Abstract

IgG antibodies mediate the clearance of target cells via the engagement of Fc gamma receptors (FcγRs) on effector cells by eliciting antibody-dependent cellular cytotoxicity and phagocytosis (ADCC and ADCP, respectively). Because (i) the IgG Fc domain binds to multiple FcγRs with varying affinities; (ii) even low Fc:FcγRs affinity interactions can play a significant role when antibodies are engaged in high avidity immune complexes and (iii) most effector cells express multiple FcγRs, the clearance mechanisms that can be mediated by individual FcγR are not well-understood. Human FcγRIIIa (hFcγRIIIa; CD16a), which exists as two polymorphic variants at position 158, hFcγRIIIaV158 and hFcγRIIIaF158, is widely considered to only trigger ADCC, especially with natural killer (NK) cells as effectors. To evaluate the role of hFcγRIIIa ligation in myeloid-derived effector cells, and in particular on macrophages and monocytes which express multiple FcγRs, we engineered an aglycosylated engineered human Fc (hFc) variant, Fc3aV, which binds exclusively to hFcγRIIIaV158. Antibodies formatted with the Fc3aV variant bind to the hFcγRIIIaV158 allotype with a somewhat lower KD than their wild type IgG1 counterparts, but not to any other hFcγR. The exceptional selectivity for hFcγRIIIaV158 was demonstrated by SPR using increased avidity, dimerized GST-fused versions of the ectodomains of hFcγRs and from the absence of binding of large immune complex (IC) to CHO cells expressing each of the hFcγRs, including notably, the FcγRIIIaF158 variant or the highly homologous FcγRIIIb. We show that even though monocyte-derived GM-CSF differentiated macrophages express hFcγRIIIa at substantially lower levels than the other two major activating receptors, namely hFcγRI or hFcγRIIa, Fc3aV-formatted Rituximab and Herceptin perform ADCP toward CD20- and Her2-expressing cancer cells, respectively, at a level comparable to that of the respective wild-type antibodies. We further show that hFcγRIIIa activation plays a significant role on ADCC by human peripheral monocytes. Our data highlight the utility of Fc3aV and other similarly engineered exquisitely selective, aglycosylated Fc variants toward other hFcγRs as tools for the detailed molecular understanding of hFcγR biology.

Highlights

  • Antibodies regulate a variety of immune responses by engaging Fcγ receptors (FcγRs) expressed on various leukocytes such as macrophages, granulocytes, dendritic cells (DCs), natural killer (NK) cells, and B cells

  • NK cells which are considered to be the most important contributor to ADCC with peripheral blood mononuclear cells (PBMCs) as effector cells, express only hFcγRIIIa [2]. hFcγRIIIa is expressed in intermediate (CD14hi/CD16+) and non-classical (CD14dim/CD16hi) blood monocytes which are capable of inducing ADCC primarily via TNFα secretion [9, 10]

  • Using antibodies equipped with the Fc3aV variant, we showed that exclusive engagement of hFcγRIIIa results in potent ADCP with gmMφ and established the role of this receptor on ADCC with monocytes as effectors

Read more

Summary

Introduction

Antibodies regulate a variety of immune responses by engaging Fcγ receptors (FcγRs) expressed on various leukocytes such as macrophages, granulocytes, dendritic cells (DCs), natural killer (NK) cells, and B cells. Immune complexes (ICs) formed by antibodies binding to multivalent antigens such as viruses and antibody-opsonized cancer or infected cells, induce activating FcγR-mediated immunoreceptor tyrosine-based activation motif (ITAM) signaling in effector cells, thereby inducing antibodydependent cellular cytotoxicity and phagocytosis (ADCC/P) [1, 2]. The activation of immune cells upon binding to ICs is regulated by the immunoreceptor tyrosine-based inhibitory motif (ITIM) signaling of the inhibitory FcγRIIb [3, 4]. The finding that higher affinity to hFcγRIIIa may lead to greater therapeutic potency stimulated extensive efforts to engineer hFc domains with improved binding hFcγRIIIa via either site-directed mutagenesis or glycoengineering, the latter accomplished primarily by completely abolishing or by reducing fucosylation [18,19,20,21]. The finding that higher affinity to hFcγRIIIa may lead to greater therapeutic potency stimulated extensive efforts to engineer hFc domains with improved binding hFcγRIIIa via either site-directed mutagenesis or glycoengineering, the latter accomplished primarily by completely abolishing or by reducing fucosylation [18,19,20,21]. hFc defucosylated antibodies have up to 50-fold enhanced affinity to hFcγRIIIa and three defucosylated antibodies, anti-CCR4 mogamulizumab, anti-IL5Ra benralizumab, and anti-CD20 obinutuzumab, have been evaluated for multiple therapeutic indications and approved for clinical use [21, 22]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.