Engineering multivalent antibodies to target heregulin-induced HER3 signaling in breast cancer cells

MAbs. 2014 Mar-Apr;6(2):340-53. doi: 10.4161/mabs.27658. Epub 2013 Dec 26.

Abstract

The use of antibodies in therapy and diagnosis has undergone an unprecedented expansion during the past two decades. This is due in part to innovations in antibody engineering that now offer opportunities for the production of "second generation" antibodies with multiple specificities or altered valencies. The targeting of individual components of the human epidermal growth factor receptor (HER)3-PI3K signaling axis, including the preferred heterodimerization partner HER2, is known to have limited anti-tumor effects. The efficacy of antibodies or small molecule tyrosine kinase inhibitors (TKIs) in targeting this axis is further reduced by the presence of the HER3 ligand, heregulin. To address these shortcomings, we performed a comparative analysis of two distinct approaches toward reducing the proliferation and signaling in HER2 overexpressing tumor cells in the presence of heregulin. These strategies both involve the use of engineered antibodies in combination with the epidermal growth factor receptor (EGFR)/HER2 specific TKI, lapatinib. In the first approach, we generated a bispecific anti-HER2/HER3 antibody that, in the presence of lapatinib, is designed to sequester HER3 into inactive HER2-HER3 dimers that restrain HER3 interactions with other possible dimerization partners. The second approach involves the use of a tetravalent anti-HER3 antibody with the goal of inducing efficient HER3 internalization and degradation. In combination with lapatinib, we demonstrate that although the multivalent HER3 antibody is more effective than its bivalent counterpart in reducing heregulin-mediated signaling and growth, the bispecific HER2/HER3 antibody has increased inhibitory activity. Collectively, these observations provide support for the therapeutic use of bispecifics in combination with TKIs to recruit HER3 into complexes that are functionally inert.

Keywords: HER2; HER3; antibody engineering; bispecific antibody; receptor internalization.

Publication types

  • Comparative Study
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antibodies, Monoclonal, Humanized / administration & dosage
  • Antibodies, Monoclonal, Humanized / pharmacokinetics
  • Breast Neoplasms / immunology
  • Breast Neoplasms / therapy*
  • CHO Cells
  • Cell Line, Tumor
  • Cricetulus
  • Drug Synergism
  • Drug Therapy, Combination
  • Epitopes / genetics
  • ErbB Receptors / metabolism
  • Female
  • Humans
  • Immunotherapy / methods*
  • Lapatinib
  • Mice
  • Mice, Inbred BALB C
  • Mice, SCID
  • Neuregulin-1 / metabolism
  • Protein Binding / drug effects
  • Protein Engineering
  • Protein Kinase Inhibitors / pharmacology
  • Protein Stability
  • Quinazolines / pharmacology
  • Receptor, ErbB-2 / immunology
  • Receptor, ErbB-2 / metabolism*
  • Receptor, ErbB-3 / immunology
  • Receptor, ErbB-3 / metabolism*
  • Recombinant Fusion Proteins / administration & dosage
  • Recombinant Fusion Proteins / genetics
  • Signal Transduction* / drug effects
  • Trastuzumab

Substances

  • Antibodies, Monoclonal, Humanized
  • Epitopes
  • Neuregulin-1
  • Protein Kinase Inhibitors
  • Quinazolines
  • Recombinant Fusion Proteins
  • Lapatinib
  • EGFR protein, human
  • ERBB2 protein, human
  • ERBB3 protein, human
  • ErbB Receptors
  • Receptor, ErbB-2
  • Receptor, ErbB-3
  • pertuzumab
  • Trastuzumab