The epileptic landscape of IDH mutant gliomas

Autor: Michael Drumm, Wenxia Wang, Kirsten Bell Bell Burdett, Thomas Sears, Rodrigo Javier, Kristen Cotton, Brynna Webb, Dusten Unruh, Jordain Walshon, Alicia Steffens, Kathleen McCortney, Omar Bushara, Alyssa Weston, Suning He, John Finan, Colin Franz, Jonathan Kurz, Jessica Templer, Geoff Swanson, Craig M. Horbinski
Rok vydání: 2022
Předmět:
Zdroj: Journal of Clinical Oncology. 40:2064-2064
ISSN: 1527-7755
0732-183X
Popis: 2064 Background: Tumor-associated epilepsy (TAE) is a frequent complication of diffusely infiltrative gliomas. TAE not only impairs quality-of-life, but it can even be life-threatening. Furthermore, glioma cells have been shown to proliferate and migrate faster when exposed to firing neurons. We previously showed that isocitrate dehydrogenase 1 mutant (IDHmut) gliomas were more likely to cause preoperative seizures than IDH wildtype (IDHwt) gliomas, and that the chemical product of IDHmut, D2HG, increased synchronized network bursts in cultured neurons (PMID: 28404805). But the mechanism whereby this occurs, whether IDHmut inhibitors can block this, and whether seizure risk also varies by IDHmut status postoperatively, are unknown. Methods: Methods are embedded in Results. Results: We discovered that exogenous 3 mM D2HG had a 150% greater effect on the firing rate of cultured mouse cortical neurons when nonneoplastic glia cells were present versus when they were absent ( P=0.002). Although a recently published study suggested that D2HG causes seizures through mTOR activation (PMID: 34994387), we found that D2HG reduced neuronal mTOR activity in neuronal-glial cocultures by 54% ( P=0.0004). Patch clamp analyses showed that, while D2HG does not directly activate glutamate receptors, it does act as a glutamate transport substrate, thus potentially interfering with the ability of glial cells to take up glutamate released into the synaptic cleft. Coculture with patient-derived IDHmut glioma cells increased the firing rate of human cortical neuron/astrocyte spheroids by up to 272%, and an IDHmut inhibitor currently being tested in clinical trials, AG881, reduced the excitatory effect of IDHmut glioma cells on spheroids by 79% ( P=0.0008). Using a novel in vivo model of TAE, wherein EEG recordings were taken of immunocompetent mice engrafted with an isogenic pair of Sleeping Beauty transposase-engineered mouse glioma lines (NRAS/ATRX/TP53mut ± IDHmut), we found that IDHmut gliomas produced 7.6-fold more epileptiform spikes than IDHwt gliomas ( P=0.004). RNA-Seq analysis of the peritumoral mouse brain tissue showed that this increase in spikes was associated with significantly increased expression of key genes known to be upregulated in epilepsy, including SLC12A5, THSB1, VEGFA, FOSL2, and SYNPO. Treatment with 5 mg/kg AG881 by daily oral gavage reduced those spikes in IDHmut-engrafted mice by 51% within three days ( P=0.027), whereas vehicle control had no effect ( P=0.33). Among 247 patients with grade 2–4 adult-type diffuse gliomas, multivariable time-to-event analysis showed that postoperative seizure risk increased with preoperative seizures, subtotal resection, and IDHmut astrocytoma. Conclusions: Together, these data show that (i) the D2HG product of IDHmut gliomas increases neuronal excitation in a glial-dependent manner; (ii) IDHmut also affects postoperative seizure risk; (iii) IDHmut inhibitors may improve TAE control.
Databáze: OpenAIRE