Glioma genetic profiles associated with electrophysiologic hyperexcitability

Author:

Tobochnik Steven12ORCID,Dorotan Maria Kristina C2,Ghosh Hia S3,Lapinskas Emily2,Vogelzang Jayne4,Reardon David A5,Ligon Keith L46,Bi Wenya Linda3,Smirnakis Stelios M12,Lee Jong Woo2

Affiliation:

1. Department of Neurology, VA Boston Healthcare System , Boston, Massachusetts , USA

2. Department of Neurology, Brigham and Women’s Hospital , Boston, Massachusetts , USA

3. Department of Neurosurgery, Brigham and Women’s Hospital , Boston, Massachusetts , USA

4. Department of Pathology, Dana-Farber Cancer Institute , Boston, Massachusetts , USA

5. Department of Medical Oncology, Center for Neuro-Oncology, Dana-Farber Cancer Institute , Boston, Massachusetts , USA

6. Department of Pathology, Brigham and Women’s Hospital , Boston, Massachusetts , USA

Abstract

Abstract Background Distinct genetic alterations determine glioma aggressiveness, however, the diversity of somatic mutations contributing to peritumoral hyperexcitability and seizures over the course of the disease is uncertain. This study aimed to identify tumor somatic mutation profiles associated with clinically significant hyperexcitability. Methods A single center cohort of adults with WHO grades 1–4 glioma and targeted exome sequencing (n = 1716) was analyzed and cross-referenced with a validated EEG database to identify the subset of individuals who underwent continuous EEG monitoring (n = 206). Hyperexcitability was defined by the presence of lateralized periodic discharges and/or electrographic seizures. Cross-validated discriminant analysis models trained exclusively on recurrent somatic mutations were used to identify variants associated with hyperexcitability. Results The distribution of WHO grades and tumor mutational burdens were similar between patients with and without hyperexcitability. Discriminant analysis models classified the presence or absence of EEG hyperexcitability with an overall accuracy of 70.9%, regardless of IDH1 R132H inclusion. Predictive variants included nonsense mutations in ATRX and TP53, indel mutations in RBBP8 and CREBBP, and nonsynonymous missense mutations with predicted damaging consequences in EGFR, KRAS, PIK3CA, TP53, and USP28. This profile improved estimates of hyperexcitability in a multivariate analysis controlling for age, sex, tumor location, integrated pathologic diagnosis, recurrence status, and preoperative epilepsy. Predicted somatic mutation variants were over-represented in patients with hyperexcitability compared to individuals without hyperexcitability and those who did not undergo continuous EEG. Conclusion These findings implicate diverse glioma somatic mutations in cancer genes associated with peritumoral hyperexcitability. Tumor genetic profiling may facilitate glioma-related epilepsy prognostication and management.

Funder

Department of Veterans Affairs, Veterans Health Administration

National Institute of Neurological Disorders and Stroke

Publisher

Oxford University Press (OUP)

Subject

Cancer Research,Neurology (clinical),Oncology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3