Metabolic memory underlying minimal residual disease in breast cancer

Abstract Tumor relapse from treatment‐resistant cells (minimal residual disease, MRD) underlies most breast cancer‐related deaths. Yet, the molecular characteristics defining their malignancy have largely remained elusive. Here, we integrated multi‐omics data from a tractable organoid system with a...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Ksenija Radic Shechter, Eleni Kafkia, Katharina Zirngibl, Sylwia Gawrzak, Ashna Alladin, Daniel Machado, Christian Lüchtenborg, Daniel C Sévin, Britta Brügger, Kiran R Patil, Martin Jechlinger
Formato: article
Lenguaje:EN
Publicado: Wiley 2021
Materias:
Acceso en línea:https://doaj.org/article/c0276c0d71bf4b688df33f54bc4ee1f4
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:c0276c0d71bf4b688df33f54bc4ee1f4
record_format dspace
spelling oai:doaj.org-article:c0276c0d71bf4b688df33f54bc4ee1f42021-11-11T11:30:47ZMetabolic memory underlying minimal residual disease in breast cancer1744-429210.15252/msb.202010141https://doaj.org/article/c0276c0d71bf4b688df33f54bc4ee1f42021-10-01T00:00:00Zhttps://doi.org/10.15252/msb.202010141https://doaj.org/toc/1744-4292Abstract Tumor relapse from treatment‐resistant cells (minimal residual disease, MRD) underlies most breast cancer‐related deaths. Yet, the molecular characteristics defining their malignancy have largely remained elusive. Here, we integrated multi‐omics data from a tractable organoid system with a metabolic modeling approach to uncover the metabolic and regulatory idiosyncrasies of the MRD. We find that the resistant cells, despite their non‐proliferative phenotype and the absence of oncogenic signaling, feature increased glycolysis and activity of certain urea cycle enzyme reminiscent of the tumor. This metabolic distinctiveness was also evident in a mouse model and in transcriptomic data from patients following neo‐adjuvant therapy. We further identified a marked similarity in DNA methylation profiles between tumor and residual cells. Taken together, our data reveal a metabolic and epigenetic memory of the treatment‐resistant cells. We further demonstrate that the memorized elevated glycolysis in MRD is crucial for their survival and can be targeted using a small‐molecule inhibitor without impacting normal cells. The metabolic aberrances of MRD thus offer new therapeutic opportunities for post‐treatment care to prevent breast tumor recurrence.Ksenija Radic ShechterEleni KafkiaKatharina ZirngiblSylwia GawrzakAshna AlladinDaniel MachadoChristian LüchtenborgDaniel C SévinBritta BrüggerKiran R PatilMartin JechlingerWileyarticleglycolysismetabolic modelingmulti‐omics integrationoncogenic memoryorganoidsBiology (General)QH301-705.5Medicine (General)R5-920ENMolecular Systems Biology, Vol 17, Iss 10, Pp n/a-n/a (2021)
institution DOAJ
collection DOAJ
language EN
topic glycolysis
metabolic modeling
multi‐omics integration
oncogenic memory
organoids
Biology (General)
QH301-705.5
Medicine (General)
R5-920
spellingShingle glycolysis
metabolic modeling
multi‐omics integration
oncogenic memory
organoids
Biology (General)
QH301-705.5
Medicine (General)
R5-920
Ksenija Radic Shechter
Eleni Kafkia
Katharina Zirngibl
Sylwia Gawrzak
Ashna Alladin
Daniel Machado
Christian Lüchtenborg
Daniel C Sévin
Britta Brügger
Kiran R Patil
Martin Jechlinger
Metabolic memory underlying minimal residual disease in breast cancer
description Abstract Tumor relapse from treatment‐resistant cells (minimal residual disease, MRD) underlies most breast cancer‐related deaths. Yet, the molecular characteristics defining their malignancy have largely remained elusive. Here, we integrated multi‐omics data from a tractable organoid system with a metabolic modeling approach to uncover the metabolic and regulatory idiosyncrasies of the MRD. We find that the resistant cells, despite their non‐proliferative phenotype and the absence of oncogenic signaling, feature increased glycolysis and activity of certain urea cycle enzyme reminiscent of the tumor. This metabolic distinctiveness was also evident in a mouse model and in transcriptomic data from patients following neo‐adjuvant therapy. We further identified a marked similarity in DNA methylation profiles between tumor and residual cells. Taken together, our data reveal a metabolic and epigenetic memory of the treatment‐resistant cells. We further demonstrate that the memorized elevated glycolysis in MRD is crucial for their survival and can be targeted using a small‐molecule inhibitor without impacting normal cells. The metabolic aberrances of MRD thus offer new therapeutic opportunities for post‐treatment care to prevent breast tumor recurrence.
format article
author Ksenija Radic Shechter
Eleni Kafkia
Katharina Zirngibl
Sylwia Gawrzak
Ashna Alladin
Daniel Machado
Christian Lüchtenborg
Daniel C Sévin
Britta Brügger
Kiran R Patil
Martin Jechlinger
author_facet Ksenija Radic Shechter
Eleni Kafkia
Katharina Zirngibl
Sylwia Gawrzak
Ashna Alladin
Daniel Machado
Christian Lüchtenborg
Daniel C Sévin
Britta Brügger
Kiran R Patil
Martin Jechlinger
author_sort Ksenija Radic Shechter
title Metabolic memory underlying minimal residual disease in breast cancer
title_short Metabolic memory underlying minimal residual disease in breast cancer
title_full Metabolic memory underlying minimal residual disease in breast cancer
title_fullStr Metabolic memory underlying minimal residual disease in breast cancer
title_full_unstemmed Metabolic memory underlying minimal residual disease in breast cancer
title_sort metabolic memory underlying minimal residual disease in breast cancer
publisher Wiley
publishDate 2021
url https://doaj.org/article/c0276c0d71bf4b688df33f54bc4ee1f4
work_keys_str_mv AT ksenijaradicshechter metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT elenikafkia metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT katharinazirngibl metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT sylwiagawrzak metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT ashnaalladin metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT danielmachado metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT christianluchtenborg metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT danielcsevin metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT brittabrugger metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT kiranrpatil metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
AT martinjechlinger metabolicmemoryunderlyingminimalresidualdiseaseinbreastcancer
_version_ 1718439163934539776