Microbiology & Immunology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 12
  • Item
    Thumbnail Image
    Landscape of human antibody recognition of the SARS-CoV-2 receptor binding domain
    Wheatley, AK ; Pymm, P ; Esterbauer, R ; Dietrich, MH ; Lee, WS ; Drew, D ; Kelly, HG ; Chan, L-J ; Mordant, FL ; Black, KA ; Adair, A ; Tan, H-X ; Juno, JA ; Wragg, KM ; Amarasena, T ; Lopez, E ; Selva, KJ ; Haycroft, ER ; Cooney, JP ; Venugopal, H ; Tan, LL ; Neill, MTO ; Allison, CC ; Cromer, D ; Davenport, MP ; Bowen, RA ; Chung, AW ; Pellegrini, M ; Liddament, MT ; Glukhova, A ; Subbarao, K ; Kent, SJ ; Tham, W-H (CELL PRESS, 2021-10-12)
    Potent neutralizing monoclonal antibodies are one of the few agents currently available to treat COVID-19. SARS-CoV-2 variants of concern (VOCs) that carry multiple mutations in the viral spike protein can exhibit neutralization resistance, potentially affecting the effectiveness of some antibody-based therapeutics. Here, the generation of a diverse panel of 91 human, neutralizing monoclonal antibodies provides an in-depth structural and phenotypic definition of receptor binding domain (RBD) antigenic sites on the viral spike. These RBD antibodies ameliorate SARS-CoV-2 infection in mice and hamster models in a dose-dependent manner and in proportion to in vitro, neutralizing potency. Assessing the effect of mutations in the spike protein on antibody recognition and neutralization highlights both potent single antibodies and stereotypic classes of antibodies that are unaffected by currently circulating VOCs, such as B.1.351 and P.1. These neutralizing monoclonal antibodies and others that bind analogous epitopes represent potentially useful future anti-SARS-CoV-2 therapeutics.
  • Item
    Thumbnail Image
    Tear antibodies to SARS-CoV-2: implications for transmission
    Selva, K ; Davis, S ; Haycroft, E ; Lee, WS ; Lopez, E ; Reynaldi, A ; Davenport, M ; Kent, H ; Juno, J ; Chung, A ; Kent, S ( 2021)

    Objectives

    SARS-CoV-2 can be transmitted by aerosols and the ocular surface may be an important route of transmission. Little is known about protective antibody responses to SARS-CoV-2 in tears after infection or vaccination. We analysed SARS-CoV-2 specific IgG and IgA responses in human tears after either COVID-19 infection or vaccination.

    Methods

    We recruited 16 subjects with COVID-19 infection an average of 7 months previously and 15 subjects before and 2 weeks after Comirnaty (Pfizer-BioNtech) vaccination. Plasma, saliva and basal tears were collected. Pre-pandemic plasma, saliva and basal tears from 11 individuals were included as healthy controls. Antibody responses to 5 SARS-CoV-2 antigens were measured via multiplex.

    Results

    IgG antibodies to Spike and Nucleoprotein were detected in tears, saliva and plasma from subjects with prior SARS-CoV-2 infection in comparison to uninfected controls. While RBD-specific antibodies were detected in plasma, minimal RBD-specific antibodies were detected in tears and saliva. In contrast, high levels of IgG antibodies to Spike and RBD, but not Nucleoprotein, were induced in tears, saliva and plasma of subjects receiving 2 doses of the Comirnaty vaccine. Increased levels of IgA1 and IgA2 antibodies to SARS-CoV-2 antigens were detected in plasma following infection or vaccination, but were unchanged in tears and saliva.

    Conclusion

    Both infection and vaccination induce SARS-CoV-2-specific IgG antibodies in tears. RBD-specific IgG antibodies in tears were induced by vaccination but were not present 7 months post-infection. This suggests neutralising antibodies may be low in the tears late following infection.
  • Item
    Thumbnail Image
    Tear antibodies to SARS-CoV-2: implications for transmission
    Selva, KJ ; Davis, SK ; Haycroft, ER ; Lee, WS ; Lopez, E ; Reynaldi, A ; Davenport, MP ; Kent, HE ; Juno, JA ; Chung, AW ; Kent, SJ (WILEY, 2021)
    OBJECTIVES: SARS-CoV-2 can be transmitted by aerosols, and the ocular surface may be an important route of transmission. Little is known about protective antibody responses to SARS-CoV-2 in tears after infection or vaccination. We analysed the SARS-CoV-2-specific IgG and IgA responses in human tears after either COVID-19 infection or vaccination. METHODS: We measured the antibody responses in 16 subjects with COVID-19 infection for an average of 7 months before, and 15 subjects before and 2 weeks post-Comirnaty (Pfizer-BioNtech) vaccination. Plasma, saliva and basal tears were collected. Eleven pre-pandemic individuals were included as healthy controls. RESULTS: IgG antibodies to spike and nucleoprotein were detected in tears, saliva and plasma from subjects with prior SARS-CoV-2 infection in comparison with uninfected controls. While receptor-binding domain (RBD)-specific antibodies were detected in plasma, minimal RBD-specific antibodies were detected in tears and saliva. By contrast, high levels of IgG antibodies to spike and RBD, but not nucleoprotein, were induced in tears, saliva and plasma of subjects receiving 2 doses of the Comirnaty vaccine. Increased levels of IgA1 and IgA2 antibodies to SARS-CoV-2 antigens were detected in plasma following infection or vaccination but were unchanged in tears and saliva. Comirnaty vaccination induced high neutralising Abs in the plasma, but limited neutralising antibodies were detected in saliva or tears. CONCLUSION: Both infection and vaccination induce SARS-CoV-2-specific IgG antibodies in tears. RBD-specific IgG antibodies in tears were induced by vaccination but were not present 7 months post-infection. This suggests the neutralising antibodies may be low in the tears late following infection.
  • Item
    Thumbnail Image
    Simultaneous evaluation of antibodies that inhibit SARS-CoV-2 variants via multiplex assay
    Lopez, E ; Haycroft, ER ; Adair, A ; Mordant, FL ; O'Neill, MT ; Pymm, P ; Redmond, SJ ; Lee, WS ; Gherardin, NA ; Wheatley, AK ; Juno, JA ; Selva, KJ ; Davis, SK ; Grimley, SL ; Harty, L ; Purcell, DFJ ; Subbarao, K ; Godfrey, D ; Kent, SJ ; Tham, W-H ; Chung, AW (AMER SOC CLINICAL INVESTIGATION INC, 2021-08-23)
    The SARS-CoV-2 receptor binding domain (RBD) is both the principal target of neutralizing antibodies and one of the most rapidly evolving domains, which can result in the emergence of immune escape mutations, limiting the effectiveness of vaccines and antibody therapeutics. To facilitate surveillance, we developed a rapid, high-throughput, multiplex assay able to assess the inhibitory response of antibodies to 24 RBD natural variants simultaneously. We demonstrate how this assay can be implemented as a rapid surrogate assay for functional cell-based serological methods to measure the SARS-CoV-2 neutralizing capacity of antibodies at the angiotensin-converting enzyme 2-RBD (ACE2-RBD) interface. We describe the enhanced affinity of RBD variants N439K, S477N, Q493L, S494P, and N501Y to the ACE2 receptor and demonstrate the ability of this assay to bridge a major gap for SARS-CoV-2 research, informing selection of complementary monoclonal antibody candidates and the rapid identification of immune escape to emerging RBD variants following vaccination or natural infection.
  • Item
    Thumbnail Image
    Simultaneous evaluation of antibodies that inhibit SARS-CoV-2 RBD variants with a novel competitive multiplex assay
    Lopez, E ; Haycroft, E ; Adair, A ; Mordant, F ; O’Neill, M ; Pymm, P ; Redmond, S ; Gherardin, N ; Wheatley, A ; Juno, J ; Selva, K ; Davis, S ; Harty, L ; Purcell, DFJ ; Subbarao, K ; Godfrey, D ; Kent, S ; Tham, W-H ; Chung, A ( 2021)

    ABSTRACT

    The SARS-CoV-2 Receptor Binding Domain (RBD) is both the principal target of neutralizing antibodies, and one of the most rapidly evolving domains, which can result in the emergence of immune escape mutations limiting the effectiveness of vaccines and antibody therapeutics. To facilitate surveillance, we developed a rapid, high-throughput, multiplex assay able to assess the inhibitory response of antibodies to 24 RBD natural variants simultaneously. We demonstrate that immune escape can occur through two mechanisms, antibodies that fail to recognize mutations, along with antibodies that have reduced inhibitory capacity due to enhanced variant RBD-ACE2 affinity. A competitive approach where antibodies simultaneously compete with ACE2 for binding to the RBD may therefore more accurately reflect the physiological dynamics of infection. We describe the enhanced affinity of RBD variants N439K, S477N, Q493L, S494P and N501Y to the ACE2 receptor, and demonstrate the ability of this assay to bridge a major gap for SARS-CoV-2 research; informing selection of complementary monoclonal antibody candidates and the rapid identification of immune escape to emerging RBD variants following vaccination or natural infection.
  • Item
    Thumbnail Image
    Decay of Fc-dependent antibody functions after mild to moderate COVID-19
    Lee, WS ; Selva, KJ ; Davis, SK ; Wines, BD ; Reynaldi, A ; Esterbauer, R ; Kelly, HG ; Haycroft, ER ; Tan, H-X ; Juno, JA ; Wheatley, AK ; Hogarth, PM ; Cromer, D ; Davenport, MP ; Chung, AW ; Kent, SJ (CELL PRESS, 2021-06-15)
    The capacity of antibodies to engage with immune cells via the Fc region is important in preventing and controlling many infectious diseases. The evolution of such antibodies during convalescence from coronavirus disease 2019 (COVID-19) is largely unknown. We develop assays to measure Fc-dependent antibody functions against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S)-expressing cells in serial samples from subjects primarily with mild-moderate COVID-19 up to 149 days post-infection. We find that S-specific antibodies capable of engaging Fcγ receptors decay over time, with S-specific antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent phagocytosis (ADP) activity within plasma declining accordingly. Although there is significant decay in ADCC and ADP activity, they remain readily detectable in almost all subjects at the last time point studied (94%) in contrast with neutralization activity (70%). Although it remains unclear the degree to which Fc effector functions contribute to protection against SARS-CoV-2 re-infection, our results indicate that antibodies with Fc effector functions persist longer than neutralizing antibodies.
  • Item
    Thumbnail Image
    Decay of Fc-dependent antibody functions after mild to moderate COVID-19
    Lee, WS ; Selva, KJ ; Davis, S ; Wines, B ; Reynaldi, A ; Esterbauer, R ; Kelly, H ; Haycroft, E ; Tan, H-X ; Juno, J ; Wheatley, A ; Hogarth, M ; Cromer, D ; Davenport, M ; Chung, A ; Kent, S ( 2020)
    The capacity of antibodies to engage with innate and adaptive immune cells via the Fc region is important in preventing and controlling many infectious diseases, and is likely critical in SARS-CoV-2 infection. The evolution of such antibodies during convalescence from COVID-19 is largely unknown. We developed novel assays to measure Fc-dependent antibody functions against SARS-CoV-2 spike (S)-expressing cells in serial samples from a cohort of 53 subjects primarily with mild-moderate COVID-19, out to a maximum of 149 days post-infection. We found that S-specific antibodies capable of engaging dimeric FcγRIIa and FcγRIIIa decayed linearly over time. S-specific antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent phagocytosis (ADP) activity within plasma declined linearly as well, in line with the decay of S-specific IgG. Although there was significant decay in S-specific plasma ADCC and ADP activity, they remained readily detectable by all assays in 94% of our cohort at the last timepoint studied, in contrast with neutralisation activity which was only detectable in 70% of our cohort by the last timepoint. Our results suggest that Fc effector functions such as ADCC and ADP could contribute to the durability of SARS-CoV-2 immunity, particularly late in convalescence when neutralising antibodies have waned. Understanding the protective potential of antibody Fc effector functions is critical for defining the durability of immunity generated by infection or vaccination.
  • Item
    Thumbnail Image
    Distinct systems serology features in children, elderly and COVID patients
    Selva, K ; van de Sandt, C ; Lemke, M ; Lee, C ; Shoffner, S ; Chua, B ; Nguyen, THO ; Rowntree, L ; Hensen, L ; Koutsakos, M ; Wong, CY ; Jackson, D ; Flanagan, K ; Crowe, J ; Cheng, A ; Doolan, D ; Amanat, F ; Krammer, F ; Chappell, K ; Modhiran, N ; Watterson, D ; Young, P ; Wines, B ; Hogarth, M ; Esterbauer, R ; Kelly, H ; Tan, H-X ; Juno, J ; Wheatley, A ; Kent, S ; Arnold, K ; Kedzierska, K ; Chung, A ( 2020)
    SARS-CoV-2, the pandemic coronavirus that causes COVID-19, has infected millions worldwide, causing unparalleled social and economic disruptions. COVID-19 results in higher pathogenicity and mortality in the elderly compared to children. Examining baseline SARS-CoV-2 cross-reactive coronavirus immunological responses, induced by circulating human coronaviruses, is critical to understand such divergent clinical outcomes. The cross-reactivity of coronavirus antibody responses of healthy children (n=89), adults (n=98), elderly (n=57), and COVID-19 patients (n=19) were analysed by systems serology. While moderate levels of cross-reactive SARS-CoV-2 IgG, IgM, and IgA were detected in healthy individuals, we identified serological signatures associated with SARS-CoV-2 antigen-specific Fcγ receptor binding, which accurately distinguished COVID-19 patients from healthy individuals and suggested that SARS-CoV-2 induces qualitative changes to antibody Fc upon infection, enhancing Fcγ receptor engagement. Vastly different serological signatures were observed between healthy children and elderly, with markedly higher cross-reactive SARS-CoV-2 IgA and IgG observed in elderly, whereas children displayed elevated SARS-CoV-2 IgM, including receptor binding domain-specific IgM with higher avidity. These results suggest that less-experienced humoral immunity associated with higher IgM, as observed in children, may have the potential to induce more potent antibodies upon SARS-CoV-2 infection. These key insights will inform COVID-19 vaccination strategies, improved serological diagnostics and therapeutics.
  • Item
    Thumbnail Image
    Systems serology detects functionally distinct coronavirus antibody features in children and elderly
    Selva, KJ ; van de Sandt, CE ; Lemke, MM ; Lee, CY ; Shoffner, SK ; Chua, BY ; Davis, SK ; Nguyen, THO ; Rowntree, LC ; Hensen, L ; Koutsakos, M ; Wong, CY ; Mordant, F ; Jackson, DC ; Flanagan, KL ; Crowe, J ; Tosif, S ; Neeland, MR ; Sutton, P ; Licciardi, P ; Crawford, NW ; Cheng, AC ; Doolan, DL ; Amanat, F ; Krammer, F ; Chappell, K ; Modhiran, N ; Watterson, D ; Young, P ; Lee, WS ; Wines, BD ; Hogarth, PM ; Esterbauer, R ; Kelly, HG ; Tan, H-X ; Juno, JA ; Wheatley, AK ; Kent, SJ ; Arnold, KB ; Kedzierska, K ; Chung, AW (NATURE PORTFOLIO, 2021-04-01)
    The hallmarks of COVID-19 are higher pathogenicity and mortality in the elderly compared to children. Examining baseline SARS-CoV-2 cross-reactive immunological responses, induced by circulating human coronaviruses (hCoVs), is needed to understand such divergent clinical outcomes. Here we show analysis of coronavirus antibody responses of pre-pandemic healthy children (n = 89), adults (n = 98), elderly (n = 57), and COVID-19 patients (n = 50) by systems serology. Moderate levels of cross-reactive, but non-neutralizing, SARS-CoV-2 antibodies are detected in pre-pandemic healthy individuals. SARS-CoV-2 antigen-specific Fcγ receptor binding accurately distinguishes COVID-19 patients from healthy individuals, suggesting that SARS-CoV-2 infection induces qualitative changes to antibody Fc, enhancing Fcγ receptor engagement. Higher cross-reactive SARS-CoV-2 IgA and IgG are observed in healthy elderly, while healthy children display elevated SARS-CoV-2 IgM, suggesting that children have fewer hCoV exposures, resulting in less-experienced but more polyreactive humoral immunity. Age-dependent analysis of COVID-19 patients, confirms elevated class-switched antibodies in elderly, while children have stronger Fc responses which we demonstrate are functionally different. These insights will inform COVID-19 vaccination strategies, improved serological diagnostics and therapeutics.
  • Item
    Thumbnail Image
    Evolution of immune responses to SARS-CoV-2 in mild-moderate COVID-19
    Wheatley, AK ; Juno, JA ; Wang, JJ ; Selva, KJ ; Reynaldi, A ; Tan, H-X ; Lee, WS ; Wragg, KM ; Kelly, HG ; Esterbauer, R ; Davis, SK ; Kent, HE ; Mordant, FL ; Schlub, TE ; Gordon, DL ; Khoury, DS ; Subbarao, K ; Cromer, D ; Gordon, TP ; Chung, AW ; Davenport, MP ; Kent, SJ (NATURE PORTFOLIO, 2021-02-19)
    The durability of infection-induced SARS-CoV-2 immunity has major implications for reinfection and vaccine development. Here, we show a comprehensive profile of antibody, B cell and T cell dynamics over time in a cohort of patients who have recovered from mild-moderate COVID-19. Binding and neutralising antibody responses, together with individual serum clonotypes, decay over the first 4 months post-infection. A similar decline in Spike-specific CD4+ and circulating T follicular helper frequencies occurs. By contrast, S-specific IgG+ memory B cells consistently accumulate over time, eventually comprising a substantial fraction of circulating the memory B cell pool. Modelling of the concomitant immune kinetics predicts maintenance of serological neutralising activity above a titre of 1:40 in 50% of convalescent participants to 74 days, although there is probably additive protection from B cell and T cell immunity. This study indicates that SARS-CoV-2 immunity after infection might be transiently protective at a population level. Therefore, SARS-CoV-2 vaccines might require greater immunogenicity and durability than natural infection to drive long-term protection.