eLife (May 2023)

Noncovalent antibody catenation on a target surface greatly increases the antigen-binding avidity

  • Jinyeop Song,
  • Bo-Seong Jeong,
  • Seong-Woo Kim,
  • Seong-Bin Im,
  • Seonghoon Kim,
  • Chih-Jen Lai,
  • Wonki Cho,
  • Jae U Jung,
  • Myung-Ju Ahn,
  • Byung-Ha Oh

DOI
https://doi.org/10.7554/eLife.81646
Journal volume & issue
Vol. 12

Abstract

Read online

Immunoglobulin G (IgG) antibodies are widely used for diagnosis and therapy. Given the unique dimeric structure of IgG, we hypothesized that, by genetically fusing a homodimeric protein (catenator) to the C-terminus of IgG, reversible catenation of antibody molecules could be induced on a surface where target antigen molecules are abundant, and that it could be an effective way to greatly enhance the antigen-binding avidity. A thermodynamic simulation showed that quite low homodimerization affinity of a catenator, e.g. dissociation constant of 100 μM, can enhance nanomolar antigen-binding avidity to a picomolar level, and that the fold enhancement sharply depends on the density of the antigen. In a proof-of-concept experiment where antigen molecules are immobilized on a biosensor tip, the C-terminal fusion of a pair of weakly homodimerizing proteins to three different antibodies enhanced the antigen-binding avidity by at least 110 or 304 folds from the intrinsic binding avidity. Compared with the mother antibody, Obinutuzumab(Y101L) which targets CD20, the same antibody with fused catenators exhibited significantly enhanced binding to SU-DHL5 cells. Together, the homodimerization-induced antibody catenation would be a new powerful approach to improve antibody applications, including the detection of scarce biomarkers and targeted anticancer therapies.

Keywords