investigator_user investigator user funding collaborators pending menu bell message arrow_up arrow_down filter layers globe marker add arrow close download edit facebook info linkedin minus plus save share search sort twitter remove user-plus user-minus
  • Project leads
  • Collaborators

Inhibitor-sensitive and -resistant EGFR mutants from lung cancer and glioblastoma

Michael J Eck

0 Collaborator(s)

Funding source

National Institutes of Health (NIH)
The epidermal growth factor receptor tyrosine kinase (EGFR) is one of the most commonly activated oncoproteins in lung adenocarcinoma, glioblastoma and other cancers. The identification of cancer-associated EGFR mutants, and their predictive power to select patients for treatment with EGFR inhibitors including erlotinib and gefitinib, has led to a major advance in cancer treatment. The collaborative efforts of the Eck and Meyerson laboratories for the past seven years have focused on understanding the structure-function relationship of cancer-derived EGFR mutants with the overarching goal of improved targeted therapies for cancers bearing these mutations. Refinement of our understanding of the mechanisms of action of mutant EGFR, and the spectrum of response of specific mutants to EGFR inhibitors, will permit more efficient development and application of EGFR-directed therapies. In this renewal, we will undertake the following Specific Aims: Aim 1) Characterize the response of novel EGFR mutants, identified by genomic studies, to low- molecular weight ATP-competitive enzymatic inhibitors and to antibody therapies, Aim 2) Perform structural and functional studies of the exon 20 insertion mutants of EGFR, which are resistant to gefitinib, erlotinib and cetuximab, to ascertain potential therapeutic approaches, andAim 3) Analyze the substrate specificity of wild- type and cancer-derived EGFR mutants through peptide library array experiments, mass spectrometry-based phosphoproteomics, and crystal structure determination. Execution of these aims will help define the pathogenesis of EGFR-driven tumors and will thus provide a critically important mechanistic foundation for clinical trias targeting EGFR in a variety of cancer types using known agents. Additionally, our efforts will structural and mechanistic foundation for development of next-generation EGFR inhibitors that can be effective in patients whose tumors carry EGFR mutants that are resistant to currently available inhibitors.

Related projects