An In vivo Tumor Model Exploiting Metabolic Response as a Biomarker for Targeted Drug Development

C Cullinane, DS Dorow, M Kansara, N Conus, D Binns… - Cancer research, 2005 - AACR
C Cullinane, DS Dorow, M Kansara, N Conus, D Binns, RJ Hicks, LK Ashman, GA McArthur
Cancer research, 2005AACR
In vivo models that recapitulate oncogene-dependent tumorigenesis will greatly facilitate
development of molecularly targeted anticancer therapies. We have developed a model
based on activating mutations in c-KIT in gastrointestinal stromal tumors (GISTs). This model
comprises murine tumors of FDC-P1 cell lines expressing c-KIT mutations that render the
tumors either responsive (V560G) or resistant (D816V) to the small-molecule c-KIT inhibitor,
imatinib. Clinically, GIST response to imatinib is associated with rapid reduction in …
Abstract
In vivo models that recapitulate oncogene-dependent tumorigenesis will greatly facilitate development of molecularly targeted anticancer therapies. We have developed a model based on activating mutations in c-KIT in gastrointestinal stromal tumors (GISTs). This model comprises murine tumors of FDC-P1 cell lines expressing c-KIT mutations that render the tumors either responsive (V560G) or resistant (D816V) to the small-molecule c-KIT inhibitor, imatinib. Clinically, GIST response to imatinib is associated with rapid reduction in fluorodeoxyglucose (FDG) uptake on positron emission tomography (PET), preceding changes in conventional response criteria by several weeks. Using the FDC-P1 model in small animal PET, FDG uptake into tumors expressing the c-KIT V560G mutation was significantly reduced as early as 4 hours after imatinib treatment. In contrast, no change in FDG uptake was observed in resistant c-KIT D816V-expressing tumors after 48 hours of imatinib treatment. Consistent with the PET results, expression of the glucose transporter, GLUT1, was significantly reduced in V560G tumors at 4 hours, preceding changes in markers of proliferation by several hours. In vitro, imatinib treatment of V560G cells resulted in a reduction of glucose transporter numbers at the cell surface and decreased glucose uptake well before changes in cell viability. Notably, decreased ambient glucose concentrations enhanced the cytotoxic effect of imatinib. Taken together, these data account for the rapidity and significance of the PET response to imatinib and suggest that metabolic effects may contribute to imatinib cytotoxicity. Further, the FDC-P1 model represents a very useful paradigm for molecularly targeted drug development.
AACR