Cargando…

Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer

BACKGROUND: Chemotherapy is the standard treatment for breast cancer; however, the response to chemotherapy is disappointingly low. Here, we investigated the alternative therapeutic efficacy of novel combination treatment with necroptosis-inducing small molecules to overcome chemotherapeutic resista...

Descripción completa

Detalles Bibliográficos
Autores principales: Lee, Kyung-Min, Lee, Hyebin, Han, Dohyun, Moon, Woo Kyung, Kim, Kwangsoo, Oh, Hyeon Jeong, Choi, Jinwoo, Hwang, Eun Hye, Kang, Seong Eun, Im, Seock-Ah, Lee, Kyung-Hun, Ryu, Han Suk
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7687715/
https://www.ncbi.nlm.nih.gov/pubmed/33239070
http://dx.doi.org/10.1186/s13058-020-01367-7
_version_ 1783613581121552384
author Lee, Kyung-Min
Lee, Hyebin
Han, Dohyun
Moon, Woo Kyung
Kim, Kwangsoo
Oh, Hyeon Jeong
Choi, Jinwoo
Hwang, Eun Hye
Kang, Seong Eun
Im, Seock-Ah
Lee, Kyung-Hun
Ryu, Han Suk
author_facet Lee, Kyung-Min
Lee, Hyebin
Han, Dohyun
Moon, Woo Kyung
Kim, Kwangsoo
Oh, Hyeon Jeong
Choi, Jinwoo
Hwang, Eun Hye
Kang, Seong Eun
Im, Seock-Ah
Lee, Kyung-Hun
Ryu, Han Suk
author_sort Lee, Kyung-Min
collection PubMed
description BACKGROUND: Chemotherapy is the standard treatment for breast cancer; however, the response to chemotherapy is disappointingly low. Here, we investigated the alternative therapeutic efficacy of novel combination treatment with necroptosis-inducing small molecules to overcome chemotherapeutic resistance in tyrosine aminoacyl-tRNA synthetase (YARS)-positive breast cancer. METHODS: Pre-chemotherapeutic needle biopsy of 143 invasive ductal carcinomas undergoing the same chemotherapeutic regimen was subjected to proteomic analysis. Four different machine learning algorithms were employed to determine signature protein combinations. Immunoreactive markers were selected using three common candidate proteins from the machine-learning algorithms and verified by immunohistochemistry using 123 cases of independent needle biopsy FFPE samples. The regulation of chemotherapeutic response and necroptotic cell death was assessed using lentiviral YARS overexpression and depletion 3D spheroid formation assay, viability assays, LDH release assay, flow cytometry analysis, and transmission electron microscopy. The ROS-induced metabolic dysregulation and phosphorylation of necrosome complex by YARS were assessed using oxygen consumption rate analysis, flow cytometry analysis, and 3D cell viability assay. The therapeutic roles of SMAC mimetics (LCL161) and a pan-BCL2 inhibitor (ABT-263) were determined by 3D cell viability assay and flow cytometry analysis. Additional biologic process and protein-protein interaction pathway analysis were performed using Gene Ontology annotation and Cytoscape databases. RESULTS: YARS was selected as a potential biomarker by proteomics-based machine-learning algorithms and was exclusively associated with good response to chemotherapy by subsequent immunohistochemical validation. In 3D spheroid models of breast cancer cell lines, YARS overexpression significantly improved chemotherapy response via phosphorylation of the necrosome complex. YARS-induced necroptosis sequentially mediated mitochondrial dysfunction through the overproduction of ROS in breast cancer cell lines. Combination treatment with necroptosis-inducing small molecules, including a SMAC mimetic (LCL161) and a pan-BCL2 inhibitor (ABT-263), showed therapeutic efficacy in YARS-overexpressing breast cancer cells. CONCLUSIONS: Our results indicate that, before chemotherapy, an initial screening of YARS protein expression should be performed, and YARS-positive breast cancer patients might consider the combined treatment with LCL161 and ABT-263; this could be a novel stepwise clinical approach to apply new targeted therapy in breast cancer patients in the future.
format Online
Article
Text
id pubmed-7687715
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-76877152020-11-30 Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer Lee, Kyung-Min Lee, Hyebin Han, Dohyun Moon, Woo Kyung Kim, Kwangsoo Oh, Hyeon Jeong Choi, Jinwoo Hwang, Eun Hye Kang, Seong Eun Im, Seock-Ah Lee, Kyung-Hun Ryu, Han Suk Breast Cancer Res Research Article BACKGROUND: Chemotherapy is the standard treatment for breast cancer; however, the response to chemotherapy is disappointingly low. Here, we investigated the alternative therapeutic efficacy of novel combination treatment with necroptosis-inducing small molecules to overcome chemotherapeutic resistance in tyrosine aminoacyl-tRNA synthetase (YARS)-positive breast cancer. METHODS: Pre-chemotherapeutic needle biopsy of 143 invasive ductal carcinomas undergoing the same chemotherapeutic regimen was subjected to proteomic analysis. Four different machine learning algorithms were employed to determine signature protein combinations. Immunoreactive markers were selected using three common candidate proteins from the machine-learning algorithms and verified by immunohistochemistry using 123 cases of independent needle biopsy FFPE samples. The regulation of chemotherapeutic response and necroptotic cell death was assessed using lentiviral YARS overexpression and depletion 3D spheroid formation assay, viability assays, LDH release assay, flow cytometry analysis, and transmission electron microscopy. The ROS-induced metabolic dysregulation and phosphorylation of necrosome complex by YARS were assessed using oxygen consumption rate analysis, flow cytometry analysis, and 3D cell viability assay. The therapeutic roles of SMAC mimetics (LCL161) and a pan-BCL2 inhibitor (ABT-263) were determined by 3D cell viability assay and flow cytometry analysis. Additional biologic process and protein-protein interaction pathway analysis were performed using Gene Ontology annotation and Cytoscape databases. RESULTS: YARS was selected as a potential biomarker by proteomics-based machine-learning algorithms and was exclusively associated with good response to chemotherapy by subsequent immunohistochemical validation. In 3D spheroid models of breast cancer cell lines, YARS overexpression significantly improved chemotherapy response via phosphorylation of the necrosome complex. YARS-induced necroptosis sequentially mediated mitochondrial dysfunction through the overproduction of ROS in breast cancer cell lines. Combination treatment with necroptosis-inducing small molecules, including a SMAC mimetic (LCL161) and a pan-BCL2 inhibitor (ABT-263), showed therapeutic efficacy in YARS-overexpressing breast cancer cells. CONCLUSIONS: Our results indicate that, before chemotherapy, an initial screening of YARS protein expression should be performed, and YARS-positive breast cancer patients might consider the combined treatment with LCL161 and ABT-263; this could be a novel stepwise clinical approach to apply new targeted therapy in breast cancer patients in the future. BioMed Central 2020-11-25 2020 /pmc/articles/PMC7687715/ /pubmed/33239070 http://dx.doi.org/10.1186/s13058-020-01367-7 Text en © The Author(s) 2020 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research Article
Lee, Kyung-Min
Lee, Hyebin
Han, Dohyun
Moon, Woo Kyung
Kim, Kwangsoo
Oh, Hyeon Jeong
Choi, Jinwoo
Hwang, Eun Hye
Kang, Seong Eun
Im, Seock-Ah
Lee, Kyung-Hun
Ryu, Han Suk
Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer
title Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer
title_full Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer
title_fullStr Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer
title_full_unstemmed Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer
title_short Combined the SMAC mimetic and BCL2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-tRNA synthase-positive breast cancer
title_sort combined the smac mimetic and bcl2 inhibitor sensitizes neoadjuvant chemotherapy by targeting necrosome complexes in tyrosine aminoacyl-trna synthase-positive breast cancer
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7687715/
https://www.ncbi.nlm.nih.gov/pubmed/33239070
http://dx.doi.org/10.1186/s13058-020-01367-7
work_keys_str_mv AT leekyungmin combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT leehyebin combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT handohyun combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT moonwookyung combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT kimkwangsoo combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT ohhyeonjeong combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT choijinwoo combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT hwangeunhye combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT kangseongeun combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT imseockah combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT leekyunghun combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer
AT ryuhansuk combinedthesmacmimeticandbcl2inhibitorsensitizesneoadjuvantchemotherapybytargetingnecrosomecomplexesintyrosineaminoacyltrnasynthasepositivebreastcancer