4.8 Article

CRAF gene fusions in pediatric low-grade gliomas define a distinct drug response based on dimerization profiles

Journal

ONCOGENE
Volume 36, Issue 45, Pages 6348-6358

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/onc.2017.276

Keywords

-

Funding

  1. Kids' Brain Tumor Cure Foundation Pediatric Low-Grade Astrocytoma Foundation
  2. Voices Against Brain Cancer
  3. Children's Brain Tumor Foundation
  4. Damon-Runyon Sohn Pediatric Fellowship Award
  5. Hyundai Scholar Grant
  6. Bear Necessities Pediatric Cancer Foundation
  7. Rally Foundation for Childhood Cancer Research
  8. Thea's Star of HopeNIH National Center for Advancing Translational Sciences Award [TL1TR000138]
  9. National Institute of Health grant [R01NS085336]

Ask authors/readers for more resources

Pediatric low-grade gliomas (PLGGs) are commonly associated with BRAF gene fusions that aberrantly activate the mitogenactivated protein kinase (MAPK) signaling pathway. This has led to PLGG clinical trials utilizing RAF- and MAPK pathway-targeted therapeutics. Whole-genome profiling of PLGGs has also identified rare gene fusions involving another RAF isoform, CRAF/RAF1, in PLGGs and cancers occuring in adults. Whereas BRAF fusions primarily dysregulate MAPK signaling, the CRAF fusions QKI-RAF1 and SRGAP3-RAF1 aberrantly activate both the MAPK and phosphoinositide-3 kinase/mammalian target of rapamycin (PI3K/mTOR) signaling pathways. Although ATP-competitive, first-generation RAF inhibitors (vemurafenib/PLX4720, RAFi) cause paradoxical activation of the MAPK pathway in BRAF-fusion tumors, inhibition can be achieved with 'paradox breaker' RAFi, such as PLX8394. Here we report that, unlike BRAF fusions, CRAF fusions are unresponsive to both generations of RAFi, vemurafenib and PLX8394, highlighting a distinct responsiveness of CRAF fusions to clinically relevant RAFi. Whereas PLX8394 decreased BRAF-fusion dimerization, CRAF-fusion dimerization is unaffected primarily because of robust protein-protein interactions mediated by the N-terminal non-kinase fusion partner, such as QKI. The pan-RAF dimer inhibitor, LY3009120, could suppress CRAF-fusion oncogenicity by inhibiting dimer-mediated signaling. In addition, as CRAF fusions activate both the MAPK and PI3K/mTOR signaling pathways, we identify combinatorial inhibition of the MAPK/mTOR pathway as a potential therapeutic strategy for CRAF-fusiondriven tumors. Overall, we define a mechanistic distinction between PLGG-associated BRAF-and CRAF/RAF1 fusions in response to RAFi, highlighting the importance of molecularly classifying PLGG patients for targeted therapy. Furthermore, our study uncovers an important contribution of the non-kinase fusion partner to oncogenesis and potential therapeutic strategies against PLGG-associated CRAF fusions and possibly pan-cancer CRAF fusions.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available