1
|
Wang SF, Chang YL, Tzeng YD, Wu CL, Wang YZ, Tseng LM, Chen S, Lee HC. Mitochondrial stress adaptation promotes resistance to aromatase inhibitor in human breast cancer cells via ROS/calcium up-regulated amphiregulin-estrogen receptor loop signaling. Cancer Lett 2021; 523:82-99. [PMID: 34610415 DOI: 10.1016/j.canlet.2021.09.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/12/2021] [Accepted: 09/30/2021] [Indexed: 10/20/2022]
Abstract
Many breast cancer patients harbor high estrogen receptor (ER) expression in tumors that can be treated with endocrine therapy, which includes aromatase inhibitors (AI); unfortunately, resistance often occurs. Mitochondrial dysfunction has been thought to contribute to progression and to be related to hormone receptor expression in breast tumors. Mitochondrial alterations in AI-resistant breast cancer have not yet been defined. In this study, we characterized mitochondrial alterations and their roles in AI resistance. MCF-7aro AI-resistant breast cancer cells were shown to have significant changes in mitochondria. Low expressions of mitochondrial genes and proteins could be poor prognostic factors for breast cancer patients. Long-term mitochondrial inhibitor treatments-mediated mitochondrial stress adaptation could induce letrozole resistance. ERα-amphiregulin (AREG) loop signaling was activated and contributed to mitochondrial stress adaptation-mediated letrozole resistance. The up-regulation of AREG-epidermal growth factor receptor (EGFR) crosstalk activated the PI3K/Akt/mTOR and ERK pathways and was responsible for ERα activation. Moreover, mitochondrial stress adaptation-increased intracellular levels of reactive oxygen species (ROS) and calcium were shown to induce AREG expression and secretion. In conclusion, our results support the claim that mitochondrial stress adaptation contributes to AI resistance via ROS/calcium-mediated AREG-ERα loop signaling and provide possible treatment targets for overcoming AI resistance.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yuh-Lih Chang
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan; Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yen-Dun Tzeng
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | - Chun-Ling Wu
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yuan-Zhong Wang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, CA, 91010, USA
| | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, CA, 91010, USA.
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan; Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
2
|
Petrossian K, Kanaya N, Lo C, Hsu PY, Nguyen D, Yang L, Yang L, Warden C, Wu X, Pillai R, Bernal L, Huang CS, Kruper L, Yuan Y, Somlo G, Mortimer J, Chen S. ERα-mediated cell cycle progression is an important requisite for CDK4/6 inhibitor response in HR+ breast cancer. Oncotarget 2018; 9:27736-27751. [PMID: 29963233 PMCID: PMC6021239 DOI: 10.18632/oncotarget.25552] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/19/2018] [Indexed: 01/01/2023] Open
Abstract
While ER has multiple biological effects, ER-cyclin D1-CDK4/6-RB is a critical pathway for the action of estrogen on the cell cycle, especially for breast cancers that rely on estrogen for growth. The latest and most efficient CDK4/6 inhibitors target the phosphorylation of retinoblastoma (RB) tumor suppressor gene; thus, altering levels of many cell cycle molecules. Estrogen receptor (ER)+/HER2- breast cancers have shown great progression free survival when CDK4/6 inhibitors are combined with endocrine therapies. Here we report the mechanism of antiestrogen (fulvestrant) combination with CDK4/6 inhibitors is due to synergism in the suppression of ER-mediated cell cycle progression. Furthermore, we performed single cell analysis of cells from an estrogen dependent/hormone receptor-positive patient derived xenograft (PDX) tumor model treated with palbociclib. These single cells expressed various levels of ER and RB which are involved in cell cycle regulation; and the response to palbociclib treatment relies not only on the ER-cyclin D1-CDK4/6-RB pathway but it is also dependent on elevated levels of ER and/or RB. Our preclinical studies show that palbociclib response is dependent on cells with ER, which is directly involved in cell cycle progression in hormone receptor positive (HR+) breast cancer.
Collapse
Affiliation(s)
- Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiao Lo
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Pei-Yin Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Duc Nguyen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lixin Yang
- Molecular Pathology Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lu Yang
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Charles Warden
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Raju Pillai
- Molecular Pathology Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lauren Bernal
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiun-Sheng Huang
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Laura Kruper
- Department of Surgery, City of Hope Medical Center, Duarte, CA, United States
| | - Yuan Yuan
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - George Somlo
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - Joanne Mortimer
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| |
Collapse
|
3
|
Wawrzynow B, Zylicz A, Zylicz M. Chaperoning the guardian of the genome. The two-faced role of molecular chaperones in p53 tumor suppressor action. Biochim Biophys Acta Rev Cancer 2018; 1869:161-174. [DOI: 10.1016/j.bbcan.2017.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 12/17/2022]
|
4
|
Hsu PY, Wu VS, Kanaya N, Petrossian K, Hsu HK, Nguyen D, Schmolze D, Kai M, Liu CY, Lu H, Chu P, Vito CA, Kruper L, Mortimer J, Chen S. Dual mTOR Kinase Inhibitor MLN0128 Sensitizes HR +/HER2 + Breast Cancer Patient-Derived Xenografts to Trastuzumab or Fulvestrant. Clin Cancer Res 2017; 24:395-406. [PMID: 29079660 DOI: 10.1158/1078-0432.ccr-17-1983] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/24/2017] [Accepted: 10/23/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Therapeutic strategies against hormonal receptor-positive (HR+)/HER2+ breast cancers with poor response to trastuzumab need to be optimized.Experimental Design: Two HR+/HER2+ patient-derived xenograft (PDX) models named as COH-SC1 and COH-SC31 were established to explore targeted therapies for HER2+ breast cancers. RNA sequencing and RPPA (reverse phase protein array) analyses were conducted to decipher molecular features of the two PDXs and define the therapeutic strategy of interest, validated by in vivo drug efficacy examination and in vitro cell proliferation analysis.Results: Estrogen acted as a growth driver of trastuzumab-resistant COH-SC31 tumors but an accelerator in the trastuzumab-sensitive COH-SC1 model. In vivo trastuzumab efficacy examination further confirmed the consistent responses between PDXs and the corresponding tumors. Integrative omics analysis revealed that mammalian target of rapamycin (mTOR) and ERα signaling predominantly regulate tumor growth of the two HR+/HER2+ PDXs. Combination of the dual mTOR complex inhibitor MLN0128 and anti-HER2 trastuzumab strongly suppressed tumor growth of COH-SC1 PDX accompanied by increasing ER-positive cell population in vivo Instead, MLN0128 in combination with antiestrogen fulvestrant significantly halted the growth of HR+/HER2+ cancer cells in vitro and trastuzumab-resistant COH-SC31 as well as trastuzumab-sensitive COH-SC1 tumors in vivoConclusions: Compared with the standard trastuzumab treatment, this study demonstrates alternative therapeutic strategies against HR+/HER2+ tumors through establishment of two PDXs coupled with integrative omics analyses and in vivo drug efficacy examination. This work presents a prototype of future "co-clinical" trials to tailor personalized medicine in clinical practice. Clin Cancer Res; 24(2); 395-406. ©2017 AACR.
Collapse
Affiliation(s)
- Pei-Yin Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Victoria Shang Wu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Hang-Kai Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Duc Nguyen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Daniel Schmolze
- Department of Pathology, City of Hope Medical Center, Duarte, California
| | - Masaya Kai
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Chun-Yu Liu
- Department of Oncology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hannah Lu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Peiguo Chu
- Department of Pathology, City of Hope Medical Center, Duarte, California
| | - Courtney A Vito
- Department of Surgery, City of Hope Medical Center, Duarte, California
| | - Laura Kruper
- Department of Surgery, City of Hope Medical Center, Duarte, California
| | - Joanne Mortimer
- Department of Medical Oncology and Therapeutic Research, City of Hope Medical Center, Duarte, California
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California.
| |
Collapse
|
5
|
Huang D, Yang F, Wang Y, Guan X. Mechanisms of resistance to selective estrogen receptor down-regulator in metastatic breast cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:148-156. [PMID: 28344099 DOI: 10.1016/j.bbcan.2017.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/18/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023]
Abstract
Based on the prominent role estrogen receptor (ER) plays in breast cancer, endocrine therapy has been developed to block the ER pathway and has shown great effectiveness. Fulvestrant, the first selective ER down-regulator (SERD), was demonstrated to completely suppress ERα and notably efficient. However, resistance to fulvestrant occurs, either intrinsic or acquired during the treatment. Several potential mechanisms inducing fulvestrant resistance have been proposed, composed of activated ERα-independent compensatory growth factor signaling, stimulated downstream kinases, altered cell cycle mediators, etcetera. Experimentally, combinations of fulvestrant with targeted treatments were reported to eliminate the resistance and improve the effect of fulvestrant. Meanwhile, some clinical trials associated with the targeted combination therapies are in progress. This review focuses on the underlying mechanisms that contribute to fulvestrant resistance in ER-positive breast cancer and provides an overview of combined fulvestrant with targeted agents to shed light on optimal therapies for patients with ER-positive breast cancer.
Collapse
Affiliation(s)
- Doudou Huang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Fang Yang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Yucai Wang
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China.
| |
Collapse
|
6
|
SGK3 sustains ERα signaling and drives acquired aromatase inhibitor resistance through maintaining endoplasmic reticulum homeostasis. Proc Natl Acad Sci U S A 2017; 114:E1500-E1508. [PMID: 28174265 DOI: 10.1073/pnas.1612991114] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Many estrogen receptor alpha (ERα)-positive breast cancers initially respond to aromatase inhibitors (AIs), but eventually acquire resistance. Here, we report that serum- and glucocorticoid-inducible kinase 3 (SGK3), a kinase transcriptionally regulated by ERα in breast cancer, sustains ERα signaling and drives acquired AI resistance. SGK3 is up-regulated and essential for endoplasmic reticulum (EnR) homeostasis through preserving sarcoplasmic/EnR calcium ATPase 2b (SERCA2b) function in AI-resistant cells. We have further found that EnR stress response down-regulates ERα expression through the protein kinase RNA-like EnR kinase (PERK) arm, and SGK3 retains ERα expression and signaling by preventing excessive EnR stress. Our study reveals regulation of ERα expression mediated by the EnR stress response and the feed-forward regulation between SGK3 and ERα in breast cancer. Given SGK3 inhibition reduces AI-resistant cell survival by eliciting excessive EnR stress and also depletes ERα expression/function, we propose SGK3 inhibition as a potential effective treatment of acquired AI-resistant breast cancer.
Collapse
|
7
|
SERPINA1 is a direct estrogen receptor target gene and a predictor of survival in breast cancer patients. Oncotarget 2016; 6:25815-27. [PMID: 26158350 PMCID: PMC4694868 DOI: 10.18632/oncotarget.4441] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 06/19/2015] [Indexed: 01/02/2023] Open
Abstract
Of all breast cancer patients, about 70% are ER+ and 10% are ER+/HER2+. The ER+/HER2+ patients have a worse outcome compared to ER+/HER2- patients. Currently there is a lack of effective prognosis biomarkers for the prediction of outcome in ER+/HER2+ patients. Genome-wide differences in ER binding between the endocrine-responsive and endocrine-resistant cells were discovered using ChIP-seq, and combined with gene expression microarray data to identify direct ER target genes. These genes were correlated to survival outcome using publicly available breast cancer patient cohorts. We found the expression of the gene SERPINA1 to have a significant predictive value for the overall survival (OS) of ER+ patients in the TCGA cohort, and validated this finding in the Curtis cohort. SERPINA1 also has a significant predictive value for the OS of ER+/HER2+ patients in the TCGA cohort, with validation in the Bild cohort. The expression of SERPINA1 can be suppressed by fulvestrant and HER2 siRNA. Our results indicate that ER is constitutively activated, resulting in an E2-independent ER binding to the SERPINA1 gene and upregulation of SERPINA1 expression. Importantly, results of survival correlation suggests that high expression of SERPINA1 could be predictive for a better clinical outcome of ER+ and ER+/HER2+ patients.
Collapse
|
8
|
Huyan T, Li Q, Dong DD, Yang H, Zhang J, Huang QS, Yin DC, Shang P. Heat shock protein 90 inhibitors induce functional inhibition of human natural killer cells in a dose-dependent manner. Immunopharmacol Immunotoxicol 2015; 38:77-86. [PMID: 26642940 DOI: 10.3109/08923973.2015.1119159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heat shock protein 90 (Hsp90) is a ubiquitously expressed ATP-dependent molecular chaperone across all species that helps to the correct the folding of many proteins related to important signaling pathways. Tumor cells expressing Hsp90 have more ATP-binding affinity than normal cells. Many correlative inhibitors have been developed to promising anti-tumor strategies and have been evaluated in clinical trials. However, the effect of Hsp90 inhibitors on immunocytes cannot be ignored. Natural killer (NK) cells are key components of the innate immune system that play a pivotal role in tumor surveillance. The present study has investigated the potential effect of four Hsp90 inhibitors (NVP-AUY922, BIIB021, 17-DMAG, and SNX-2112) on human primary NK cells. The viability, cytotoxicity, apoptosis, phenotype, and cytokine secretion of NK cells after inhibitor treatment were assessed. The results of this study demonstrated that the inhibitors had negative effects on NK cell activity in a dose-dependent manner. The four inhibitors significantly reduced the cytotoxicity of the NK cells by decreasing viability, inducing apoptosis and down-regulating the expression of cytokines and functional receptors. These findings suggest that more attention should be given to the effect of Hsp90 inhibitors on NK cell function during clinical trials and also represent a potential immunosuppressant strategy.
Collapse
Affiliation(s)
- Ting Huyan
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| | - Qi Li
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| | - Dan-Dan Dong
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| | - Hui Yang
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| | - Jian Zhang
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| | - Qing-Sheng Huang
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| | - Da-Chuan Yin
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| | - Peng Shang
- a Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi , People's Republic of China
| |
Collapse
|
9
|
Chen Z, Yuan YC, Wang Y, Liu Z, Chan HJ, Chen S. Down-regulation of programmed cell death 4 (PDCD4) is associated with aromatase inhibitor resistance and a poor prognosis in estrogen receptor-positive breast cancer. Breast Cancer Res Treat 2015; 152:29-39. [PMID: 26026468 DOI: 10.1007/s10549-015-3446-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 05/23/2015] [Indexed: 10/23/2022]
Abstract
Progression or recurrence due to resistance to aromatase inhibitors (AIs) is a significant clinical problem for a considerable number of patients with breast cancer. Programmed cell death 4 (PDCD4), a tumor suppressor protein, is targeted for degradation during tumor progression. In the current study, we aimed to examine PDCD4 expression and regulation in AI-resistant breast cancer cells, and its association with survival in patients with estrogen receptor (ER)-positive breast cancer. We determined PDCD4 expression levels in AI-resistant breast cancer cell lines and ER-positive breast cancer tumors, investigated the regulation of PDCD4 in AI-resistant breast cancer cell lines, and carried out a Kaplan-Meier survival analysis in two independent cohorts that included a total of 420 patients with ER-positive breast cancer. We found that PDCD4 expression was down-regulated in AI-resistant breast cancer cells, and this down-regulation was inversely correlated with activation of HER2 signaling. Moreover, lower expression of PDCD4 was significantly associated with HER2 positive status in ER-positive breast tumors. Down-regulation of PDCD4 was mediated through up-regulation of HER2 via the mitogen-activated protein kinase (MAPK), protein kinase B (PKB/AKT), and miR-21 in AI-resistant breast cancer cells. MiR-21 inhibitor and the ER down-regulator fulvestrant induced PDCD4 expression and decreased cell proliferation in AI-resistant breast cancer cells. Furthermore, forced overexpression of PDCD4 resensitized AI-resistant cells to AI or hormone deprivation. Finally, we identified that down-regulation of PDCD4 was associated with a lower rate of disease-free survival in patients with ER-positive breast cancer and high histologic grade of breast tumors. In summary, our study shows that expression of PDCD4 is down-regulated by HER2 signaling in AI-resistant breast cancer. Down-regulation of PDCD4 is associated with AI resistance and a poor prognosis in patients with ER-positive breast cancer.
Collapse
Affiliation(s)
- Zhike Chen
- Department of Cancer Biology, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Yate-Ching Yuan
- Bioinformatics Core, Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Yuanzhong Wang
- Department of Cancer Biology, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Zheng Liu
- Bioinformatics Core, Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Hei Jason Chan
- Department of Cancer Biology, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, United States.
| |
Collapse
|
10
|
Vilquin P, Donini CF, Villedieu M, Grisard E, Corbo L, Bachelot T, Vendrell JA, Cohen PA. MicroRNA-125b upregulation confers aromatase inhibitor resistance and is a novel marker of poor prognosis in breast cancer. Breast Cancer Res 2015; 17:13. [PMID: 25633049 PMCID: PMC4342894 DOI: 10.1186/s13058-015-0515-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/08/2015] [Indexed: 12/14/2022] Open
Abstract
Introduction Increasing evidence indicates that microRNAs (miRNAs) are important players in oncogenesis. Considering the widespread use of aromatase inhibitors (AIs) in endocrine therapy as a first-line treatment for postmenopausal estrogen receptor α–positive breast cancer patients, identifying deregulated expression levels of miRNAs in association with AI resistance is of utmost importance. Methods To gain further insight into the molecular mechanisms underlying the AI resistance, we performed miRNA microarray experiments using a new model of acquired resistance to letrozole (Res-Let cells), obtained by long-term exposure of aromatase-overexpressing MCF-7 cells (MCF-7aro cells) to letrozole, and a model of acquired anastrozole resistance (Res-Ana cells). Three miRNAs (miR-125b, miR-205 and miR-424) similarly deregulated in both AI-resistant cell lines were then investigated in terms of their functional role in AI resistance development and breast cancer cell aggressiveness and their clinical relevance using a cohort of 65 primary breast tumor samples. Results We identified the deregulated expression of 33 miRNAs in Res-Let cells and of 18 miRNAs in Res-Ana cells compared with the sensitive MCF-7aro cell line. The top-ranked Kyoto Encyclopedia of Genes and Genomes pathways delineated by both miRNA signatures converged on the AKT/mTOR pathway, which was found to be constitutively activated in both AI-resistant cell lines. We report for the first time, to our knowledge, that ectopic overexpression of either miR-125b or miR-205, or the silencing of miR-424 expression, in the sensitive MCF-7aro cell line was sufficient to confer resistance to letrozole and anastrozole, to target and activate the AKT/mTOR pathway and to increase the formation capacity of stem-like and tumor-initiating cells possessing self-renewing properties. Increasing miR-125b expression levels was also sufficient to confer estrogen-independent growth properties to the sensitive MCF-7aro cell line. We also found that elevated miR-125b expression levels were a novel marker for poor prognosis in breast cancer and that targeting miR-125b in Res-Let cells overcame letrozole resistance. Conclusion This study highlights that acquisition of specific deregulated miRNAs is a newly discovered alternative mechanism developed by AI-resistant breast cancer cells to achieve constitutive activation of the AKT/mTOR pathway and to develop AI resistance. It also highlights that miR-125b is a new biomarker of poor prognosis and a candidate therapeutic target in AI-resistant breast cancers. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0515-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paul Vilquin
- ISPB, Faculté de Pharmacie, 8 Avenue Rockefeller, 69008, Lyon, France. .,Université Lyon 1, 8 Avenue Rockefeller, 69008, Lyon, France. .,INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69008, Lyon, France.
| | - Caterina F Donini
- ISPB, Faculté de Pharmacie, 8 Avenue Rockefeller, 69008, Lyon, France. .,Université Lyon 1, 8 Avenue Rockefeller, 69008, Lyon, France. .,INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69008, Lyon, France. .,Unité Cancer et Environnement, Centre Léon Bérard-Université Lyon 1, 28 Rue Laennec, 69008, Lyon, France.
| | - Marie Villedieu
- ISPB, Faculté de Pharmacie, 8 Avenue Rockefeller, 69008, Lyon, France. .,Université Lyon 1, 8 Avenue Rockefeller, 69008, Lyon, France. .,INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69008, Lyon, France.
| | - Evelyne Grisard
- ISPB, Faculté de Pharmacie, 8 Avenue Rockefeller, 69008, Lyon, France. .,Université Lyon 1, 8 Avenue Rockefeller, 69008, Lyon, France. .,INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69008, Lyon, France.
| | - Laura Corbo
- Université Lyon 1, 8 Avenue Rockefeller, 69008, Lyon, France. .,INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69008, Lyon, France.
| | | | - Julie A Vendrell
- ISPB, Faculté de Pharmacie, 8 Avenue Rockefeller, 69008, Lyon, France. .,Université Lyon 1, 8 Avenue Rockefeller, 69008, Lyon, France. .,INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69008, Lyon, France.
| | - Pascale A Cohen
- ISPB, Faculté de Pharmacie, 8 Avenue Rockefeller, 69008, Lyon, France. .,Université Lyon 1, 8 Avenue Rockefeller, 69008, Lyon, France. .,INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69008, Lyon, France. .,Unité Cancer et Environnement, Centre Léon Bérard-Université Lyon 1, 28 Rue Laennec, 69008, Lyon, France. .,ProfileXpert, SFR Lyon-Est, 69008, Lyon, France. .,ISPBL-Faculté de Pharmacie de Lyon, 8 Avenue Rockefeller, 69373, Lyon, Cedex 08, France.
| |
Collapse
|
11
|
Solárová Z, Mojžiš J, Solár P. Hsp90 inhibitor as a sensitizer of cancer cells to different therapies (review). Int J Oncol 2014; 46:907-26. [PMID: 25501619 DOI: 10.3892/ijo.2014.2791] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/22/2014] [Indexed: 11/06/2022] Open
Abstract
Hsp90 is a molecular chaperone that maintains the structural and functional integrity of various client proteins involved in signaling and many other functions of cancer cells. The natural inhibitors, ansamycins influence the Hsp90 chaperone function by preventing its binding to client proteins and resulting in their proteasomal degradation. N- and C-terminal inhibitors of Hsp90 and their analogues are widely tested as potential anticancer agents in vitro, in vivo as well as in clinical trials. It seems that Hsp90 competitive inhibitors target different tumor types at nanomolar concentrations and might have therapeutic benefit. On the contrary, some Hsp90 inhibitors increased toxicity and resistance of cancer cells induced by heat shock response, and through the interaction of survival signals, that occured as side effects of treatments, could be very effectively limited via combination of therapies. The aim of our review was to collect the data from experimental and clinical trials where Hsp90 inhibitor was combined with other therapies in order to prevent resistance as well as to potentiate the cytotoxic and/or antiproliferative effects.
Collapse
Affiliation(s)
- Zuzana Solárová
- Department of Pharmacology, Faculty of Medicine, P.J. Šafárik University, 040 01 Košice, Slovak Republic
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, P.J. Šafárik University, 040 01 Košice, Slovak Republic
| | - Peter Solár
- Laboratory of Cell Biology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University, 040 01 Košice, Slovak Republic
| |
Collapse
|
12
|
Fox EM, Kuba MG, Miller TW, Davies BR, Arteaga CL. Autocrine IGF-I/insulin receptor axis compensates for inhibition of AKT in ER-positive breast cancer cells with resistance to estrogen deprivation. Breast Cancer Res 2014; 15:R55. [PMID: 23844554 PMCID: PMC3979036 DOI: 10.1186/bcr3449] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 05/29/2013] [Accepted: 07/11/2013] [Indexed: 12/12/2022] Open
Abstract
Introduction Estrogen receptor α-positive (ER+) breast cancers adapt to hormone deprivation and acquire resistance to antiestrogen therapies. Upon acquisition of hormone independence, ER+ breast cancer cells increase their dependence on the phosphatidylinositol-3 kinase (PI3K)/AKT pathway. We examined the effects of AKT inhibition and its compensatory upregulation of insulin-like growth factor (IGF)-I/InsR signaling in ER+ breast cancer cells with acquired resistance to estrogen deprivation. Methods Inhibition of AKT using the catalytic inhibitor AZD5363 was examined in four ER+ breast cancer cell lines resistant to long-term estrogen deprivation (LTED) by western blotting and proliferation assays. Feedback upregulation and activation of receptor tyrosine kinases (RTKs) was examined by western blotting, real-time qPCR, ELISAs, membrane localization of AKT PH-GFP by immunofluorescence and phospho-RTK arrays. For studies in vivo, athymic mice with MCF-7 xenografts were treated with AZD5363 and fulvestrant with either the ATP-competitive IGF-IR/InsR inhibitor AZD9362 or the fibroblast growth factor receptor (FGFR) inhibitor AZD4547. Results Treatment with AZD5363 reduced phosphorylation of the AKT/mTOR substrates PRAS40, GSK3α/β and S6K while inducing hyperphosphorylation of AKT at T308 and S473. Inhibition of AKT with AZD5363 suppressed growth of three of four ER+ LTED lines and prevented emergence of hormone-independent MCF-7, ZR75-1 and MDA-361 cells. AZD5363 suppressed growth of MCF-7 xenografts in ovariectomized mice and a patient-derived luminal B xenograft unresponsive to tamoxifen or fulvestrant. Combined treatment with AZD5363 and fulvestrant suppressed MCF-7 xenograft growth better than either drug alone. Inhibition of AKT with AZD5363 resulted in upregulation and activation of RTKs, including IGF-IR and InsR, upregulation of FoxO3a and ERα mRNAs as well as FoxO- and ER-dependent transcription of IGF-I and IGF-II ligands. Inhibition of IGF-IR/InsR or PI3K abrogated AKT PH-GFP membrane localization and T308 P-AKT following treatment with AZD5363. Treatment with IGFBP-3 blocked AZD5363-induced P-IGF-IR/InsR and T308 P-AKT, suggesting that receptor phosphorylation was dependent on increased autocrine ligands. Finally, treatment with the dual IGF-IR/InsR inhibitor AZD9362 enhanced the anti-tumor effect of AZD5363 in MCF-7/LTED cells and MCF-7 xenografts in ovariectomized mice devoid of estrogen supplementation. Conclusions These data suggest combinations of AKT and IGF-IR/InsR inhibitors would be an effective treatment strategy against hormone-independent ER+ breast cancer.
Collapse
|
13
|
HSP90 empowers evolution of resistance to hormonal therapy in human breast cancer models. Proc Natl Acad Sci U S A 2014; 111:18297-302. [PMID: 25489079 DOI: 10.1073/pnas.1421323111] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The efficacy of hormonal therapies for advanced estrogen receptor-positive breast cancers is limited by the nearly inevitable development of acquired resistance. Efforts to block the emergence of resistance have met with limited success, largely because the mechanisms underlying it are so varied and complex. Here, we investigate a new strategy aimed at the very processes by which cancers evolve resistance. From yeast to vertebrates, heat shock protein 90 (HSP90) plays a unique role among molecular chaperones by promoting the evolution of heritable new traits. It does so by regulating the folding of a diverse portfolio of metastable client proteins, many of which mediate adaptive responses that allow organisms to adapt and thrive in the face of diverse challenges, including those posed by drugs. Guided by our previous work in pathogenic fungi, in which very modest HSP90 inhibition impairs resistance to mechanistically diverse antifungals, we examined the effect of similarly modest HSP90 inhibition on the emergence of resistance to antiestrogens in breast cancer models. Even though this degree of inhibition fell below the threshold for proteotoxic activation of the heat-shock response and had no overt anticancer activity on its own, it dramatically impaired the emergence of resistance to hormone antagonists both in cell culture and in mice. Our findings strongly support the clinical testing of combined hormone antagonist-low-level HSP90 inhibitor regimens in the treatment of metastatic estrogen receptor-positive breast cancer. At a broader level, they also provide promising proof of principle for a generalizable strategy to combat the pervasive problem of rapidly emerging resistance to molecularly targeted therapeutics.
Collapse
|
14
|
|
15
|
Vilquin P, Villedieu M, Grisard E, Larbi SB, Ghayad SE, Heudel PE, Bachelot T, Corbo L, Treilleux I, Vendrell JA, Cohen PA. Molecular characterization of anastrozole resistance in breast cancer: Pivotal role of the Akt/mTOR pathway in the emergence ofde novoor acquired resistance and importance of combining the allosteric Akt inhibitor MK-2206 with an aromatase inhibitor. Int J Cancer 2013; 133:1589-602. [DOI: 10.1002/ijc.28182] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 03/07/2013] [Indexed: 01/01/2023]
Affiliation(s)
| | | | | | | | - Sandra E. Ghayad
- INSERM U1052; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon; Lyon; France
| | | | | | - Laura Corbo
- INSERM U1052; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon; Lyon; France
| | | | | | | |
Collapse
|
16
|
Dias MC, Furtado KS, Rodrigues MAM, Barbisan LF. Effects of Ginkgo biloba on chemically-induced mammary tumors in rats receiving tamoxifen. Altern Ther Health Med 2013; 13:93. [PMID: 23634930 PMCID: PMC3655872 DOI: 10.1186/1472-6882-13-93] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 04/24/2013] [Indexed: 11/26/2022]
Abstract
Background Ginkgo biloba extract (GbE) is used extensively by breast cancer patients undergoing treatment with Tamoxifen (TAM). Thus, the present study investigated the effects of GbE in female Sprague–Dawley (SD) rats bearing chemically-induced mammary tumors and receiving TAM. Methods Animals bearing mammary tumors (≥1 cm in diameter) were divided into four groups: TAM [10 mg/kg, intragastrically (i.g.)], TAM plus GbE [50 and 100 mg/kg, intraperitoneally (i.p.)] or an untreated control group. After 4 weeks, the therapeutic efficacy of the different treatments was evaluated by measuring the tumor volume (cm3) and the proportions of each tumor that were alive, necrotic or degenerative (mm2). In addition, labeling indexes (LI%) were calculated for cell proliferation (PCNA LI%) and apoptosis (cleaved caspase-3 LI%), expression of estrogen receptor-alpha (ER-α) and p63 biomarkers. Results Overall, the tumor volume and the PCNA LI% within live tumor areas were reduced by 83% and 99%, respectively, in all TAM-treated groups when compared to the untreated control group. GbE treatment (100 mg/kg) reduced the proportions of live (24.8%) and necrotic areas (2.9%) (p = 0.046 and p = 0.038, respectively) and significantly increased the proportion of degenerative areas (72.9%) (p = 0.004) in mammary tumors when compared to the group treated only with TAM. The expression of ER-α, p63 and cleaved caspase-3 in live tumor tissues was not modified by GbE treatment. Conclusions Co-treatment with 100 mg/kg GbE presented a slightly beneficial effect on the therapeutic efficacy of TAM in female SD rats bearing mammary tumors.
Collapse
|
17
|
Oskay Halacli S, Halacli B, Altundag K. The significance of heat shock proteins in breast cancer therapy. Med Oncol 2013; 30:575. [PMID: 23606238 DOI: 10.1007/s12032-013-0575-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 04/04/2013] [Indexed: 12/16/2022]
Abstract
The evalutionary conserved heat shock proteins are involved basically life protecting mechanisms against harmful extracellular effects such as primarily heat shock response. Normally, the expression of these proteins is increased for cellular adaptation to high temperature. This increase is also important in the etiology of breast cancer. Overexpression of heat shock proteins is associated with reduced disease-free survival in breast cancer. However, increased expression of these proteins is related to acquired resistance of traditional chemotherapeutic drugs in use in breast cancer treatment. In this review, we discuss the multiple roles of heatshock proteins in resistance and where we are to overcome this in clinical practice.
Collapse
Affiliation(s)
- Sevil Oskay Halacli
- Department of Medical Oncology, Hacettepe University Cancer Institute, Sihhiye, Ankara 06100, Turkey
| | | | | |
Collapse
|