Augmented antibody-based anticancer therapeutics boost neutrophil cytotoxicity

Niels Heemskerk, Mandy Gruijs, A. Robin Temming, Marieke H. Heineke, Dennis Y. Gout, Tessa Hellingman, Cornelis W. Tuk, Paula J. Winter, Suzanne Lissenberg-Thunnissen, Arthur E. H. Bentlage, Marco de Donatis, Marijn Bögels, Thies Rösner, Thomas Valerius, Jantine E. Bakema, Gestur Vidarsson, Marjolein van Egmond

Research output: Contribution to journalArticleAcademicpeer-review

27 Citations (Scopus)

Abstract

Most clinically used anticancer mAbs are of the IgG isotype, which can eliminate tumor cells through NK cell-mediated antibody-dependent cellular cytotoxicity and macrophage-mediated antibody-dependent phagocytosis. IgG, however, ineffectively recruits neutrophils as effector cells. IgA mAbs induce migration and activation of neutrophils through the IgA Fc receptor (FcαRI) but are unable to activate NK cells and have poorer half-life. Here, we combined the agonistic activity of IgG mAbs and FcαRI targeting in a therapeutic bispecific antibody format. The resulting TrisomAb molecules recruited NK cells, macrophages, and neutrophils as effector cells for eradication of tumor cells in vitro and in vivo. Moreover, TrisomAb had long in vivo half-life and strongly decreased B16F10gp75 tumor outgrowth in mice. Importantly, neutrophils of colorectal cancer patients effectively eliminated tumor cells in the presence of anti-EGFR TrisomAb but were less efficient in mediating killing in the presence of IgG anti-EGFR mAb (cetuximab). The clinical application of TrisomAb may provide potential alternatives for cancer patients who do not benefit from current IgG mAb therapy.
Original languageEnglish
Article numbere134680
JournalJournal of clinical investigation
Volume131
Issue number6
DOIs
Publication statusPublished - 15 Mar 2021

Cite this