A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancer

Breast cancers of the luminal B subtype are frequent tumors with high proliferation and poor prognosis. Epigenetic alterations have been found in breast tumors and in biological fluids. We aimed to profile the cell-free DNA (cfDNA) methylome of metastatic luminal B breast cancer (LBBC) patients usin...

Full description

Bibliographic Details
Main Authors: Aitor Rodriguez-Casanova, Nicolas Costa-Fraga, Clara Castro-Carballeira, Miriam González-Conde, Carmen Abuin, Aida Bao-Caamano, Tomás García-Caballero, Elena Brozos-Vazquez, Carmela Rodriguez-López, Victor Cebey, Patricia Palacios, Juan F. Cueva, Rafael López-López, Clotilde Costa, Angel Díaz-Lagares
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-10-01
Series:Frontiers in Cell and Developmental Biology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fcell.2022.1016955/full
_version_ 1811234540646039552
author Aitor Rodriguez-Casanova
Aitor Rodriguez-Casanova
Aitor Rodriguez-Casanova
Nicolas Costa-Fraga
Nicolas Costa-Fraga
Clara Castro-Carballeira
Miriam González-Conde
Miriam González-Conde
Carmen Abuin
Aida Bao-Caamano
Aida Bao-Caamano
Tomás García-Caballero
Elena Brozos-Vazquez
Carmela Rodriguez-López
Victor Cebey
Patricia Palacios
Juan F. Cueva
Juan F. Cueva
Rafael López-López
Rafael López-López
Rafael López-López
Clotilde Costa
Clotilde Costa
Angel Díaz-Lagares
Angel Díaz-Lagares
author_facet Aitor Rodriguez-Casanova
Aitor Rodriguez-Casanova
Aitor Rodriguez-Casanova
Nicolas Costa-Fraga
Nicolas Costa-Fraga
Clara Castro-Carballeira
Miriam González-Conde
Miriam González-Conde
Carmen Abuin
Aida Bao-Caamano
Aida Bao-Caamano
Tomás García-Caballero
Elena Brozos-Vazquez
Carmela Rodriguez-López
Victor Cebey
Patricia Palacios
Juan F. Cueva
Juan F. Cueva
Rafael López-López
Rafael López-López
Rafael López-López
Clotilde Costa
Clotilde Costa
Angel Díaz-Lagares
Angel Díaz-Lagares
author_sort Aitor Rodriguez-Casanova
collection DOAJ
description Breast cancers of the luminal B subtype are frequent tumors with high proliferation and poor prognosis. Epigenetic alterations have been found in breast tumors and in biological fluids. We aimed to profile the cell-free DNA (cfDNA) methylome of metastatic luminal B breast cancer (LBBC) patients using an epigenomic approach to discover potential noninvasive biomarkers. Plasma cfDNA was analyzed using the Infinium MethylationEpic array in a cohort of 14 women, including metastatic LBBC patients and nontumor controls. The methylation levels of cfDNA and tissue samples were validated with droplet digital PCR. The methylation and gene expression data of 582 primary luminal breast tumors and 79 nontumor tissues were obtained from The Cancer Genome Atlas (TCGA). We found an episignature of 1,467 differentially methylated CpGs that clearly identified patients with LBBC. Among the genes identified, the promoter hypermethylation of WNT1 was validated in cfDNA, showing an area under the ROC curve (AUC) of 0.86 for the noninvasive detection of metastatic LBBC. Both paired cfDNA and primary/metastatic breast tumor samples showed hypermethylation of WNT1. TCGA analysis revealed significant WNT1 hypermethylation in the primary tumors of luminal breast cancer patients, with a negative association between WNT1 methylation and gene expression. In this proof-of-principle study, we discovered an episignature associated with metastatic LBBC using a genome-wide cfDNA methylation approach. We also identified the promoter hypermethylation of WNT1 in cfDNA as a potential noninvasive biomarker for luminal breast cancer. Our results support the use of EPIC arrays to identify new epigenetic noninvasive biomarkers in breast cancer.
first_indexed 2024-04-12T11:38:05Z
format Article
id doaj.art-eed70f27ab04426b97329ccbc70f0e54
institution Directory Open Access Journal
issn 2296-634X
language English
last_indexed 2024-04-12T11:38:05Z
publishDate 2022-10-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Cell and Developmental Biology
spelling doaj.art-eed70f27ab04426b97329ccbc70f0e542022-12-22T03:34:47ZengFrontiers Media S.A.Frontiers in Cell and Developmental Biology2296-634X2022-10-011010.3389/fcell.2022.10169551016955A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancerAitor Rodriguez-Casanova0Aitor Rodriguez-Casanova1Aitor Rodriguez-Casanova2Nicolas Costa-Fraga3Nicolas Costa-Fraga4Clara Castro-Carballeira5Miriam González-Conde6Miriam González-Conde7Carmen Abuin8Aida Bao-Caamano9Aida Bao-Caamano10Tomás García-Caballero11Elena Brozos-Vazquez12Carmela Rodriguez-López13Victor Cebey14Patricia Palacios15Juan F. Cueva16Juan F. Cueva17Rafael López-López18Rafael López-López19Rafael López-López20Clotilde Costa21Clotilde Costa22Angel Díaz-Lagares23Angel Díaz-Lagares24Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainRoche-Chus Joint Unit, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), Santiago de Compostela, SpainUniversidade de Santiago de Compostela (USC), Santiago de Compostela, SpainEpigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainUniversidade de Santiago de Compostela (USC), Santiago de Compostela, SpainDepartment of Oncology, Marqués de Valdecilla University Hospital, Santander, SpainRoche-Chus Joint Unit, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), Santiago de Compostela, SpainCentro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, SpainRoche-Chus Joint Unit, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), Santiago de Compostela, SpainEpigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainUniversidade de Santiago de Compostela (USC), Santiago de Compostela, SpainDepartment of Morphological Sciences, University of Santiago de Compostela and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, SpainTranslational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainTranslational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainTranslational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainTranslational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainCentro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, SpainTranslational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainRoche-Chus Joint Unit, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), Santiago de Compostela, SpainCentro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, SpainTranslational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainRoche-Chus Joint Unit, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), Santiago de Compostela, SpainCentro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, SpainEpigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, SpainCentro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, SpainBreast cancers of the luminal B subtype are frequent tumors with high proliferation and poor prognosis. Epigenetic alterations have been found in breast tumors and in biological fluids. We aimed to profile the cell-free DNA (cfDNA) methylome of metastatic luminal B breast cancer (LBBC) patients using an epigenomic approach to discover potential noninvasive biomarkers. Plasma cfDNA was analyzed using the Infinium MethylationEpic array in a cohort of 14 women, including metastatic LBBC patients and nontumor controls. The methylation levels of cfDNA and tissue samples were validated with droplet digital PCR. The methylation and gene expression data of 582 primary luminal breast tumors and 79 nontumor tissues were obtained from The Cancer Genome Atlas (TCGA). We found an episignature of 1,467 differentially methylated CpGs that clearly identified patients with LBBC. Among the genes identified, the promoter hypermethylation of WNT1 was validated in cfDNA, showing an area under the ROC curve (AUC) of 0.86 for the noninvasive detection of metastatic LBBC. Both paired cfDNA and primary/metastatic breast tumor samples showed hypermethylation of WNT1. TCGA analysis revealed significant WNT1 hypermethylation in the primary tumors of luminal breast cancer patients, with a negative association between WNT1 methylation and gene expression. In this proof-of-principle study, we discovered an episignature associated with metastatic LBBC using a genome-wide cfDNA methylation approach. We also identified the promoter hypermethylation of WNT1 in cfDNA as a potential noninvasive biomarker for luminal breast cancer. Our results support the use of EPIC arrays to identify new epigenetic noninvasive biomarkers in breast cancer.https://www.frontiersin.org/articles/10.3389/fcell.2022.1016955/fullDNA methylationEPIC Arraycell-free DNAliquid biopsymetastasisluminal B
spellingShingle Aitor Rodriguez-Casanova
Aitor Rodriguez-Casanova
Aitor Rodriguez-Casanova
Nicolas Costa-Fraga
Nicolas Costa-Fraga
Clara Castro-Carballeira
Miriam González-Conde
Miriam González-Conde
Carmen Abuin
Aida Bao-Caamano
Aida Bao-Caamano
Tomás García-Caballero
Elena Brozos-Vazquez
Carmela Rodriguez-López
Victor Cebey
Patricia Palacios
Juan F. Cueva
Juan F. Cueva
Rafael López-López
Rafael López-López
Rafael López-López
Clotilde Costa
Clotilde Costa
Angel Díaz-Lagares
Angel Díaz-Lagares
A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancer
Frontiers in Cell and Developmental Biology
DNA methylation
EPIC Array
cell-free DNA
liquid biopsy
metastasis
luminal B
title A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancer
title_full A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancer
title_fullStr A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancer
title_full_unstemmed A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancer
title_short A genome-wide cell-free DNA methylation analysis identifies an episignature associated with metastatic luminal B breast cancer
title_sort genome wide cell free dna methylation analysis identifies an episignature associated with metastatic luminal b breast cancer
topic DNA methylation
EPIC Array
cell-free DNA
liquid biopsy
metastasis
luminal B
url https://www.frontiersin.org/articles/10.3389/fcell.2022.1016955/full
work_keys_str_mv AT aitorrodriguezcasanova agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aitorrodriguezcasanova agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aitorrodriguezcasanova agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT nicolascostafraga agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT nicolascostafraga agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT claracastrocarballeira agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT miriamgonzalezconde agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT miriamgonzalezconde agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT carmenabuin agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aidabaocaamano agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aidabaocaamano agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT tomasgarciacaballero agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT elenabrozosvazquez agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT carmelarodriguezlopez agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT victorcebey agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT patriciapalacios agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT juanfcueva agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT juanfcueva agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT rafaellopezlopez agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT rafaellopezlopez agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT rafaellopezlopez agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT clotildecosta agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT clotildecosta agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT angeldiazlagares agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT angeldiazlagares agenomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aitorrodriguezcasanova genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aitorrodriguezcasanova genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aitorrodriguezcasanova genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT nicolascostafraga genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT nicolascostafraga genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT claracastrocarballeira genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT miriamgonzalezconde genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT miriamgonzalezconde genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT carmenabuin genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aidabaocaamano genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT aidabaocaamano genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT tomasgarciacaballero genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT elenabrozosvazquez genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT carmelarodriguezlopez genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT victorcebey genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT patriciapalacios genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT juanfcueva genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT juanfcueva genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT rafaellopezlopez genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT rafaellopezlopez genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT rafaellopezlopez genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT clotildecosta genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT clotildecosta genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT angeldiazlagares genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer
AT angeldiazlagares genomewidecellfreednamethylationanalysisidentifiesanepisignatureassociatedwithmetastaticluminalbbreastcancer