Abstract

The Cancer Genome Atlas (TCGA) projects have advanced our understanding of the driver mutations, genetic backgrounds, and key pathways activated across cancer types. Analysis of TCGA datasets have mostly focused on somatic mutations and translocations, with less emphasis placed on gene amplifications. Here we describe a bioinformatics screening strategy to identify putative cancer driver genes amplified across TCGA datasets. We carried out GISTIC2 analysis of TCGA datasets spanning 14 cancer subtypes and identified 461 genes that were amplified in two or more datasets. The list was narrowed to 73 cancer-associated genes with potential “druggable” properties. The majority of the genes were localized to 14 amplicons spread across the genome. To identify potential cancer driver genes, we analyzed gene copy number and mRNA expression data from individual patient samples and identified 40 putative cancer driver genes linked to diverse oncogenic processes. Oncogenic activity was further validated by siRNA/shRNA knockdown and by referencing the Project Achilles datasets. The amplified genes represented a number of gene families, including epigenetic regulators, cell cycle-associated genes, DNA damage response/repair genes, metabolic regulators, and genes linked to the Wnt, Notch, Hedgehog, JAK/STAT, NF-KB and MAPK signaling pathways. Among the 40 putative driver genes were known driver genes, such as EGFR, ERBB2 and PIK3CA. Wild-type KRAS was amplified in several cancer types, and KRAS-amplified cancer cell lines were most sensitive to KRAS shRNA, suggesting that KRAS amplification was an independent oncogenic event. A number of MAP kinase adapters were co-amplified with their receptor tyrosine kinases, such as the FGFR adapter FRS2 and the EGFR family adapter GRB7. The ubiquitin-like ligase DCUN1D1 and the histone methyltransferase NSD3 were also identified as novel putative cancer driver genes. We discuss the patient tailoring implications for existing cancer drug targets and we further discuss potential novel opportunities for drug discovery efforts.

Highlights

  • Recent advancements in DNA sequencing technology have enabled the sequencing of whole cancer genomes and identification of commonly mutated, amplified, and deleted genes across cancer types

  • The gene list was cross-referenced with the Cancer Genes database, which showed that less than 25% of the 461 genes were linked to oncogenesis

  • We carried out a GISTIC2 analysis of gene amplifications in The Cancer Genome Atlas (TCGA) datasets and identified a number of amplified genes with cancer driver activity

Read more

Summary

Introduction

Recent advancements in DNA sequencing technology have enabled the sequencing of whole cancer genomes and identification of commonly mutated, amplified, and deleted genes across cancer types. Further analysis revealed pathway-specific genetic driver mutations in breast cancer subtypes, such as BRCA1/2 alterations and PIK3CA alterations in basal-like and luminal breast cancers, respectively [4]. Eleven genes were commonly mutated, including TP53, oxidative stress response genes and squamous differentiation genes [1]. These studies have shed light into the major genetic drivers of cancer subtypes and have identified potentially druggable pathways linked to these subtypes. Since gene amplification is an important mechanism of carcinogenesis, we sought to mine the TCGA datasets to identify novel targets and drivers amplified across cancer types

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call