A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.

A key constraint in genomic testing in oncology is that matched normal specimens are not commonly obtained in clinical practice. Thus, while well-characterized genomic alterations do not require normal tissue for interpretation, a significant number of alterations will be unknown in whether they are...

Full description

Bibliographic Details
Main Authors: James X Sun, Yuting He, Eric Sanford, Meagan Montesion, Garrett M Frampton, Stéphane Vignot, Jean-Charles Soria, Jeffrey S Ross, Vincent A Miller, Phil J Stephens, Doron Lipson, Roman Yelensky
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2018-02-01
Series:PLoS Computational Biology
Online Access:http://europepmc.org/articles/PMC5832436?pdf=render
id doaj-2c3540ac65e64877b4ea2ae6249ea3de
record_format Article
spelling doaj-2c3540ac65e64877b4ea2ae6249ea3de2020-11-25T02:20:15ZengPublic Library of Science (PLoS)PLoS Computational Biology1553-734X1553-73582018-02-01142e100596510.1371/journal.pcbi.1005965A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.James X SunYuting HeEric SanfordMeagan MontesionGarrett M FramptonStéphane VignotJean-Charles SoriaJeffrey S RossVincent A MillerPhil J StephensDoron LipsonRoman YelenskyA key constraint in genomic testing in oncology is that matched normal specimens are not commonly obtained in clinical practice. Thus, while well-characterized genomic alterations do not require normal tissue for interpretation, a significant number of alterations will be unknown in whether they are germline or somatic, in the absence of a matched normal control. We introduce SGZ (somatic-germline-zygosity), a computational method for predicting somatic vs. germline origin and homozygous vs. heterozygous or sub-clonal state of variants identified from deep massively parallel sequencing (MPS) of cancer specimens. The method does not require a patient matched normal control, enabling broad application in clinical research. SGZ predicts the somatic vs. germline status of each alteration identified by modeling the alteration's allele frequency (AF), taking into account the tumor content, tumor ploidy, and the local copy number. Accuracy of the prediction depends on the depth of sequencing and copy number model fit, which are achieved in our clinical assay by sequencing to high depth (>500x) using MPS, covering 394 cancer-related genes and over 3,500 genome-wide single nucleotide polymorphisms (SNPs). Calls are made using a statistic based on read depth and local variability of SNP AF. To validate the method, we first evaluated performance on samples from 30 lung and colon cancer patients, where we sequenced tumors and matched normal tissue. We examined predictions for 17 somatic hotspot mutations and 20 common germline SNPs in 20,182 clinical cancer specimens. To assess the impact of stromal admixture, we examined three cell lines, which were titrated with their matched normal to six levels (10-75%). Overall, predictions were made in 85% of cases, with 95-99% of variants predicted correctly, a significantly superior performance compared to a basic approach based on AF alone. We then applied the SGZ method to the COSMIC database of known somatic variants in cancer and found >50 that are in fact more likely to be germline.http://europepmc.org/articles/PMC5832436?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author James X Sun
Yuting He
Eric Sanford
Meagan Montesion
Garrett M Frampton
Stéphane Vignot
Jean-Charles Soria
Jeffrey S Ross
Vincent A Miller
Phil J Stephens
Doron Lipson
Roman Yelensky
spellingShingle James X Sun
Yuting He
Eric Sanford
Meagan Montesion
Garrett M Frampton
Stéphane Vignot
Jean-Charles Soria
Jeffrey S Ross
Vincent A Miller
Phil J Stephens
Doron Lipson
Roman Yelensky
A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.
PLoS Computational Biology
author_facet James X Sun
Yuting He
Eric Sanford
Meagan Montesion
Garrett M Frampton
Stéphane Vignot
Jean-Charles Soria
Jeffrey S Ross
Vincent A Miller
Phil J Stephens
Doron Lipson
Roman Yelensky
author_sort James X Sun
title A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.
title_short A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.
title_full A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.
title_fullStr A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.
title_full_unstemmed A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.
title_sort computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal.
publisher Public Library of Science (PLoS)
series PLoS Computational Biology
issn 1553-734X
1553-7358
publishDate 2018-02-01
description A key constraint in genomic testing in oncology is that matched normal specimens are not commonly obtained in clinical practice. Thus, while well-characterized genomic alterations do not require normal tissue for interpretation, a significant number of alterations will be unknown in whether they are germline or somatic, in the absence of a matched normal control. We introduce SGZ (somatic-germline-zygosity), a computational method for predicting somatic vs. germline origin and homozygous vs. heterozygous or sub-clonal state of variants identified from deep massively parallel sequencing (MPS) of cancer specimens. The method does not require a patient matched normal control, enabling broad application in clinical research. SGZ predicts the somatic vs. germline status of each alteration identified by modeling the alteration's allele frequency (AF), taking into account the tumor content, tumor ploidy, and the local copy number. Accuracy of the prediction depends on the depth of sequencing and copy number model fit, which are achieved in our clinical assay by sequencing to high depth (>500x) using MPS, covering 394 cancer-related genes and over 3,500 genome-wide single nucleotide polymorphisms (SNPs). Calls are made using a statistic based on read depth and local variability of SNP AF. To validate the method, we first evaluated performance on samples from 30 lung and colon cancer patients, where we sequenced tumors and matched normal tissue. We examined predictions for 17 somatic hotspot mutations and 20 common germline SNPs in 20,182 clinical cancer specimens. To assess the impact of stromal admixture, we examined three cell lines, which were titrated with their matched normal to six levels (10-75%). Overall, predictions were made in 85% of cases, with 95-99% of variants predicted correctly, a significantly superior performance compared to a basic approach based on AF alone. We then applied the SGZ method to the COSMIC database of known somatic variants in cancer and found >50 that are in fact more likely to be germline.
url http://europepmc.org/articles/PMC5832436?pdf=render
work_keys_str_mv AT jamesxsun acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT yutinghe acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT ericsanford acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT meaganmontesion acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT garrettmframpton acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT stephanevignot acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT jeancharlessoria acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT jeffreysross acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT vincentamiller acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT philjstephens acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT doronlipson acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT romanyelensky acomputationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT jamesxsun computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT yutinghe computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT ericsanford computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT meaganmontesion computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT garrettmframpton computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT stephanevignot computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT jeancharlessoria computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT jeffreysross computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT vincentamiller computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT philjstephens computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT doronlipson computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
AT romanyelensky computationalapproachtodistinguishsomaticvsgermlineoriginofgenomicalterationsfromdeepsequencingofcancerspecimenswithoutamatchednormal
_version_ 1724872594113429504