Your browser doesn't support javascript.
loading
CRAF gene fusions in pediatric low-grade gliomas define a distinct drug response based on dimerization profiles.
Jain, P; Fierst, T M; Han, H J; Smith, T E; Vakil, A; Storm, P B; Resnick, A C; Waanders, A J.
Affiliation
  • Jain P; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA.
  • Fierst TM; Cell and Molecular Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Han HJ; Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Smith TE; Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Vakil A; Department of Neurosurgery, Temple University School of Medicine, Philadelphia, PA, USA.
  • Storm PB; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA.
  • Resnick AC; Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Waanders AJ; Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
Oncogene ; 36(45): 6348-6358, 2017 11 09.
Article in En | MEDLINE | ID: mdl-28806393
ABSTRACT
Pediatric low-grade gliomas (PLGGs) are commonly associated with BRAF gene fusions that aberrantly activate the mitogen-activated 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-fusion-driven 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.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Proto-Oncogene Proteins c-raf / Glioma Type of study: Prognostic_studies Limits: Adolescent / Animals / Child / Child, preschool / Humans Language: En Journal: Oncogene Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2017 Type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Proto-Oncogene Proteins c-raf / Glioma Type of study: Prognostic_studies Limits: Adolescent / Animals / Child / Child, preschool / Humans Language: En Journal: Oncogene Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2017 Type: Article Affiliation country: United States