Characterization of MET Exon 14 Skipping Alterations (in NSCLC) and Identification of Potential Therapeutic Targets Using Whole Transcriptome Sequencing

Autor: So Yeon Kim, MD, Jun Yin, MD, Stephen Bohlman, MD, PhD, Phillip Walker, PhD, Sanja Dacic, MD, PhD, Chul Kim, MD, Hina Khan, MD, Stephen V. Liu, MD, Patrick C. Ma, MD, Misako Nagasaka, MD, PhD, Karen L. Reckamp, MD, Jim Abraham, PhD, Dipesh Uprety, MD, Feng Wang, PhD, Joanne Xiu, PhD, Jian Zhang, PhD, Haiying Cheng, MD, PhD, Balazs Halmos, MD
Jazyk: angličtina
Rok vydání: 2022
Předmět:
Zdroj: JTO Clinical and Research Reports, Vol 3, Iss 9, Pp 100381- (2022)
Druh dokumentu: article
ISSN: 2666-3643
DOI: 10.1016/j.jtocrr.2022.100381
Popis: Introduction: Genomic alterations in the juxtamembrane exon 14 splice sites in NSCLC lead to increased MET stability and oncogenesis. We present the largest cohort study of MET Exon 14 (METex14) using whole transcriptome sequencing. Methods: A total of 21,582 NSCLC tumor samples underwent complete genomic profiling with next-generation sequencing of DNA (592 Gene Panel, NextSeq, whole exome sequencing, NovaSeq) and RNA (NovaSeq, whole transcriptome sequencing). Clinicopathologic information including programmed death-ligand 1 and tumor mutational burden were collected and RNA expression for mutation subtypes and MET amplification were quantified. Immunogenic signatures and potential pathways of invasion were characterized using single-sample gene set enrichment analysis and mRNA gene signatures. Results: A total of 533tumors (2.47%) with METex14 were identified. The most common alterations were point mutations (49.5%) at donor splice sites. Most alterations translated to increased MET expression, with MET co-amplification resulting in synergistic increase in expression (q < 0.05). Common coalterations were amplifications of MDM2 (19.0% versus 1.8% wild-type [WT]), HMGA2 (13.2% versus 0.98% WT), and CDK4 (10.0% versus 1.5% WT) (q < 0.05). High programmed death-ligand 1 > 50% (52.5% versus 27.3% WT, q < 0.0001) and lower proportion of high tumor mutational burden (>10 mutations per megabase, 8.3% versus 36.7% WT, p < 0.0001) were associated with METex14, which were also enriched in both immunogenic signatures and immunosuppressive checkpoints. Pathways associated with METex14 included angiogenesis and apical junction pathways (q < 0.05). Conclusions: METex14 splicing alterations and MET co-amplification translated to higher and synergistic MET expression at the transcriptomic level. High frequencies of MDM2 and CDK4 co-amplifications and association with multiple immunosuppressive checkpoints and angiogenic pathways provide insight into potential actionable targets for combination strategies in METex14 NSCLC.
Databáze: Directory of Open Access Journals