1
|
Siatis KE, Giannopoulou E, Manou D, Sarantis P, Karamouzis MV, Raftopoulou S, Fasseas K, Alzahrani FM, Kalofonos HP, Theocharis AD. Resistance to hormone therapy in breast cancer cells promotes autophagy and EGFR signaling pathway. Am J Physiol Cell Physiol 2023; 325:C708-C720. [PMID: 37575061 PMCID: PMC10625825 DOI: 10.1152/ajpcell.00199.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
Abstract
Breast cancer is the leading cause of cancer deaths for women worldwide. Endocrine therapies represent the cornerstone for hormone-dependent breast cancer treatment. However, in many cases, endocrine resistance is induced with poor prognosis for patients. In the current study, we have developed MCF-7 cell lines resistant to fulvestrant (MCF-7Fulv) and tamoxifen (MCF-7Tam) aiming at investigating mechanisms underlying resistance. Both resistant cell lines exerted lower proliferation capacity in two-dimensional (2-D) cultures but retain estrogen receptor α (ERα) expression and proliferate independent of the presence of estrogens. The established cell lines tend to be more aggressive exhibiting advanced capacity to form colonies, increased expression of epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and heterodimerization of ERBB family receptors and activation of EGFR downstream pathways like MEK/ERK1/2 and PI3K/AKT. Tyrosine kinase inhibitors tested against resistant MCF-7Fulv and MCF-7Tam cells showed moderate efficacy to inhibit cell proliferation, except for lapatinib, which concomitantly inhibits both EGFR and HER2 receptors and strongly reduced cell proliferation. Furthermore, increased autophagy was observed in resistant MCF-7Fulv and MCF-7Tam cells as shown by the presence of autophagosomes and increased Beclin-1 levels. The increased autophagy in resistant cells is not associated with increased apoptosis, suggesting a cytoprotective role for autophagy that may favor cells' survival and aggressiveness. Thus, by exploiting those underlying mechanisms, new targets could be established to overcome endocrine resistance.NEW & NOTEWORTHY The development of resistance to hormone therapy caused by both fulvestrant and tamoxifen promotes autophagy with concomitant apoptosis evasion, rendering cells capable of surviving and growing. The fact that resistance also triggers ERBB family signaling pathways, which are poorly inhibited by tyrosine kinase inhibitors might attribute to cells' aggressiveness. It is obvious that the development of endocrine therapy resistance involves a complex interplay between deregulated ERBB signaling and autophagy that may be considered in clinical practice.
Collapse
Affiliation(s)
- Konstantinos E Siatis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Rio, Greece
- Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, Rio, Greece
| | - Efstathia Giannopoulou
- Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, Rio, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Rio, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Raftopoulou
- Electron Microscopy Laboratory, Faculty of Crop Production, Agricultural University of Athens, Athens, Greece
| | - Konstantinos Fasseas
- Electron Microscopy Laboratory, Faculty of Crop Production, Agricultural University of Athens, Athens, Greece
| | - Fatimah Mohammed Alzahrani
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Haralabos P Kalofonos
- Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, Rio, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Rio, Greece
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Hany D, Vafeiadou V, Picard D. CRISPR-Cas9 screen reveals a role of purine synthesis for estrogen receptor α activity and tamoxifen resistance of breast cancer cells. SCIENCE ADVANCES 2023; 9:eadd3685. [PMID: 37172090 PMCID: PMC10181187 DOI: 10.1126/sciadv.add3685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
In breast cancer, resistance to endocrine therapies that target estrogen receptor α (ERα), such as tamoxifen and fulvestrant, remains a major clinical problem. Whether and how ERα+ breast cancers switch from being estrogen-dependent to estrogen-independent remains unclear. With a genome-wide CRISPR-Cas9 knockout screen, we identified previously unknown biomarkers and potential therapeutic targets of endocrine resistance. We demonstrate that high levels of PAICS, an enzyme involved in the de novo biosynthesis of purines, can shift the balance of ERα activity to be more estrogen-independent and tamoxifen-resistant. We find that this may be due to elevated activities of cAMP-activated protein kinase A and mTOR, kinases known to phosphorylate ERα specifically and to stimulate its activity. Genetic or pharmacological targeting of PAICS sensitizes tamoxifen-resistant cells to tamoxifen. Addition of purines renders them more resistant. On the basis of these findings, we propose the combined targeting of PAICS and ERα as a new, effective, and potentially safe therapeutic regimen.
Collapse
Affiliation(s)
- Dina Hany
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
- On leave from: Department of Pharmacology and Therapeutics Faculty of Pharmacy, Pharos University in Alexandria, Alexandria 21311, Egypt
| | - Vasiliki Vafeiadou
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
| |
Collapse
|
3
|
Andrade de Oliveira K, Sengupta S, Yadav AK, Clarke R. The complex nature of heterogeneity and its roles in breast cancer biology and therapeutic responsiveness. Front Endocrinol (Lausanne) 2023; 14:1083048. [PMID: 36909339 PMCID: PMC9997040 DOI: 10.3389/fendo.2023.1083048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Heterogeneity is a complex feature of cells and tissues with many interacting components. Depending on the nature of the research context, interacting features of cellular, drug response, genetic, molecular, spatial, temporal, and vascular heterogeneity may be present. We describe the various forms of heterogeneity with examples of their interactions and how they play a role in affecting cellular phenotype and drug responses in breast cancer. While cellular heterogeneity may be the most widely described and invoked, many forms of heterogeneity are evident within the tumor microenvironment and affect responses to the endocrine and cytotoxic drugs widely used in standard clinical care. Drug response heterogeneity is a critical determinant of clinical response and curative potential and also is multifaceted when encountered. The interactive nature of some forms of heterogeneity is readily apparent. For example, the process of metastasis has the properties of both temporal and spatial heterogeneity within the host, whereas each individual metastatic deposit may exhibit cellular, genetic, molecular, and vascular heterogeneity. This review describes the many forms of heterogeneity, their integrated activities, and offers some insights into how heterogeneity may be understood and studied in the future.
Collapse
Affiliation(s)
- Karla Andrade de Oliveira
- The Hormel Institute, University of Minnesota, Austin, MN, United States
- Department of Biochemistry and Pharmacology, Universidade Federal do Piaui, Piauí, Brazil
| | - Surojeet Sengupta
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Anil Kumar Yadav
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN, United States
- *Correspondence: Robert Clarke,
| |
Collapse
|
4
|
Abstract
Signaling via extracellular regulated kinase 1/2 (ERK1/2) and p90 ribosomal S6 kinase (RSK), a downstream effector, mediates numerous processes. For example, ERK1/2-RSK signaling is essential for estrogen homeostasis in the mammary gland and uterus to maintain physiological responsiveness. This review will focus on the coordination of ERK1/2-RSK2 and estrogen signaling through estrogen receptor alpha (ERα). The interrelationship and the feedback mechanisms between these pathways occurs at the level of transcription, translation, and posttranslational modification. Identifying how ERK1/2-RSK2 and estrogen signaling cooperate in homeostasis and disease may lead to novel therapeutic approaches in estrogen-dependent disorders.
Collapse
Affiliation(s)
- Deborah A Lannigan
- Correspondence: Deborah A. Lannigan, PhD, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
5
|
PEGylated Protamine Letrozole Nanoparticles: A Promising Strategy to Combat Human Breast Cancer via MCF-7 Cell Lines. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4438518. [PMID: 35722457 PMCID: PMC9205697 DOI: 10.1155/2022/4438518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 04/15/2022] [Accepted: 05/13/2022] [Indexed: 12/02/2022]
Abstract
The objective of the study was to develop PEGylated protamine letrozole nanoparticles to combat human breast cancer by modifying the release pattern of letrozole. Breast cancer is amongst the most prevalent diseases in women due to overactivity of human epidermal growth factor receptor 2 (HER2). PEG-protamine letrozole nanoparticle formulation was designed and optimized to alter the release pattern of the drug. The size, morphology, and structure of PEG-protamine letrozole NP were characterized by FTIR, XRD, Zetasizer, and SEM analysis. The result showed the PEG-protamine letrozole nanoparticles were irregular in shape and have size ranging from 258 nm to 388 nm, polydispersity index 0.114 to 0.45, zeta potential of 11.2 mV, and entrapment efficiency 89.93%. XRD studies have confirmed that the crystal structure of letrozole has become amorphous. The drug release study maintained the prolonged release for 72 hours. Moreover, the PEG-protamine letrozole NPs displayed a strong anticancer action compared to MCF-7 cells with an IC50 70 μM for letrozole and 50 μM for PEG-protamine letrozole NPs. Overall, our results indicate that letrozole PEG-protamine NPs alter the release profile of letrozole, which could be an excellent approach for overcoming letrozole resistance in human breast cancer.
Collapse
|
6
|
Choi Y. Estrogen Receptor β Expression and Its Clinical Implication in Breast Cancers: Favorable or Unfavorable? J Breast Cancer 2022; 25:75-93. [PMID: 35380018 PMCID: PMC9065353 DOI: 10.4048/jbc.2022.25.e9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 02/03/2022] [Indexed: 11/30/2022] Open
Abstract
There are two estrogen receptor (ER) genes (ESR1/ERα and ESR2/ERβ) in humans. Of those. ERβ, the second ER isotype identified in 1996, is differentially expressed in different phenotypes and molecular subtypes of breast cancer (BCa), and is highly expressed in ERα-negative BCa and triple-negative BCa (TNBC). This review summarizes the potential clinical relevance of ERβ in BCa and the challenges associated with studies on the role of ERβ in BCa. The experimental and clinical studies evaluating clinical outcomes and associations with clinical characteristics and responses to endocrine therapy on targeting ERβ reviewed herein indicate that ERβ is a clinically important biomarker in BCa. The reviewed studies also suggest that each ERβ isoform has a distinct role in BCa subtypes and the potential of novel- targeted therapies in BCa, especially ERα-negative BCa and TNBC. However, the findings of many studies on ERβ are inconsistent, and the exact role of ERβ in BCa remains elusive; this may potentially be attributed to the complexity of ERβ isoforms, but also to the lack of standardized testing protocol. Thus, successful clinical application of ERβ requires the development of standardized, reproducible, and objective measurement methods for ERβ that can be widely and routinely applied in clinical setting.
Collapse
Affiliation(s)
- Young Choi
- Department of Pathology, Yale School of Medicine, Hartsdale, NY, USA
| |
Collapse
|
7
|
Nagalingam A, Siddharth S, Parida S, Muniraj N, Avtanski D, Kuppusamy P, Elsey J, Arbiser JL, Győrffy B, Sharma D. Hyperleptinemia in obese state renders luminal breast cancers refractory to tamoxifen by coordinating a crosstalk between Med1, miR205 and ErbB. NPJ Breast Cancer 2021; 7:105. [PMID: 34389732 PMCID: PMC8363746 DOI: 10.1038/s41523-021-00314-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022] Open
Abstract
Obese women with hormone receptor-positive breast cancer exhibit poor response to therapy and inferior outcomes. However, the underlying molecular mechanisms by which obesity/hyperleptinemia may reduce the efficacy of hormonal therapy remain elusive. Obese mice with hyperleptinemia exhibit increased tumor progression and respond poorly to tamoxifen compared to non-obese mice. Exogenous leptin abrogates tamoxifen-mediated growth inhibition and potentiates breast tumor growth even in the presence of tamoxifen. Mechanistically, leptin induces nuclear translocation of phosphorylated-ER and increases the expression of ER-responsive genes, while reducing tamoxifen-mediated gene repression by abrogating tamoxifen-induced recruitment of corepressors NCoR, SMRT, and Mi2 and potentiating coactivator binding. Furthermore, in silico analysis revealed that coactivator Med1 potentially associates with 48 (out of 74) obesity-signature genes. Interestingly, leptin upregulates Med1 expression by decreasing miR-205, and increases its functional activation via phosphorylation, which is mediated by activation of Her2 and EGFR. It is important to note that Med1 silencing abrogates the negative effects of leptin on tamoxifen efficacy. In addition, honokiol or adiponectin treatment effectively inhibits leptin-induced Med1 expression and improves tamoxifen efficacy in hyperleptinemic state. These studies uncover the mechanistic insights how obese/hyperleptinemic state may contribute to poor response to tamoxifen implicating leptin-miR205-Med1 and leptin-Her2-EGFR-Med1 axes, and present bioactive compound honokiol and adipocytokine adiponectin as agents that can block leptin’s negative effect on tamoxifen.
Collapse
Affiliation(s)
- Arumugam Nagalingam
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Sumit Siddharth
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Sheetal Parida
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Nethaji Muniraj
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Dimiter Avtanski
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Division of Endocrinology, Department of Medicine, Lenox Hill Hospital, New York, NY, USA
| | | | - Justin Elsey
- Department of Dermatology, Emory School of Medicine, Atlanta Veterans Administration Medical Center, Atlanta, GA, USA
| | - Jack L Arbiser
- Department of Dermatology, Emory School of Medicine, Atlanta Veterans Administration Medical Center, Atlanta, GA, USA
| | - Balázs Győrffy
- MTA TTK Momentum Cancer Biomarker Research Group, Budapest, Hungary.,Semmelweis University, Department of Bioinformatics and 2nd Department of Pediatrics, Budapest, Hungary
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| |
Collapse
|
8
|
Yuseran H, Hartoyo E, Nurseta T, Kalim H. Genistein inhibits the proliferation of human choriocarcinoma cells via the downregulation of estrogen receptor-α phosphorylation at serine 118. CLINICAL NUTRITION OPEN SCIENCE 2021. [DOI: 10.1016/j.yclnex.2020.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
9
|
Ludwik KA, Lannigan DA. RSK2 and ERα comrades-in-arms in homeostasis and transformation. Mol Cell Oncol 2020; 7:1825916. [PMID: 33235919 PMCID: PMC7671069 DOI: 10.1080/23723556.2020.1825916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The physiological response to estrogen differs according to the developmental stage. We show, in the adult, estrogen-responsiveness is driven by ERK1/2 (extracellular signal-regulated kinase 1/2) whereas its downstream effector, RSK2 (p90 ribosomal S6 kinase 2), prevents continuous ERK1/2 activity through regulation of oxidative stress. Bioinformatic analysis revealed RSK2 association with breast cancer risk and oral contraceptives.
Collapse
Affiliation(s)
- Katarzyna A Ludwik
- Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Deborah A Lannigan
- Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA.,Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.,Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
10
|
MST2 silencing induces apoptosis and inhibits tumor growth for estrogen receptor alpha-positive MCF-7 breast cancer. Toxicol Appl Pharmacol 2020; 408:115257. [PMID: 33007383 DOI: 10.1016/j.taap.2020.115257] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/27/2020] [Indexed: 12/14/2022]
Abstract
Mammalian sterile 20-like kinase 1/2 (MST1/2) plays an important role in cell growth and apoptosis and functions as a tumor suppressor. Previously, we showed that MST2 overexpression activates Estrogen receptor alpha (ERα) in human breast cancer MCF-7 cells in the absence of a ligand. Here, we examined the role of MST2 in the growth of ER-positive MCF-7 cells. Cell cycle, apoptosis, and mammosphere formation assay method were implemented to detect the biological effects of MST2 ablation on the growth of MCF-7 cells in vitro. The effect of MST2-siRNA on MCF-7 cells tumor growth in vivo was studied in tumor-bearing mouse model. Kaplan-Meier plotter analysis was used to determine the effect of MST2 on overall survival in breast cancer patients. MST2 overexpression increased cell viability marginally. The ablation of MST2 using siRNA dramatically suppressed the viability of the MCF-7 cells, but not ER-negative MDA-MB-231 breast cancer cells. Furthermore, MST2 knockdown increased caspase-dependent apoptosis and led to decreased mammosphere formation. Treatment of MCF-7 tumor-bearing mice with MST2 siRNA significantly inhibited tumor growth. The tumor weight was reduced further when tamoxifen was added. Patients with ER-positive breast cancer with low MST2 expression had better overall survival than did those with high MST2 expression in Kaplan-Meier survival analyses using public datasets. Our results provide new insight into the role of MST2, a key component of the Hippo signaling pathway, in mediating breast cancer progression.
Collapse
|
11
|
Kochi M, Hinoi T, Niitsu H, Miguchi M, Saito Y, Sada H, Sentani K, Sakamoto N, Oue N, Tashiro H, Sotomaru Y, Yasui W, Ohdan H. Oncogenic mutation in RAS-RAF axis leads to increased expression of GREB1, resulting in tumor proliferation in colorectal cancer. Cancer Sci 2020; 111:3540-3549. [PMID: 32629543 PMCID: PMC7541019 DOI: 10.1111/cas.14558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/27/2020] [Accepted: 06/30/2020] [Indexed: 12/22/2022] Open
Abstract
BRAFV600E mutation accounts for up to 90% of all BRAF mutations in human colorectal cancer (CRC), and constitutively activates the MEK‐MAPK pathway. It is recognized that neutralizing mAbs for epidermal growth factor receptor alone are not effective for CRC with BRAFV600E mutation. Therefore, there is increasing interest in identification of the possible therapeutic targets in downstream of BRAF mutation in CRCs. To address this, we studied genome engineered mouse models for colonic neoplasia that has BrafV600E mutation on the basis of Apc inactivation, induced in 2 distinct Cre mouse models, CDX2P‐G22Cre and CDX2P‐CreERT2 mice. We carried out oligonucleotide microarray analysis for colonic neoplasia generated in these mouse models, and compared gene expression profiles among Kras/Braf WT, Kras‐mutated, and Braf‐mutated mouse colon tumors to seek new molecular targets corresponding to the KRAS‐BRAF‐MAPK axis. We found that the expression of the growth regulation by estrogen in breast cancer protein 1 (Greb1) was the most upregulated gene in Braf‐mutated mouse tumors compared to Kras/Braf WT counterparts. The silencing of GREB1 significantly reduced the proliferation and tumorigenesis of CRC cell lines, whereas the overexpression of GREB1 promoted cell proliferation. Although GREB1 was first identified as a hormone‐responsive gene mediating estrogen‐stimulated cell proliferation in endometriosis, breast, and ovarian cancers, these results suggest that RAS‐RAF‐MAPK signaling upregulates GREB1 expression in CRC, resulting in cellular proliferation. Thus, GREB1 is a possible therapeutic target for CRCs with BrafV600E mutation.
Collapse
Affiliation(s)
- Masatoshi Kochi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takao Hinoi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan.,Department of Clinical and Molecular Genetics, Hiroshima University Hospital, Hiroshima, Japan
| | - Hiroaki Niitsu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Masashi Miguchi
- Department of Gastroenterological, Breast and Transplant Surgery, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | - Yasufumi Saito
- Department of Surgery, Chugoku Rosai Hospital, Hiroshima, Japan
| | - Haruki Sada
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan
| | - Kazuhiro Sentani
- Department of Molecular Pathology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Naoya Sakamoto
- Department of Molecular Pathology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Naohide Oue
- Department of Molecular Pathology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Hirotaka Tashiro
- Department of Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan
| | - Yusuke Sotomaru
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Wataru Yasui
- Department of Molecular Pathology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
12
|
Ferreira Almeida C, Oliveira A, João Ramos M, Fernandes PA, Teixeira N, Amaral C. Estrogen receptor-positive (ER +) breast cancer treatment: Are multi-target compounds the next promising approach? Biochem Pharmacol 2020; 177:113989. [PMID: 32330493 DOI: 10.1016/j.bcp.2020.113989] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
Endocrine therapy is currently the main therapeutic approach for estrogen receptor-positive (ER+) breast cancer, the most frequent subtype of breast cancer in women worldwide. For this subtype of tumors, the current clinical treatment includes aromatase inhibitors (AIs) and anti-estrogenic compounds, such as Tamoxifen and Fulvestrant, being AIs the first-line treatment option for post-menopausal women. Moreover, the recent guidelines also suggest the use of these compounds by pre-menopausal women after suppressing ovaries function. However, besides its therapeutic efficacy, the prolonged use of this type of therapies may lead to the development of several adverse effects, as well as, endocrine resistance, limiting the effectiveness of such treatments. In order to surpass this issues and clinical concerns, during the last years, several studies have been suggesting alternative therapeutic approaches, considering the function of aromatase, ERα and ERβ. Here, we review the structural and functional features of these three targets and their importance in ER+ breast cancer treatment, as well as, the current treatment strategies used in clinic, emphasizing the importance of the development of multi-target compounds able to simultaneously modulate these key targets, as a novel and promising therapeutic strategy for this type of cancer.
Collapse
Affiliation(s)
- Cristina Ferreira Almeida
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Ana Oliveira
- UCIBIO.REQUIMTE, Computational Biochemistry Laboratory, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Maria João Ramos
- UCIBIO.REQUIMTE, Computational Biochemistry Laboratory, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Pedro A Fernandes
- UCIBIO.REQUIMTE, Computational Biochemistry Laboratory, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Natércia Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| |
Collapse
|
13
|
Effects of phosphorylated estrogen receptor alpha on apoptosis in human endometrial epithelial cells. Anat Sci Int 2019; 95:240-250. [PMID: 31823335 DOI: 10.1007/s12565-019-00515-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/29/2019] [Indexed: 01/15/2023]
Abstract
It is known that the activities of estrogen receptor α (ERα) can be modulated by epidermal growth factor (EGF) through the phosphatidylinostitol 3-kinase-alpha serine/threonine protein kinase (PI3K-AKT) pathway by phosphorylation. To clarify how ERα functions are regulated in endometrial cells during menstrual cycle, molecules related to phosphorylation of ERα (pERα) were examined. It was found that the expression of phosphorylated AKT on serine 473 (pAKT-Ser473) was increased during the proliferative phase, but decreased in the secretory phase. Although the expression of pAKT on threonine 308 in the proliferative phase was only identified in the wall of arterioles, it was strongly expressed in the cytoplasm of endometrial glandular cells after entering the secretory phase. Further observations revealed that while the expression of pERα-Ser104 was constant, pERα-Ser118 was expressed following a cyclic pattern similar to that of the pAKT-Ser473. Following treatment with specific inhibitors for EGFR-PI3K-AKT pathway, it was found that while the expression of pERα-Ser118 and pERα-Ser167 was inhibited, the induced apoptosis could be antagonized by the addition of estrogen, indicating that a mitochondrial pathway is involved. Therefore, pAKT and pERα or ERα could act cooperatively on coiled arterioles and endometrial cells in order to control menstrual cycle.
Collapse
|
14
|
Pancholi S, Leal MF, Ribas R, Simigdala N, Schuster E, Chateau-Joubert S, Zabaglo L, Hills M, Dodson A, Gao Q, Johnston SR, Dowsett M, Cosulich SC, Maragoni E, Martin LA. Combination of mTORC1/2 inhibitor vistusertib plus fulvestrant in vitro and in vivo targets oestrogen receptor-positive endocrine-resistant breast cancer. Breast Cancer Res 2019; 21:135. [PMID: 31801615 PMCID: PMC6894349 DOI: 10.1186/s13058-019-1222-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/30/2019] [Indexed: 01/14/2023] Open
Abstract
Background Endocrine therapies are still the main strategy for the treatment of oestrogen receptor-positive (ER+) breast cancers (BC), but resistance remains problematic. Cross-talk between ER and PI3K/AKT/mTORC has been associated with ligand-independent transcription of ER. We have previously reported the anti-proliferative effects of the combination of everolimus (an mTORC1 inhibitor) with endocrine therapy in resistance models, but potential routes of escape via AKT signalling can lead to resistance; therefore, the use of dual mTORC1/2 inhibitors has met with significant interest. Methods To address this, we tested the effect of vistusertib, a dual mTORC1 and mTORC2 inhibitor, in a panel of endocrine-resistant and endocrine-sensitive ER+ BC cell lines, with varying PTEN, PIK3CA and ESR1 mutation status. End-points included proliferation, cell signalling, cell cycle and effect on ER-mediated transcription. Two patient-derived xenografts (PDX) modelling endocrine resistance were used to assess the efficacy of vistusertib, fulvestrant or the combination on tumour progression, and biomarker studies were conducted using immunohistochemistry and RNA-seq technologies. Results Vistusertib caused a dose-dependent decrease in proliferation of all the cell lines tested and reduced abundance of mTORC1, mTORC2 and cell cycle markers, but caused an increase in abundance of EGFR, IGF1R and ERBB3 in a context-dependent manner. ER-mediated transcription showed minimal effect of vistusertib. Combined therapy of vistusertib with fulvestrant showed synergy in two ER+ PDX models of resistance to endocrine therapy and delayed tumour progression after cessation of therapy. Conclusions These data support the notion that models of acquired endocrine resistance may have a different sensitivity to mTOR inhibitor/endocrine therapy combinations.
Collapse
Affiliation(s)
- Sunil Pancholi
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Mariana Ferreira Leal
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Ricardo Ribas
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Nikiana Simigdala
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Eugene Schuster
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | | | - Lila Zabaglo
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Margaret Hills
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Andrew Dodson
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Qiong Gao
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | | | - Mitch Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, SW3 6JJ, UK
| | | | | | - Lesley-Ann Martin
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK.
| |
Collapse
|
15
|
Kruger DT, Alexi X, Opdam M, Schuurman K, Voorwerk L, Sanders J, van der Noort V, Boven E, Zwart W, Linn SC. IGF-1R pathway activation as putative biomarker for linsitinib therapy to revert tamoxifen resistance in ER-positive breast cancer. Int J Cancer 2019; 146:2348-2359. [PMID: 31490549 PMCID: PMC7065127 DOI: 10.1002/ijc.32668] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/26/2019] [Accepted: 08/14/2019] [Indexed: 12/18/2022]
Abstract
Preclinical studies indicate that activated IGF-1R can drive endocrine resistance in ER-positive (ER+) breast cancer, but its clinical relevance is unknown. We studied the effect of IGF-1R signaling on tamoxifen benefit in patients and we searched for approaches to overcome IGF-1R-mediated tamoxifen failure in cell lines. Primary tumor blocks from postmenopausal ER+ breast cancer patients randomized between adjuvant tamoxifen versus nil were recollected. Immunohistochemistry for IGF-1R, p-IGF-1R/InsR, p-ERα(Ser118), p-ERα(Ser167) and PI3K/MAPK pathway proteins was performed. Multivariate Cox models were employed to assess tamoxifen efficacy. The association between p-IGF-1R/InsR and PI3K/MAPK pathway activation in MCF-7 and T47D cells was analyzed with Western blots. Cell proliferation experiments were performed under various growth-stimulating and -inhibiting conditions. Patients with ER+, IGF-1R-positive breast cancer without p-IGF-1R/InsR staining (n = 242) had tamoxifen benefit (HR 0.41, p = 0.0038), while the results for p-IGF-1R/InsR-positive patients (n = 125) were not significant (HR 0.95, p = 0.3). High p-ERα(Ser118) or p-ERα(Ser167) expression was associated with less tamoxifen benefit. In MCF-7 cells, IGF-1R stimulation increased phosphorylation of PI3K/MAPK proteins and ERα(Ser167) regardless of IGF-1R overexpression. This could be abrogated by the dual IGF-1R/InsR inhibitor linsitinib, but not by the IGF-IR-selective antibody 1H7. In MCF-7 and T47D cells, stimulation of the IGF-1R/InsR pathway resulted in cell proliferation regardless of tamoxifen. Abrogation of cell growth was regained by addition of linsitinib. In conclusion, p-IGF-1R/InsR positivity in ER+ breast cancer is associated with reduced benefit from adjuvant tamoxifen in postmenopausal patients. In cell lines, stimulation rather than overexpression of IGF-1R is driving tamoxifen resistance to be abrogated by linsitinib.
Collapse
Affiliation(s)
- Dinja T Kruger
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, The Netherlands.,Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Xanthippi Alexi
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Karianne Schuurman
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leonie Voorwerk
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Vincent van der Noort
- Division of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Epie Boven
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Medical Oncology, The National Cancer Institute, Amsterdam, The Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
16
|
Szijgyarto Z, Flach KD, Opdam M, Palmieri C, Linn SC, Wesseling J, Ali S, Bliss JM, Cheang MCU, Zwart W, Coombes RC. Dissecting the predictive value of MAPK/AKT/estrogen-receptor phosphorylation axis in primary breast cancer to treatment response for tamoxifen over exemestane: a Translational Report of the Intergroup Exemestane Study (IES)-PathIES. Breast Cancer Res Treat 2019; 175:149-163. [PMID: 30680659 PMCID: PMC6491661 DOI: 10.1007/s10549-018-05110-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/18/2018] [Indexed: 01/06/2023]
Abstract
PURPOSE The prognostic and predictive values of the MAPK/AKT/ERα phosphorylation axis (pT202/T204MAPK, pT308AKT, pS473AKT, pS118ERα and pS167ERα) in primary tumours were assessed to determine whether these markers can differentiate between patient responses for switching adjuvant endocrine therapy after 2-3 years from tamoxifen to exemestane and continued tamoxifen monotherapy in the Intergroup Exemestane Study (IES). METHODS Of the 4724 patients in IES, 1506 were managed in a subset of centres (N = 89) participating in PathIES. These centres recruited 1282 (85%, 1282/1506) women into PathIES of whom 1036 had phospho-marker data. All phospho-markers were analysed by immunohistochemistry staining. Multivariable Cox proportional hazards models of the phospho-markers for disease-free survival (DFS) and overall survival (OS) were adjusted for clinicopathological factors. Treatment effects on the biomarker expression were determined by interaction tests. Benjamini-Hochberg adjustment for multiple testing with a false discovery rate of 10% was applied (pBH). RESULTS Phospho-T202/T204MAPK, pS118ERα and pS167ERα were all found to be correlated (pBH = 0.0002). These markers were not associated with either DFS or OS when controlling for the established clinicopathological factors. Interaction terms between the phospho-markers and treatment strategies for either DFS or OS were not statistically significant (pBH > 0.05 for all). CONCLUSIONS This PathIES study confirmed previously described associations between the phosphorylation site markers of AKT, MAPK and ERα activity in postmenopausal breast cancer patients. No prognostic correlations between the phosphorylation markers and clinical outcome were found, nor were they predictive for clinical outcomes among patients who switched therapy over those treated with tamoxifen alone.
Collapse
Affiliation(s)
- Zsolt Szijgyarto
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG UK
| | - Koen D. Flach
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Carlo Palmieri
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3BX UK
- Academic Department of Medical Oncology, Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, CH63 4JY UK
- Department of Cancer and Surgery, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN UK
| | - Sabine C. Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Department of Medical Onology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Simak Ali
- Department of Cancer and Surgery, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN UK
| | - Judith M. Bliss
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG UK
| | - Maggie Chon U. Cheang
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG UK
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - R. Charles Coombes
- Department of Cancer and Surgery, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN UK
| |
Collapse
|
17
|
Liu XY, Ma D, Xu XE, Jin X, Yu KD, Jiang YZ, Shao ZM. Genomic Landscape and Endocrine-Resistant Subgroup in Estrogen Receptor-Positive, Progesterone Receptor-Negative, and HER2-Negative Breast Cancer. Am J Cancer Res 2018; 8:6386-6399. [PMID: 30613307 PMCID: PMC6299689 DOI: 10.7150/thno.29164] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/08/2018] [Indexed: 02/01/2023] Open
Abstract
Estrogen receptor-positive, progesterone receptor-negative, and human epidermal growth factor receptor 2 (HER2)-negative (ER+PR-HER2-) breast cancer comprise a special type of breast cancer that constitutes ~10% of all breast cancer patients. ER+PR-HER2- tumor benefits less from endocrine therapy, while its genomic features remain elusive. In this study, we systematically assessed the multiomic landscape and endocrine responsiveness of ER+PR-HER2- breast cancer. Methods: This study incorporated five cohorts. The first and second cohorts were from the Surveillance, Epidemiology, and End Results database (n=130,856) and Molecular Taxonomy of Breast Cancer International Consortium (n=1,055) for analyzing survival outcomes and endocrine responsiveness. The third cohort was from The Cancer Genome Atlas (n=630) for multiomic analysis and endocrine-resistant subgroup exploration. The fourth cohort, from the MD Anderson database (n=92), was employed to assist gene selection. The fifth cohort was a prospective observational cohort from Fudan University Shanghai Cancer Center (n=245) that was utilized to validate the gene-defined subgroup by immunohistochemistry (IHC). Results: Clinically, ER+PR-HER2- tumors showed lower endocrine responsiveness than did ER+PR+HER2- tumors. Genomically, copy number loss or promoter methylation of PR genes occurred in 75% of ER+PR-HER2- tumors, collectively explaining PR loss. ER+PR-HER2- tumors had higher TP53 (30.3% vs. 17.0%) and lower PIK3CA mutation rates (25.8% vs. 42.7%) and exhibited more ZNF703 (21.5% vs. 13.6%) and RPS6KB1 (18.5% vs. 7.8%) amplification events than ER+PR+HER2- tumors. Among ER+PR-HER2- tumors, nearly 20% were of the PAM50-defined non-luminal-like subgroup and manifested lower endocrine sensitivity scores and enriched biosynthesis, metabolism and DNA replication pathways. We further identified the non-luminal-like subgroup using three IHC markers, GATA3, CK5, and EGFR. These IHC-defined non-luminal-like (GATA3-negative, CK5-positive and/or EGFR-positive) tumors received limited benefit from adjuvant endocrine therapy. Conclusion: ER+PR-HER2- breast cancer consists of clinically and genomically distinct groups that may require different treatment strategies. The non-luminal-like subgroup was associated with reduced benefit from endocrine therapy.
Collapse
|
18
|
Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol Ther 2018; 186:1-24. [DOI: 10.1016/j.pharmthera.2017.12.012] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
19
|
Ishida N, Baba M, Hatanaka Y, Hagio K, Okada H, Hatanaka KC, Togashi K, Matsuno Y, Yamashita H. PIK3CA mutation, reduced AKT serine 473 phosphorylation, and increased ERα serine 167 phosphorylation are positive prognostic indicators in postmenopausal estrogen receptor-positive early breast cancer. Oncotarget 2018; 9:17711-17724. [PMID: 29707142 PMCID: PMC5915150 DOI: 10.18632/oncotarget.24845] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 02/28/2018] [Indexed: 12/12/2022] Open
Abstract
Although endocrine therapy is the most important treatment option in estrogen receptor (ER)-positive breast cancer, new strategies, such as molecular targeted agents together with endocrine therapy are required to improve survival. PIK3CA is the most frequent mutated gene in ER-positive early breast cancers, and PIK3CA mutation status is reported to affect activation of AKT and ERα. Moreover, recent studies demonstrate that patients had a better prognosis when tumors expressed ER, androgen receptor (AR), and vitamin D receptor (VDR). In this study, we examined expression of AR and VDR, phosphorylation of AKT serine (Ser) 473 (AKT phospho-Ser473) and ERα Ser167 (ERα phospho-Ser167) by immunohistochemistry in ER-positive, HER2-negative early breast cancer. PIK3CA gene mutations were also detected in genomic DNA extracted from tumor blocks. Correlations between these biological markers, clinicopathological factors and prognosis were analyzed. Levels of AKT phospho-Ser473 were significantly higher in premenopausal women than in postmenopausal women. In contrast, AR expression was significantly higher in postmenopausal women than in premenopausal women. PIK3CA mutations were detected in 47% in premenopausal women and 47% in postmenopausal women. Postmenopausal women with PIK3CA wild-type tumors had significantly worse disease-free survival than patients with PIK3CA mutant tumors. Low levels of AKT phospho-Ser473 and high levels of ERα phospho-Ser167 were strongly associated with increased disease-free survival in postmenopausal women. Evaluation of ERα activation, in addition to PIK3CA mutation status, might be helpful in identifying patients who are likely to benefit from endocrine therapy alone versus those who are not in postmenopausal ER-positive early breast cancer.
Collapse
Affiliation(s)
- Naoko Ishida
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Motoi Baba
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Yutaka Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
- Research Division of Companion Diagnostics, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Kanako Hagio
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Hiromi Okada
- Department of Surgical Pathology, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Kanako C. Hatanaka
- Research Division of Companion Diagnostics, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Kenichi Togashi
- Roche Diagnostics K.K., Konan, Minato-ku, Tokyo 108-0075, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
- Research Division of Companion Diagnostics, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Hiroko Yamashita
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| |
Collapse
|
20
|
Ludwik KA, McDonald OG, Brenin DR, Lannigan DA. ERα-Mediated Nuclear Sequestration of RSK2 Is Required for ER + Breast Cancer Tumorigenesis. Cancer Res 2018; 78:2014-2025. [PMID: 29351904 DOI: 10.1158/0008-5472.can-17-2063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/02/2017] [Accepted: 01/16/2018] [Indexed: 11/16/2022]
Abstract
Although ribosomal protein S6 kinase A3 (RSK2) activation status positively correlates with patient responses to antiestrogen hormonal therapies, the mechanistic basis for these observations is unknown. Using multiple in vitro and in vivo models of estrogen receptor-positive (ER+) breast cancer, we report that ERα sequesters active RSK2 into the nucleus to promote neoplastic transformation and facilitate metastatic tumor growth. RSK2 physically interacted with ERα through its N terminus to activate a proneoplastic transcriptional network critical to the ER+ lineage in the mammary gland, thereby providing a gene signature that effectively stratified patient tumors according to ERα status. ER+ tumor growth was strongly dependent on nuclear RSK2, and transgenic mice engineered to stably express nuclear RSK2 in the mammary gland developed high-grade ductal carcinoma in situ Mammary cells isolated from the transgenic model and introduced systemically successfully disseminated and established metastatic lesions. Antiestrogens disrupted the interaction between RSK2 and ERα, driving RSK2 into the cytoplasm and impairing tumor formation. These findings establish RSK2 as an obligate participant of ERα-mediated transcriptional programs, tumorigenesis, and divergent patient responses to antiestrogen therapies.Significance: Nuclear accumulation of active RSK drives a protumorigenic transcriptional program and renders ER+ breast cancer susceptible to endocrine-based therapies. Cancer Res; 78(8); 2014-25. ©2018 AACR.
Collapse
Affiliation(s)
- Katarzyna A Ludwik
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Oliver G McDonald
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David R Brenin
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Deborah A Lannigan
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee. .,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
21
|
Zeng X, Che X, Liu YP, Qu XJ, Xu L, Zhao CY, Zheng CL, Hou KZ, Teng Y. FEN1 knockdown improves trastuzumab sensitivity in human epidermal growth factor 2-positive breast cancer cells. Exp Ther Med 2017; 14:3265-3272. [PMID: 28912877 DOI: 10.3892/etm.2017.4873] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 04/28/2017] [Indexed: 12/18/2022] Open
Abstract
Trastuzumab has been widely applied as a treatment for human epidermal growth factor 2 (HER2)-overexpressing breast cancer. However, the therapeutic efficacy of trastuzumab is limited. Flap endonuclease 1 (FEN1) is a multifunctional endonuclease that has a crucial role in DNA recombination and repair. Inhibition of FEN1 is associated with the reversal of anticancer drug resistance. However, it is unclear whether FEN1 is involved in trastuzumab resistance. In the present study, it was demonstrated that trastuzumab increases the expression of FEN1, and FEN1 knockdown significantly enhanced the sensitivity of BT474 cells to trastuzumab (P<0.05). It was also revealed that trastuzumab induced HER receptor activation, increased binding with FEN1 and estrogen receptor α (ERα), and upregulated ERα-target gene transcription (P<0.05). Upon silencing of FEN1 expression with siRNA, activation of HER receptor and FEN1 binding to ERα were decreased, and trastuzumab-induced ERα target gene upregulation was partially ameliorated (P<0.05). These results suggest that FEN1 may mediate trastuzumab resistance via inducing HER receptor activation and enhancing ERα-target gene transcription. The findings of the present study indicate a novel role of FEN1 in trastuzumab resistance, suggesting that targeting FEN1 may enhance the efficiency of trastuzumab as a treatment for HER2-positive breast cancer.
Collapse
Affiliation(s)
- Xue Zeng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yun-Peng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiu-Juan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lu Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chen-Yang Zhao
- Central Laboratory, The Fourth Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Chun-Lei Zheng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ke-Zuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuee Teng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
22
|
Cheng KKW, Dickson A, Gujam FJA, McMillan DC, Edwards J. The relationship between oestrogen receptor-alpha phosphorylation and the tumour microenvironment in patients with primary operable ductal breast cancer. Histopathology 2016; 70:782-797. [PMID: 27891654 DOI: 10.1111/his.13134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/09/2016] [Accepted: 11/23/2016] [Indexed: 01/26/2023]
Abstract
AIMS Although the role of phosphorylation of oestrogen receptor (ER) at serines 118 (p-S118) and 167 (p-S167) has been studied, the relationship between p-S118, p-S167 and the tumour microenvironment in ER-positive primary operable ductal breast cancers have not been investigated. The aims of this study are to investigate (i) the relationship between p-S118/p-S167 and the tumour microenvironment, and (ii) the effect of p-S118/167 on survival and recurrence in ER-positive primary operable ductal breast cancers. METHODS AND RESULTS Patients presenting at three Glasgow hospitals between 1995 and 1998 with invasive ductal ER-positive primary breast cancers were studied (n = 294). Immunohistochemical staining of p-S118 and p-S167 was performed and their association with clinicopathological characteristics, cancer-specific survival (CSS) and recurrence-free interval (RFI) were examined. In the whole cohort, tumour size (P < 0.05) and microvessel density (P < 0.05) were associated with high p-S118 while increased micovessel density (P < 0.05), apoptosis (P < 0.05), general inflammatory infiltrate measured using the Klintrup-Makinen score (P < 0.05) and macrophage infiltrate (P < 0.05) were found to be associated with high p-S167. Only high p-S167 was associated with shorter CSS (P < 0.005) and shorter RFI in the whole cohort (P = 0.001) and separately in the luminal A (P < 0.05) and B tumours (P < 0.05). CONCLUSIONS This study showed that both p-S118 and p-S167 were associated with several microenvironmental factors, including increased microvessel density. In particular, p-S167 was associated with reduced RFI and CSS in the whole cohort and RFI in luminal A and B tumours and could possibly be employed to predict response to kinase inhibitors.
Collapse
Affiliation(s)
| | - Ashley Dickson
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Fadia J A Gujam
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences, University of Glasgow, Royal Infirmary, Glasgow, UK
| | - Donald C McMillan
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences, University of Glasgow, Royal Infirmary, Glasgow, UK
| | - Joanne Edwards
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
23
|
Post-relapse survival in patients with the early and late distant recurrence in estrogen receptor-positive HER2-negative breast cancer. Breast Cancer 2016; 24:473-482. [PMID: 27628678 DOI: 10.1007/s12282-016-0730-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Few studies have been performed on post-relapse survival in patients with the early and late distant recurrence in estrogen receptor (ER)-positive, HER2-negative breast cancer. METHODS A total of 205 patients with the early distant recurrence and 134 patients with the late distant recurrence of ER-positive, HER2-negative breast cancer who had undergone breast surgery or neoadjuvant chemotherapy between January 2000 and December 2004 were registered from nine institutions. Prognostic factors for post-relapse survival in patients with the early and late recurrence were analyzed. RESULTS Post-relapse survival was significantly longer in patients with the late recurrence than in patients with the early recurrence. Predictive factors for post-relapse survival in patients with the early recurrence were lack of adjuvant chemotherapy, a long disease-free interval, and long durations of endocrine therapies and chemotherapies after relapse. In patients with the late recurrence, post-relapse survival was significantly improved for those individuals with one metastatic organ at relapse and individuals who were treated with the first-line and subsequent endocrine therapies for prolonged periods. Moreover, ER expression in primary breast tumors of late recurrence patients was significantly higher with a duration of the first-line endocrine therapy >6 months than in those with a duration ≤6 months. CONCLUSION Predictors for prognosis after relapse differed between patients with the early and late distant recurrence. Endocrine responsiveness after relapse is a key factor for improved post-relapse survival, and it is thus important to establish whether metastatic tumors are endocrine-resistant in ER-positive, HER2-negative recurrent breast cancer.
Collapse
|
24
|
Abstract
INTRODUCTION The p90 ribosomal S6 kinases (RSK) are a family of Ser/Thr protein kinases that are downstream effectors of MEK1/2-ERK1/2. Increased RSK activation is implicated in the etiology of multiple pathologies, including numerous types of cancers, cardiovascular disease, liver and lung fibrosis, and infections. AREAS COVERED The review summarizes the patent and scientific literature on small molecule modulators of RSK and their potential use as therapeutics. The patents were identified using World Intellectual Property Organization and United States Patent and Trademark Office databases. The compounds described are predominantly RSK inhibitors, but a RSK activator is also described. The majority of the inhibitors are not RSK-specific. EXPERT OPINION Based on the overwhelming evidence that RSK is involved in a number of diseases that have high mortalities it seems surprising that there are no RSK modulators that have pharmacokinetic properties suitable for in vivo use. MEK1/2 inhibitors are in the clinic, but the efficacy of these compounds appears to be limited by their side effects. We hypothesize that targeting the downstream effectors of MEK1/2, like RSK, are an untapped source of drug targets and that they will generate less side effects than MEK1/2 inhibitors because they regulate fewer effectors.
Collapse
Affiliation(s)
- Katarzyna A Ludwik
- a Department of Pathology, Microbiology & Immunology , Vanderbilt University , Nashville , TN , USA
| | - Deborah A Lannigan
- a Department of Pathology, Microbiology & Immunology , Vanderbilt University , Nashville , TN , USA.,b Department of Cancer Biology , Vanderbilt University , Nashville , TN , USA
| |
Collapse
|
25
|
Chan HJ, Petrossian K, Chen S. Structural and functional characterization of aromatase, estrogen receptor, and their genes in endocrine-responsive and -resistant breast cancer cells. J Steroid Biochem Mol Biol 2016; 161:73-83. [PMID: 26277097 PMCID: PMC4752924 DOI: 10.1016/j.jsbmb.2015.07.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 11/22/2022]
Abstract
Aromatase and estrogen receptor α (ER) are two key proteins for the proliferation of endocrine-responsive and -resistant breast cancers. Aromatase is an enzyme involved in the conversion of androgen (such as testosterone) to estrogen (such as 17β-estradiol). It is also a very effective therapeutic target for the treatment of endocrine-responsive breast cancer. Comparing endocrine-responsive and -resistant breast cancer, aromatase protein levels do not change significantly. Aromatase activity; however, can be increased via PI3K/Akt/IGFR signaling pathways in endocrine resistant cells. The activity of aromatase has been reported to be modulated by phosphorylation. The ER is an important steroid nuclear receptor in the proliferation of both endocrine-responsive and -resistant cells. Although the mutation or amplification of ER can cause endocrine resistance, it is not commonly found. Some point mutations and translocation events have been characterized and shown to promote estrogen-independent growth. Phosphorylation by cross-talk with growth factor pathways is one of the main mechanisms for ligand-independent activation of ER. Taken together, both ER and aromatase are important in ER-dependent breast cancer and the development of endocrine resistance.
Collapse
Affiliation(s)
- Hei Jason Chan
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States.
| |
Collapse
|
26
|
Flach KD, Zwart W. The first decade of estrogen receptor cistromics in breast cancer. J Endocrinol 2016; 229:R43-56. [PMID: 26906743 DOI: 10.1530/joe-16-0003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 02/23/2016] [Indexed: 02/03/2023]
Abstract
The advent of genome-wide transcription factor profiling has revolutionized the field of breast cancer research. Estrogen receptor α (ERα), the major drug target in hormone receptor-positive breast cancer, has been known as a key transcriptional regulator in tumor progression for over 30 years. Even though this function of ERα is heavily exploited and widely accepted as an Achilles heel for hormonal breast cancer, only since the last decade we have been able to understand how this transcription factor is functioning on a genome-wide scale. Initial ChIP-on-chip (chromatin immunoprecipitation coupled with tiling array) analyses have taught us that ERα is an enhancer-associated factor binding to many thousands of sites throughout the human genome and revealed the identity of a number of directly interacting transcription factors that are essential for ERα action. More recently, with the development of massive parallel sequencing technologies and refinements thereof in sample processing, a genome-wide interrogation of ERα has become feasible and affordable with unprecedented data quality and richness. These studies have revealed numerous additional biological insights into ERα behavior in cell lines and especially in clinical specimens. Therefore, what have we actually learned during this first decade of cistromics in breast cancer and where may future developments in the field take us?
Collapse
Affiliation(s)
- Koen D Flach
- Division of Molecular PathologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Molecular PathologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Das N, Datta N, Chatterjee U, Ghosh MK. Estrogen receptor alpha transcriptionally activates casein kinase 2 alpha: A pivotal regulator of promyelocytic leukaemia protein (PML) and AKT in oncogenesis. Cell Signal 2016; 28:675-87. [PMID: 27012497 DOI: 10.1016/j.cellsig.2016.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/17/2016] [Indexed: 01/17/2023]
Abstract
Protein kinase CK2α is frequently upregulated in different cancers. Alteration of CK2α expression and its activity is sufficient to induce dramatic changes in cell fate. It has been established that CK2α induces oncogenesis through modulation of both AKT and PML. CK2α has been found to be overexpressed in breast cancer. In contrary, statistical reports have shown low level of PML. However, the regulation of CK2α gene expression is not fully understood. In the current study, we found that CK2α and activated AKT positively correlate with ERα, whereas PML follows an inverse correlation in human breast cancer tissues. Modulation of ERα signalling leads to recruitment of activated ERα on the ERE sites of CK2α promoter, resulting in CK2α transactivation. Furthermore, the DMBA induced tumours in rat showed elevated level of active CK2α. Consequently it mediates enhancement of AKT activity and PML degradation, resulting in increased cellular proliferation, migration and metastasis. Syngeneic ERα overexpressing stable mouse 4T1 cells produce larger primary tumours and metastatic lung nodules in mice, corroborating our in vitro findings. Hence, our study provides a novel route of ERα dependent CK2α mediated oncogenesis that causes upregulation and consequent AKT activation along with degradation of tumour suppressor PML.
Collapse
Affiliation(s)
- Nilanjana Das
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, West Bengal, India.
| | - Neerajana Datta
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, West Bengal, India.
| | - Uttara Chatterjee
- Division of Pathology, Park Clinic, 4, Gorky Terrace, Kolkata 700017, India.
| | - Mrinal Kanti Ghosh
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, West Bengal, India.
| |
Collapse
|
28
|
Anbalagan M, Rowan BG. Estrogen receptor alpha phosphorylation and its functional impact in human breast cancer. Mol Cell Endocrinol 2015; 418 Pt 3:264-72. [PMID: 25597633 DOI: 10.1016/j.mce.2015.01.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/08/2015] [Accepted: 01/12/2015] [Indexed: 02/08/2023]
Abstract
Estrogen receptor α (ERα) is a member of the nuclear receptor superfamily of transcription factors that regulates cell proliferation, differentiation and homeostasis in various tissues. Sustained exposure to estrogen/estradiol (E2) increases the risk of breast, endometrial and ovarian cancers. ERα function is also regulated by phosphorylation through various kinase signaling pathways that will impact various ERα functions including chromatin interaction, coregulator recruitment and gene expression, as well impact breast tumor growth/morphology and breast cancer patient response to endocrine therapy. However, many of the previously characterized ERα phosphorylation sites do not fully explain the impact of receptor phosphorylation on ERα function. This review discusses work from our laboratory toward understanding a role of ERα site-specific phosphorylation in ERα function and breast cancer. The key findings discussed in this review are: (1) the effect of site specific ERα phosphorylation on temporal recruitment of ERα and unique coactivator complexes to specific genes; (2) the impact of stable disruption of ERα S118 and S167 phosphorylation in breast cancer cells on eliciting unique gene expression profiles that culminate in significant effects on breast cancer growth/morphology/migration/invasion; (3) the Src kinase signaling pathway that impacts ERα phosphorylation to alter ERα function; and (4) circadian disruption by light exposure at night leading to elevated ERK1/2 and Src kinase and phosphorylation of ERα, concomitant with tamoxifen resistance in breast tumor models. Results from these studies demonstrate that even changes to single ERα phosphorylation sites can have a profound impact on ERα function in breast cancer. Future work will extend beyond single site phosphorylation analysis toward identification of specific patterns/profiles of ERα phosphorylation under different physiological/pharmacological conditions to understand how common phosphorylation profiles in breast cancer program specific physiological endpoints such as growth, apoptosis, migration/invasion, and endocrine therapy response.
Collapse
Affiliation(s)
- Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Brian G Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
29
|
mTORC1 directly phosphorylates and activates ERα upon estrogen stimulation. Oncogene 2015; 35:3535-43. [PMID: 26522726 PMCID: PMC4853282 DOI: 10.1038/onc.2015.414] [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: 05/11/2015] [Revised: 09/09/2015] [Accepted: 09/28/2015] [Indexed: 01/08/2023]
Abstract
Breast cancer is the leading cause of cancer-related deaths among women. Approximately 75% of breast cancers are estrogen receptor α (ERα) positive, underscoring the dependence of cancer cells on estrogen for growth and survival. Patients treated with endocrine therapy often develop resistance, either de novo or acquired, which in some cases is caused by aberrations within the growth factor signaling pathways. The mechanistic target of rapamycin complex 1 (mTORC1) has emerged as a critical node in estrogenic signaling. We have previously shown that mTORC1 can phosphorylate and activate ERα on S167 via its effector the 40S ribosomal S6 kinase 1 (S6K1). Presently, we have uncovered a direct link between mTORC1 and ERα. We found that ERα binds to regulatory-associated protein of mTOR (Raptor) and causes it to translocate to the nucleus upon estrogen stimulation. Additionally, we identified mTOR as the kinase that phosphorylates ERα on S104/106 and activates transcription of ER target genes. Our findings show a direct link between mTORC1 and ERα, which further implicates mTORC1 signaling in the pathogenesis of ER-positive breast cancer and provides rationale for FDA-approved use of mTORC1 inhibitors in combination with endocrine agents for treatment of this disease.
Collapse
|
30
|
Wang C, Huang Y, Sheng J, Huang H, Zhou J. Estrogen receptor alpha inhibits RLR-mediated immune response via ubiquitinating TRAF3. Cell Signal 2015; 27:1977-83. [PMID: 26186972 DOI: 10.1016/j.cellsig.2015.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/13/2015] [Accepted: 07/13/2015] [Indexed: 12/20/2022]
Abstract
RIG-I-like receptors (RLRs) function as key sentinel receptor for invading viruses. Moderate activation of RLR signaling is critical for efficient viral clearance without harmful immunopathology. Estrogen receptor alpha (ERα) is a member of the nuclear receptor superfamily of ligand-activated transcription factors and is involved in the regulation of innate immune responses. However, the effects of ERα on RLR signaling and the molecular mechanisms are poorly understood. In this study, we identify ERα as a negative regulator of RLR-triggered antiviral immune responses. The expression level of ERα is upregulated following RLR activation in macrophages. In the absence of ligand, VSV infection phosphorylates ERα at serine 167. ERα inhibits VSV-induced IRF3 activation. We further demonstrate that ERα directly interacts with TRAF3 and promotes K48-linked proteasomal degradation of TRAF3. Consistently, ERα inhibits VSV-triggered IFN-β production in macrophages in a ligand independent mechanism. Thus, ERα functions as a negative feedback regulator of RLR-triggered antiviral immune responses. These findings also provide the insights that separate the immune effects of ERα from its ligand-induced hormonal effects.
Collapse
Affiliation(s)
- Changxing Wang
- The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang, China; Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yue Huang
- The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang, China
| | - Jianzhong Sheng
- The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang, China
| | - Hefeng Huang
- The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang, China
| | - Jun Zhou
- The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang, China; Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
31
|
Légaré S, Cavallone L, Mamo A, Chabot C, Sirois I, Magliocco A, Klimowicz A, Tonin PN, Buchanan M, Keilty D, Hassan S, Laperrière D, Mader S, Aleynikova O, Basik M. The Estrogen Receptor Cofactor SPEN Functions as a Tumor Suppressor and Candidate Biomarker of Drug Responsiveness in Hormone-Dependent Breast Cancers. Cancer Res 2015; 75:4351-63. [PMID: 26297734 DOI: 10.1158/0008-5472.can-14-3475] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 07/10/2015] [Indexed: 11/16/2022]
Abstract
The treatment of breast cancer has benefitted tremendously from the generation of estrogen receptor-α (ERα)-targeted therapies, but disease relapse continues to pose a challenge due to intrinsic or acquired drug resistance. In an effort to delineate potential predictive biomarkers of therapy responsiveness, multiple groups have identified several uncharacterized cofactors and interacting partners of ERα, including Split Ends (SPEN), a transcriptional corepressor. Here, we demonstrate a role for SPEN in ERα-expressing breast cancers. SPEN nonsense mutations were detectable in the ERα-expressing breast cancer cell line T47D and corresponded to undetectable protein levels. Further analysis of 101 primary breast tumors revealed that 23% displayed loss of heterozygosity at the SPEN locus and that 3% to 4% harbored somatically acquired mutations. A combination of in vitro and in vivo functional assays with microarray-based pathway analyses showed that SPEN functions as a tumor suppressor to regulate cell proliferation, tumor growth, and survival. We also found that SPEN binds ERα in a ligand-independent manner and negatively regulates the transcription of ERα targets. Moreover, we demonstrate that SPEN overexpression sensitizes hormone receptor-positive breast cancer cells to the apoptotic effects of tamoxifen, but has no effect on responsiveness to fulvestrant. Consistent with these findings, two independent datasets revealed that high SPEN protein and RNA expression in ERα-positive breast tumors predicted favorable outcome in patients treated with tamoxifen alone. Together, our data suggest that SPEN is a novel tumor-suppressor gene that may be clinically useful as a predictive biomarker of tamoxifen response in ERα-positive breast cancers.
Collapse
Affiliation(s)
- Stéphanie Légaré
- Department of Surgery and Oncology, McGill University, Montréal, Québec, Canada. Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - Luca Cavallone
- Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - Aline Mamo
- Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - Catherine Chabot
- Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - Isabelle Sirois
- Department of Surgery and Oncology, McGill University, Montréal, Québec, Canada. Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - Anthony Magliocco
- Department of Pathology, University of Calgary, Calgary, Alberta, Canada
| | | | - Patricia N Tonin
- Department of Human Genetics, McGill University and The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada. Department of Medicine, McGill University and The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Marguerite Buchanan
- Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - Dana Keilty
- Department of Surgery and Oncology, McGill University, Montréal, Québec, Canada. Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - Saima Hassan
- Department of Surgery and Oncology, McGill University, Montréal, Québec, Canada. Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | - David Laperrière
- Institut de recherche en immunologie et cancérologie, IRIC, Montréal, Québec, Canada
| | - Sylvie Mader
- Institut de recherche en immunologie et cancérologie, IRIC, Montréal, Québec, Canada. Department de Biochimie, Université de Montréal, Montréal, Québec, Canada
| | - Olga Aleynikova
- Department of Pathology, Jewish General Hospital, Montréal, Quebec, Canada
| | - Mark Basik
- Department of Surgery and Oncology, McGill University, Montréal, Québec, Canada. Department of Oncology and Surgery, Lady Davis Institute for Medical Research, Montréal, Québec, Canada.
| |
Collapse
|
32
|
Wang Y, Niu XL, Guo XQ, Yang J, Li L, Qu Y, Xiu Hu C, Mao LQ, Wang D. IL6 induces TAM resistance via kinase-specific phosphorylation of ERα in OVCA cells. J Mol Endocrinol 2015; 54:351-61. [PMID: 25943392 DOI: 10.1530/jme-15-0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/01/2015] [Indexed: 01/16/2023]
Abstract
About 40-60% of ovarian cancer (OVCA) cases express ERα, but only a small proportion of patients respond clinically to anti-estrogen treatment with estrogen receptor (ER) antagonist tamoxifen (TAM). The mechanism of TAM resistance in the course of OVCA progression remains unclear. However, IL6 plays a critical role in the development and progression of OVCA. Our recent results indicated that IL6 secreted by OVCA cells may promote the resistance of these cells to TAM via ER isoforms and steroid hormone receptor coactivator-1. Here we demonstrate that both exogenous (a relatively short period of treatment with recombinant IL6) and endogenous IL6 (generated as a result of transfection with a plasmid encoding sense IL6) increases expression of pERα-Ser118 and pERα-Ser167 in non-IL6-expressing A2780 cells, while deleting endogenous IL6 expression in IL6-overexpressing CAOV-3 cells (by transfection with a plasmid encoding antisense IL6) reduces expression of pERα-Ser118 and pERα-Ser167, indicating that IL6-induced TAM resistance may also be associated with increased expression of pERα-Ser118 and pERα-Ser167 in OVCA cells. Results of further investigation indicate that IL6 phosphorylates ERα at Ser118 and Ser167 by triggering activation of MEK/ERK and phosphotidylinositol 3 kinase/Akt signaling, respectively, to activate the ER pathway and thereby induce OVCA cells resistance to TAM. These results indicate that IL6 secreted by OVCA cells may also contribute to the refractoriness of these cells to TAM via the crosstalk between ER and IL6-mediated intracellular signal transduction cascades. Overexpression of IL6 not only plays an important role in OVCA progression but also promotes TAM resistance. Our results indicate that TAM-IL6-targeted adjunctive therapy may lead to a more effective intervention than TAM alone.
Collapse
Affiliation(s)
- Yue Wang
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Xiu Long Niu
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Xiao Qin Guo
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Jing Yang
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Ling Li
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Ye Qu
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Cun Xiu Hu
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Li Qun Mao
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Dan Wang
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental HazardTianjin, People's Republic of ChinaDepartment of Pathogenic Biology and ImmunologyLogistics College of Chinese People's Armed Police Forces, Dongli District, Huizhi Ring Road, Number 1, Tianjin 300309, People's Republic of ChinaDepartment of Infectious DiseasesAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of PharmacologyLogistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of ChinaDepartment of Gynecology and ObstetricsAffiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| |
Collapse
|
33
|
Sun J, Kil C, Stankewich MC, Yao Z, Li J, Vortmeyer AO. A 10-minute prototype assay for tissue degradation monitoring in clinical specimens. Exp Mol Pathol 2015; 99:86-94. [PMID: 26033311 DOI: 10.1016/j.yexmp.2015.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 05/27/2015] [Indexed: 02/04/2023]
Abstract
We recently identified alpha II spectrin as a Tissue Degradation Indicator (TDI) and demonstrated that intrinsic spectrin-breakdown levels reliably reveal tissue degradation status in biospecimens. With the present study, we introduce an in vitro biological assay to mimic the endogenous spectrin-breakdown process and serve as degradation monitor (DM). By initiating the DM at the time of specimen collection and by attaching the DM to respective specimens, specimen degradation can be assessed by DM readout without specimen consumption. Using a protease inhibitory assay and protease-targeted immunoassays, we identified calpain as the protease responsible for degradation-induced spectrin breakdown. To recapitulate spectrin degradation in vitro, we developed several enzymatic assays in test tubes by incubating recombinant spectrins and synthetic Fluorescence Resonance Energy Transfer (FRET)-based spectrin peptides with purified human and porcine calpains. The in vitro assays reliably performed in different environments for a limited time due to loss of calpain activity. To maintain longer calpain activity, we introduced cultured cells as calpain providers into the in vitro assays. Under a variety of degradative conditions, including 4°C, 13°C, 23°C, 29°C, 37°C, freezing, and freeze-thaw steps, we compared the use of this prototype DM to the intrinsic spectrin cleavage assay (ISCA) in specimen degradation assessment using animal models. A strong correlation (r=0.9895) was detected between the DM-revealed degradation and the ISCA-revealed degradation. Notably, the DM-based degradation assessment takes only 10min and does not jeopardize the tissue itself, whereas the ISCA-based degradation assessment needs to sacrifice tissues and takes several hours to accomplish. Our data suggests the application of an in vitro degradation monitor for fast, real time, and non-invasive assessment of specimen degradation. This observation could lead to a transformative product dedicated to biospecimen quality control. This study also addresses critical, yet unmet needs for developing a universal standard for specimen degradation measurement.
Collapse
Affiliation(s)
- Jia Sun
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, PR China; Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Catherine Kil
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Michael C Stankewich
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Zhi Yao
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, PR China
| | - Jie Li
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, United States.
| | - Alexander O Vortmeyer
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
34
|
Hayashi SI, Kimura M. Mechanisms of hormonal therapy resistance in breast cancer. Int J Clin Oncol 2015; 20:262-7. [PMID: 25652907 DOI: 10.1007/s10147-015-0788-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 02/07/2023]
Abstract
Whilst estrogen receptor (ER)-positive breast cancers are preferentially treated with hormone therapy, approximately one-third of them relapse. The mechanisms of refractoriness have been investigated by numerous studies but have not been fully clarified. Hormonal therapy resistance, particularly aromatase inhibitor (AI) resistance, may be related to the acquisition of alternative intracellular ER signaling. We have been investing the mechanisms using cancer specimens and cell lines by monitoring the transcription activity of ERs. AI refractory specimens showed diverse ER activity in the adenovirus estrogen receptor element-green fluorescent protein (ERE-GFP) assay and varied sensitivity to anti-estrogens, indicating the existence of multiple resistant mechanisms. We established six different types of cell lines mimicking AI resistance from ERE-GFP-introduced ER-positive cell lines. They revealed that multiple and alternative ER activating pathways were involved in the resistance, such as phosphorylation-dependent or androgen metabolite-dependent mechanisms. The response to fulvestrant and mammalian target of rapamycin inhibitor also varied among individual resistant cell lines. These results indicate that further subclassification of ER-positive breast cancer is extremely important to decide the therapeutic management of not only hormonal therapy but also new molecular target therapy.
Collapse
Affiliation(s)
- Shin-ichi Hayashi
- Department of Molecular and Functional Dynamics, and Center for Regulatory Epigenomics and Diseases, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan,
| | | |
Collapse
|
35
|
Dhimolea E, Tiniakos DG, Chantzi ΝΙ, Goutas N, Vassilaros SD, Mitsiou DJ, Alexis ΜN. Estrogen receptors β1 and β2 are associated with distinct responses of estrogen receptor α-positive breast carcinoma to adjuvant endocrine therapy. Cancer Lett 2014; 358:37-42. [PMID: 25524554 DOI: 10.1016/j.canlet.2014.12.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 02/07/2023]
Abstract
Our purpose was to assess whether and how ERβ1 and/or ERβ2 expression status could predict response of early stage ERα-positive breast carcinoma to adjuvant endocrine therapy (AET). ERβ1 and ERβ2 expression were determined using immunohistochemistry. ERβ1- and ERβ2-positivity were derived from receiver operating characteristic analysis and the median percentage of immunostained tumor cells, respectively. Patients with recurrent disease were grouped according to whether they relapsed within 4 years or after 4 years from surgery. The predictive significance of ERβ1 and ERβ2 was determined using Kaplan-Meier survival analysis and Cox proportional hazards regression analysis. ERβ1-positivity in the first-4-year relapse patient group was lower and ERβ2-positivity in the post-4-year relapse group was higher compared with no-relapse group. ERβ1-positivity was associated with lower tumor size and longer first-4-year disease-free survival, while ERβ2-positivity was associated with shorter post-4-year disease-free survival. Cox multivariate analysis including ERβ1, ERβ2 and established clinico-pathological variables showed that ERβ1-positivity was an independent predictor of lower first-4-year risk of relapse. Thus, low ERβ1 expression and high ERβ2 expression are markers for identification of AET-treated ERα-positive breast carcinoma patients at risk of early and late relapse, respectively.
Collapse
Affiliation(s)
- Eugen Dhimolea
- Institute of Biology, Medicinal Chemistry & Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Av, 11635 Athens, Greece
| | - Dina G Tiniakos
- Laboratory of Histology & Embryology, Medical School, University of Athens, 75 M. Asias str., 11527 Athens, Greece
| | - Νiki Ι Chantzi
- Laboratory of Histology & Embryology, Medical School, University of Athens, 75 M. Asias str., 11527 Athens, Greece
| | - Nikolaos Goutas
- Department of Pathology, Eugenideio Infirmary, 20 Papadiamantopoulou str., 11528 Athens, Greece
| | | | - Dimitra J Mitsiou
- Institute of Biology, Medicinal Chemistry & Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Av, 11635 Athens, Greece
| | - Μichael N Alexis
- Institute of Biology, Medicinal Chemistry & Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Av, 11635 Athens, Greece.
| |
Collapse
|
36
|
Petrović N, Mandušić V, Dimitrijević B, Roganović J, Lukić S, Todorović L, Stanojević B. Higher miR-21 expression in invasive breast carcinomas is associated with positive estrogen and progesterone receptor status in patients from Serbia. Med Oncol 2014; 31:977. [PMID: 24781337 DOI: 10.1007/s12032-014-0977-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/21/2014] [Indexed: 02/07/2023]
Abstract
MicroRNAs play essential role in breast carcinoma progression and invasion. Our principal goals were to assess clinicopathological and prognostic correlations of microRNA-21 (miR-21) expression levels in a group of 39 Serbian breast cancer patients with invasive lobular (ILC), ductal (IDC), or mixed (ILC-IDC) breast carcinomas and in order to discover the role of miR-21 in potential novel form of stratification of the patients with different estrogen receptor (ER) and progesterone receptor (PR) status. MiR-21 expression levels were measured by stem-loop real-time RT-PCR using TaqMan technology. ER, PR, human epidermal growth factor 2 receptor (Her-2), and proliferative index (Ki-67) were evaluated by immunohistochemistry. MiR-21 levels do not vary among ILC, IDC, and ILC-IDC subgroups. MiR-21 expression levels varied significantly in the age, tumor size, Ki-67, and different grade (p = 0.030, p = 0.036, p = 0.027 and p = 0.032, respectively) subgroups. ER+ and PR+ showed higher miR-21 levels than their negative receptor status paired groups ER- and PR- with p = 0.012 and p = 0.018, respectively. MiR-21 positively correlated with ER and PR status (p = 0.018, ρ = 0.379 and p = 0.034, ρ = 0.345, respectively). Our findings suggest that miR-21 emulates transitional form of expression and that the levels of expression might be useful for stratification of the patients with different receptor status with the purpose to seek for new therapy approaches especially for the patients with the lack of response to conventional endocrine therapy.
Collapse
Affiliation(s)
- Nina Petrović
- Department for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11000, Belgrade, Serbia,
| | | | | | | | | | | | | |
Collapse
|
37
|
Moerkens M, Zhang Y, Wester L, van de Water B, Meerman JHN. Epidermal growth factor receptor signalling in human breast cancer cells operates parallel to estrogen receptor α signalling and results in tamoxifen insensitive proliferation. BMC Cancer 2014; 14:283. [PMID: 24758408 PMCID: PMC4021213 DOI: 10.1186/1471-2407-14-283] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/08/2014] [Indexed: 02/07/2023] Open
Abstract
Background Tamoxifen resistance is a major problem in the treatment of estrogen receptor (ER) α -positive breast cancer patients. Although the mechanisms behind tamoxifen resistance are still not completely understood, clinical data suggests that increased expression of receptor tyrosine kinases is involved. Here, we studied the estrogen and anti-estrogen sensitivity of human breast cancer MCF7 cells that have a moderate, retroviral-mediated, ectopic expression of epidermal growth factor receptor (MCF7-EGFR). Methods Proliferation of MCF7-EGFR and parental cells was induced by 17β-estradiol (E2), epidermal growth factor (EGF) or a combination of these. Inhibition of proliferation under these conditions was investigated with 4-hydroxy-tamoxifen (TAM) or fulvestrant at 10-12 to 10-6 M. Cells were lysed at different time points to determine the phosphorylation status of EGFR, MAPK1/3, AKT and the expression of ERα. Knockdown of target genes was established using smartpool siRNAs. Transcriptomics analysis was done 6 hr after stimulation with growth factors using Affymetrix HG-U133 PM array plates. Results While proliferation of parental MCF7 cells could only be induced by E2, proliferation of MCF7-EGFR cells could be induced by either E2 or EGF. Treatment with TAM or fulvestrant did significantly inhibit proliferation of MCF7-EGFR cells stimulated with E2 alone. EGF treatment of E2/TAM treated cells led to a marked cell proliferation thereby overruling the anti-estrogen-mediated inhibition of cell proliferation. Under these conditions, TAM however did still inhibit ERα- mediated transcription. While siRNA-mediated knock-down of EGFR inhibited the EGF- driven proliferation under TAM/E2/EGF condition, knock down of ERα did not. The TAM resistant cell proliferation mediated by the conditional EGFR-signaling may be dependent on the PI3K/Akt pathway but not the MEK/MAPK pathway, since a MEK inhibitor (U0126), did not block the proliferation. Transcriptomic analysis under the various E2/TAM/EGF conditions revealed that E2 and EGF dependent transcription have little overlap and rather operate in a parallel fashion. Conclusions Our data indicate that enhanced EGFR-driven signalling is sufficient to overrule the TAM- mediated inhibition of E2-driven cell proliferation. This may have profound implications for the anti-estrogen treatment of ER-positive breast cancers that have increased levels of EGFR.
Collapse
Affiliation(s)
| | | | | | | | - John H N Meerman
- Leiden Academic Centre for Drug Research (LACDR), Department of Toxicology, Leiden University, Einsteinweg 55, 2333 CC Leiden The Netherlands.
| |
Collapse
|
38
|
Fujiki N, Konno H, Kaneko Y, Gohno T, Hanamura T, Imami K, Ishihama Y, Nakanishi K, Niwa T, Seino Y, Yamaguchi Y, Hayashi SI. Estrogen response element-GFP (ERE-GFP) introduced MCF-7 cells demonstrated the coexistence of multiple estrogen-deprivation resistant mechanisms. J Steroid Biochem Mol Biol 2014; 139:61-72. [PMID: 24128438 DOI: 10.1016/j.jsbmb.2013.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/17/2013] [Accepted: 08/20/2013] [Indexed: 10/26/2022]
Abstract
The acquisition of estrogen-deprivation resistance and estrogen receptor (ER) signal-independence in ER-positive breast cancer is one of the crucial steps in advancing the aggressiveness of breast cancer; however, this has not yet been elucidated in detail. To address this issue, we established several estrogen-deprivation-resistant (EDR) breast cancer cell lines from our unique MCF-7 cells, which had been stably transfected with an ERE-GFP reporter plasmid. Three cell lines with high ER activity and another 3 cell lines with no ER activity were established from cell cloning by monitoring GFP expression in living cells. The former three ERE-GFP-positive EDR cell lines showed the overexpression of ER and high expression of several ER-target genes. Further analysis of intracellular signaling factors revealed a marked change in the phosphorylation status of ERα on Ser167 and Akt on Thr308 by similar mechanisms reported previously; however, we could not find any changes in MAP-kinase factors. Comprehensive phospho-proteomic analysis also indicated the possible contribution of the Akt pathway to the phosphorylation of ERα. On the other hand, constitutive activation of c-Jun N-terminal kinase (JNK) was observed in ERE-GFP-negative EDR cells, and the growth of these cells was inhibited by a JNK inhibitor. An IGF1R-specific inhibitor diminished the phosphorylation of JNK, which suggested that a novel signaling pathway, IGF1R-JNK, may be important for the proliferation of ER-independent MCF-7 cells. These results indicate that ER-positive breast cancer cells can acquire resistance by more than two mechanisms at a time, which suggests that multiple mechanisms may occur simultaneously. This finding also implies that breast cancers with different resistance mechanisms can concomitantly occur and mingle in an individual patient, and may be a cause of the recurrence of cancer.
Collapse
Affiliation(s)
- Natsu Fujiki
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Moy I, Lin Z, Rademaker AW, Reierstad S, Khan SA, Bulun SE. Expression of estrogen-related gene markers in breast cancer tissue predicts aromatase inhibitor responsiveness. PLoS One 2013; 8:e77543. [PMID: 24223121 PMCID: PMC3819361 DOI: 10.1371/journal.pone.0077543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 09/03/2013] [Indexed: 01/27/2023] Open
Abstract
Aromatase inhibitors (AIs) are the most effective class of drugs in the endocrine treatment of breast cancer, with an approximate 50% treatment response rate. Our objective was to determine whether intratumoral expression levels of estrogen-related genes are predictive of AI responsiveness in postmenopausal women with breast cancer. Primary breast carcinomas were obtained from 112 women who received AI therapy after failing adjuvant tamoxifen therapy and developing recurrent breast cancer. Tumor ERα and PR protein expression were analyzed by immunohistochemistry (IHC). Messenger RNA (mRNA) levels of 5 estrogen-related genes–AKR1C3, aromatase, ERα, and 2 estradiol/ERα target genes, BRCA1 and PR–were measured by real-time PCR. Tumor protein and mRNA levels were compared with breast cancer progression rates to determine predictive accuracy. Responsiveness to AI therapy–defined as the combined complete response, partial response, and stable disease rates for at least 6 months–was 51%; rates were 56% in ERα-IHC-positive and 14% in ERα-IHC-negative tumors. Levels of ERα, PR, or BRCA1 mRNA were independently predictive for responsiveness to AI. In cross-validated analyses, a combined measurement of tumor ERα and PR mRNA levels yielded a more superior specificity (36%) and identical sensitivity (96%) to the current clinical practice (ERα/PR-IHC). In patients with ERα/PR-IHC-negative tumors, analysis of mRNA expression revealed either non-significant trends or statistically significant positive predictive values for AI responsiveness. In conclusion, expression levels of estrogen-related mRNAs are predictive for AI responsiveness in postmenopausal women with breast cancer, and mRNA expression analysis may improve patient selection.
Collapse
Affiliation(s)
- Irene Moy
- Northwestern University, Department of Obstetrics and Gynecology, Chicago, Illinois, United States of America
| | - Zhihong Lin
- Northwestern University, Department of Obstetrics and Gynecology, Chicago, Illinois, United States of America
| | - Alfred W. Rademaker
- Northwestern University, Department of Preventive Medicine, Chicago, Illinois, United States of America
| | - Scott Reierstad
- Northwestern University, Department of Obstetrics and Gynecology, Chicago, Illinois, United States of America
| | - Seema A. Khan
- Northwestern University, Department of Surgery, Chicago, Illinois, United States of America
| | - Serdar E. Bulun
- Northwestern University, Department of Obstetrics and Gynecology, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
40
|
Activation of Akt, mTOR, and the estrogen receptor as a signature to predict tamoxifen treatment benefit. Breast Cancer Res Treat 2012; 137:397-406. [PMID: 23242584 PMCID: PMC3539073 DOI: 10.1007/s10549-012-2376-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 12/04/2012] [Indexed: 01/13/2023]
Abstract
The frequent alterations of the PI3K/Akt/mTOR-growth signaling pathway are proposed mechanisms for resistance to endocrine therapy in breast cancer, partly through regulation of estrogen receptor α (ER) activity. Reliable biomarkers for treatment prediction are required for improved individualized treatment. We performed a retrospective immunohistochemical analysis of primary tumors from 912 postmenopausal patients with node-negative breast cancer, randomized to either tamoxifen or no adjuvant treatment. Phosphorylated (p) Akt-serine (s) 473, p-mTOR-s2448, and ER phosphorylations-s167 and -s305 were evaluated as potential biomarkers of prognosis and tamoxifen treatment efficacy. High expression of p-mTOR indicated a reduced response to tamoxifen, most pronounced in the ER+/progesterone receptor (PgR) + subgroup (tamoxifen vs. no tamoxifen: hazard ratio (HR), 0.86; 95 % confidence interval (CI), 0.31–2.38; P = 0.78), whereas low p-mTOR expression predicted tamoxifen benefit (HR, 0.29; 95 % CI, 0.18–0.49; P = 0.000002). In addition, nuclear p-Akt-s473 as well as p-ER at -s167 and/or -s305 showed interaction with tamoxifen efficacy with borderline statistical significance. A combination score of positive pathway markers including p-Akt, p-mTOR, and p-ER showed significant association with tamoxifen benefit (test for interaction; P = 0.029). Cross-talk between growth signaling pathways and ER-signaling has been proposed to affect tamoxifen response in hormone receptor-positive breast cancer. The results support this hypothesis, as an overactive pathway was significantly associated with reduced response to tamoxifen. A clinical pre-treatment test for cross-talk markers would be a step toward individualized adjuvant endocrine treatment with or without the addition of PI3K/Akt/mTOR pathway inhibitors.
Collapse
|
41
|
Moon HG, Yi JK, Kim HS, Lee HY, Lee KM, Yi M, Ahn S, Shin HC, Ju JH, Shin I, Han W, Noh DY. Phosphorylation of p90RSK is associated with increased response to neoadjuvant chemotherapy in ER-positive breast cancer. BMC Cancer 2012; 12:585. [PMID: 23216670 PMCID: PMC3523086 DOI: 10.1186/1471-2407-12-585] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 11/30/2012] [Indexed: 12/30/2022] Open
Abstract
Background The clinical implication of Ras/Raf/ERK pathway activity in breast cancer tissue and its association with response to chemotherapy is controversial. We aimed to explore the value of p90RSK phosphorylation, a downstram molecule of the pathway, in predicting chemotherapy response in breast cancer. Methods The expression of phosphorylated p90RSK (phospho-p90RSK) and chemotherapy response was measured in 11 breast cancer cell lines and 21 breast cancer tissues. The predictive value of phospho-p90RSK was validated in core needle biopsy specimens of 112 locally advanced breast cancer patients who received anthracycline and taxane-based neoadjuvant chemotherapy. Results In 11 breast cancer cell lines, the relative expression of phospho-p90RSK was inversely correlated with cell survival after doxorubicin treatment (p = 0.021). Similar association was observed in fresh tissues from 21 breast cancer patients in terms of clinical response. In paraffin-embedded, formalin-fixed tissues from core needle biopsy tissues from 112 patients, positive phospho-p90RSK expression was associated with greater tumor shrinkage and smaller post-chemotherapy tumor size. The association between phospho-p90RSK expression and chemotherapy response was more evident in estrogen receptor(ER)-positive tumors. The expression of phosphor-p90RSK did not show a significant relationship with the incidence of pCR. P90RSK silencing using siRNA did not affect the cancer cell’s response to doxorubicin, and the expression of phospho-p90RSK was highly correlated with other Ras/Raf/ERK pathway activation. Conclusion Our results suggest that phospho-p90RSK expression, which reflects the tumor’s Ras/Raf/ERK/p90RSK pathway activation can be a potential predictive marker for chemotherapy response in ER-positive breast cancer which needs further independent validation.
Collapse
Affiliation(s)
- Hyeong-Gon Moon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Metodiev M, Alldridge L. Phosphoproteomics: A possible route to novel biomarkers of breast cancer. Proteomics Clin Appl 2012; 2:181-94. [PMID: 21136824 DOI: 10.1002/prca.200780011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Proteomics is rapidly transforming the way that cancer and other pathologies are investigated. The ability to identify hundreds of proteins and to compare their abundance in different clinical samples presents a unique opportunity for direct identification of novel disease markers. Furthermore, recent advances allow us to analyse and compare PTMs. This gives an additional dimension for defining a new class of protein biomarker based not only on abundance and expression but also on the occurrence of covalent modifications specific to a disease state or therapy response. Such modifications are often a consequence of the activation/inactivation of a particular disease related pathway. In this review we evaluate the available information on breast cancer related protein-phosphorylation events, illustrating the rationale for investigating this PTM as a target for breast cancer research with eventual clinical relevance. We present a critical survey of the published experimental strategies to study protein phosphorylation on a system wide scale and highlight recent specific advances in breast cancer phosphoproteomics. Finally we discuss the feasibility of establishing novel biomarkers for breast cancer based on the detection of patterns of specific protein phosphorylation events.
Collapse
Affiliation(s)
- Metodi Metodiev
- Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex, UK
| | | |
Collapse
|
43
|
Bobustuc GC, Smith JS, Maddipatla S, Jeudy S, Limaye A, Isley B, Caparas MLM, Constantino SM, Shah N, Baker CH, Srivenugopal KS, Baidas S, Konduri SD. MGMT inhibition restores ERα functional sensitivity to antiestrogen therapy. Mol Med 2012; 18:913-29. [PMID: 22549111 DOI: 10.2119/molmed.2012.00010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 04/26/2012] [Indexed: 11/06/2022] Open
Abstract
Antiestrogen therapy resistance remains a huge stumbling block in the treatment of breast cancer. We have found significant elevation of O(6) methylguanine DNA methyl transferase (MGMT) expression in a small sample of consecutive patients who have failed tamoxifen treatment. Here, we show that tamoxifen resistance is accompanied by upregulation of MGMT. Further we show that administration of the MGMT inhibitor, O(6)-benzylguanine (BG), at nontoxic doses, leads to restoration of a favorable estrogen receptor alpha (ERα) phosphorylation phenotype (high p-ERα Ser167/low p-ERα Ser118), which has been reported to correlate with sensitivity to endocrine therapy and improved survival. We also show BG to be a dual inhibitor of MGMT and ERα. In tamoxifen-resistant breast cancer cells, BG alone or in combination with antiestrogen (tamoxifen [TAM]/ICI 182,780 [fulvestrant, Faslodex]) therapy enhances p53 upregulated modulator of apoptosis (PUMA) expression, cytochrome C release and poly (ADP-ribose) polymerase (PARP) cleavage, all indicative of apoptosis. In addition, BG increases the expression of p21(cip1/waf1). We also show that BG, alone or in combination therapy, curtails the growth of tamoxifen-resistant breast cancer in vitro and in vivo. In tamoxifen-resistant MCF7 breast cancer xenografts, BG alone or in combination treatment causes significant delay in tumor growth. Immunohistochemistry confirms that BG increases p21(cip1/waf1) and p-ERα Ser167 expression and inhibits MGMT, ERα, p-ERα Ser118 and ki-67 expression. Collectively, our results suggest that MGMT inhibition leads to growth inhibition of tamoxifen-resistant breast cancer in vitro and in vivo and resensitizes tamoxifen-resistant breast cancer cells to antiestrogen therapy. These findings suggest that MGMT inhibition may provide a novel therapeutic strategy for overcoming antiestrogen resistance.
Collapse
Affiliation(s)
- George C Bobustuc
- MD Anderson Cancer Center Orlando, Orlando, Florida 32827, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Beelen K, Zwart W, Linn SC. Can predictive biomarkers in breast cancer guide adjuvant endocrine therapy? Nat Rev Clin Oncol 2012; 9:529-41. [PMID: 22825374 DOI: 10.1038/nrclinonc.2012.121] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Personalized medicine for oestrogen receptor-α (ERα)-positive breast cancer requires predictive biomarkers for broad endocrine resistance as well as biomarkers capable of predicting resistance to a specific agent. In addition, biomarkers could be used to select patients that might benefit from the addition of treatments that do not target ERα. However, biomarker identification studies seem to be far from consistent and identified biomarkers seldom face an introduction into clinical practice. Importantly, most of the studies that seek to identify biomarkers have been performed using material from consecutive series of patients treated with tamoxifen (the most commonly prescribed ERα antagonist). Consequently, the predictive value of any biomarker identified is confounded by its prognostic value. Another important issue is the lack of differentiation between premenopausal and postmenopausal patients with breast cancer. The hormonal environment of a tumour in patients who are premenopausal is intrinsically distinct from those arising in postmenopausal women. Biomarkers of different biological mechanisms might enable the prediction of either broad endocrine resistance or resistance to a specific agent in each of these patient subtypes. Ultimately, improvements to study design are needed to establish the clinical validity of the most promising biomarkers to predict benefit from endocrine therapy.
Collapse
Affiliation(s)
- Karin Beelen
- Department of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | |
Collapse
|
45
|
Leung YK, Lee MT, Lam HM, Tarapore P, Ho SM. Estrogen receptor-beta and breast cancer: translating biology into clinical practice. Steroids 2012; 77:727-37. [PMID: 22465878 PMCID: PMC3356459 DOI: 10.1016/j.steroids.2012.03.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 03/15/2012] [Accepted: 03/16/2012] [Indexed: 02/07/2023]
Abstract
Estrogen receptor (ER) β was discovered over a decade ago. The design of most studies on this receptor was based on knowledge of its predecessor, ERα. Although breast cancer (BCa) has been a main focus of ERβ research, its precise roles in breast carcinogenesis remain elusive. Data from in vitro models have not always matched those from observational or clinical studies. Several inherent factors may contribute to these discrepancies: (a) several ERβ spliced variants are expressed at the protein level, and isoform-specific antibodies are unavailable for some variants; (b) post-translational modifications of the receptor regulate receptor functions; (c) the role of the receptor differs significantly depending on the type of ligands, cis-elements, and co-regulators that interact with the receptor; and (d) the diversity of distribution of the receptor among intracellular organelles of BCa cells. This review addresses the gaps in knowledge in ERβ research as it pertains to BCa regarding the following questions: (1) is ERβ a tumor suppressor in BCa?; (2) do ERβ isoforms play differential roles in breast carcinogenesis?; (3) do nuclear signaling and extranuclear ERβ signaling differ in BCa?; (4) what are the consequences of post-translational modifications of ERβ in BCa?; (5) how do co-regulators and interacting proteins increase functional diversity of ERβ?; and (6) how do the types of ligand and regulatory cis-elements affect the action of ERβ in BCa?. Insights gained from these key questions in ERβ research should help in prevention, diagnosis/prognosis, and treatment of BCa.
Collapse
Affiliation(s)
- Yuet-Kin Leung
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA.
| | | | | | | | | |
Collapse
|
46
|
Anbalagan M, Huderson B, Murphy L, Rowan BG. Post-translational modifications of nuclear receptors and human disease. NUCLEAR RECEPTOR SIGNALING 2012; 10:e001. [PMID: 22438791 PMCID: PMC3309075 DOI: 10.1621/nrs.10001] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 08/19/2011] [Indexed: 12/12/2022]
Abstract
Nuclear receptors (NR) impact a myriad of physiological processes including homeostasis, reproduction, development, and metabolism. NRs are regulated by post-translational modifications (PTM) that markedly impact receptor function. Recent studies have identified NR PTMs that are involved in the onset and progression of human diseases, including cancer. The majority of evidence linking NR PTMs with disease has been demonstrated for phosphorylation, acetylation and sumoylation of androgen receptor (AR), estrogen receptor α (ERα), glucocorticoid receptor (GR) and peroxisome proliferator activated receptor γ (PPARγ). Phosphorylation of AR has been associated with hormone refractory prostate cancer and decreased disease-specific survival. AR acetylation and sumoylation increased growth of prostate cancer tumor models. AR phosphorylation reduced the toxicity of the expanded polyglutamine AR in Kennedy's Disease as a consequence of reduced ligand binding. A comprehensive evaluation of ERα phosphorylation in breast cancer revealed several sites associated with better clinical outcome to tamoxifen therapy, whereas other phosphorylation sites were associated with poorer clinical outcome. ERα acetylation and sumoylation may also have predictive value for breast cancer. GR phosphorylation and acetylation impact GR responsiveness to glucocorticoids that are used as anti-inflammatory drugs. PPARγ phosphorylation can regulate the balance between growth and differentiation in adipose tissue that is linked to obesity and insulin resistance. Sumoylation of PPARγ is linked to repression of inflammatory genes important in patients with inflammatory diseases. NR PTMs provide an additional measure of NR function that can be used as both biomarkers of disease progression, and predictive markers for patient response to NR-directed treatments.
Collapse
Affiliation(s)
- Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | |
Collapse
|
47
|
Nagalingam A, Tighiouart M, Ryden L, Joseph L, Landberg G, Saxena NK, Sharma D. Med1 plays a critical role in the development of tamoxifen resistance. Carcinogenesis 2012; 33:918-30. [PMID: 22345290 DOI: 10.1093/carcin/bgs105] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Understanding the molecular pathways that contribute to the development of tamoxifen resistance is a critical research priority as acquired tamoxifen resistance is the principal cause of poor prognosis and death of patients with originally good prognosis hormone-responsive breast tumors. In this report, we provide evidence that Med1, an important subunit of mediator coactivator complex, is spontaneously upregulated during acquired tamoxifen-resistance development potentiating agonist activities of tamoxifen. Phosphorylated Med1 and estrogen receptor (ER) are abundant in tamoxifen-resistant breast cancer cells due to persistent activation of extracellular signal-regulated kinases. Mechanistically, phosphorylated Med1 exhibits nuclear accumulation, increased interaction with ER and higher tamoxifen-induced recruitment to ER-responsive promoters, which is abrogated by inhibition of Med1 phosphorylation. Stable knockdown of Med1 in tamoxifen-resistant cells not only reverses tamoxifen resistance in vitro but also in vivo. Finally, higher expression levels of Med1 in the tumor significantly correlated with tamoxifen resistance in ER-positive breast cancer patients on adjuvant tamoxifen monotherapy. In silico analysis of breast cancer, utilizing published profiling studies showed that Med1 is overexpressed in aggressive subsets. These findings provide what we believe is the first evidence for a critical role for Med1 in tamoxifen resistance and identify this coactivator protein as an essential effector of the tamoxifen-induced breast cancer growth.
Collapse
Affiliation(s)
- Arumugam Nagalingam
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Room 145, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Zhang F, Feng F, Yang P, Li Z, You J, Xie W, Gao X, Yang J. Four-and-a-half-LIM protein 1 down-regulates estrogen receptor α activity through repression of AKT phosphorylation in human breast cancer cell. Int J Biochem Cell Biol 2011; 44:320-6. [PMID: 22094188 DOI: 10.1016/j.biocel.2011.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 10/30/2011] [Accepted: 11/02/2011] [Indexed: 11/18/2022]
Abstract
The Four-and-a-half LIM protein 1 (FHL-1) is a member of LIM-only protein family. It plays important roles in proliferation and apoptosis regulation of certain hepatocellular carcinoma and human breast cancer. Estrogen receptor α (ERα) is involved in the development and progression of human breast cancer. IGF/PI3K/AKT signaling pathway also plays certain roles in the program and regulation of human breast cancer and ovary cancer. However, the biological function of FHL-1 in regulation of human breast cancer and in the cross-talk of estrogen and IGF signaling pathway remains largely unknown. In this paper, we show that FHL-1 protein interacts with ERα and AKT. FHL-1 represses the translation and transcription of estrogen receptor-responsive genes through down-regulating AKT activation. In addition, FHL-1 is not only an ERα-interacting co-regulation protein, but also decreases the phosphorylation of AKT and ERα. Depression of endogenous FHL-1 by FHL-1 targeted small interfering RNA enhances the expression of these proteins and phosphorylation of AKT and ERα. These data suggest that FHL-1 may regulate ER signaling function through regulation of AKT activation besides the physical and functional interaction with ERα. By establishing a linkage role of the FHL-1 between the estrogen ERα signaling pathway and IGF/PI3K/AKT signaling pathway, this study identifies that FHL-1 proteins may be a useful molecular target for human breast cancer therapy.
Collapse
Affiliation(s)
- Fan Zhang
- Tumor Center, Chinese PLA General Hospital, Beijing 100853, PR China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM, Corbo L. Cracking the estrogen receptor's posttranslational code in breast tumors. Endocr Rev 2011; 32:597-622. [PMID: 21680538 DOI: 10.1210/er.2010-0016] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estrogen signaling pathways, because of their central role in regulating the growth and survival of breast tumor cells, have been identified as suitable and efficient targets for cancer therapies. Agents blocking estrogen activity are already widely used clinically, and many new molecules have entered clinical trials, but intrinsic or acquired resistance to treatment limits their efficacy. The basic molecular studies underlying estrogen signaling have defined the critical role of estrogen receptors (ER) in many aspects of breast tumorigenesis. However, important knowledge gaps remain about the role of posttranslational modifications (PTM) of ER in initiation and progression of breast carcinogenesis. Whereas major attention has been focused on the phosphorylation of ER, many other PTM (such as acetylation, ubiquitination, sumoylation, methylation, and palmitoylation) have been identified as events modifying ER expression and stability, subcellular localization, and sensitivity to hormonal response. This article will provide an overview of the current and emerging knowledge on ER PTM, with a particular focus on their deregulation in breast cancer. We also discuss their clinical relevance and the functional relationship between PTM. A thorough understanding of the complete picture of these modifications in ER carcinogenesis might not only open new avenues for identifying new markers for prognosis or prediction of response to endocrine therapy but also could promote the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Muriel Le Romancer
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment Cheney D, 28 rue Laennec, 69373 Lyon Cedex 08, France.
| | | | | | | | | | | |
Collapse
|
50
|
Bai Y, Tolles J, Cheng H, Siddiqui S, Gopinath A, Pectasides E, Camp RL, Rimm DL, Molinaro AM. Quantitative assessment shows loss of antigenic epitopes as a function of pre-analytic variables. J Transl Med 2011; 91:1253-61. [PMID: 21519325 PMCID: PMC3145004 DOI: 10.1038/labinvest.2011.75] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pre-analytic variables, specifically cold ischemic time, have been implicated as key variables in the measurement of proteins by immunohistochemistry. To determine the significance and magnitude of antigenic loss due to pre-analytic variables, we have compared protein antigenicity in core needle biopsies, with essentially no cold ischemic time, with that in routinely processed tumor resection specimens. Two cohorts of matched core needle biopsies and tumor resections were collected with 20 matched pairs and 14 matched pairs, respectively. Both series were analyzed by quantitative immunofluorescence using the AQUA® method. Epitopes phospho-ERK, total ERK, phospho-AKT, total AKT, phospho-S6K1, total S6K1, estrogen receptor (ER), Ki67, cytokeratin and GAPDH were assessed. Detection levels for all phospho-epitopes were significantly decreased in tumor resections compared with biopsies while no significant change was seen in the corresponding total proteins. Of the other four proteins examined, ER and cytokeratin showed significant loss of antigenicity. This data suggest that measurement of phospho-protein antigenicity in formalin-fixed tissue by immunological methods is dramatically affected by pre-analytic variables. This study suggests that core needle biopsies are more accurate for assessment of tissue biomarkers.
Collapse
Affiliation(s)
- Yalai Bai
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Juliana Tolles
- Division of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| | - Huan Cheng
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Summar Siddiqui
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Arun Gopinath
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Eirini Pectasides
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Robert L. Camp
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - David L. Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Annette M. Molinaro
- Division of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| |
Collapse
|