4.6 Article

Optimal therapeutic targeting by HDAC inhibition in biopsy-derived treatment-naive diffuse midline glioma models

Journal

NEURO-ONCOLOGY
Volume 23, Issue 3, Pages 376-386

Publisher

OXFORD UNIV PRESS INC
DOI: 10.1093/neuonc/noaa249

Keywords

diffuse intrinsic pontine glioma (DIPG); diffuse midline glioma; H3 K27M-mutant (DMG); histone deacetylase inhibitor (HDACi); quisinostat; romidepsin

Funding

  1. National Cancer Institute [R01 CA114567]
  2. Seattle Run of Hope
  3. Pediatric Brain Tumor Research Fund Guild of Seattle Children's Hospital
  4. McKenna Claire Foundation
  5. Unravel Pediatric Cancer
  6. Team Cozzi Foundation
  7. Love for Lucy
  8. Julianna Sayler Foundation
  9. Grousemont Foundation
  10. Avery Huffman Defeat DIPG Foundation
  11. Michael Mosier Defeat DIPG Foundation
  12. ChadTough Foundation
  13. Kellen Joyce Heart of a Warrior Research Fund

Ask authors/readers for more resources

Diffuse midline gliomas, including diffuse intrinsic pontine gliomas, have poor prognosis with few effective treatment options. Studies have identified potential drugs targeting epigenetic regulation, such as HDAC inhibitors like Quisinostat and romidepsin. These inhibitors showed efficacy in inhibiting tumor growth in vivo and may serve as potential biomarkers for DMG.
Background. Diffuse midline gliomas (DMGs), including diffuse intrinsic pontine gliomas (DIPGs), have a dismal prognosis, with less than 2% surviving 5 years postdiagnosis. The majority of DIPGs and all DMGs harbor mutations altering the epigenetic regulatory histone tail (H3 K27M). Investigations addressing DMG epigenetics have identified a few promising drugs, including the HDAC inhibitor (HDACi) panobinostat. Here, we use clinically relevant DMG models to identify and validate other effective HDACi and their biomarkers of response. Methods. HDAC inhibitors were tested across biopsy-derived treatment-naive in vitro and in vivo DMG models with biologically relevant radiation resistance. RNA sequencing was performed to define and compare drug efficacy and to map predictive biomarkers of response. Results. Quisinostat and romidepsin showed efficacy with low nanomolar half-maximal inhibitory concentration (IC50) values (similar to 50 and similar to 5 nM, respectively). Comparative transcriptome analyses across quisinostat, romidepsin, and panobinostat showed a greater degree of shared biological effects between quisinostat and panobinostat, and less overlap with romidepsin. However, some transcriptional changes were consistent across all 3 drugs at similar biologically effective doses, such as overexpression of troponinT1 slow skeletal type (TNNT1) and downregulation of collagen type 20 alpha 1 chain (COL20A1), identifying these as potential vulnerabilities or on-target biomarkers in DMG. Quisinostat and romidepsin significantly (P < 0.0001) inhibited in vivo tumor growth. Conclusions. Our data highlight the utility of treatment-naive biopsy-derived models; establishes quisinostat and romidepsin as effective in vivo; illuminates potential mechanisms and/or biomarkers of DMG cell lethality due to HDAC inhibition; and emphasizes the need for brain tumor-penetrant versions of potentially efficacious agents.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available