Your browser doesn't support javascript.
loading
Microfluidic antibody profiling after repeated SARS-CoV-2 vaccination links antibody affinity and concentration to impaired immunity and variant escape in patients on anti-CD20 therapy.
Priddey, Ashley; Chen-Xu, Michael Xin Hua; Cooper, Daniel James; MacMillan, Serena; Meisl, Georg; Xu, Catherine K; Hosmillo, Myra; Goodfellow, Ian G; Kollyfas, Rafael; Doffinger, Rainer; Bradley, John R; Mohorianu, Irina I; Jones, Rachel; Knowles, Tuomas P J; Smith, Rona; Kosmoliaptsis, Vasilis.
  • Priddey A; Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom.
  • Chen-Xu MXH; Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom.
  • Cooper DJ; Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
  • MacMillan S; Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom.
  • Meisl G; Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
  • Xu CK; Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom.
  • Hosmillo M; Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom.
  • Goodfellow IG; Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom.
  • Kollyfas R; Department of Pathology, Division of Virology, University of Cambridge, Cambridge, United Kingdom.
  • Doffinger R; Department of Pathology, Division of Virology, University of Cambridge, Cambridge, United Kingdom.
  • Bradley JR; Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom.
  • Mohorianu II; Department of Clinical Biochemistry and Immunology, Addenbrooke's Hospital, Cambridge, United Kingdom.
  • Jones R; Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom.
  • Knowles TPJ; Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
  • Smith R; Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom.
  • Kosmoliaptsis V; Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom.
Front Immunol ; 14: 1296148, 2023.
Article en En | MEDLINE | ID: mdl-38259440
ABSTRACT

Background:

Patients with autoimmune/inflammatory conditions on anti-CD20 therapies, such as rituximab, have suboptimal humoral responses to vaccination and are vulnerable to poorer clinical outcomes following SARS-CoV-2 infection. We aimed to examine how the fundamental parameters of antibody responses, namely, affinity and concentration, shape the quality of humoral immunity after vaccination in these patients.

Methods:

We performed in-depth antibody characterisation in sera collected 4 to 6 weeks after each of three vaccine doses to wild-type (WT) SARS-CoV-2 in rituximab-treated primary vasculitis patients (n = 14) using Luminex and pseudovirus neutralisation assays, whereas we used a novel microfluidic-based immunoassay to quantify polyclonal antibody affinity and concentration against both WT and Omicron (B.1.1.529) variants. We performed comparative antibody profiling at equivalent timepoints in healthy individuals after three antigenic exposures to WT SARS-CoV-2 (one infection and two vaccinations; n = 15) and in convalescent patients after WT SARS-CoV-2 infection (n = 30).

Results:

Rituximab-treated patients had lower antibody levels and neutralisation titres against both WT and Omicron SARS-CoV-2 variants compared to healthy individuals. Neutralisation capacity was weaker against Omicron versus WT both in rituximab-treated patients and in healthy individuals. In the rituximab cohort, this was driven by lower antibody affinity against Omicron versus WT [median (range) KD 21.6 (9.7-38.8) nM vs. 4.6 (2.3-44.8) nM, p = 0.0004]. By contrast, healthy individuals with hybrid immunity produced a broader antibody response, a subset of which recognised Omicron with higher affinity than antibodies in rituximab-treated patients [median (range) KD 1.05 (0.45-1.84) nM vs. 20.25 (13.2-38.8) nM, p = 0.0002], underpinning the stronger serum neutralisation capacity against Omicron in the former group. Rituximab-treated patients had similar anti-WT antibody levels and neutralisation titres to unvaccinated convalescent individuals, despite two more exposures to SARS-CoV-2 antigen. Temporal profiling of the antibody response showed evidence of affinity maturation in healthy convalescent patients after a single SARS-CoV-2 infection, which was not observed in rituximab-treated patients, despite repeated vaccination.

Discussion:

Our results enrich previous observations of impaired humoral immune responses to SARS-CoV-2 in rituximab-treated patients and highlight the significance of quantitative assessment of serum antibody affinity and concentration in monitoring anti-viral immunity, viral escape, and the evolution of the humoral response.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Enfermedades Autoinmunes / COVID-19 Tipo de estudio: Qualitative_research Límite: Humans Idioma: En Año: 2023 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Enfermedades Autoinmunes / COVID-19 Tipo de estudio: Qualitative_research Límite: Humans Idioma: En Año: 2023 Tipo del documento: Article