ERK inhibition overcomes acquired resistance to MEK inhibitors

G Hatzivassiliou, B Liu, C O'Brien, JM Spoerke… - Molecular cancer …, 2012 - AACR
G Hatzivassiliou, B Liu, C O'Brien, JM Spoerke, KP Hoeflich, PM Haverty, R Soriano…
Molecular cancer therapeutics, 2012AACR
Abstract The RAS/RAF/MEK pathway is activated in more than 30% of human cancers, most
commonly via mutation in the K-ras oncogene and also via mutations in BRAF. Several
allosteric mitogen-activated protein/extracellular signal–regulated kinase (MEK) inhibitors,
aimed at treating tumors with RAS/RAF pathway alterations, are in clinical development.
However, acquired resistance to these inhibitors has been documented both in preclinical
and clinical samples. To identify strategies to overcome this resistance, we have derived …
Abstract
The RAS/RAF/MEK pathway is activated in more than 30% of human cancers, most commonly via mutation in the K-ras oncogene and also via mutations in BRAF. Several allosteric mitogen-activated protein/extracellular signal–regulated kinase (MEK) inhibitors, aimed at treating tumors with RAS/RAF pathway alterations, are in clinical development. However, acquired resistance to these inhibitors has been documented both in preclinical and clinical samples. To identify strategies to overcome this resistance, we have derived three independent MEK inhibitor–resistant cell lines. Resistance to allosteric MEK inhibitors in these cell lines was consistently linked to acquired mutations in the allosteric binding pocket of MEK. In one cell line, concurrent amplification of mutant K-ras was observed in conjunction with MEK allosteric pocket mutations. Clonal analysis showed that both resistance mechanisms occur in the same cell and contribute to enhanced resistance. Importantly, in all cases the MEK-resistant cell lines retained their addiction to the mitogen-activated protein kinase (MAPK) pathway, as evidenced by their sensitivity to a selective inhibitor of the ERK1/2 kinases. These data suggest that tumors with acquired MEK inhibitor resistance remain dependent on the MAPK pathway and are therefore sensitive to inhibitors that act downstream of the mutated MEK target. Importantly, we show that dual inhibition of MEK and ERK by small molecule inhibitors was synergistic and acted to both inhibit the emergence of resistance, as well as to overcome acquired resistance to MEK inhibitors. Therefore, our data provide a rationale for cotargeting multiple nodes within the MAPK signaling cascade in K-ras mutant tumors to maximize therapeutic benefit for patients. Mol Cancer Ther; 11(5); 1143–54. ©2012 AACR.
AACR