , Niu, S. , Song, C. , Zhang, Z. , Lu, G. et al., Structural and Functional Basis of SARS‐CoV‐2 Entry by Using Human ACE2. Cell 2020. 181: 894–904 e899.
    1. Walls, A. C. , Park, Y. J. , Tortorici, M. A. , Wall, A. , McGuire, A. T. and Veesler, D. . Structure, Function, and Antigenicity of the SARS‐CoV‐2 Spike Glycoprotein. Cell 2020. 181: 281–292 e286.
    1. Amanat, F. , Stadlbauer, D. , Strohmeier, S. , Nguyen, T. H. O. , Chromikova, V. , McMahon, M. , Jiang, K. et al., A serological assay to detect SARS‐CoV‐2 seroconversion in humans. Nat. Med. 2020. 26: 1033–1036.
    1. Okba, N. M. A. , Muller, M. A. , Li, W. , Wang, C. , GeurtsvanKessel, C. H. , Corman, V. M. , Lamers, M. M. et al., Severe Acute Respiratory Syndrome Coronavirus 2‐Specific Antibody Responses in Coronavirus Disease Patients. Emerg. Infect. Dis. 2020. 26: 1478–1488.
    1. GeurtsvanKessel, C. H. , Okba, N. M. A. , Igloi, Z. , Bogers, S. , Embregts, C. W. E. , Laksono, B. M. , Leijten, L. et al., An evaluation of COVID‐19 serological assays informs future diagnostics and exposure assessment. Nat. Commun. 2020. 11: 3436.
    1. Stadlbauer, D. , Amanat, F. , Chromikova, V. , Jiang, K. , Strohmeier, S. , Arunkumar, G. A. , Tan, J. et al., SARS‐CoV‐2 Seroconversion in Humans: A Detailed Protocol for a Serological Assay, Antigen Production, and Test Setup. Curr Protoc Microbiol 2020. 57: e100.
    1. Remy, M. M. , Alfter, M. , Chiem, M. N. , Barbani, M. T. , Engler, O. B. and Suter‐Riniker, F. . Effective chemical virus inactivation of patient serum compatible with accurate serodiagnosis of infections. Clin. Microbiol. Infect. 2019. 25: 907 e907‐907 e912.
    1. Scott‐Taylor, T. H. , Axinia, S. C. , Amin, S. and Pettengell, R. . Immunoglobulin G; structure and functional implications of different subclass modifications in initiation and resolution of allergy. Immun Inflamm Dis 2018. 6: 13–33.
    1. Hsueh, P. R. , Huang, L. M. , Chen, P. J. , Kao, C. L. and Yang, P. C. . Chronological evolution of IgM, IgA, IgG and neutralisation antibodies after infection with SARS‐associated coronavirus. Clin. Microbiol. Infect. 2004. 10: 1062–1066.
    1. Wajnberg, A. , Amanat, F. , Firpo, A. , Altman, D. , Bailey, M. , Mansour, M. , McMahon, M. et al., SARS‐CoV‐2 infection induces robust, neutralizing antibody responses that are sTable for at least three months. medRxiv 2020. 2020.2007.2014.20151126.
    1. Wang, W. , Xu, Y. , Gao, R. , Lu, R. , Han, K. , Wu, G. and Tan, W. . Detection of SARS‐CoV‐2 in Different Types of Clinical Specimens. JAMA 2020.
    1. Blanco, J. L. , Ambrosioni, J. , Garcia, F. , Martinez, E. , Soriano, A. , Mallolas, J. and Miro, J. M. , Investigators C‐iH: COVID‐19 in patients with HIV: clinical case series. Lancet HIV 2020. 7: e314‐e316.
    1. Cooper, T. J. , Woodward, B. L. , Alom, S. and Harky, A. . Coronavirus disease 2019 (COVID‐19) outcomes in HIV/AIDS patients: a systematic review. HIV Med. 2020.
    1. Group, R. C. , Horby, P. , Lim, W. S. , Emberson, J. R. , Mafham, M. , Bell, J. L. , Linsell, L. et al., Dexamethasone in Hospitalized Patients with Covid‐19 ‐ Preliminary Report. N. Engl. J. Med. 2020.
    1. Klein, S. L. and Flanagan, K. L. . Sex differences in immune responses. Nat. Rev. Immunol. 2016. 16: 626–638.
    1. Jin, J. M. , Bai, P. , He, W. , Wu, F. , Liu, X. F. , Han, D. M. , Liu, S. and Yang, J. K. . Gender Differences in Patients With COVID‐19: Focus on Severity and Mortality. Front Public Health 2020. 8:152.
    1. Wenham, C. , Smith, J. and Morgan, R. , Gender, Group C‐W . COVID‐19: the gendered impacts of the outbreak. Lancet 2020. 395: 846–848.
    1. Laffont, S. , Rouquie, N. , Azar, P. , Seillet, C. , Plumas, J. , Aspord, C. and Guery, J. C. . X‐Chromosome complement and estrogen receptor signaling independently contribute to the enhanced TLR7‐mediated IFN‐alpha production of plasmacytoid dendritic cells from women. J. Immunol. 2014. 193: 5444–5452.
    1. Klein, S. L. , Hodgson, A. and Robinson, D. P. . Mechanisms of sex disparities in influenza pathogenesis. J. Leukoc. Biol. 2012. 92: 67–73.
    1. Gudbjartsson, D. F. , Helgason, A. , Jonsson, H. , Magnusson, O. T. , Melsted, P. , Norddahl, G. L. , Saemundsdottir, J. et al., Spread of SARS‐CoV‐2 in the Icelandic Population. N. Engl. J. Med. 2020. 382: 2302–2315.
    1. Behrens, G. M. , Cossmann, A. , Stankov, M. V. , Schulte, B. , Streeck, H. , Foerster, R. , Bosnjak, B. et al., Strategic anti‐SARS‐CoV‐2 serology testing in a low prevalence pandemic: The COVID‐19 Contact (CoCo) Study in health care professionals. medRxiv 2020. 2020.2008.2006.20169250.
    1. Pollan, M. , Perez‐Gomez, B. , Pastor‐Barriuso, R. , Oteo, J. , Hernan, M. A. , Perez‐Olmeda, M. , Sanmartin, J. L. et al., Prevalence of SARS‐CoV‐2 in Spain (ENE‐COVID): a nationwide, population‐based seroepidemiological study. Lancet 2020.
    1. Stringhini, S. , Wisniak, A. , Piumatti, G. , Azman, A. S. , Lauer, S. A. , Baysson, H. , De Ridder, D. et al., Seroprevalence of anti‐SARS‐CoV‐2 IgG antibodies in Geneva, Switzerland (SEROCoV‐POP): a population‐based study. Lancet 2020. 396: 313–319.
    1. Long, Q. X. , Liu, B. Z. , Deng, H. J. , Wu, G. C. , Deng, K. , Chen, Y. K. , Liao, P. et al., Antibody responses to SARS‐CoV‐2 in patients with COVID‐19. Nat. Med. 2020. 26: 845–848.
    1. Addetia, A. , Crawford, K. H. , Dingens, A. , Zhu, H. , Roychoudhury, P. , Huang, M. , Jerome, K. R. et al., Neutralizing antibodies correlate with protection from SARS‐CoV‐2 in humans during a fishery vessel outbreak with high attack rate. medRxiv 2020. 2020.2008.2013.20173161.
    1. Grifoni, A. , Weiskopf, D. , Ramirez, S. I. , Mateus, J. , Dan, J. M. , Moderbacher, C. R. , Rawlings, S. A. et al., Targets of T Cell Responses to SARS‐CoV‐2 Coronavirus in Humans with COVID‐19 Disease and Unexposed Individuals. Cell 2020. 181: 1489–1501 e1415.
    1. Braun, J. , Loyal, L. , Frentsch, M. , Wendisch, D. , Georg, P. , Kurth, F. , Hippenstiel, S. et al., Presence of SARS‐CoV‐2 reactive T cells in COVID‐19 patients and healthy donors. medRxiv 2020. 2020.2004.2017.20061440.
    1. Gangaev, A. , Ketelaars, S. L. C. , Patiwael, S. , Dopler, A. , Isaeva, O. I. , Hoefakker, K. , De Biasi, S. et al., Profound CD8 T cell responses towards the SARS‐CoV‐2 ORF1ab in COVID‐19 patients. Researchsquare 2020.
    1. Le Bert, N. , Tan, A. T. , Kunasegaran, K. , Tham, C. Y. L. , Hafezi, M. , Chia, A. , Chng, M. H. Y. et al., SARS‐CoV‐2‐specific T cell immunity in cases of COVID‐19 and SARS, and uninfected controls. Nature 2020.
    1. Oguntuyo, K. Y. , Stevens, C. S. , Hung, C. ‐. T. , Ikegame, S. , Acklin, J. A. , Kowdle, S. S. , Carmichael, J. C. et al., Quantifying absolute neutralization titers against SARS‐CoV‐2 by a standardized virus neutralization assay allows for cross‐cohort comparisons of COVID‐19 sera. medRxiv 2020. 2020, 2008.2013.20157222.

Source: PubMed

3
Prenumerera