Your browser doesn't support javascript.
loading
Targeting the HER3 pseudokinase domain with small molecule inhibitors.
Colomba, Audrey; Claus, Jeroen; Gao, Forson; George, Roger; Fornili, Arianna; Cameron, Angus J M.
Afiliación
  • Colomba A; Alzheimer Research UK- UCL Drug Discovery Institute, University College London, London, United Kingdom.
  • Claus J; Phospho Biomedical Animation, The Greenhouse Studio 6, Unit 17, Millmead Industrial Estate, London, United Kingdom.
  • Gao F; Phospho Biomedical Animation, The Greenhouse Studio 6, Unit 17, Millmead Industrial Estate, London, United Kingdom; Section of Structural Biology, Department of Medicine, Imperial College London, London, United Kingdom.
  • George R; Structural Biology Science Technology platform, The Francis Crick Institute, London, United Kingdom.
  • Fornili A; Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London, United Kingdom.
  • Cameron AJM; Section of Structural Biology, Department of Medicine, Imperial College London, London, United Kingdom; Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, London, United Kingdom. Electronic address: a.cameron@qmul.ac.uk.
Methods Enzymol ; 667: 455-505, 2022.
Article en En | MEDLINE | ID: mdl-35525551
ABSTRACT
HER3 is a potent oncogenic growth factor receptor belonging to the human epidermal growth factor (HER/EGFR) family of receptor tyrosine kinases. In contrast to other EGFR family members, HER3 is a pseudokinase, lacking functional kinase activity. As such, efforts to develop small molecule tyrosine kinase inhibitors against this family member have been limited. In response to HER3-specific growth factors such as neuregulin (NRG, also known as heregulin or HRG), HER3 must couple with catalytically active family members, including its preferred partner HER2. Dimerization of the intracellular HER2HER3 kinase domains is a critical part of the activation mechanism and HER3 plays a specialized role as an allosteric activator of the active HER2 kinase partner. Intriguingly, many pseudokinases retain functionally important nucleotide binding capacity, despite loss of kinase activity. We demonstrated that occupation of the nucleotide pocket of the pseudokinase HER3 retains functional importance for growth factor signaling through oncogenic HER2HER3 heterodimers. Mutation of the HER3 nucleotide pocket both disrupts signaling and disrupts HER2HER3 dimerization. Conversely, ATP competitive drugs which bind to HER3, but not HER2, can stabilize HER2HER3 dimers, induce signaling and promote cell growth in breast cancer models. This indicates a nucleotide-dependent conformational role for the HER3 kinase domain. Critically, our recent proof-of-concept work demonstrated that HER3-directed small molecule inhibitors can also disrupt HER2HER3 dimerization and signaling, supporting the prospect that HER3 can be a direct drug target despite its lack of intrinsic activity. In this chapter we will describe methods for identifying and validating small molecule inhibitors against the HER3 pseudokinase.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Receptor ErbB-2 / Receptor ErbB-3 Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Revista: Methods Enzymol Año: 2022 Tipo del documento: Article País de afiliación: Reino Unido

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Receptor ErbB-2 / Receptor ErbB-3 Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Revista: Methods Enzymol Año: 2022 Tipo del documento: Article País de afiliación: Reino Unido