Login / Signup

Cooperativity between H3.3K27M and PDGFRA poses multiple therapeutic vulnerabilities in human iPSC-derived diffuse midline glioma avatars.

Kasey R SkinnerTomoyuki KogaShunichiro MikiRobert F GruenerFlorina-Nicoleta GrigoreEmma H ToriiDavis M SeeligYuta SuzukiDaisuke KawauchiBenjamin LinDenise M MalickiClark C ChenEtty N BenvenisteRakesh P PatelBraden C McFarlandRong Stephanie HuangChris JonesAlan MackayC Ryan MillerFrank B Furnari
Published in: bioRxiv : the preprint server for biology (2023)
Diffuse midline glioma (DMG) is a leading cause of brain tumor death in children. In addition to hallmark H3.3K27M mutations, significant subsets also harbor alterations of other genes, such as TP53 and PDGFRA . Despite the prevalence of H3.3K27M, the results of clinical trials in DMG have been mixed, possibly due to the lack of models recapitulating its genetic heterogeneity. To address this gap, we developed human iPSC-derived tumor models harboring TP53 R248Q with or without heterozygous H3.3K27M and/or PDGFRA D842V overexpression. The combination of H3.3K27M and PDGFRA D842V resulted in more proliferative tumors when gene-edited neural progenitor (NP) cells were implanted into mouse brains compared to NP with either mutation alone. Transcriptomic comparison of tumors and their NP cells of origin identified conserved JAK/STAT pathway activation across genotypes as characteristic of malignant transformation. Conversely, integrated genome-wide epigenomic and transcriptomic analyses, as well as rational pharmacologic inhibition, revealed targetable vulnerabilities unique to the TP53 R248Q ; H3.3K27M; PDGFRA D842V tumors and related to their aggressive growth phenotype. These include AREG -mediated cell cycle control, altered metabolism, and vulnerability to combination ONC201/trametinib treatment. Taken together, these data suggest that cooperation between H3.3K27M and PDGFRA influences tumor biology, underscoring the need for better molecular stratification in DMG clinical trials.
Keyphrases