Cargando…

Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data

BACKGROUND: Protein biomarkers of cancer progression and response to therapy are increasingly important for improving personalized medicine. Advanced quantitative pathology platforms enable measurement of protein expression in tissues at the single-cell level. However, this rich quantitative cell-by...

Descripción completa

Detalles Bibliográficos
Autores principales: Yi, Misung, Zhan, Tingting, Peck, Amy R., Hooke, Jeffrey A., Kovatich, Albert J., Shriver, Craig D., Hu, Hai, Sun, Yunguang, Rui, Hallgeir, Chervoneva, Inna
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10363294/
https://www.ncbi.nlm.nih.gov/pubmed/37481512
http://dx.doi.org/10.1186/s12859-023-05408-8
_version_ 1785076593687789568
author Yi, Misung
Zhan, Tingting
Peck, Amy R.
Hooke, Jeffrey A.
Kovatich, Albert J.
Shriver, Craig D.
Hu, Hai
Sun, Yunguang
Rui, Hallgeir
Chervoneva, Inna
author_facet Yi, Misung
Zhan, Tingting
Peck, Amy R.
Hooke, Jeffrey A.
Kovatich, Albert J.
Shriver, Craig D.
Hu, Hai
Sun, Yunguang
Rui, Hallgeir
Chervoneva, Inna
author_sort Yi, Misung
collection PubMed
description BACKGROUND: Protein biomarkers of cancer progression and response to therapy are increasingly important for improving personalized medicine. Advanced quantitative pathology platforms enable measurement of protein expression in tissues at the single-cell level. However, this rich quantitative cell-by-cell biomarker information is most often not exploited. Instead, it is reduced to a single mean across the cells of interest or converted into a simple proportion of binary biomarker-positive or -negative cells. RESULTS: We investigated the utility of retaining all quantitative information at the single-cell level by considering the values of the quantile function (inverse of the cumulative distribution function) estimated from a sample of cell signal intensity levels in a tumor tissue. An algorithm was developed for selecting optimal cutoffs for dichotomizing cell signal intensity distribution quantiles as predictors of continuous, categorical or survival outcomes. The proposed algorithm was used to select optimal quantile biomarkers of breast cancer progression based on cancer cells’ cell signal intensity levels of nuclear protein Ki-67, Proliferating cell nuclear antigen, Programmed cell death 1 ligand 2, and Progesterone receptor. The performance of the resulting optimal quantile biomarkers was validated and compared to the standard cancer compartment mean signal intensity markers using an independent external validation cohort. For Ki-67, the optimal quantile biomarker was also compared to established biomarkers based on percentages of Ki67-positive cells. For proteins significantly associated with PFS in the external validation cohort, the optimal quantile biomarkers yielded either larger or similar effect size (hazard ratio for progression-free survival) as compared to cancer compartment mean signal intensity biomarkers. CONCLUSION: The optimal quantile protein biomarkers yield generally improved prognostic value as compared to the standard protein expression markers. The proposed methodology has a broad application to single-cell data from genomics, transcriptomics, proteomics, or metabolomics studies at the single cell level. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12859-023-05408-8.
format Online
Article
Text
id pubmed-10363294
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-103632942023-07-24 Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data Yi, Misung Zhan, Tingting Peck, Amy R. Hooke, Jeffrey A. Kovatich, Albert J. Shriver, Craig D. Hu, Hai Sun, Yunguang Rui, Hallgeir Chervoneva, Inna BMC Bioinformatics Research BACKGROUND: Protein biomarkers of cancer progression and response to therapy are increasingly important for improving personalized medicine. Advanced quantitative pathology platforms enable measurement of protein expression in tissues at the single-cell level. However, this rich quantitative cell-by-cell biomarker information is most often not exploited. Instead, it is reduced to a single mean across the cells of interest or converted into a simple proportion of binary biomarker-positive or -negative cells. RESULTS: We investigated the utility of retaining all quantitative information at the single-cell level by considering the values of the quantile function (inverse of the cumulative distribution function) estimated from a sample of cell signal intensity levels in a tumor tissue. An algorithm was developed for selecting optimal cutoffs for dichotomizing cell signal intensity distribution quantiles as predictors of continuous, categorical or survival outcomes. The proposed algorithm was used to select optimal quantile biomarkers of breast cancer progression based on cancer cells’ cell signal intensity levels of nuclear protein Ki-67, Proliferating cell nuclear antigen, Programmed cell death 1 ligand 2, and Progesterone receptor. The performance of the resulting optimal quantile biomarkers was validated and compared to the standard cancer compartment mean signal intensity markers using an independent external validation cohort. For Ki-67, the optimal quantile biomarker was also compared to established biomarkers based on percentages of Ki67-positive cells. For proteins significantly associated with PFS in the external validation cohort, the optimal quantile biomarkers yielded either larger or similar effect size (hazard ratio for progression-free survival) as compared to cancer compartment mean signal intensity biomarkers. CONCLUSION: The optimal quantile protein biomarkers yield generally improved prognostic value as compared to the standard protein expression markers. The proposed methodology has a broad application to single-cell data from genomics, transcriptomics, proteomics, or metabolomics studies at the single cell level. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12859-023-05408-8. BioMed Central 2023-07-22 /pmc/articles/PMC10363294/ /pubmed/37481512 http://dx.doi.org/10.1186/s12859-023-05408-8 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Yi, Misung
Zhan, Tingting
Peck, Amy R.
Hooke, Jeffrey A.
Kovatich, Albert J.
Shriver, Craig D.
Hu, Hai
Sun, Yunguang
Rui, Hallgeir
Chervoneva, Inna
Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data
title Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data
title_full Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data
title_fullStr Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data
title_full_unstemmed Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data
title_short Selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data
title_sort selection of optimal quantile protein biomarkers based on cell-level immunohistochemistry data
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10363294/
https://www.ncbi.nlm.nih.gov/pubmed/37481512
http://dx.doi.org/10.1186/s12859-023-05408-8
work_keys_str_mv AT yimisung selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT zhantingting selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT peckamyr selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT hookejeffreya selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT kovatichalbertj selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT shrivercraigd selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT huhai selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT sunyunguang selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT ruihallgeir selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata
AT chervonevainna selectionofoptimalquantileproteinbiomarkersbasedoncelllevelimmunohistochemistrydata