Proteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer

Abstract Precise biomarkers are needed to guide better diagnostics and therapeutics for basal-like breast cancer, for which DNA-dependent protein kinase catalytic subunit (DNA-PKcs) has been recently reported by the Clinical Proteomic Tumor Analysis Consortium as the most specific biomarker. We eval...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Karama Asleh, Nazia Riaz, Angela S. Cheng, Dongxia Gao, Samuel C. Y. Leung, Meenakshi Anurag, Torsten O. Nielsen
Formato: article
Lenguaje:EN
Publicado: Nature Portfolio 2021
Materias:
Acceso en línea:https://doaj.org/article/50da77342af4406d9748983cb7210dc1
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:50da77342af4406d9748983cb7210dc1
record_format dspace
spelling oai:doaj.org-article:50da77342af4406d9748983cb7210dc12021-12-02T14:54:51ZProteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer10.1038/s41523-021-00320-x2374-4677https://doaj.org/article/50da77342af4406d9748983cb7210dc12021-09-01T00:00:00Zhttps://doi.org/10.1038/s41523-021-00320-xhttps://doaj.org/toc/2374-4677Abstract Precise biomarkers are needed to guide better diagnostics and therapeutics for basal-like breast cancer, for which DNA-dependent protein kinase catalytic subunit (DNA-PKcs) has been recently reported by the Clinical Proteomic Tumor Analysis Consortium as the most specific biomarker. We evaluated DNA-PKcs expression in clinically-annotated breast cancer tissue microarrays and correlated results with immune biomarkers (training set: n = 300; validation set: n = 2401). Following a pre-specified study design per REMARK criteria, we found that high expression of DNA-PKcs was significantly associated with stromal and CD8 + tumor infiltrating lymphocytes. Within the basal-like subtype, tumors with low DNA-PKcs and high tumor-infiltrating lymphocytes displayed the most favourable survival. DNA-PKcs expression by immunohistochemistry identified estrogen receptor-positive cases with a basal-like gene expression subtype. Non-silent mutations in PRKDC were significantly associated with poor outcomes. Integrating DNA-PKcs expression with validated immune biomarkers could guide patient selection for DNA-PKcs targeting strategies, DNA-damaging agents, and their combination with an immune-checkpoint blockade.Karama AslehNazia RiazAngela S. ChengDongxia GaoSamuel C. Y. LeungMeenakshi AnuragTorsten O. NielsenNature PortfolioarticleNeoplasms. Tumors. Oncology. Including cancer and carcinogensRC254-282ENnpj Breast Cancer, Vol 7, Iss 1, Pp 1-13 (2021)
institution DOAJ
collection DOAJ
language EN
topic Neoplasms. Tumors. Oncology. Including cancer and carcinogens
RC254-282
spellingShingle Neoplasms. Tumors. Oncology. Including cancer and carcinogens
RC254-282
Karama Asleh
Nazia Riaz
Angela S. Cheng
Dongxia Gao
Samuel C. Y. Leung
Meenakshi Anurag
Torsten O. Nielsen
Proteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer
description Abstract Precise biomarkers are needed to guide better diagnostics and therapeutics for basal-like breast cancer, for which DNA-dependent protein kinase catalytic subunit (DNA-PKcs) has been recently reported by the Clinical Proteomic Tumor Analysis Consortium as the most specific biomarker. We evaluated DNA-PKcs expression in clinically-annotated breast cancer tissue microarrays and correlated results with immune biomarkers (training set: n = 300; validation set: n = 2401). Following a pre-specified study design per REMARK criteria, we found that high expression of DNA-PKcs was significantly associated with stromal and CD8 + tumor infiltrating lymphocytes. Within the basal-like subtype, tumors with low DNA-PKcs and high tumor-infiltrating lymphocytes displayed the most favourable survival. DNA-PKcs expression by immunohistochemistry identified estrogen receptor-positive cases with a basal-like gene expression subtype. Non-silent mutations in PRKDC were significantly associated with poor outcomes. Integrating DNA-PKcs expression with validated immune biomarkers could guide patient selection for DNA-PKcs targeting strategies, DNA-damaging agents, and their combination with an immune-checkpoint blockade.
format article
author Karama Asleh
Nazia Riaz
Angela S. Cheng
Dongxia Gao
Samuel C. Y. Leung
Meenakshi Anurag
Torsten O. Nielsen
author_facet Karama Asleh
Nazia Riaz
Angela S. Cheng
Dongxia Gao
Samuel C. Y. Leung
Meenakshi Anurag
Torsten O. Nielsen
author_sort Karama Asleh
title Proteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer
title_short Proteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer
title_full Proteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer
title_fullStr Proteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer
title_full_unstemmed Proteomics-derived basal biomarker DNA-PKcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer
title_sort proteomics-derived basal biomarker dna-pkcs is associated with intrinsic subtype and long-term clinical outcomes in breast cancer
publisher Nature Portfolio
publishDate 2021
url https://doaj.org/article/50da77342af4406d9748983cb7210dc1
work_keys_str_mv AT karamaasleh proteomicsderivedbasalbiomarkerdnapkcsisassociatedwithintrinsicsubtypeandlongtermclinicaloutcomesinbreastcancer
AT naziariaz proteomicsderivedbasalbiomarkerdnapkcsisassociatedwithintrinsicsubtypeandlongtermclinicaloutcomesinbreastcancer
AT angelascheng proteomicsderivedbasalbiomarkerdnapkcsisassociatedwithintrinsicsubtypeandlongtermclinicaloutcomesinbreastcancer
AT dongxiagao proteomicsderivedbasalbiomarkerdnapkcsisassociatedwithintrinsicsubtypeandlongtermclinicaloutcomesinbreastcancer
AT samuelcyleung proteomicsderivedbasalbiomarkerdnapkcsisassociatedwithintrinsicsubtypeandlongtermclinicaloutcomesinbreastcancer
AT meenakshianurag proteomicsderivedbasalbiomarkerdnapkcsisassociatedwithintrinsicsubtypeandlongtermclinicaloutcomesinbreastcancer
AT torstenonielsen proteomicsderivedbasalbiomarkerdnapkcsisassociatedwithintrinsicsubtypeandlongtermclinicaloutcomesinbreastcancer
_version_ 1718389382182862848