Abstract

AbstractWe have identified a rare healthy FcγRIIIB (CD16B)-null donor completely lacking FCGR3B RNA and protein expression and dissected the role of the different neutrophil Fcγ receptors in the response to therapeutic anti-CD20 monoclonal antibodies. We observed that polymorphonuclear neutrophils (PMNs) from FcγRIIIB wild-type (WT) individuals or the null donor were more effectively activated by chronic lymphocytic leukemia (CLL) B-cell targets opsonized with glycoengineered anti-CD20 antibodies compared with fully core-fucosylated anti-CD20 antibodies, suggesting the presence and role of FcγRIIIA (CD16A) on PMNs. Indeed, we demonstrated by reverse-transcription polymerase chain reaction, flow cytometry, and western blot analysis that PMNs from FcγRIIIB WT donors and the null individual express low levels of FcγRIIIA on their surfaces. FcγRIIIA is a functional and activating molecule on these cells, because anti-CD16 F(ab′)2 antibodies alone were able to activate highly purified PMNs from the FcγRIIIB-null donor. Use of blocking anti-CD16 and anti-CD32 antibodies showed that FcγRIIIA is also a major mediator of phagocytosis of CD20-opsonized beads by FcγRIIIB WT and null PMNs. In contrast, trogocytosis of antibody-opsonized CLL B cells by PMNs was mediated primarily by FcγRIIIB in WT PMNs and by FcγRIIA in null PMNs. We conclude that FcγRIIIA is an important player in PMN functions, whereas FcγRIIIB is dispensable for activation and phagocytosis. We discuss the clinical implications of these findings.

Highlights

  • Unconjugated immunoglobulin G1 (IgG1) therapeutic antibodies, such as anti-CD20, act in vivo primarily through Fcg receptors (FcgRs) (FcgRIII [CD16], FcgRII [CD32], and FcgRI [CD64]) and immune-mediated mechanisms, including antibodydependent cellular cytotoxicity (ADCC) by natural killer (NK) cells and phagocytosis by macrophages.[1]

  • Several previous FcgRIIIB-null donors have been described in the literature who have a deletion of CD16B and CD32C genes due to homologous recombination.[27,28,29,36]

  • The availability of a rare natural knockout for FcgRIIIB allowed us to investigate in more detail the role of this molecule in Polymorphonuclear neutrophils (PMNs) functions induced by therapeutic anti-CD20 antibodies

Read more

Summary

Introduction

Unconjugated immunoglobulin G1 (IgG1) therapeutic antibodies, such as anti-CD20, act in vivo primarily through Fcg receptors (FcgRs) (FcgRIII [CD16], FcgRII [CD32], and FcgRI [CD64]) and immune-mediated mechanisms, including antibodydependent cellular cytotoxicity (ADCC) by natural killer (NK) cells and phagocytosis by macrophages.[1]. Low-fucose–containing therapeutic antibodies (eg, anti-CD20 obinutuzumab [OBZ]) are known to bind more efficiently than fully core-fucosylated MAbs to FcgRIIIA and FcgRIIIB as a result of the high degree of homology (.95%) between the extracellular portions of these 2 FcgRs. On the other hand, fucose content does not affect binding to the other FcgRs: FcgRI and FcgRIIA/ B.6,12-15. Glycoengineered anti-CD20 antibodies have been shown to activate neutrophils more efficiently than the unmodified versions of the same antibodies, suggesting that FcgRIIIB plays an important role in neutrophil activation.[6] PMNs express high levels of FcgRIIIB and FcgRIIA, and both of these FcgRs have been implicated in antibody-mediated PMN activation.[16] FcgRI is expressed weakly on resting neutrophils but is induced only after PMN activation, in particular following stimulation with IFN-g.17

Methods
Results
Conclusion
Full Text
Published version (Free)

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