Abstract

Multistage tumorigenesis is a dynamic process characterized by the accumulation of mutations. Thus, a tumor mass is composed of genetically divergent cell subclones. With the advancement of next-generation sequencing (NGS), mathematical models have been recently developed to decompose tumor subclonal architecture from a collective genome sequencing data. Most of the methods focused on single-nucleotide variants (SNVs). However, somatic copy number aberrations (CNAs) also play critical roles in carcinogenesis. Therefore, further modeling subclonal CNAs composition would hold the promise to improve the analysis of tumor heterogeneity and cancer evolution. To address this issue, we developed a two-way mixture Poisson model, named CloneDeMix for the deconvolution of read-depth information. It can infer the subclonal copy number, mutational cellular prevalence (MCP), subclone composition, and the order in which mutations occurred in the evolutionary hierarchy. The performance of CloneDeMix was systematically assessed in simulations. As a result, the accuracy of CNA inference was nearly 93% and the MCP was also accurately restored. Furthermore, we also demonstrated its applicability using head and neck cancer samples from TCGA. Our results inform about the extent of subclonal CNA diversity, and a group of candidate genes that probably initiate lymph node metastasis during tumor evolution was also discovered. Most importantly, these driver genes are located at 11q13.3 which is highly susceptible to copy number change in head and neck cancer genomes. This study successfully estimates subclonal CNAs and exhibit the evolutionary relationships of mutation events. By doing so, we can track tumor heterogeneity and identify crucial mutations during evolution process. Hence, it facilitates not only understanding the cancer development but finding potential therapeutic targets. Briefly, this framework has implications for improved modeling of tumor evolution and the importance of inclusion of subclonal CNAs.

Highlights

  • Cancer, a dynamic disease, is characterized by unusual cells with somatic mutations

  • We demonstrated the performance of the algorithm with simulation data and applied it to a head and neck cancer dataset from The Cancer Genome Atlas (TCGA) and primary esophageal squamous cell carcinoma (ESCC) [19]

  • We developed CloneDeMix for the deconvolution of tumor progression through high-throughput DNA sequencing data

Read more

Summary

Introduction

The hypothesis for the somatic evolution of cancer was proposed in the 1970s [1] It states that all tumor cells descend from a single founder cell, and cells with some advantageous mutations become more competitive than normal cells for growth and clonal expansion. This hypothesis could be formed through random drift. To construct a phylogenetic tree, the mutations in each cancer cell should be measured to infer evolutionary relationships among various cells For addressing this concern, the current technology of single-cell sequencing seems appropriate [3, 4]. The cellular prevalence of each subclone have to be measured through the relative read count information of the variants

Objectives
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