AUTHOR=Ghanem Paola , Fatteh Maria , Kamson David Olayinka , Balan Archana , Chang Michael , Tao Jessica , Blakeley Jaishri , The Johns Hopkins Molecular Tumor Board Investigators , Canzoniero Jenna , Grossman Stuart A. , Marrone Kristen , Schreck Karisa C. , Anagnostou Valsamo , Pratilas Christine , Botsis Taxiarchis , Xian Rena , Gocke Chris , Ming-Lin Tseh , Halper-Stromberg Eitan , Zou Ying , Hardart Kent , Spiker Jonathan , Kreimeyer Kory , He Ting , Fiallos Katie , Petry Dana , Visvanathan Kala , Wolff Antonio , Santa-Maria Cesar , Nunez Raquel , Meyer Christian , Laterra John , Stearns Vered , Smith Karen , Armstrong Deborah , Karchin Rachel , Karaindrou Katerina , Zandi Lily , Majcherska Marta , Too Faith , Makell Monique , Lehman Jennifer , Wanchoo Timsy , Wehr Jaime , Conroy Michael , Shiqing Teh. Selina TITLE=Druggable genomic landscapes of high-grade gliomas JOURNAL=Frontiers in Medicine VOLUME=10 YEAR=2023 URL=https://www.frontiersin.org/journals/medicine/articles/10.3389/fmed.2023.1254955 DOI=10.3389/fmed.2023.1254955 ISSN=2296-858X ABSTRACT=Background

Despite the putatively targetable genomic landscape of high-grade gliomas, the long-term survival benefit of genomically-tailored targeted therapies remains discouraging.

Methods

Using 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).

Results

Among 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).

Conclusion

While 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.