Your browser doesn't support javascript.
loading
Multifunctional NK Cell-Engaging Antibodies Targeting EGFR and NKp30 Elicit Efficient Tumor Cell Killing and Proinflammatory Cytokine Release.
Klausz, Katja; Pekar, Lukas; Boje, Ammelie Svea; Gehlert, Carina Lynn; Krohn, Steffen; Gupta, Tushar; Xiao, Yanping; Krah, Simon; Zaynagetdinov, Rinat; Lipinski, Britta; Toleikis, Lars; Poetzsch, Sven; Rabinovich, Brian; Peipp, Matthias; Zielonka, Stefan.
Afiliação
  • Klausz K; Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany.
  • Pekar L; Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany.
  • Boje AS; Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany.
  • Gehlert CL; Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany.
  • Krohn S; Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany.
  • Gupta T; Protein Engineering and Antibody Technologies, EMD Serono Research & Development Institute, Inc., Billerica, MA.
  • Xiao Y; Department of Oncology and Immuno-oncology, EMD Serono Research & Development Institute, Inc., Billerica, MA.
  • Krah S; Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany.
  • Zaynagetdinov R; Department of Oncology and Immuno-oncology, EMD Serono Research & Development Institute, Inc., Billerica, MA.
  • Lipinski B; Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany.
  • Toleikis L; Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany; and.
  • Poetzsch S; Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany.
  • Rabinovich B; Strategic Innovation, Merck Healthcare KGaA, Darmstadt, Germany.
  • Peipp M; Department of Oncology and Immuno-oncology, EMD Serono Research & Development Institute, Inc., Billerica, MA.
  • Zielonka S; Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany; matthias.peipp@uksh.de stefan.zielonka@merckgroup.com.
J Immunol ; 209(9): 1724-1735, 2022 11 01.
Article em En | MEDLINE | ID: mdl-36104113
ABSTRACT
In this work, we have generated novel Fc-comprising NK cell engagers (NKCEs) that bridge human NKp30 on NK cells to human epidermal growth factor receptor (EGFR) on tumor cells. Camelid-derived VHH single-domain Abs specific for human NKp30 and a humanized Fab derived from the EGFR-specific therapeutic Ab cetuximab were used as binding arms. By combining camelid immunization with yeast surface display, we were able to isolate a diverse panel of NKp30-specific VHHs against different epitopes on NKp30. Intriguingly, NKCEs built with VHHs that compete for binding to NKp30 with B7-H6, the natural ligand of NKp30, were significantly more potent in eliciting tumor cell lysis of EGFR-positive tumor cells than NKCEs harboring VHHs that target different epitopes on NKp30 from B7-H6. We demonstrate that the NKCEs can be further improved with respect to killing capabilities by concomitant engagement of FcγRIIIa and that soluble B7-H6 does not impede cytolytic capacities of all scrutinized NKCEs at significantly higher B7-H6 concentrations than observed in cancer patients. Moreover, we show that physiological processes requiring interactions between membrane-bound B7-H6 and NKp30 on NK cells are unaffected by noncompeting NKCEs still eliciting tumor cell killing at low picomolar concentrations. Ultimately, the NKCEs generated in this study were significantly more potent in eliciting NK cell-mediated tumor cell lysis than cetuximab and elicited a robust release of proinflammatory cytokines, both features which might be beneficial for antitumor therapy.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Citocinas / Receptor 3 Desencadeador da Citotoxicidade Natural Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Citocinas / Receptor 3 Desencadeador da Citotoxicidade Natural Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article