TP53 pathway alterations drive radioresistance in diffuse intrinsic pontine gliomas (DIPG)

C Werbrouck, CCS Evangelista, MJ Lobón-Iglesias… - Clinical Cancer …, 2019 - AACR
C Werbrouck, CCS Evangelista, MJ Lobón-Iglesias, E Barret, G Le Teuff, J Merlevede…
Clinical Cancer Research, 2019AACR
Purpose: Diffuse intrinsic pontine gliomas (DIPG) are the most severe pediatric brain tumors.
Although accepted as the standard therapeutic, radiotherapy is only efficient transiently and
not even in every patient. The goal of the study was to identify the underlying molecular
determinants of response to radiotherapy in DIPG. Experimental Design: We assessed in
vitro response to ionizing radiations in 13 different DIPG cellular models derived from
treatment-naïve stereotactic biopsies reflecting the genotype variability encountered in …
Purpose
Diffuse intrinsic pontine gliomas (DIPG) are the most severe pediatric brain tumors. Although accepted as the standard therapeutic, radiotherapy is only efficient transiently and not even in every patient. The goal of the study was to identify the underlying molecular determinants of response to radiotherapy in DIPG.
Experimental Design
We assessed in vitro response to ionizing radiations in 13 different DIPG cellular models derived from treatment-naïve stereotactic biopsies reflecting the genotype variability encountered in patients at diagnosis and correlated it to their principal molecular alterations. Clinical and radiologic response to radiotherapy of a large cohort of 73 DIPG was analyzed according to their genotype. Using a kinome-wide synthetic lethality RNAi screen, we further identified target genes that can sensitize DIPG cells to ionizing radiations.
Results
We uncover TP53 mutation as the main driver of increased radioresistance and validated this finding in four isogenic pairs of TP53WT DIPG cells with or without TP53 knockdown. In an integrated clinical, radiological, and molecular study, we show that TP53MUT DIPG patients respond less to irradiation, relapse earlier after radiotherapy, and have a worse prognosis than their TP53WT counterparts. Finally, a kinome-wide synthetic lethality RNAi screen identifies CHK1 as a potential target, whose inhibition increases response to radiation specifically in TP53MUT cells.
Conclusions
Here, we demonstrate that TP53 mutations are driving DIPG radioresistance both in patients and corresponding cellular models. We suggest alternative treatment strategies to mitigate radioresistance with CHK1 inhibitors. These findings will allow to consequently refine radiotherapy schedules in DIPG.
AACR