Academic Journal

DDRE-32. THERAPEUTIC TARGETING OF A NOVEL METABOLIC ADDICTION IN DIFFUSE MIDLINE GLIOMA

Bibliographic Details
Title: DDRE-32. THERAPEUTIC TARGETING OF A NOVEL METABOLIC ADDICTION IN DIFFUSE MIDLINE GLIOMA
Authors: Pal, Sharmistha, Kaplan, Jakub P, Stopka, Sylwia A, Regan, Michael S, Hunsel, Bradley R, Kann, Benjamin H, Agar, Nathalie Y R, Stiles, Charles D, Cooney, Tabitha M, Mueller, Sabine, Chowdhury, Dipanjan, Kaelin, William G, McBrayer, Samuel K, Haas-Kogan, Daphne
Superior Title: Neurooncol Adv
Publisher Information: Oxford University Press
Publication Year: 2021
Collection: PubMed Central (PMC)
Subject Terms: Supplement Abstracts
Description: Diffuse midline glioma (DMG) is a uniformly fatal pediatric cancer that is in need of urgent “outside the box” therapeutic approaches. Recent studies show that tumor cells adapt to stresses created by oncogenic mutations and these oncogene-induced adaptations create vulnerabilities that can be exploited to therapeutic ends. To uncover these oncogene-induced vulnerabilities in DMGs we conducted a genome-wide CRIPSR knockout screen in three DMG lines. The top common DMG dependency pathway that we discovered is de novo pyrimidine biosynthesis. Under normal conditions pyrimidine nucleotide needs are met through the salvage pathway. However, in DMG tumorigenesis, pyrimidine nucleotide synthesis is rewired such that the cells become dependent on the de novo biosynthesis pathway. De novo pyrimidine synthesis is catalyzed by CAD, DHODH and UMPS; all three genes are identified as dependencies in our screen and have been validated using shRNA mediated gene knockdown. Interestingly, DMG cells did not exhibit a dependency on the de novo purine biosynthesis pathway. Using a small molecule inhibitor of DHODH, BAY2402234 [currently studied in phase I trial for myeloid malignancies (NCT03404726)], we have demonstrated and validated, (i) efficacy and specificity of de novo pyrimidine synthesis inhibition in vitro in DMG cells; (ii) de novo pyrimidine addiction is not attributable to cell proliferation; (iii) DHODH inhibition induces apoptosis by hindering replication and inciting DNA damage; (iv) DHODH and ATR inhibition act synergistically to induce DMG cell death; and (v) critical in vivo efficacy. The in vivo experiment documents that BAY2402234 crosses the blood-brain barrier, is present in the brain at therapeutically relevant concentrations, suppresses de novo pyrimidine biosynthesis in intracranial DMG tumors in mice, and prolongs survival of orthotopic DMG tumor bearing mice. Taken together, our studies have identified a novel metabolic vulnerability that can be translated for the treatment of DMG patients.
Document Type: text
Language: English
Relation: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC7992269/; http://dx.doi.org/10.1093/noajnl/vdab024.054
DOI: 10.1093/noajnl/vdab024.054
Availability: https://doi.org/10.1093/noajnl/vdab024.054
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC7992269/
Rights: © The Author(s) 2021. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology. ; http://creativecommons.org/licenses/by-nc/4.0/ ; This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
Accession Number: edsbas.EBCE082
Database: BASE
Description
Description not available.