Cargando…

Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models

OBJECTIVE: To investigate the clinical implications of 17β-estradiol (E2) in estrogen receptor α (ERα)-negative female cancer progression as well as the underlying biological mechanisms. METHODS: Clinical data from 306 locally-advanced cervical cancer (stage IIB-IVA) patients were analyzed in order...

Descripción completa

Detalles Bibliográficos
Autores principales: Kozasa, Katsumi, Mabuchi, Seiji, Matsumoto, Yuri, Kuroda, Hiromasa, Yokoi, Eriko, Komura, Naoko, Kawano, Mahiru, Takahashi, Ryoko, Sasano, Tomoyuki, Shimura, Kotaro, Kodama, Michiko, Hashimoto, Kae, Sawada, Kenjiro, Nagasaka, Kazunori, Kimura, Tadashi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6443012/
https://www.ncbi.nlm.nih.gov/pubmed/30956772
http://dx.doi.org/10.18632/oncotarget.26711
_version_ 1783407786580770816
author Kozasa, Katsumi
Mabuchi, Seiji
Matsumoto, Yuri
Kuroda, Hiromasa
Yokoi, Eriko
Komura, Naoko
Kawano, Mahiru
Takahashi, Ryoko
Sasano, Tomoyuki
Shimura, Kotaro
Kodama, Michiko
Hashimoto, Kae
Sawada, Kenjiro
Nagasaka, Kazunori
Kimura, Tadashi
author_facet Kozasa, Katsumi
Mabuchi, Seiji
Matsumoto, Yuri
Kuroda, Hiromasa
Yokoi, Eriko
Komura, Naoko
Kawano, Mahiru
Takahashi, Ryoko
Sasano, Tomoyuki
Shimura, Kotaro
Kodama, Michiko
Hashimoto, Kae
Sawada, Kenjiro
Nagasaka, Kazunori
Kimura, Tadashi
author_sort Kozasa, Katsumi
collection PubMed
description OBJECTIVE: To investigate the clinical implications of 17β-estradiol (E2) in estrogen receptor α (ERα)-negative female cancer progression as well as the underlying biological mechanisms. METHODS: Clinical data from 306 locally-advanced cervical cancer (stage IIB-IVA) patients were analyzed in order to investigate the relationships between age, serum E2 levels, and treatment outcomes. Clinical samples, ERα-negative cervical and breast cancer cell lines, and mouse xenograft models of cervical and breast cancers were employed in order to elucidate the mechanisms responsible for the E2- and pregnancy-mediated progression of cervical and breast cancers, with a focus on the role of myeloid-derived suppressor cells (MDSC). RESULTS: Younger patients with elevated E2 levels showed significantly shorter progression-free survival (P = 0.040) and overall survival (P = 0.039). The exogenous E2 treatment stimulated the mobilization of MDSC from bone marrow and directly augmented their suppressive activities, leading to the progression of ERα-negative cervical and breast cancers. The co-administration of an anti-Gr-1 neutralizing antibody with E2 prevented the E2-mediated induction of MDSC, and attenuated E2-mediated tumor growth in cervical and breast cancer xenografts. Significantly increased MDSC numbers and enhanced tumor growth were observed during pregnancy in mice with cervical or breast cancer. Significantly increased MDSC numbers were also observed during pregnancy in cervical cancer patients. CONCLUSIONS: E2 facilitates the progression of ERα-negative cervical or breast cancer under non-pregnant and pregnant conditions by inducing MDSC. MDSC inhibition therapy may have therapeutic efficacy in premenopausal or pregnant female cancer patients.
format Online
Article
Text
id pubmed-6443012
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-64430122019-04-05 Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models Kozasa, Katsumi Mabuchi, Seiji Matsumoto, Yuri Kuroda, Hiromasa Yokoi, Eriko Komura, Naoko Kawano, Mahiru Takahashi, Ryoko Sasano, Tomoyuki Shimura, Kotaro Kodama, Michiko Hashimoto, Kae Sawada, Kenjiro Nagasaka, Kazunori Kimura, Tadashi Oncotarget Research Paper OBJECTIVE: To investigate the clinical implications of 17β-estradiol (E2) in estrogen receptor α (ERα)-negative female cancer progression as well as the underlying biological mechanisms. METHODS: Clinical data from 306 locally-advanced cervical cancer (stage IIB-IVA) patients were analyzed in order to investigate the relationships between age, serum E2 levels, and treatment outcomes. Clinical samples, ERα-negative cervical and breast cancer cell lines, and mouse xenograft models of cervical and breast cancers were employed in order to elucidate the mechanisms responsible for the E2- and pregnancy-mediated progression of cervical and breast cancers, with a focus on the role of myeloid-derived suppressor cells (MDSC). RESULTS: Younger patients with elevated E2 levels showed significantly shorter progression-free survival (P = 0.040) and overall survival (P = 0.039). The exogenous E2 treatment stimulated the mobilization of MDSC from bone marrow and directly augmented their suppressive activities, leading to the progression of ERα-negative cervical and breast cancers. The co-administration of an anti-Gr-1 neutralizing antibody with E2 prevented the E2-mediated induction of MDSC, and attenuated E2-mediated tumor growth in cervical and breast cancer xenografts. Significantly increased MDSC numbers and enhanced tumor growth were observed during pregnancy in mice with cervical or breast cancer. Significantly increased MDSC numbers were also observed during pregnancy in cervical cancer patients. CONCLUSIONS: E2 facilitates the progression of ERα-negative cervical or breast cancer under non-pregnant and pregnant conditions by inducing MDSC. MDSC inhibition therapy may have therapeutic efficacy in premenopausal or pregnant female cancer patients. Impact Journals LLC 2019-03-08 /pmc/articles/PMC6443012/ /pubmed/30956772 http://dx.doi.org/10.18632/oncotarget.26711 Text en Copyright: © 2019 Kozasa et al. http://creativecommons.org/licenses/by/3.0/ This article is distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0/) (CC-BY), which permits unrestricted use and redistribution provided that the original author and source are credited.
spellingShingle Research Paper
Kozasa, Katsumi
Mabuchi, Seiji
Matsumoto, Yuri
Kuroda, Hiromasa
Yokoi, Eriko
Komura, Naoko
Kawano, Mahiru
Takahashi, Ryoko
Sasano, Tomoyuki
Shimura, Kotaro
Kodama, Michiko
Hashimoto, Kae
Sawada, Kenjiro
Nagasaka, Kazunori
Kimura, Tadashi
Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models
title Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models
title_full Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models
title_fullStr Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models
title_full_unstemmed Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models
title_short Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models
title_sort estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6443012/
https://www.ncbi.nlm.nih.gov/pubmed/30956772
http://dx.doi.org/10.18632/oncotarget.26711
work_keys_str_mv AT kozasakatsumi estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT mabuchiseiji estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT matsumotoyuri estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT kurodahiromasa estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT yokoieriko estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT komuranaoko estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT kawanomahiru estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT takahashiryoko estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT sasanotomoyuki estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT shimurakotaro estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT kodamamichiko estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT hashimotokae estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT sawadakenjiro estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT nagasakakazunori estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels
AT kimuratadashi estrogenstimulatesfemalecancerprogressionbyinducingmyeloidderivedsuppressivecellsinvestigationsonpregnantandnonpregnantexperimentalmodels