Microbiology & Immunology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 23
  • Item
    No Preview Available
    Durable reprogramming of neutralizing antibody responses following Omicron breakthrough infection
    Lee, WS ; Tan, H-X ; Reynaldi, A ; Esterbauer, R ; Koutsakos, M ; Nguyen, J ; Amarasena, T ; Kent, HE ; Aggarwal, A ; Turville, SG ; Taiaroa, G ; Kinsella, P ; Liew, KC ; Tran, T ; Williamson, DA ; Cromer, D ; Davenport, MP ; Kent, SJ ; Juno, JA ; Khoury, DS ; Wheatley, AK (AMER ASSOC ADVANCEMENT SCIENCE, 2023-07)
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infection of vaccinated individuals is increasingly common with the circulation of highly immune evasive and transmissible Omicron variants. Here, we report the dynamics and durability of recalled spike-specific humoral immunity following Omicron BA.1 or BA.2 breakthrough infection, with longitudinal sampling up to 8 months after infection. Both BA.1 and BA.2 infections robustly boosted neutralization activity against the infecting strain while expanding breadth against BA.4, although neutralization activity was substantially reduced for the more recent XBB and BQ.1.1 strains. Cross-reactive memory B cells against both ancestral and Omicron spike were predominantly expanded by infection, with limited recruitment of de novo Omicron-specific B cells or antibodies. Modeling of neutralization titers predicts that protection from symptomatic reinfection against antigenically similar strains will be durable but is undermined by new emerging strains with further neutralization escape.
  • Item
    No Preview Available
    Robust immunity to influenza vaccination in haematopoietic stem cell transplant recipients following reconstitution of humoral and adaptive immunity
    Zhang, W ; Rowntree, LC ; Muttucumaru, R ; Damelang, T ; Aban, M ; Hurt, AC ; Auladell, M ; Esterbauer, R ; Wines, B ; Hogarth, M ; Turner, SJ ; Wheatley, AK ; Kent, SJ ; Patil, S ; Avery, S ; Morrissey, O ; Chung, AW ; Koutsakos, M ; Nguyen, THO ; Cheng, AC ; Kotsimbos, TC ; Kedzierska, K (WILEY, 2023)
    OBJECTIVES: Influenza causes significant morbidity and mortality, especially in high-risk populations. Although current vaccination regimens are the best method to combat annual influenza disease, vaccine efficacy can be low in high-risk groups, such as haematopoietic stem cell transplant (HSCT) recipients. METHODS: We comprehensively assessed humoral immunity, antibody landscapes, systems serology and influenza-specific B-cell responses, together with their phenotypes and isotypes, to the inactivated influenza vaccine (IIV) in HSCT recipients in comparison to healthy controls. RESULTS: Inactivated influenza vaccine significantly increased haemagglutination inhibition (HAI) titres in HSCT recipients, similar to healthy controls. Systems serology revealed increased IgG1 and IgG3 antibody levels towards the haemagglutinin (HA) head, but not to neuraminidase, nucleoprotein or HA stem. IIV also increased frequencies of total, IgG class-switched and CD21loCD27+ influenza-specific B cells, determined by HA probes and flow cytometry. Strikingly, 40% of HSCT recipients had markedly higher antibody responses towards A/H3N2 vaccine strain than healthy controls and showed cross-reactivity to antigenically drifted A/H3N2 strains by antibody landscape analysis. These superior humoral responses were associated with a greater time interval after HSCT, while multivariant analyses revealed the importance of pre-existing immune memory. Conversely, in HSCT recipients who did not respond to the first dose, the second IIV dose did not greatly improve their humoral response, although 50% of second-dose patients reached a seroprotective HAI titre for at least one of vaccine strains. CONCLUSIONS: Our study demonstrates efficient, although time-dependent, immune responses to IIV in HSCT recipients, and provides insights into influenza vaccination strategies targeted to immunocompromised high-risk groups.
  • Item
    No Preview Available
    Cutting Edge: SARS-CoV-2 Infection Induces Robust Germinal Center Activity in the Human Tonsil
    Tan, H-X ; Wragg, KM ; Kelly, HG ; Esterbauer, R ; Dixon, BJ ; Lau, JSY ; Flanagan, KL ; van de Sandt, CE ; Kedzierska, K ; McMahon, JH ; Wheatley, AK ; Juno, JA ; Kent, SJ (AMER ASSOC IMMUNOLOGISTS, 2022-05-15)
    Understanding the generation of immunity to SARS-CoV-2 in lymphoid tissues draining the site of infection has implications for immunity to SARS-CoV-2. We performed tonsil biopsies under local anesthesia in 19 subjects who had recovered from SARS-CoV-2 infection 24-225 d previously. The biopsies yielded >3 million cells for flow cytometric analysis in 17 subjects. Total and SARS-CoV-2 spike-specific germinal center B cells, and T follicular helper cells, were readily detectable in human tonsils early after SARS-CoV-2 infection, as assessed by flow cytometry. Responses were higher in samples within 2 mo of infection but still detectable in some subjects out to 7 mo following infection. We conclude the tonsils are a secondary lymphoid organ that develop germinal center responses to SARS-CoV-2 infection and could play a role in the long-term development of immunity.
  • Item
    Thumbnail Image
    Lung-resident memory B cells established after pulmonary influenza infection display distinct transcriptional and phenotypic profiles
    Tan, H-X ; Juno, JA ; Esterbauer, R ; Kelly, HG ; Wragg, KM ; Konstandopoulos, P ; Alcantara, S ; Alvarado, C ; Jones, R ; Starkey, G ; Wang, BZ ; Yoshino, O ; Tiang, T ; Grayson, ML ; Opdam, H ; D'Costa, R ; Vago, A ; Mackay, LK ; Gordon, CL ; Masopust, D ; Groom, JR ; Kent, SJ ; Wheatley, AK (AMER ASSOC ADVANCEMENT SCIENCE, 2022-01)
    Recent studies have established that memory B cells, largely thought to be circulatory in the blood, can take up long-term residency in inflamed tissues, analogous to widely described tissue-resident T cells. The dynamics of recruitment and retention of memory B cells to tissues and their immunological purpose remains unclear. Here, we characterized tissue-resident memory B cells (BRM) that are stably maintained in the lungs of mice after pulmonary influenza infection. Influenza-specific BRM were localized within inducible bronchus-associated lymphoid tissues (iBALTs) and displayed transcriptional signatures distinct from classical memory B cells in the blood or spleen while showing partial overlap with memory B cells in lung-draining lymph nodes. We identified lung-resident markers, including elevated expression of CXCR3, CCR6, and CD69, on hemagglutinin (HA)- and nucleoprotein (NP)-specific lung BRM. We found that CCR6 facilitates increased recruitment and/or retention of BRM in lungs and differentiation into antibody-secreting cells upon recall. Although expression of CXCR3 and CCR6 was comparable in total and influenza-specific memory B cells isolated across tissues of human donors, CD69 expression was higher in memory B cells from lung and draining lymph nodes of human organ donors relative to splenic and PBMC-derived populations, indicating that mechanisms underpinning BRM localization may be evolutionarily conserved. Last, we demonstrate that human memory B cells in lungs are transcriptionally distinct to populations in lung-draining lymph nodes or PBMCs. These data suggest that BRM may constitute a discrete component of B cell immunity, positioned at the lung mucosa for rapid humoral response against respiratory viral infections.
  • Item
    No Preview Available
    Establishment and recall of SARS-CoV-2 spike epitope-specific CD4+ T cell memory
    Wragg, KM ; Lee, WS ; Koutsakos, M ; Tan, H-X ; Amarasena, T ; Reynaldi, A ; Gare, G ; Konstandopoulos, P ; Field, KR ; Esterbauer, R ; Kent, HE ; Davenport, MP ; Wheatley, AK ; Kent, SJ ; Juno, JA (NATURE PORTFOLIO, 2022-05)
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination elicit CD4+ T cell responses to the spike protein, including circulating follicular helper T (cTFH) cells that correlate with neutralizing antibodies. Using a novel HLA-DRB1*15:01/S751 tetramer to track spike-specific CD4+ T cells, we show that primary infection or vaccination induces robust S751-specific CXCR5- and cTFH cell memory responses. Secondary exposure induced recall of CD4+ T cells with a transitory CXCR3+ phenotype, and drove expansion of cTFH cells transiently expressing ICOS, CD38 and PD-1. In both contexts, cells exhibited a restricted T cell antigen receptor repertoire, including a highly public clonotype and considerable clonotypic overlap between CXCR5- and cTFH populations. Following a third vaccine dose, the rapid re-expansion of spike-specific CD4+ T cells contrasted with the comparatively delayed increase in antibody titers. Overall, we demonstrate that stable pools of cTFH and memory CD4+ T cells established by infection and/or vaccination are efficiently recalled upon antigen reexposure and may contribute to long-term protection against SARS-CoV-2.
  • Item
    No Preview Available
    Phenotypic and functional characterization of pharmacologically expanded Vγ9Vδ2 T cells in pigtail macaques
    Barber-Axthelm, IM ; Wragg, KM ; Esterbauer, R ; Amarasena, TH ; Barber-Axthelm, VRB ; Wheatley, AK ; Gibbon, AM ; Kent, SJ ; Juno, JA (CELL PRESS, 2023-03-17)
    While gaining interest as treatment for cancer and infectious disease, the clinical efficacy of Vγ9Vδ2 T cell-based immunotherapeutics has to date been limited. An improved understanding of γδ T cell heterogeneity across lymphoid and non-lymphoid tissues, before and after pharmacological expansion, is required. Here, we describe the phenotype and tissue distribution of Vγ9Vδ2 T cells at steady state and following in vivo pharmacological expansion in pigtail macaques. Intravenous phosphoantigen administration with subcutaneous rhIL-2 drove robust expansion of Vγ9Vδ2 T cells in blood and pulmonary mucosa, while expansion was confined to the pulmonary mucosa following intratracheal antigen administration. Peripheral blood Vγ9Vδ2 T cell expansion was polyclonal, and associated with a significant loss of CCR6 expression due to IL-2-mediated receptor downregulation. Overall, we show the tissue distribution and phenotype of in vivo pharmacologically expanded Vγ9Vδ2 T cells can be altered based on the antigen administration route, with implications for tissue trafficking and the clinical efficacy of Vγ9Vδ2 T cell immunotherapeutics.
  • 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
    Induction of vaginal-resident HIV-specific CD8 T cells with mucosal prime-boost immunization
    Tan, H-X ; Wheatley, AK ; Esterbauer, R ; Jegaskanda, S ; Glass, JJ ; Masopust, D ; De Rose, R ; Kent, SJ (NATURE PUBLISHING GROUP, 2018-05)
    Tissue-resident memory (TRM) CD8 T cells survey a range of non-lymphoid mucosal tissues where they rapidly mediate clearance of viral infections at the entry portals. Vaccines that establish CD8 TRM cells in the cervicovaginal mucosa hold promise for effective immunity against sexually transmitted HIV. We demonstrate that HIV-specific CD8 TRM cells can be established in the murine vaginal mucosa using a combined intranasal and intravaginal mucosal immunization with recombinant influenza-HIV vectors. Using in situ tetramer immunofluorescence microscopy, we found that this mucosally administered prime-boost immunization also resulted in the durable seeding of CD8 T cells in the frontline vaginal epithelial compartment as opposed to the vaginal submucosa. Upon cognate antigen recognition within the vaginal mucosa, these HIV-specific CD8 TRM cells rapidly initiated a tissue-wide state of immunity. The activation of HIV-specific CD8 TRM cells resulted in the upregulation of endothelial vessel addressin expression and substantial recruitment of both adaptive and innate immune cells in the vaginal mucosa. These findings suggest that the epithelial localization of HIV-specific CD8 TRM cell populations and their capacity to rapidly activate both arms of the immune system could significantly augment frontline defenses against vaginal HIV infection.
  • Item
    Thumbnail Image
    Protective efficacy of the anti-HIV broadly neutralizing antibody PGT121 in the context of semen exposure
    Parsons, MS ; Kristensen, AB ; Selva, KJ ; Lee, WS ; Amarasena, T ; Esterbauer, R ; Wheatley, AK ; Bavinton, BR ; Kelleher, AD ; Grulich, AE ; Khoury, G ; Juno, JA ; Kent, SJ (ELSEVIER, 2021-08)
    BACKGROUND: HIV-1 infections occur following viral exposure at anogenital mucosal surfaces in the presence of semen. Semen contains immunosuppressive and pro-inflammatory factors. Semen from HIV-1-infected donors contains anti-HIV-1 antibodies. We assessed if passively infused anti-HIV-1 neutralizing antibody conferred protection from rectal SHIVSF162P3 challenge at semen exposed mucosae. METHODS: We pooled seminal plasma from HIV-1-infected donors. The pool was screened by ELISA for antibodies against HIV-1SF162 gp140. The ability of seminal plasma to inhibit macaque NK cells from responding to direct and antibody-dependent stimulation was assessed. The ability of seminal plasma to inhibit macaque granulocytes from mediating oxidative burst was also assessed. To demonstrate viral infectivity in the presence of seminal plasma, macaques (n = 4) were rectally challenged with SHIVSF162P3 following exposure to 2.5 mL of seminal plasma. To evaluate if anti-HIV-1 neutralizing antibody confers protection against rectal SHIV challenge at semen exposed mucosae, eight macaques were intravenously infused with PGT121, either wild type (n = 4) or the Fc receptor binding deficient LALA variant (n = 4), and rectally challenged with SHIVSF162P3 following exposure to 2.5 mL of seminal plasma. FINDINGS: Anti-HIV-1SF162 gp140 antibodies were detected in seminal plasma. Seminal plasma inhibited direct and antibody-dependent NK cell activation and granulocyte oxidative burst in vitro. Rectal SHIVSF162P3 challenge of control macaques following seminal plasma exposure resulted in infection of all animals. All macaques infused with wild type or LALA PGT121 and challenged with SHIVSF162P3 following seminal plasma exposure were protected. INTERPRETATION: PGT121 conferred protection against rectal SHIVSF162P3 challenge at semen exposed mucosae. Future research should investigate if semen alters protection conferred by antibodies more dependent on non-neutralizing functions. FUNDING: This work was supported by a grant from the Australian National Health and Medical Research Council (APP1124680).
  • 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.