Abstract

The evolution of cancer genomes within a single tumor creates mixed cell populations with divergent somatic mutational landscapes. Inference of tumor subpopulations has been disproportionately focused on the assessment of somatic point mutations, whereas computational methods targeting evolutionary dynamics of copy number alterations (CNA) and loss of heterozygosity (LOH) in whole-genome sequencing data remain underdeveloped. We present a novel probabilistic model, TITAN, to infer CNA and LOH events while accounting for mixtures of cell populations, thereby estimating the proportion of cells harboring each event. We evaluate TITAN on idealized mixtures, simulating clonal populations from whole-genome sequences taken from genomically heterogeneous ovarian tumor sites collected from the same patient. In addition, we show in 23 whole genomes of breast tumors that the inference of CNA and LOH using TITAN critically informs population structure and the nature of the evolving cancer genome. Finally, we experimentally validated subclonal predictions using fluorescence in situ hybridization (FISH) and single-cell sequencing from an ovarian cancer patient sample, thereby recapitulating the key modeling assumptions of TITAN.

Highlights

  • Tumor progression follows the principles of clonal evolution (Nowell 1976)

  • We present a rigorous evaluation of TITAN including: (1) single-cell sequencing and fluorescence in situ hybridization (FISH) experimental validation of predictions on WGS data from a highgrade serous ovarian tumor; (2) systematically engineered in silico www.genome.org mixtures with WGS data from multiple intrapatient samples; (3) artificially embedded copy number alterations (CNA) and loss of heterozygosity (LOH) events in diploid chromosomes; and (4) 23 triple negative breast cancer (TNBC) genomes

  • The model is predicated on four main assumptions: (1) joint analysis of allelic ratio and tumor sequence coverage at ;1–3 million heterozygous germline SNP loci reflects the underlying somatic genotype of the tumor; (2) segmental regions of CNA and LOH span tens to thousands of contiguous SNP loci; (3) the observed sequencing signal is an aggregated measure of heterogeneous cellular populations, including normal and tumor subpopulations (Fig. 2A,D); and (4) sets of genetic aberrations observed at similar cellular prevalences possibly co-occurred in the same clone

Read more

Summary

Introduction

Accumulation of genomic alterations is patterned by phylogenetic branching, creating a substrate for natural selection. This leads to the emergence of distinct cell populations (clones) with divergent genotypes and associated phenotypes (Aparicio and Caldas 2013). We define a clone as a population of cells related by descent from a unitary origin and uniquely identified by the complement of fixed genetic marks comprising its clonal genotype. We define the cellular prevalence of a somatic mutation as the proportion of cells harboring an aberration in the overall (bulk) tumor cell population (Aparicio and Caldas 2013). The dynamics of cellular prevalence of a mutation are reflective of growth

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