Back to Search Start Over

H3.3-G34 mutations impair DNA repair and promote cGAS/STING-mediated immune responses in pediatric high-grade glioma models

Authors :
Santiago Haase
Kaushik Banerjee
Anzar A. Mujeeb
Carson S. Hartlage
Fernando M. Núñez
Felipe J. Núñez
Mahmoud S. Alghamri
Padma Kadiyala
Stephen Carney
Marcus N. Barissi
Ayman W. Taher
Emily K. Brumley
Sarah Thompson
Justin T. Dreyer
Caitlin T. Alindogan
Maria B. Garcia-Fabiani
Andrea Comba
Sriram Venneti
Visweswaran Ravikumar
Carl Koschmann
Ángel M. Carcaboso
Maria Vinci
Arvind Rao
Jennifer S. Yu
Pedro R. Lowenstein
Maria G. Castro
Source :
The Journal of Clinical Investigation, Vol 132, Iss 22 (2022)
Publication Year :
2022
Publisher :
American Society for Clinical Investigation, 2022.

Abstract

Pediatric high-grade gliomas (pHGGs) are the leading cause of cancer-related deaths in children in the USA. Sixteen percent of hemispheric pediatric and young adult HGGs encode Gly34Arg/Val substitutions in the histone H3.3 (H3.3-G34R/V). The mechanisms by which H3.3-G34R/V drive malignancy and therapeutic resistance in pHGGs remain unknown. Using a syngeneic, genetically engineered mouse model (GEMM) and human pHGG cells encoding H3.3-G34R, we demonstrate that this mutation led to the downregulation of DNA repair pathways. This resulted in enhanced susceptibility to DNA damage and inhibition of the DNA damage response (DDR). We demonstrate that genetic instability resulting from improper DNA repair in G34R-mutant pHGG led to the accumulation of extrachromosomal DNA, which activated the cyclic GMP–AMP synthase/stimulator of IFN genes (cGAS/STING) pathway, inducing the release of immune-stimulatory cytokines. We treated H3.3-G34R pHGG–bearing mice with a combination of radiotherapy (RT) and DNA damage response inhibitors (DDRi) (i.e., the blood-brain barrier–permeable PARP inhibitor pamiparib and the cell-cycle checkpoint CHK1/2 inhibitor AZD7762), and these combinations resulted in long-term survival for approximately 50% of the mice. Moreover, the addition of a STING agonist (diABZl) enhanced the therapeutic efficacy of these treatments. Long-term survivors developed immunological memory, preventing pHGG growth upon rechallenge. These results demonstrate that DDRi and STING agonists in combination with RT induced immune-mediated therapeutic efficacy in G34-mutant pHGG.

Subjects

Subjects :
Oncology
Therapeutics
Medicine

Details

Language :
English
ISSN :
15588238
Volume :
132
Issue :
22
Database :
Directory of Open Access Journals
Journal :
The Journal of Clinical Investigation
Publication Type :
Academic Journal
Accession number :
edsdoj.86291c23bf8d4ccda5764eae5949072f
Document Type :
article
Full Text :
https://doi.org/10.1172/JCI154229