Frontiers in Medicine (Dec 2023)

Druggable genomic landscapes of high-grade gliomas

  • Paola Ghanem,
  • Paola Ghanem,
  • Maria Fatteh,
  • Maria Fatteh,
  • David Olayinka Kamson,
  • Archana Balan,
  • Archana Balan,
  • Michael Chang,
  • Jessica Tao,
  • Jessica Tao,
  • Jaishri Blakeley,
  • Jaishri Blakeley,
  • The Johns Hopkins Molecular Tumor Board Investigators,
  • Jenna Canzoniero,
  • Jenna Canzoniero,
  • Stuart A. Grossman,
  • Stuart A. Grossman,
  • Kristen Marrone,
  • Karisa C. Schreck,
  • Karisa C. Schreck,
  • Karisa C. Schreck,
  • Valsamo Anagnostou,
  • Valsamo Anagnostou,
  • Christine Pratilas,
  • Taxiarchis Botsis,
  • Rena Xian,
  • Chris Gocke,
  • Tseh Ming-Lin,
  • Eitan Halper-Stromberg,
  • Ying Zou,
  • Kent Hardart,
  • Jonathan Spiker,
  • Kory Kreimeyer,
  • Ting He,
  • Katie Fiallos,
  • Dana Petry,
  • Kala Visvanathan,
  • Antonio Wolff,
  • Cesar Santa-Maria,
  • Raquel Nunez,
  • Christian Meyer,
  • John Laterra,
  • Vered Stearns,
  • Karen Smith,
  • Deborah Armstrong,
  • Rachel Karchin,
  • Katerina Karaindrou,
  • Lily Zandi,
  • Marta Majcherska,
  • Faith Too,
  • Monique Makell,
  • Jennifer Lehman,
  • Timsy Wanchoo,
  • Jaime Wehr,
  • Michael Conroy,
  • Selina Shiqing Teh.

DOI
https://doi.org/10.3389/fmed.2023.1254955
Journal volume & issue
Vol. 10

Abstract

Read online

BackgroundDespite the putatively targetable genomic landscape of high-grade gliomas, the long-term survival benefit of genomically-tailored targeted therapies remains discouraging.MethodsUsing glioblastoma (GBM) as a representative example of high-grade gliomas, we evaluated the clonal architecture and distribution of hotspot mutations in 388 GBMs from the Cancer Genome Atlas (TCGA). Mutations were matched with 54 targeted therapies, followed by a comprehensive evaluation of drug biochemical properties in reference to the drug’s clinical efficacy in high-grade gliomas. We then assessed clinical outcomes of a cohort of patients with high-grade gliomas with targetable mutations reviewed at the Johns Hopkins Molecular Tumor Board (JH MTB; n = 50).ResultsAmong 1,156 sequence alterations evaluated, 28.6% represented hotspots. While the frequency of hotspot mutations in GBM was comparable to cancer types with actionable hotspot alterations, GBMs harbored a higher fraction of subclonal mutations that affected hotspots (7.0%), compared to breast cancer (4.9%), lung cancer (4.4%), and melanoma (1.4%). In investigating the biochemical features of targeted therapies paired with recurring alterations, we identified a trend toward higher lipid solubility and lower IC50 in GBM cell lines among drugs with clinical efficacy. The drugs’ half-life, molecular weight, surface area and binding to efflux transporters were not associated with clinical efficacy. Among the JH MTB cohort of patients with IDH1 wild-type high-grade gliomas who received targeted therapies, trametinib monotherapy or in combination with dabrafenib conferred radiographic partial response in 75% of patients harboring BRAF or NF1 actionable mutations. Cabozantinib conferred radiographic partial response in two patients harboring a MET and a PDGFRA/KDR amplification. Patients with IDH1 wild-type gliomas that harbored actionable alterations who received genotype-matched targeted therapy had longer progression-free (PFS) and overall survival (OS; 7.37 and 14.72 respectively) than patients whose actionable alterations were not targeted (2.83 and 4.2 months respectively).ConclusionWhile multiple host, tumor and drug-related features may limit the delivery and efficacy of targeted therapies for patients with high-grade gliomas, genotype-matched targeted therapies confer favorable clinical outcomes. Further studies are needed to generate more data on the impact of biochemical features of targeted therapies on their clinical efficacy for high-grade gliomas.

Keywords