Determinants of EGFR-Targeted Therapeutic Biochemical Efficacy when Using Computational Modeling

Results of researches from C S Monast and M J Lazzara highlight numerous other considerations that determine biochemical efficacy beyond those reflected by equilibrium affinities. By integrating these considerations, their model also predicts minimum therapeutic combination concentrations to maximally reduce receptor phosphorylation.

B4E1D3C1-681C-4B3E-B2DF-23301F089658

Their finding that the abilities of cetuximab and gefitinib to antagonize EGFR phosphorylation are determined by processes beyond those describing equilibrium drug binding motivates new consideration of optimal design strategies for EGFR-targeted therapeutics. To the extent that EGFR phosphorylation level correlates with clinical efficacy, these results also identify possible factors that could underlie the differential effectiveness of the therapeutics among patients.

The identification of EGFR dephosphorylation kinetics as a key determinant of therapeutic biochemical efficacy is intriguing given that the activities of protein tyrosine phosphatases that regulate EGFR are altered in certain cancer settings.

Their results also predict IC50 dependence on the rates of drug and ligand association and dissociation, even for constant affinities. Their prediction that slowing EGFR gefitinib binding and dissociation cycling decreases IC50 is consistent with the observation that slower erlotinib cycling promotes growth inhibition of lung and brain cancer cells.

Reference

 Identifying Determinants of EGFR-Targeted Therapeutic Biochemical Efficacy Using Computational Modeling, CPT Pharmacometrics Syst. Pharmacol. (2014) 3, e141; doi:10.1038/psp.2014.39

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>