Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.

Driven by genomic somatic variation, tumour tissues are typically heterogeneous, yet unbiased quantitative methods are rarely used to analyse heterogeneity at the protein level. Motivated by this problem, we developed automated image segmentation of images of multiple biomarkers in Ewing sarcoma to...

Full description

Bibliographic Details
Main Authors: Claudia Bühnemann, Simon Li, Haiyue Yu, Harriet Branford White, Karl L Schäfer, Antonio Llombart-Bosch, Isidro Machado, Piero Picci, Pancras C W Hogendoorn, Nicholas A Athanasou, J Alison Noble, A Bassim Hassan
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2014-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC4171480?pdf=render
id doaj-86e31c89d9c640f7ae6a93847fb8ffb6
record_format Article
spelling doaj-86e31c89d9c640f7ae6a93847fb8ffb62020-11-24T21:48:25ZengPublic Library of Science (PLoS)PLoS ONE1932-62032014-01-0199e10710510.1371/journal.pone.0107105Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.Claudia BühnemannSimon LiHaiyue YuHarriet Branford WhiteKarl L SchäferAntonio Llombart-BoschIsidro MachadoPiero PicciPancras C W HogendoornNicholas A AthanasouJ Alison NobleA Bassim HassanDriven by genomic somatic variation, tumour tissues are typically heterogeneous, yet unbiased quantitative methods are rarely used to analyse heterogeneity at the protein level. Motivated by this problem, we developed automated image segmentation of images of multiple biomarkers in Ewing sarcoma to generate distributions of biomarkers between and within tumour cells. We further integrate high dimensional data with patient clinical outcomes utilising random survival forest (RSF) machine learning. Using material from cohorts of genetically diagnosed Ewing sarcoma with EWSR1 chromosomal translocations, confocal images of tissue microarrays were segmented with level sets and watershed algorithms. Each cell nucleus and cytoplasm were identified in relation to DAPI and CD99, respectively, and protein biomarkers (e.g. Ki67, pS6, Foxo3a, EGR1, MAPK) localised relative to nuclear and cytoplasmic regions of each cell in order to generate image feature distributions. The image distribution features were analysed with RSF in relation to known overall patient survival from three separate cohorts (185 informative cases). Variation in pre-analytical processing resulted in elimination of a high number of non-informative images that had poor DAPI localisation or biomarker preservation (67 cases, 36%). The distribution of image features for biomarkers in the remaining high quality material (118 cases, 104 features per case) were analysed by RSF with feature selection, and performance assessed using internal cross-validation, rather than a separate validation cohort. A prognostic classifier for Ewing sarcoma with low cross-validation error rates (0.36) was comprised of multiple features, including the Ki67 proliferative marker and a sub-population of cells with low cytoplasmic/nuclear ratio of CD99. Through elimination of bias, the evaluation of high-dimensionality biomarker distribution within cell populations of a tumour using random forest analysis in quality controlled tumour material could be achieved. Such an automated and integrated methodology has potential application in the identification of prognostic classifiers based on tumour cell heterogeneity.http://europepmc.org/articles/PMC4171480?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Claudia Bühnemann
Simon Li
Haiyue Yu
Harriet Branford White
Karl L Schäfer
Antonio Llombart-Bosch
Isidro Machado
Piero Picci
Pancras C W Hogendoorn
Nicholas A Athanasou
J Alison Noble
A Bassim Hassan
spellingShingle Claudia Bühnemann
Simon Li
Haiyue Yu
Harriet Branford White
Karl L Schäfer
Antonio Llombart-Bosch
Isidro Machado
Piero Picci
Pancras C W Hogendoorn
Nicholas A Athanasou
J Alison Noble
A Bassim Hassan
Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.
PLoS ONE
author_facet Claudia Bühnemann
Simon Li
Haiyue Yu
Harriet Branford White
Karl L Schäfer
Antonio Llombart-Bosch
Isidro Machado
Piero Picci
Pancras C W Hogendoorn
Nicholas A Athanasou
J Alison Noble
A Bassim Hassan
author_sort Claudia Bühnemann
title Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.
title_short Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.
title_full Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.
title_fullStr Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.
title_full_unstemmed Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.
title_sort quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2014-01-01
description Driven by genomic somatic variation, tumour tissues are typically heterogeneous, yet unbiased quantitative methods are rarely used to analyse heterogeneity at the protein level. Motivated by this problem, we developed automated image segmentation of images of multiple biomarkers in Ewing sarcoma to generate distributions of biomarkers between and within tumour cells. We further integrate high dimensional data with patient clinical outcomes utilising random survival forest (RSF) machine learning. Using material from cohorts of genetically diagnosed Ewing sarcoma with EWSR1 chromosomal translocations, confocal images of tissue microarrays were segmented with level sets and watershed algorithms. Each cell nucleus and cytoplasm were identified in relation to DAPI and CD99, respectively, and protein biomarkers (e.g. Ki67, pS6, Foxo3a, EGR1, MAPK) localised relative to nuclear and cytoplasmic regions of each cell in order to generate image feature distributions. The image distribution features were analysed with RSF in relation to known overall patient survival from three separate cohorts (185 informative cases). Variation in pre-analytical processing resulted in elimination of a high number of non-informative images that had poor DAPI localisation or biomarker preservation (67 cases, 36%). The distribution of image features for biomarkers in the remaining high quality material (118 cases, 104 features per case) were analysed by RSF with feature selection, and performance assessed using internal cross-validation, rather than a separate validation cohort. A prognostic classifier for Ewing sarcoma with low cross-validation error rates (0.36) was comprised of multiple features, including the Ki67 proliferative marker and a sub-population of cells with low cytoplasmic/nuclear ratio of CD99. Through elimination of bias, the evaluation of high-dimensionality biomarker distribution within cell populations of a tumour using random forest analysis in quality controlled tumour material could be achieved. Such an automated and integrated methodology has potential application in the identification of prognostic classifiers based on tumour cell heterogeneity.
url http://europepmc.org/articles/PMC4171480?pdf=render
work_keys_str_mv AT claudiabuhnemann quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT simonli quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT haiyueyu quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT harrietbranfordwhite quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT karllschafer quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT antoniollombartbosch quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT isidromachado quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT pieropicci quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT pancrascwhogendoorn quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT nicholasaathanasou quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT jalisonnoble quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
AT abassimhassan quantificationoftheheterogeneityofprognosticcellularbiomarkersinewingsarcomausingautomatedimageandrandomsurvivalforestanalysis
_version_ 1725892274917736448