HDAC Inhibition Enhances the In Vivo Efficacy of MEK Inhibitor Therapy in Uveal Melanoma

F Faião-Flores, MF Emmons, MA Durante… - Clinical Cancer …, 2019 - AACR
F Faião-Flores, MF Emmons, MA Durante, F Kinose, B Saha, B Fang, JM Koomen
Clinical Cancer Research, 2019AACR
Purpose: The clinical use of MEK inhibitors in uveal melanoma is limited by the rapid
acquisition of resistance. This study has used multiomics approaches and drug screens to
identify the pan-HDAC inhibitor panobinostat as an effective strategy to limit MEK inhibitor
resistance. Experimental Design: Mass spectrometry–based proteomics and RNA-Seq were
used to identify the signaling pathways involved in the escape of uveal melanoma cells from
MEK inhibitor therapy. Mechanistic studies were performed to evaluate the escape pathways …
Purpose
The clinical use of MEK inhibitors in uveal melanoma is limited by the rapid acquisition of resistance. This study has used multiomics approaches and drug screens to identify the pan-HDAC inhibitor panobinostat as an effective strategy to limit MEK inhibitor resistance.
Experimental Design: Mass spectrometry–based proteomics and RNA-Seq were used to identify the signaling pathways involved in the escape of uveal melanoma cells from MEK inhibitor therapy. Mechanistic studies were performed to evaluate the escape pathways identified, and the efficacy of the MEK-HDAC inhibitor combination was demonstrated in multiple in vivo models of uveal melanoma.
Results
We identified a number of putative escape pathways that were upregulated following MEK inhibition, including the PI3K/AKT pathway, ROR1/2, and IGF-1R signaling. MEK inhibition was also associated with increased GPCR expression, particularly the endothelin B receptor, and this contributed to therapeutic escape through ET-3–mediated YAP signaling. A screen of 289 clinical grade compounds identified HDAC inhibitors as potential candidates that suppressed the adaptive YAP and AKT signaling that followed MEK inhibition. In vivo, the MEK-HDAC inhibitor combination outperformed either agent alone, leading to a long-term decrease of tumor growth in both subcutaneous and liver metastasis models and the suppression of adaptive PI3K/AKT and YAP signaling.
Conclusions
Together, our studies have identified GPCR-mediated YAP activation and RTK-driven AKT signaling as key pathways involved in the escape of uveal melanoma cells from MEK inhibition. We further demonstrate that HDAC inhibition is a promising combination partner for MEK inhibitors in advanced uveal melanoma.
AACR