author_facet Maia, Ana
Gu, Zuguang
Koch, André
Berdiel‐Acer, Mireia
Will, Rainer
Schlesner, Matthias
Wiemann, Stefan
Maia, Ana
Gu, Zuguang
Koch, André
Berdiel‐Acer, Mireia
Will, Rainer
Schlesner, Matthias
Wiemann, Stefan
author Maia, Ana
Gu, Zuguang
Koch, André
Berdiel‐Acer, Mireia
Will, Rainer
Schlesner, Matthias
Wiemann, Stefan
spellingShingle Maia, Ana
Gu, Zuguang
Koch, André
Berdiel‐Acer, Mireia
Will, Rainer
Schlesner, Matthias
Wiemann, Stefan
Molecular Oncology
IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
Cancer Research
Genetics
Molecular Medicine
General Medicine
Oncology
author_sort maia, ana
spelling Maia, Ana Gu, Zuguang Koch, André Berdiel‐Acer, Mireia Will, Rainer Schlesner, Matthias Wiemann, Stefan 1574-7891 1878-0261 Wiley Cancer Research Genetics Molecular Medicine General Medicine Oncology http://dx.doi.org/10.1002/1878-0261.12905 <jats:p>Chemotherapy (CTX) remains the standard of care for most aggressive tumours, including breast cancer (BC). In BC chemotherapeutic regimens, the maximum tolerated dose of cytotoxic drugs is administered at regular intervals, and cancer cells can re‐grow or adapt during the resting periods between cycles. The impact of the tumour microenvironment on the fate of cancer cells after CTX remains poorly understood. Here, we show that paracrine signalling from CTX‐treated cancer cells to stromal fibroblasts can drive cancer cell recovery after cytotoxic drug withdrawal. Interferon β1 (IFNβ1) secreted by cancer cells following treatment with high doses of CTX instigates the acquisition of an anti‐viral state in stromal fibroblasts. This state is associated with an expression pattern here referred to as interferon signature (IFNS), which encompasses several interferon‐stimulated genes (ISGs), including numerous pro‐inflammatory cytokine genes. This crosstalk is an important driver of the expansion of BC cells after CTX, and IFNβ1 blockade in tumour cells abrogated their fibroblast‐dependent recovery potential. Analysis of human breast carcinomas supported a link between CTX‐induced IFNS in tumour stroma and poor response to CTX treatment. First, IFNβ1 expression in human breast carcinomas was found to inversely correlate with recurrence free survival (RFS). Second, using laser capture microdissection data sets, we show a higher expression of IFNS in the stromal tumour compartment compared to the epithelial one and this signature was found to be more prominent in more aggressive subtypes of BC (basal‐like), pointing to a pro‐tumorigenic role of this signature. Moreover, IFNS was associated with higher recurrence rates and a worse outcome in BC patients. Our study unravels a novel form of paracrine communication between cancer cells and fibroblasts that ultimately results in CTX resistance. Targeting this axis has the potential to improve CTX outcomes in patients with BC.</jats:p> IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment Molecular Oncology
doi_str_mv 10.1002/1878-0261.12905
facet_avail Online
Free
finc_class_facet Medizin
Biologie
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi8xODc4LTAyNjEuMTI5MDU
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi8xODc4LTAyNjEuMTI5MDU
institution DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
DE-L229
DE-D275
DE-Bn3
DE-Brt1
DE-D161
DE-Zwi2
DE-Gla1
DE-Zi4
imprint Wiley, 2021
imprint_str_mv Wiley, 2021
issn 1574-7891
1878-0261
issn_str_mv 1574-7891
1878-0261
language English
mega_collection Wiley (CrossRef)
match_str maia2021ifnb1secretedbybreastcancercellsundergoingchemotherapyreprogramsstromalfibroblaststosupporttumourgrowthaftertreatment
publishDateSort 2021
publisher Wiley
recordtype ai
record_format ai
series Molecular Oncology
source_id 49
title IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_unstemmed IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_full IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_fullStr IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_full_unstemmed IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_short IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_sort ifnβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
topic Cancer Research
Genetics
Molecular Medicine
General Medicine
Oncology
url http://dx.doi.org/10.1002/1878-0261.12905
publishDate 2021
physical 1308-1329
description <jats:p>Chemotherapy (CTX) remains the standard of care for most aggressive tumours, including breast cancer (BC). In BC chemotherapeutic regimens, the maximum tolerated dose of cytotoxic drugs is administered at regular intervals, and cancer cells can re‐grow or adapt during the resting periods between cycles. The impact of the tumour microenvironment on the fate of cancer cells after CTX remains poorly understood. Here, we show that paracrine signalling from CTX‐treated cancer cells to stromal fibroblasts can drive cancer cell recovery after cytotoxic drug withdrawal. Interferon β1 (IFNβ1) secreted by cancer cells following treatment with high doses of CTX instigates the acquisition of an anti‐viral state in stromal fibroblasts. This state is associated with an expression pattern here referred to as interferon signature (IFNS), which encompasses several interferon‐stimulated genes (ISGs), including numerous pro‐inflammatory cytokine genes. This crosstalk is an important driver of the expansion of BC cells after CTX, and IFNβ1 blockade in tumour cells abrogated their fibroblast‐dependent recovery potential. Analysis of human breast carcinomas supported a link between CTX‐induced IFNS in tumour stroma and poor response to CTX treatment. First, IFNβ1 expression in human breast carcinomas was found to inversely correlate with recurrence free survival (RFS). Second, using laser capture microdissection data sets, we show a higher expression of IFNS in the stromal tumour compartment compared to the epithelial one and this signature was found to be more prominent in more aggressive subtypes of BC (basal‐like), pointing to a pro‐tumorigenic role of this signature. Moreover, IFNS was associated with higher recurrence rates and a worse outcome in BC patients. Our study unravels a novel form of paracrine communication between cancer cells and fibroblasts that ultimately results in CTX resistance. Targeting this axis has the potential to improve CTX outcomes in patients with BC.</jats:p>
container_issue 5
container_start_page 1308
container_title Molecular Oncology
container_volume 15
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792348333274038278
geogr_code not assigned
last_indexed 2024-03-01T18:09:30.727Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=IFN%CE%B21+secreted+by+breast+cancer+cells+undergoing+chemotherapy+reprograms+stromal+fibroblasts+to+support+tumour+growth+after+treatment&rft.date=2021-05-01&genre=article&issn=1878-0261&volume=15&issue=5&spage=1308&epage=1329&pages=1308-1329&jtitle=Molecular+Oncology&atitle=IFN%CE%B21+secreted+by+breast+cancer+cells+undergoing+chemotherapy+reprograms+stromal+fibroblasts+to+support+tumour+growth+after+treatment&aulast=Wiemann&aufirst=Stefan&rft_id=info%3Adoi%2F10.1002%2F1878-0261.12905&rft.language%5B0%5D=eng
SOLR
_version_ 1792348333274038278
author Maia, Ana, Gu, Zuguang, Koch, André, Berdiel‐Acer, Mireia, Will, Rainer, Schlesner, Matthias, Wiemann, Stefan
author_facet Maia, Ana, Gu, Zuguang, Koch, André, Berdiel‐Acer, Mireia, Will, Rainer, Schlesner, Matthias, Wiemann, Stefan, Maia, Ana, Gu, Zuguang, Koch, André, Berdiel‐Acer, Mireia, Will, Rainer, Schlesner, Matthias, Wiemann, Stefan
author_sort maia, ana
container_issue 5
container_start_page 1308
container_title Molecular Oncology
container_volume 15
description <jats:p>Chemotherapy (CTX) remains the standard of care for most aggressive tumours, including breast cancer (BC). In BC chemotherapeutic regimens, the maximum tolerated dose of cytotoxic drugs is administered at regular intervals, and cancer cells can re‐grow or adapt during the resting periods between cycles. The impact of the tumour microenvironment on the fate of cancer cells after CTX remains poorly understood. Here, we show that paracrine signalling from CTX‐treated cancer cells to stromal fibroblasts can drive cancer cell recovery after cytotoxic drug withdrawal. Interferon β1 (IFNβ1) secreted by cancer cells following treatment with high doses of CTX instigates the acquisition of an anti‐viral state in stromal fibroblasts. This state is associated with an expression pattern here referred to as interferon signature (IFNS), which encompasses several interferon‐stimulated genes (ISGs), including numerous pro‐inflammatory cytokine genes. This crosstalk is an important driver of the expansion of BC cells after CTX, and IFNβ1 blockade in tumour cells abrogated their fibroblast‐dependent recovery potential. Analysis of human breast carcinomas supported a link between CTX‐induced IFNS in tumour stroma and poor response to CTX treatment. First, IFNβ1 expression in human breast carcinomas was found to inversely correlate with recurrence free survival (RFS). Second, using laser capture microdissection data sets, we show a higher expression of IFNS in the stromal tumour compartment compared to the epithelial one and this signature was found to be more prominent in more aggressive subtypes of BC (basal‐like), pointing to a pro‐tumorigenic role of this signature. Moreover, IFNS was associated with higher recurrence rates and a worse outcome in BC patients. Our study unravels a novel form of paracrine communication between cancer cells and fibroblasts that ultimately results in CTX resistance. Targeting this axis has the potential to improve CTX outcomes in patients with BC.</jats:p>
doi_str_mv 10.1002/1878-0261.12905
facet_avail Online, Free
finc_class_facet Medizin, Biologie
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi8xODc4LTAyNjEuMTI5MDU
imprint Wiley, 2021
imprint_str_mv Wiley, 2021
institution DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1, DE-L229, DE-D275, DE-Bn3, DE-Brt1, DE-D161, DE-Zwi2, DE-Gla1, DE-Zi4
issn 1574-7891, 1878-0261
issn_str_mv 1574-7891, 1878-0261
language English
last_indexed 2024-03-01T18:09:30.727Z
match_str maia2021ifnb1secretedbybreastcancercellsundergoingchemotherapyreprogramsstromalfibroblaststosupporttumourgrowthaftertreatment
mega_collection Wiley (CrossRef)
physical 1308-1329
publishDate 2021
publishDateSort 2021
publisher Wiley
record_format ai
recordtype ai
series Molecular Oncology
source_id 49
spelling Maia, Ana Gu, Zuguang Koch, André Berdiel‐Acer, Mireia Will, Rainer Schlesner, Matthias Wiemann, Stefan 1574-7891 1878-0261 Wiley Cancer Research Genetics Molecular Medicine General Medicine Oncology http://dx.doi.org/10.1002/1878-0261.12905 <jats:p>Chemotherapy (CTX) remains the standard of care for most aggressive tumours, including breast cancer (BC). In BC chemotherapeutic regimens, the maximum tolerated dose of cytotoxic drugs is administered at regular intervals, and cancer cells can re‐grow or adapt during the resting periods between cycles. The impact of the tumour microenvironment on the fate of cancer cells after CTX remains poorly understood. Here, we show that paracrine signalling from CTX‐treated cancer cells to stromal fibroblasts can drive cancer cell recovery after cytotoxic drug withdrawal. Interferon β1 (IFNβ1) secreted by cancer cells following treatment with high doses of CTX instigates the acquisition of an anti‐viral state in stromal fibroblasts. This state is associated with an expression pattern here referred to as interferon signature (IFNS), which encompasses several interferon‐stimulated genes (ISGs), including numerous pro‐inflammatory cytokine genes. This crosstalk is an important driver of the expansion of BC cells after CTX, and IFNβ1 blockade in tumour cells abrogated their fibroblast‐dependent recovery potential. Analysis of human breast carcinomas supported a link between CTX‐induced IFNS in tumour stroma and poor response to CTX treatment. First, IFNβ1 expression in human breast carcinomas was found to inversely correlate with recurrence free survival (RFS). Second, using laser capture microdissection data sets, we show a higher expression of IFNS in the stromal tumour compartment compared to the epithelial one and this signature was found to be more prominent in more aggressive subtypes of BC (basal‐like), pointing to a pro‐tumorigenic role of this signature. Moreover, IFNS was associated with higher recurrence rates and a worse outcome in BC patients. Our study unravels a novel form of paracrine communication between cancer cells and fibroblasts that ultimately results in CTX resistance. Targeting this axis has the potential to improve CTX outcomes in patients with BC.</jats:p> IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment Molecular Oncology
spellingShingle Maia, Ana, Gu, Zuguang, Koch, André, Berdiel‐Acer, Mireia, Will, Rainer, Schlesner, Matthias, Wiemann, Stefan, Molecular Oncology, IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment, Cancer Research, Genetics, Molecular Medicine, General Medicine, Oncology
title IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_full IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_fullStr IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_full_unstemmed IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_short IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_sort ifnβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
title_unstemmed IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment
topic Cancer Research, Genetics, Molecular Medicine, General Medicine, Oncology
url http://dx.doi.org/10.1002/1878-0261.12905