Abstract

The interaction of the Fc region of therapeutic antibodies and antibody-drug conjugates with Fcγ receptors (FcγRs) can lead to unpredictable and severe side effects. Over the last decades several strategies have been developed to overcome this drawback, including extensive Fc- and glycoengineering and antibody isotype switching. However, these approaches result in permanently Fc-silenced antibody derivates which partially or completely lack antibody-mediated effector functions. Nevertheless, for a majority of antibody-based drugs, Fc-mediated effector functions, like antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP) as well as complement-dependent cytotoxicity (CDC), represent the most substantial modes of action. We argued that a new strategy combining the beneficial properties of Fc-silencing and controlled activation of effector functions can pave the way to potent antibody therapeutics, reducing the FcγRs-mediated off-target toxicity. We present a novel Fc-tamed antibody format, where the FcγR-binding sites of antibodies are blocked by anti-isotypic masking units, hindering the association of FcγR and complement component 1 (c1q) to the Fc domain. The masking units were genetically fused to trastuzumab, including a protease-addressable peptide-liker. Our Fc-tamed antibodies demonstrated completely abolished interaction to soluble high-affinity Fcγ-Receptor I and c1q. In reporter cell-based ADCC assays, our Fc-tamed antibodies exhibited a 2,700 to 7,100-fold reduction in activation, compared to trastuzumab. Upon demasking by a tumor-associated protease, the Fc-activated antibodies demonstrated restored FcγR-binding, c1q-binding and the ability to induce potent ADCC activation. Furthermore, cell killing assays using donor-derived NK cells were performed to validate the functionality of the Fc-tamed antibody variants. To our knowledge, this approach represents the first non-permanently Fc-silenced antibody, which can be re-activated by a tumor-associated protease, eventually extending the field of novel antibody formats.

Highlights

  • In the last decades monoclonal antibodies became powerful and promising drug classes, due to their ability to selectively address cancer-related molecules, infectious cells, virus particles, immune cells and immune-checkpoint-related molecules

  • For the generation of a suitable masking unit, which targets the Fcg receptors (FcgRs)-binding site of antibodies, we focused on implementation of anti-isotypic single-chain variable fragments

  • Comparing the induction level reached by the highest concentration of TRZ-Fc4 (100 nM) with the concentration of trastuzumab required for a similar induction (0.014 nM), we found a 7,100-fold reduction in antibody-dependent cellmediated cytotoxicity (ADCC) activation

Read more

Summary

Introduction

Thereby, the different IgG subclasses (IgG1, IgG2, IgG3, IgG4) display unique FcgR and complement component binding profiles [2]. Polymorphisms of FcgRs show an immense influence on the affinity to different subclasses of IgGs, translating in reduced or enhanced efficacy of therapeutic antibodies [4, 5]. Several studies reported a correlation of Fc receptor binding-related internalization of antibodies and antibody-drug conjugates (ADCs) and adverse side effects (e.g. thrombocytopenia) [10, 15,16,17]. A prominent example is the implementation of three single point mutations in the Fc part of an anti-HER2 tubulysin (IgG1) ADC (results from clinical trial phase 1) in order to reduce FcgRrelated side effects [19]. A single point mutation (K322A) is known to limit the interaction of C1q to the IgG1

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