1
|
Ayoub NM, Al-Taani GM, Alkhalifa AE, Ibrahim DR, Shatnawi A. The Impact of the Coexpression of MET and ESR Genes on Prognosticators and Clinical Outcomes of Breast Cancer: An Analysis for the METABRIC Dataset. Breast J 2024; 2024:2582341. [PMID: 39742369 PMCID: PMC11098610 DOI: 10.1155/2024/2582341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/29/2024] [Accepted: 04/25/2024] [Indexed: 01/03/2025]
Abstract
Purpose Breast cancer is a heterogeneous disease. Exploring new prognostic and therapeutic targets in patients with breast cancer is essential. This study investigated the expression of MET, ESR1, and ESR2 genes and their association with clinicopathologic characteristics and clinical outcomes in patients with breast cancer. Methods The METABRIC dataset for breast cancer was obtained from the cBioPortal public domain. Gene expression data for MET, ESR1, and ESR2, as well as the putative copy number alterations (CNAs) for MET were retrieved. Results The MET mRNA expression levels correlated inversely with the expression levels of ESR1 and positively with the expression levels of ESR2 (r = -0.379, p < 0.001 and r = 0.066, and p=0.004, respectively). The ESR1 mRNA expression was significantly different among MET CNAs groups (p < 0.001). Patients with high MET/ESR1 coexpression had favorable clinicopathologic tumor characteristics and prognosticators compared to low MET/ESR1 coexpression in terms of greater age at diagnosis, reduced Nottingham Prognostic Index, lower tumor grade, hormone receptor positivity, HER2-negative status, and luminal subtype (p < 0.001). In contrast, patients with high MET/ESR2 coexpression had unfavorable tumor features and advanced prognosticators compared to patients with low MET/ESR2 coexpression (p < 0.001). No significant difference in overall survival was observed based on the MET/ESR coexpression status. However, when data were stratified based on the treatment type (chemotherapy and hormonal therapy), survival was significantly different based on the coexpression status of MET/ESR. Conclusions Findings from our study add to the growing evidence on the potential crosstalk between MET and estrogen receptors in breast cancer. The expression of the MET/ESR genes could be a novel prognosticator and calls for future studies to evaluate the impact of combinational treatment approaches with MET inhibitors and endocrine drugs in breast cancer.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. BOX: 3030, Irbid 22110, Jordan
| | - Ghaith M. Al-Taani
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. BOX: 3030, Irbid 22110, Jordan
| | - Dalia R. Ibrahim
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. BOX: 3030, Irbid 22110, Jordan
| | - Aymen Shatnawi
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, 70 President St., Charleston, SC 29425, USA
| |
Collapse
|
2
|
Hayat A, Carter EP, King HW, Ors A, Doe A, Teijeiro SA, Charrot S, Godinho S, Cutillas P, Mohammed H, Grose RP, Ficz G. Low HER2 expression in normal breast epithelium enables dedifferentiation and malignant transformation via chromatin opening. Dis Model Mech 2023; 16:dmm049894. [PMID: 36661191 PMCID: PMC9922733 DOI: 10.1242/dmm.049894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023] Open
Abstract
Overexpression of the HER2 protein in breast cancer patients is a predictor of poor prognosis and resistance to therapies. We used an inducible breast cancer transformation system that allows investigation of early molecular changes. HER2 overexpression to similar levels as those observed in a subtype of HER2-positive breast cancer patients induced transformation of MCF10A cells and resulted in gross morphological changes, increased anchorage-independent growth of cells, and altered the transcriptional programme of genes associated with oncogenic transformation. Global phosphoproteomic analysis during HER2 induction predominantly detected an increase in protein phosphorylation. Intriguingly, this correlated with chromatin opening, as measured by ATAC-seq on acini isolated from 3D cell culture. HER2 overexpression resulted in opening of many distal regulatory regions and promoted reprogramming-associated heterogeneity. We found that a subset of cells acquired a dedifferentiated breast stem-like phenotype, making them likely candidates for malignant transformation. Our data show that this population of cells, which counterintuitively enriches for relatively low HER2 protein abundance and increased chromatin accessibility, possesses transformational drive, resulting in increased anchorage-independent growth in vitro compared to cells not displaying a stem-like phenotype.
Collapse
Affiliation(s)
- Ateequllah Hayat
- Institute of Medical and Biomedical Education, St George’s, University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Edward P. Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Hamish W. King
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical Research, Royal Parade, Parkville, VIC 3052, Australia
| | - Aysegul Ors
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Aaron Doe
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Saul A. Teijeiro
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sarah Charrot
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Susana Godinho
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Pedro Cutillas
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Hisham Mohammed
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Richard P. Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Gabriella Ficz
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
3
|
Expression of c-MET in Estrogen Receptor Positive and HER2 Negative Resected Breast Cancer Correlated with a Poor Prognosis. J Clin Med 2022; 11:jcm11236987. [PMID: 36498560 PMCID: PMC9738605 DOI: 10.3390/jcm11236987] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction: The mesenchymal-epithelial transition factor (c-MET) receptor is overexpressed in about 14−54% of invasive breast cancers, but its prognostic value in clinical practice is still unclear. Methods: In order to investigate the relationship between c-MET expression levels and prognosis, we retrospectively reviewed the clinical features and outcomes of 105 women with estrogen receptor positive HER2 negative (ER+/HER2-) resected breast cancer. We used the Kaplan Meier method to estimate Disease Free Survival (DFS) and Breast Cancer Specific Survival (BCSS) in the subgroups of patients with high (≥50%) and low (<50%) c-MET expression. Univariate and multivariate Cox proportional regression models were performed to assess the prognostic impact of clinicopathological parameters for DFS an BCSS. Results: High c-MET values significantly correlated with tumor size, high Ki67 and low (<20%) progesterone receptor expression. At a median follow up of 60 months, patients with high c-MET tumor had significantly worse (p = 0.00026) and BCSS (p = 0.0013). Univariate analysis showed a significant association between large tumor size, elevated Ki67, c-MET values and increased risk of recurrence or death. The multivariate COX regression model showed that tumor size and high c-MET expression were independent predictors of DFS (p = 0.019 and p = 0.022). Moreover, large tumor size was associated with significantly higher risk of cancer related death at multivariate analysis (p = 0.017), while a trend towards a poorer survival was registered in the high c-MET levels cohort (p = 0.084). Conclusions: In our series, high c-MET expression correlated with poor survival outcomes. Further studies are warranted to validate the clinical relevance and applicability of c-MET as a prognostic factor in ER+/HER2- early BC.
Collapse
|
4
|
Todorova VK, Byrum SD, Gies AJ, Haynie C, Smith H, Reyna NS, Makhoul I. Circulating Exosomal microRNAs as Predictive Biomarkers of Neoadjuvant Chemotherapy Response in Breast Cancer. Curr Oncol 2022; 29:613-630. [PMID: 35200555 PMCID: PMC8870357 DOI: 10.3390/curroncol29020055] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Neoadjuvant chemotherapy (NACT) is an increasingly used approach for treatment of breast cancer. The pathological complete response (pCR) is considered a good predictor of disease-specific survival. This study investigated whether circulating exosomal microRNAs could predict pCR in breast cancer patients treated with NACT. Method: Plasma samples of 20 breast cancer patients treated with NACT were collected prior to and after the first cycle. RNA sequencing was used to determine microRNA profiling. The Cancer Genome Atlas (TCGA) was used to explore the expression patterns and survivability of the candidate miRNAs, and their potential targets based on the expression levels and copy number variation (CNV) data. Results: Three miRNAs before that NACT (miR-30b, miR-328 and miR-423) predicted pCR in all of the analyzed samples. Upregulation of miR-127 correlated with pCR in triple-negative breast cancer (TNBC). After the first NACT dose, pCR was predicted by exo-miR-141, while miR-34a, exo-miR182, and exo-miR-183 predicted non-pCR. A significant correlation between the candidate miRNAs and the overall survival, subtype, and metastasis in breast cancer, suggesting their potential role as predictive biomarkers of pCR. Conclusions: If the miRNAs identified in this study are validated in a large cohort of patients, they might serve as predictive non-invasive liquid biopsy biomarkers for monitoring pCR to NACT in breast cancer.
Collapse
Affiliation(s)
- Valentina K. Todorova
- Division of Medical Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Correspondence:
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (A.J.G.)
| | - Allen J. Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (A.J.G.)
| | - Cade Haynie
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (C.H.); (H.S.); (N.S.R.)
| | - Hunter Smith
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (C.H.); (H.S.); (N.S.R.)
| | - Nathan S. Reyna
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (C.H.); (H.S.); (N.S.R.)
| | - Issam Makhoul
- Division of Medical Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
5
|
Sjögren syndrome/scleroderma autoantigen 1 is a direct Tankyrase binding partner in cancer cells. Commun Biol 2020; 3:123. [PMID: 32170109 PMCID: PMC7070046 DOI: 10.1038/s42003-020-0851-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/21/2020] [Indexed: 12/30/2022] Open
Abstract
Sjögren syndrome/scleroderma autoantigen 1 (SSSCA1) was first described as an auto-antigen over-expressed in Sjögren’s syndrome and in scleroderma patients. SSSCA1 has been linked to mitosis and centromere association and as a potential marker candidate in diverse solid cancers. Here we characterize SSSCA1 for the first time, to our knowledge, at the molecular, structural and subcellular level. We have determined the crystal structure of a zinc finger fold, a zinc ribbon domain type 2 (ZNRD2), at 2.3 Å resolution. We show that the C-terminal domain serves a dual function as it both behaves as the interaction site to Tankyrase 1 (TNKS1) and as a nuclear export signal. We identify TNKS1 as a direct binding partner of SSSCA1, map the binding site to TNKS1 ankyrin repeat cluster 2 (ARC2) and thus define a new binding sequence. We experimentally verify and map a new nuclear export signal sequence in SSSCA1. Perdreau-Dahl et al. systematically characterise Sjögren syndrome/scleroderma autoantigen 1 at the molecular, structural and subcellular level. They show that the C-terminal domain serves a dual function as it both behaves as the interaction site to Tankyrase 1 and as a nuclear export signal.
Collapse
|
6
|
Hasan M, Marzouk MA, Adhikari S, Wright TD, Miller BP, Matossian MD, Elliott S, Wright M, Alzoubi M, Collins-Burow BM, Burow ME, Holzgrabe U, Zlotos DP, Stratford RE, Witt-Enderby PA. Pharmacological, Mechanistic, and Pharmacokinetic Assessment of Novel Melatonin-Tamoxifen Drug Conjugates as Breast Cancer Drugs. Mol Pharmacol 2019; 96:272-296. [PMID: 31221824 PMCID: PMC6666385 DOI: 10.1124/mol.119.116202] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/07/2019] [Indexed: 02/05/2023] Open
Abstract
Tamoxifen is used to prevent and treat estrogen receptor-positive (ER+) breast cancer (BC); however, its chronic use can increase uterine cancer risk and induce tamoxifen resistance. Novel melatonin-tamoxifen drug conjugates may be promising to treat BC and may help offset the adverse effects of tamoxifen usage alone due to the presence of melatonin. We synthesized and screened five drug conjugates (C2, C4, C5, C9, and C15 linked) for their effects on BC cell (MCF-7, tamoxifen-resistant MCF-7, mouse mammary carcinoma, MDA-MB-231, and BT-549) viability, migration, and binding affinity to melatonin receptor 1 (MT1R) and estrogen receptor 1 (ESR1). C4 and C5 demonstrated the most favorable pharmacological characteristics with respect to binding profiles (affinity for ESR1 and MT1R) and their potency/efficacy to inhibit BC cell viability and migration in four phenotypically diverse invasive ductal BC cell lines. C4 and C5 were further assessed for their actions against tamoxifen-resistant MCF-7 cells and a patient-derived xenograft triple-negative BC cell line (TU-BcX-4IC) and for their mechanisms of action using selective mitogen-activated protein kinase kinase MEK1/2, MEK5, and phosphoinositide 3-kinase (PI3K) inhibitors. C4 and C5 inhibited tamoxifen-resistant MCF-7 cells with equal potency (IC50 = 4-8 μM) and efficacy (∼90% inhibition of viability and migration) but demonstrated increased potency (IC50 = 80-211 μM) and efficacy (∼140% inhibition) to inhibit migration versus cell viability (IC50 = 181-304 mM; efficacy ∼80% inhibition) in TU-BcX-4IC cells. Unique pharmacokinetic profiles were observed, with C4 having greater bioavailability than C5. Further assessment of C4 and C5 demonstrates that they create novel pharmacophores within each BC cell that is context specific and involves MEK1/2/pERK1/2, MEK5/pERK5, PI3K, and nuclear factor κB. These melatonin-tamoxifen drug conjugates show promise as novel anticancer drugs and further preclinical and clinical evaluation is warranted.
Collapse
Affiliation(s)
- Mahmud Hasan
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Mohamed Akmal Marzouk
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Saugat Adhikari
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Thomas D Wright
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Benton P Miller
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Margarite D Matossian
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Steven Elliott
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Maryl Wright
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Madlin Alzoubi
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Bridgette M Collins-Burow
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Matthew E Burow
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Ulrike Holzgrabe
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Darius P Zlotos
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Robert E Stratford
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Paula A Witt-Enderby
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| |
Collapse
|
7
|
Liu J, Shelkar GP, Zhao F, Clausen RP, Dravid SM. Modulation of burst firing of neurons in nucleus reticularis of the thalamus by GluN2C-containing NMDA receptors. Mol Pharmacol 2019; 96:mol.119.116780. [PMID: 31160332 PMCID: PMC6620419 DOI: 10.1124/mol.119.116780] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/17/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
The GluN2C subunit of the NMDA receptor is enriched in the neurons in nucleus reticularis of the thalamus (nRT), but its role in regulating their function is not well understood. We found that deletion of GluN2C subunit did not affect spike frequency in response to depolarizing current injection or hyperpolarization-induced rebound burst firing of nRT neurons. D-cycloserine or CIQ (GluN2C/GluN2D positive allosteric modulator) did not affect the depolarization-induced spike frequency in nRT neurons. A newly identified highly potent and efficacious co-agonist of GluN1/GluN2C NMDA receptors, AICP, was found to reduce the spike frequency and burst firing of nRT neurons in wildtype but not GluN2C knockout. This effect was potentially due to facilitation of GluN2C-containing receptors because inhibition of NMDA receptors by AP5 did not affect spike frequency in nRT neurons. We evaluated the effect of intracerebroventricular injection of AICP. AICP did not affect basal locomotion or prepulse inhibition but facilitated MK-801-induced hyperlocomotion. This effect was observed in wildtype but not in GluN2C knockout mice demonstrating that AICP produces GluN2C-selective effects in vivo Using a chemogenetic approach we examined the role of nRT in this behavioral effect. Gq or Gi coupled DREADDs were selectively expressed in nRT neurons using cre-dependent viral vectors and PV-Cre mouse line. We found that similar to AICP effect, activation of Gq but not Gi coupled DREADD facilitated MK-801-induced hyperlocomotion. Together, these results identify a unique role of GluN2C-containing receptors in the regulation of nRT neurons and suggest GluN2C-selective in vivo targeting of NMDA receptors by AICP. SIGNIFICANCE STATEMENT: The nucleus reticularis of the thalamus composed of GABAergic neurons is termed as guardian of the gateway and is an important regulator of corticothalamic communication which may be impaired in autism, non-convulsive seizures and other conditions. We found that strong facilitation of tonic activity of GluN2C subtype of NMDA receptors using AICP, a newly identified glycine-site agonist of NMDA receptors, modulates the function of reticular thalamus neurons. AICP was also able to produce GluN2C-dependent behavioral effects in vivo. Together, these finding identify a novel mechanism and a pharmacological tool to modulate activity of reticular thalamic neurons in disease states.
Collapse
|
8
|
Selli C, Sims AH. Neoadjuvant Therapy for Breast Cancer as a Model for Translational Research. Breast Cancer (Auckl) 2019; 13:1178223419829072. [PMID: 30814840 PMCID: PMC6381436 DOI: 10.1177/1178223419829072] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 01/21/2023] Open
Abstract
Neoadjuvant therapy, where patients receive systemic therapy before surgical removal of the tumour, can downstage tumours allowing breast-conserving surgery, rather than mastectomy. In addition to its impact on surgery, the neoadjuvant setting offers a valuable opportunity to monitor individual tumour response. The effectiveness of standard and/or potential new therapies can be tested in the neoadjuvant pre-surgical setting. It can potentially help to identify markers differentiating patients that will potentially benefit from continuing with the same or a different adjuvant treatment enabling personalised treatment. Characterising the molecular response to treatment over time can more accurately identify the significant differences between baseline samples that would not be identified without post-treatment samples. In this review, we discuss the potential and challenges of using the neoadjuvant setting in translational breast cancer research, considering the implications for improving our understanding of response to treatment, predicting therapy benefit, modelling breast cancer dormancy, and the development of drug resistance.
Collapse
Affiliation(s)
- Cigdem Selli
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK
| |
Collapse
|
9
|
Selli C, Turnbull AK, Pearce DA, Li A, Fernando A, Wills J, Renshaw L, Thomas JS, Dixon JM, Sims AH. Molecular changes during extended neoadjuvant letrozole treatment of breast cancer: distinguishing acquired resistance from dormant tumours. Breast Cancer Res 2019; 21:2. [PMID: 30616553 PMCID: PMC6323855 DOI: 10.1186/s13058-018-1089-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/19/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The risk of recurrence for endocrine-treated breast cancer patients persists for many years or even decades following surgery and apparently successful adjuvant therapy. This period of dormancy and acquired resistance is inherently difficult to investigate; previous efforts have been limited to in-vitro or in-vivo approaches. In this study, sequential tumour samples from patients receiving extended neoadjuvant aromatase inhibitor therapy were characterised as a novel clinical model. METHODS Consecutive tumour samples from 62 patients undergoing extended (4-45 months) neoadjuvant aromatase inhibitor therapy with letrozole were subjected to transcriptomic and proteomic analysis, representing before (≤ 0), early (13-120 days), and long-term (> 120 days) neoadjuvant aromatase inhibitor therapy with letrozole. Patients with at least a 40% initial reduction in tumour size by 4 months of treatment were included. Of these, 42 patients with no subsequent progression were classified as "dormant", and the remaining 20 patients as "acquired resistant". RESULTS Changes in gene expression in dormant tumours begin early and become more pronounced at later time points. Therapy-induced changes in resistant tumours were common features of treatment, rather than being specific to the resistant phenotype. Comparative analysis of long-term treated dormant and resistant tumours highlighted changes in epigenetics pathways including DNA methylation and histone acetylation. The DNA methylation marks 5-methylcytosine and 5-hydroxymethylcytosine were significantly reduced in resistant tumours compared with dormant tissues after extended letrozole treatment. CONCLUSIONS This is the first patient-matched gene expression study investigating long-term aromatase inhibitor-induced dormancy and acquired resistance in breast cancer. Dormant tumours continue to change during treatment whereas acquired resistant tumours more closely resemble their diagnostic samples. Global loss of DNA methylation was observed in resistant tumours under extended treatment. Epigenetic alterations may lead to escape from dormancy and drive acquired resistance in a subset of patients, supporting a potential role for therapy targeted at these epigenetic alterations in the management of resistance to oestrogen deprivation therapy.
Collapse
Affiliation(s)
- Cigdem Selli
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.,Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040, Izmir, Turkey
| | - Arran K Turnbull
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.,Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Dominic A Pearce
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - Ang Li
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - Anu Fernando
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.,Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Jimi Wills
- Mass Spectrometry Unit, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - Lorna Renshaw
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Jeremy S Thomas
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.
| |
Collapse
|
10
|
Walter KR, Ford ME, Gregoski MJ, Kramer RM, Knight KD, Spruill L, Nogueira LM, Krisanits BA, Phan V, La Rue AC, Lilly MB, Ambs S, Chan K, Turner TF, Varner H, Singh S, Uribarri J, Garrett-Mayer E, Armeson KE, Hilton EJ, Clair MJ, Taylor MH, Abbott AM, Findlay VJ, Peterson LL, Magwood G, Turner DP. Advanced glycation end products are elevated in estrogen receptor-positive breast cancer patients, alter response to therapy, and can be targeted by lifestyle intervention. Breast Cancer Res Treat 2018; 173:559-571. [PMID: 30368741 PMCID: PMC6394600 DOI: 10.1007/s10549-018-4992-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/03/2018] [Indexed: 12/18/2022]
Abstract
Purpose Lifestyle factors associated with personal behavior can alter tumor-associated biological pathways and thereby increase cancer risk, growth, and disease recurrence. Advanced glycation end products (AGEs) are reactive metabolites produced endogenously as a by-product of normal metabolism. A Western lifestyle also promotes AGE accumulation in the body which is associated with disease phenotypes through modification of the genome, protein crosslinking/dysfunction, and aberrant cell signaling. Given the links between lifestyle, AGEs, and disease, we examined the association between dietary-AGEs and breast cancer. Methods We evaluated AGE levels in bio-specimens from estrogen receptor-positive (ER+) and estrogen receptor-negative (ER−) breast cancer patients, examined their role in therapy resistance, and assessed the ability of lifestyle intervention to reduce circulating AGE levels in ER+ breast cancer survivors. Results An association between ER status and AGE levels was observed in tumor and serum samples. AGE treatment of ER+ breast cancer cells altered ERα phosphorylation and promoted resistance to tamoxifen therapy. In a proof of concept study, physical activity and dietary intervention was shown to be viable options for reducing circulating AGE levels in breast cancer survivors. Conclusions There is a potential prognostic and therapeutic role for lifestyle derived AGEs in breast cancer. Given the potential benefits of lifestyle intervention on incidence and mortality, opportunities exist for the development of community health and nutritional programs aimed at reducing AGE exposure in order to improve breast cancer prevention and treatment outcomes. Electronic supplementary material The online version of this article (10.1007/s10549-018-4992-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katherine R Walter
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Marvella E Ford
- Department of Public Health Sciences, MUSC, Charleston, SC, USA. .,Hollings Cancer Center, MUSC, Charleston, SC, USA. .,James E. Clyburn Research Center Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Mathew J Gregoski
- Department of Exercise Science, College of Arts and Sciences, Campbell University, Buies Creek, NC, USA
| | | | | | - Laura Spruill
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Lourdes M Nogueira
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Bradley A Krisanits
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Van Phan
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Amanda C La Rue
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA.,Hollings Cancer Center, MUSC, Charleston, SC, USA.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA
| | - Michael B Lilly
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - King Chan
- Cancer Research Technology Program, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD, USA
| | | | - Heidi Varner
- Department of Public Health Sciences, MUSC, Charleston, SC, USA
| | - Shweta Singh
- Department of Public Health Sciences, MUSC, Charleston, SC, USA
| | - Jaime Uribarri
- Department of Medicine/Renal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences, MUSC, Charleston, SC, USA.,Hollings Cancer Center, MUSC, Charleston, SC, USA
| | - Kent E Armeson
- Department of Public Health Sciences, MUSC, Charleston, SC, USA.,Hollings Cancer Center, MUSC, Charleston, SC, USA
| | - Ebony J Hilton
- Department of Anesthesia and Perioperative Medicine, MUSC, Charleston, SC, USA
| | - Mark J Clair
- Department of Medicine, Division of Cardiology, MUSC, Charleston, SC, USA
| | - Marian H Taylor
- Department of Medicine, Division of Cardiology, MUSC, Charleston, SC, USA
| | | | - Victoria J Findlay
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA.,Department of Public Health Sciences, MUSC, Charleston, SC, USA
| | | | | | - David P Turner
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA. .,Department of Public Health Sciences, MUSC, Charleston, SC, USA. .,James E. Clyburn Research Center Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
11
|
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]
|
12
|
Alves MR, E Melo NC, Barros-Filho MC, do Amaral NS, Silva FIDB, Baiocchi Neto G, Soares FA, de Brot Andrade L, Rocha RM. Downregulation of AGR2, p21, and cyclin D and alterations in p53 function were associated with tumor progression and chemotherapy resistance in epithelial ovarian carcinoma. Cancer Med 2018; 7:3188-3199. [PMID: 29845750 PMCID: PMC6051166 DOI: 10.1002/cam4.1530] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/04/2018] [Accepted: 04/09/2018] [Indexed: 12/15/2022] Open
Abstract
Anterior gradient 2 protein belongs to a family of chaperone‐like proteins, namely protein disulfide isomerase. Generally, AGR2 is highly expressed in mucus‐secreting cells and endocrine organs, and in this study, we aimed to evaluate AGR2 and cell cycle molecules in epithelial ovarian cancer and its implications on prognosis. One hundred seventy‐five patient's samples that were diagnosed with primary epithelial ovarian carcinoma were selected. All the patients were treated with platinum‐taxane standard chemotherapy after surgery and CA125 serum levels were routinely determined. Four‐micrometer‐thick sections were processed by immunohistochemistry using an automated immunostainer, Ventana BenchMark AutoStainer with AGR2, cyclin D1, p21WAF1, and p53. Forty‐nine of 167 cases (29.3%) showed strong to moderate cytoplasmic marking of AGR2, and 118 (70.7%) had weak to negative expression. The absence of the AGR2 protein was observed in high‐grade serous carcinoma (P < .001) and significantly associated with disease‐free survival (DFS; P = .034). The expression of G1‐S phase‐regulatory proteins showed loss of p21 in high‐grade serous carcinoma (P < .001) and was related with poor DFS (P = .003). Strong and diffuse immunoexpression of p53 plus complete absence of p53 staining was interpreted as likely indicating a TP53 gene mutation. This result showed worse DFS alone (P = .012) and combined with low levels of AGR2 (P = .005). The expression profile of AGR2 and cell cycle proteins here presented was showed as good prognosis marker in epithelial ovarian cancer. This finding suggests AGR2 and as putative biomarker of disease progression in chemotherapy‐treated high‐grade serous carcinoma patients.
Collapse
Affiliation(s)
| | - Natalia Cruz E Melo
- Molecular Gynecology Laboratory, Gynecologic Department, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | - Rafael Malagoli Rocha
- Molecular Gynecology Laboratory, Gynecologic Department, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Bellanger A, Donini CF, Vendrell JA, Lavaud J, Machuca-Gayet I, Ruel M, Vollaire J, Grisard E, Győrffy B, Bièche I, Peyruchaud O, Coll JL, Treilleux I, Maguer-Satta V, Josserand V, Cohen PA. The critical role of the ZNF217 oncogene in promoting breast cancer metastasis to the bone. J Pathol 2017; 242:73-89. [PMID: 28207159 DOI: 10.1002/path.4882] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/10/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022]
Abstract
Bone metastasis affects >70% of patients with advanced breast cancer. However, the molecular mechanisms underlying this process remain unclear. On the basis of analysis of clinical datasets, and in vitro and in vivo experiments, we report that the ZNF217 oncogene is a crucial mediator and indicator of bone metastasis. Patients with high ZNF217 mRNA expression levels in primary breast tumours had a higher risk of developing bone metastases. MDA-MB-231 breast cancer cells stably transfected with ZNF217 (MDA-MB-231-ZNF217) showed the dysregulated expression of a set of genes with bone-homing and metastasis characteristics, which overlapped with two previously described 'osteolytic bone metastasis' gene signatures, while also highlighting the bone morphogenetic protein (BMP) pathway. The latter was activated in MDA-MB-231-ZNF217 cells, and its silencing by inhibitors (Noggin and LDN-193189) was sufficient to rescue ZNF217-dependent cell migration, invasion or chemotaxis towards the bone environment. Finally, by using non-invasive multimodal in vivo imaging, we found that ZNF217 increases the metastatic growth rate in the bone and accelerates the development of severe osteolytic lesions. Altogether, the findings of this study highlight ZNF217 as an indicator of the emergence of breast cancer bone metastasis; future therapies targeting ZNF217 and/or the BMP signalling pathway may be beneficial by preventing the development of bone metastases. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Aurélie Bellanger
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Caterina F Donini
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Unité Cancer et Environnement, Centre Léon Bérard, Lyon, France
| | - Julie A Vendrell
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jonathan Lavaud
- INSERM U1209, Institut Albert Bonniot, Grenoble, France.,Université Grenoble Alpes, Institut Albert Bonniot, Grenoble, France
| | - Irma Machuca-Gayet
- Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France.,INSERM, Unit 1033 (Faculté de Médecine Lyon Est), Lyon, France
| | - Maëva Ruel
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Julien Vollaire
- INSERM U1209, Institut Albert Bonniot, Grenoble, France.,Université Grenoble Alpes, Institut Albert Bonniot, Grenoble, France
| | - Evelyne Grisard
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary.,Second Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Ivan Bièche
- Unit of Pharmacogenetics, Department of Genetics, Institut Curie, Paris, France
| | - Olivier Peyruchaud
- Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France.,INSERM, Unit 1033 (Faculté de Médecine Lyon Est), Lyon, France
| | - Jean-Luc Coll
- INSERM U1209, Institut Albert Bonniot, Grenoble, France.,Université Grenoble Alpes, Institut Albert Bonniot, Grenoble, France
| | | | - Véronique Maguer-Satta
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Véronique Josserand
- INSERM U1209, Institut Albert Bonniot, Grenoble, France.,Université Grenoble Alpes, Institut Albert Bonniot, Grenoble, France
| | - Pascale A Cohen
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Univ. Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
14
|
De Marchi T, Foekens JA, Umar A, Martens JWM. Endocrine therapy resistance in estrogen receptor (ER)-positive breast cancer. Drug Discov Today 2016; 21:1181-8. [PMID: 27233379 DOI: 10.1016/j.drudis.2016.05.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/25/2016] [Accepted: 05/18/2016] [Indexed: 12/20/2022]
Abstract
Estrogen receptor (ER)-positive breast cancer represents the majority (∼70%) of all breast malignancies. In this subgroup of breast cancers, endocrine therapies are effective both in the adjuvant and recurrent settings, although resistance remains a major issue. Several high-throughput approaches have been used to elucidate mechanisms of resistance and to derive potential predictive markers or alternative therapies. In this review, we cover the state-of-the-art of endocrine-resistance biomarker discovery with regard to the latest technological developments, and discuss current opportunities and restrictions for their implementation into a clinical setting.
Collapse
Affiliation(s)
- Tommaso De Marchi
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John A Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Arzu Umar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Dieci MV, Griguolo G, Miglietta F, Guarneri V. The immune system and hormone-receptor positive breast cancer: Is it really a dead end? Cancer Treat Rev 2016; 46:9-19. [PMID: 27055087 DOI: 10.1016/j.ctrv.2016.03.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/21/2016] [Indexed: 01/08/2023]
Abstract
Even if breast cancer has not been traditionally considered an immunogenic tumor, recent data suggest that immunity, and its interaction with tumor cells and tumor microenvironment, might play an important role in this malignancy, in particular in triple negative and HER2+ subtypes. As no consistent data on the potential clinical relevance of tumor infiltrating lymphocytes have been produced in hormone receptor positive (HR+) HER2- breast cancer, the interest in studying immune aspects in this subtype has become less appealing. Nevertheless, some scattered evidence indicates that immunity and inflammation may be implicated in the biology of this subtype as well. In HR+ breast cancer, the interaction between tumor cells and the immune milieu might rely on different mechanisms than in other BC subtypes, involving the modulation of the tumor microenvironment by mutual interplays of endocrine factors, pro-inflammatory status and immune cells. These subtle mechanisms may require more refined methods of evaluation, such as the assessment of tumor infiltrating lymphocytes subpopulations or gene signatures. In this paper we aim to perform a comprehensive review of pre-clinical and clinical data on the interplay between the immune system and breast cancer in the HR+ subtype, to guide further research in the field.
Collapse
Affiliation(s)
- Maria Vittoria Dieci
- Dept. of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy.
| | - Gaia Griguolo
- Dept. of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Federica Miglietta
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Valentina Guarneri
- Dept. of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| |
Collapse
|
16
|
Busch S, Sims AH, Stål O, Fernö M, Landberg G. Loss of TGFβ Receptor Type 2 Expression Impairs Estrogen Response and Confers Tamoxifen Resistance. Cancer Res 2016; 75:1457-69. [PMID: 25833830 DOI: 10.1158/0008-5472.can-14-1583] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One third of the patients with estrogen receptor α (ERα)-positive breast cancer who are treated with the antiestrogen tamoxifen will either not respond to initial therapy or will develop drug resistance. Endocrine response involves crosstalk between ERα and TGFβ signaling, such that tamoxifen nonresponsiveness or resistance in breast cancer might involve aberrant TGFβ signaling. In this study, we analyzed TGFβ receptor type 2 (TGFBR2) expression and correlated it with ERα status and phosphorylation in a cohort of 564 patients who had been randomized to tamoxifen or no-adjuvant treatment for invasive breast carcinoma. We also evaluated an additional four independent genetic datasets in invasive breast cancer. In all the cohorts we analyzed, we documented an association of low TGFBR2 protein and mRNA expression with tamoxifen resistance. Functional investigations confirmed that cell cycle or apoptosis responses to estrogen or tamoxifen in ERα-positive breast cancer cells were impaired by TGFBR2 silencing, as was ERα phosphorylation, tamoxifen-induced transcriptional activation of TGFβ, and upregulation of the multidrug resistance protein ABCG2. Acquisition of low TGFBR2 expression as a contributing factor to endocrine resistance was validated prospectively in a tamoxifen-resistant cell line generated by long-term drug treatment. Collectively, our results established a central contribution of TGFβ signaling in endocrine resistance in breast cancer and offered evidence that TGFBR2 can serve as an independent biomarker to predict treatment outcomes in ERα-positive forms of this disease.
Collapse
Affiliation(s)
- Susann Busch
- Sahlgrenska Cancer Center, Gothenburg University, Gothenburg, Sweden
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh, Cancer Research UK Centre, United Kingdom
| | - Olle Stål
- Department of Clinical and Experimental Medicine, Institution of Surgery and Clinical Oncology, Linköpings Universitet, Linköping, Sweden
| | - Mårten Fernö
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Göran Landberg
- Sahlgrenska Cancer Center, Gothenburg University, Gothenburg, Sweden. Molecular Pathology, Breakthrough Breast Cancer Research Unit, University of Manchester, United Kingdom.
| |
Collapse
|
17
|
Turnbull AK, Arthur LM, Renshaw L, Larionov AA, Kay C, Dunbier AK, Thomas JS, Dowsett M, Sims AH, Dixon JM. Accurate Prediction and Validation of Response to Endocrine Therapy in Breast Cancer. J Clin Oncol 2015; 33:2270-8. [PMID: 26033813 DOI: 10.1200/jco.2014.57.8963] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Aromatase inhibitors (AIs) have an established role in the treatment of breast cancer. Response rates are only 50% to 70% in the neoadjuvant setting and lower in advanced disease. Accurate biomarkers are urgently needed to predict response in these settings and to determine which individuals will benefit from adjuvant AI therapy. PATIENTS AND METHODS Pretreatment and on-treatment (after 2 weeks and 3 months) biopsies were obtained from 89 postmenopausal women who had estrogen receptor-alpha positive breast cancer and were receiving neoadjuvant letrozole for transcript profiling. Dynamic clinical response was assessed with use of three-dimensional ultrasound measurements. RESULTS The molecular response to letrozole was characterized and a four-gene classifier of clinical response was established (accuracy of 96%) on the basis of the level of two genes before treatment (one gene [IL6ST] was associated with immune signaling, and the other [NGFRAP1] was associated with apoptosis) and the level of two proliferation genes (ASPM, MCM4) after 2 weeks of therapy. The four-gene signature was found to be 91% accurate in a blinded, completely independent validation data set of patients treated with anastrozole. Matched 2-week on-treatment biopsies were associated with improved predictive power as compared with pretreatment biopsies alone. This signature also significantly predicted recurrence-free survival (P = .029) and breast cancer -specific survival (P = .009). We demonstrate that the test can also be performed with use of quantitative polymerase chain reaction or immunohistochemistry. CONCLUSION A four-gene predictive model of clinical response to AIs by 2 weeks has been generated and validated. Deregulated immune and apoptotic responses before treatment and cell proliferation that is not reduced 2 weeks after initiation of treatment are functional characteristics of breast tumors that do not respond to AIs.
Collapse
Affiliation(s)
- Arran K Turnbull
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Laura M Arthur
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Lorna Renshaw
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Alexey A Larionov
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Charlene Kay
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Anita K Dunbier
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Jeremy S Thomas
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Mitch Dowsett
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Andrew H Sims
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand.
| | - J Michael Dixon
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
miR-451a Inhibited Cell Proliferation and Enhanced Tamoxifen Sensitive in Breast Cancer via Macrophage Migration Inhibitory Factor. BIOMED RESEARCH INTERNATIONAL 2015; 2015:207684. [PMID: 26161389 PMCID: PMC4486513 DOI: 10.1155/2015/207684] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/27/2014] [Accepted: 11/05/2014] [Indexed: 12/13/2022]
Abstract
This study aims to investigate the regulative effects of microRNA-451a (miR-451a) on cell proliferation and sensitivity to tamoxifen in breast cancer cells. In cell culture experiments, the lentiviral vectors of pHBLV-miR-451a and pHBLV-miR-451a sponge were constructed and used to transfect MCF-7 and LCC2 cells. The transfection efficiency was tested by fluorescent observation, and cell lines with stable over- or downregulated expression of miR-451a were established. The expression of miR-451a and the target gene macrophage migration inhibitory factor (MIF) were detected by real-time reverse transcriptase polymerase chain reaction and/or western blot. Moreover, MTT assay, colony formation, and Transwell invasion assays were also performed. Data showed that the recombinant lentiviral vectors were constructed correctly, and the virus titer was 1 × 10(8) CFU/mL. The stable transfected cells were obtained. Overexpression of miR-451a downregulated MIF expression in mRNA and protein levels and inhibited cell proliferation, colony formation, and invasion of breast cancer cells. Downregulation of miR-451a upregulated MIF expression and increased breast cancer cell growth, invasion, and tamoxifen sensitivity. In summary, the miR-451a/MIF pathway may play important roles in the biological properties of breast cancer cells and may be a potential therapeutic target for breast cancer.
Collapse
|
19
|
Yan S, Jiao X, Zou H, Li K. Prognostic significance of c-Met in breast cancer: a meta-analysis of 6010 cases. Diagn Pathol 2015; 10:62. [PMID: 26047809 PMCID: PMC4458003 DOI: 10.1186/s13000-015-0296-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/12/2015] [Indexed: 12/21/2022] Open
Abstract
Background The prognostic value of c-Met in breast cancer remains controversial. A meta-analysis of the impact of c-Met in breast cancer was performed by searching published data. Methods Published studies analyzing overall survival (OS) or relapse free survival (RFS) according to c-Met expression were searched. The principal outcome measures were hazard ratios (HRs) for RFS or OS according to c-Met expression. Combined HRs were calculated using fixed- or random- effects models according to the heterogeneity. Results Twenty-one studies involving 6,010 patients met our selection criteria. The impact of c-Met on RFS and OS was investigated in 12 and 17 studies, respectively. The meta-analysis results showed that c-Met overexpression significantly predicted poor RFS and OS in unselected breast cancer. Subgroup analysis indicated that c-Met overexpression was correlated with poor RFS and OS in Western patients, but was not associated with RFS or OS in Asian patients. C-Met was associated with poor OS in lymph node negative breast cancer and with poor RFS in hormone-receptor positive and triple negative breast cancer, but was not associated with prognosis in human epidermal growth factor receptor (HER)-2 positive breast cancer. Conclusions C-Met overexpression is an adverse prognostic marker in breast cancer, except among Asian and HER-2 positive patients. Virtual slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1869780799156041
Collapse
Affiliation(s)
- Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| | - Xin Jiao
- Department of Respiratory Medicine, Shenyang Chest Hospital, Shenyang, 110044, China.
| | - Huawei Zou
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| | - Kai Li
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
20
|
Notas G, Pelekanou V, Kampa M, Alexakis K, Sfakianakis S, Laliotis A, Askoxilakis J, Tsentelierou E, Tzardi M, Tsapis A, Castanas E. Tamoxifen induces a pluripotency signature in breast cancer cells and human tumors. Mol Oncol 2015; 9:1744-59. [PMID: 26115764 DOI: 10.1016/j.molonc.2015.05.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 05/20/2015] [Indexed: 01/01/2023] Open
Abstract
Tamoxifen is the treatment of choice in estrogen receptor alpha breast cancer patients that are eligible for adjuvant endocrine therapy. However, ∼50% of ERα-positive tumors exhibit intrinsic or rapidly acquire resistance to endocrine treatment. Unfortunately, prediction of de novo resistance to endocrine therapy and/or assessment of relapse likelihood remain difficult. While several mechanisms regulating the acquisition and the maintenance of endocrine resistance have been reported, there are several aspects of this phenomenon that need to be further elucidated. Altered metabolic fate of tamoxifen within patients and emergence of tamoxifen-resistant clones, driven by evolution of the disease phenotype during treatment, appear as the most compelling hypotheses so far. In addition, tamoxifen was reported to induce pluripotency in breast cancer cell lines, in vitro. In this context, we have performed a whole transcriptome analysis of an ERα-positive (T47D) and a triple-negative breast cancer cell line (MDA-MB-231), exposed to tamoxifen for a short time frame (hours), in order to identify how early pluripotency-related effects of tamoxifen may occur. Our ultimate goal was to identify whether the transcriptional actions of tamoxifen related to induction of pluripotency are mediated through specific ER-dependent or independent mechanisms. We report that even as early as 3 hours after the exposure of breast cancer cells to tamoxifen, a subset of ERα-dependent genes associated with developmental processes and pluripotency are induced and this is accompanied by specific phenotypic changes (expression of pluripotency-related proteins). Furthermore we report an association between the increased expression of pluripotency-related genes in ERα-positive breast cancer tissues samples and disease relapse after tamoxifen therapy. Finally we describe that in a small group of ERα-positive breast cancer patients, with disease relapse after surgery and tamoxifen treatment, ALDH1A1 (a marker of pluripotency in epithelial cancers which is absent in normal breast tissue) is increased in relapsing tumors, with a concurrent modification of its intra-cellular localization. Our data could be of value in the discrimination of patients susceptible to develop tamoxifen resistance and in the selection of optimized patient-tailored therapies.
Collapse
Affiliation(s)
- George Notas
- Laboratories of Experimental Endocrinology, University of Crete School of Medicine, Heraklion, Greece; Institute of Applied Computational Mathematics, Foundation of Research and Technology (FORTH), Heraklion, Greece.
| | - Vassiliki Pelekanou
- Laboratories of Experimental Endocrinology, University of Crete School of Medicine, Heraklion, Greece; Laboratories of Pathology, University of Crete School of Medicine, Heraklion, Greece
| | - Marilena Kampa
- Laboratories of Experimental Endocrinology, University of Crete School of Medicine, Heraklion, Greece
| | - Konstantinos Alexakis
- Laboratories of Experimental Endocrinology, University of Crete School of Medicine, Heraklion, Greece
| | - Stelios Sfakianakis
- Institute of Computer Science, Foundation of Research and Technology (FORTH), Heraklion, Greece
| | - Aggelos Laliotis
- Department of Surgical Oncology, University Hospital, Heraklion, Greece
| | - John Askoxilakis
- Department of Surgical Oncology, University Hospital, Heraklion, Greece
| | | | - Maria Tzardi
- Laboratories of Pathology, University of Crete School of Medicine, Heraklion, Greece
| | - Andreas Tsapis
- Laboratories of Experimental Endocrinology, University of Crete School of Medicine, Heraklion, Greece; INSERM U976, Hôpital Saint Louis, Paris, France; University Paris Diderot, Paris, France
| | - Elias Castanas
- Laboratories of Experimental Endocrinology, University of Crete School of Medicine, Heraklion, Greece.
| |
Collapse
|
21
|
Liu MZ, McLeod HL, He FZ, Chen XP, Zhou HH, Shu Y, Zhang W. Epigenetic perspectives on cancer chemotherapy response. Pharmacogenomics 2014; 15:699-715. [PMID: 24798726 DOI: 10.2217/pgs.14.41] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Epigenetic programs are now widely recognized as being critical to the biological processes of cancer genesis. However, it has not been comprehensively understood how and to what degree they can influence anticancer drugs responses. The development of drugs targeting epigenetic regulation has generated great enthusiasm, with a growing number in clinical development. We highlight here that epigenetic modifications can be involved in the regulation of genes responsible for the absorption, distribution, metabolism and excretion of drugs and for the pathological progression of cancer, thereby affecting anticancer drug responses. The major epigenetic regulatory mechanisms are reviewed, including DNA methylation, miRNA regulation and histone modification, with the aim of promoting rational use of anticancer drugs in the clinic and epigenetic drug development.
Collapse
Affiliation(s)
- Mou-Ze Liu
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, PR China
| | | | | | | | | | | | | |
Collapse
|
22
|
A functional interplay between ZNF217 and estrogen receptor alpha exists in luminal breast cancers. Mol Oncol 2014; 8:1441-57. [PMID: 24973012 DOI: 10.1016/j.molonc.2014.05.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/26/2014] [Accepted: 05/26/2014] [Indexed: 01/15/2023] Open
Abstract
We aimed at highlighting the role of ZNF217, a Krüppel-like finger protein, in Estrogen Receptor-α (ERα)-positive (ER+) and luminal breast cancers. Here we report for the first time that ZNF217 and ERα proteins bind to each other in both breast cancer cells and breast tumour samples, via the ERα hinge domain and the ZNF217 C-terminal domain. ZNF217 enhances the recruitment of ERα to its estrogen response elements (ERE) and the ERα-dependent transcription of the GREB1 estrogen-regulated gene. The prognostic power of ZNF217 mRNA expression levels is most discriminatory in breast cancers classified with a "good prognosis", particularly the Luminal-A subclass. A new immunohistochemistry ZNF217 index, based on nuclear and cytoplasmic ZNF217 staining, also allowed the identification of intermediate/poor relapse-free survivors in the Luminal-A subgroup. ZNF217 confers tamoxifen resistance in ER+ breast cancer cells and is a predictor of relapse under endocrine therapy in patients with ER+ breast cancer. ZNF217 thus allows the re-stratification of patients with ER+ breast cancers considered as cancers with good prognosis where no other biomarkers are currently available and widely used. Here we propose a model in ER+ breast cancer where ZNF217-driven aggressiveness incorporates ZNF217 as a positive enhancer of ERα direct genomic activity and where ZNF217 possesses its highest discriminatory prognostic value.
Collapse
|
23
|
Jiang X, Shapiro DJ. The immune system and inflammation in breast cancer. Mol Cell Endocrinol 2014; 382:673-682. [PMID: 23791814 PMCID: PMC4919022 DOI: 10.1016/j.mce.2013.06.003] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 02/07/2023]
Abstract
During different stages of tumor development the immune system can either identify and destroy tumors, or promote their growth. Therapies targeting the immune system have emerged as a promising treatment modality for breast cancer, and immunotherapeutic strategies are being examined in preclinical and clinical models. However, our understanding of the complex interplay between cells of the immune system and breast cancer cells is incomplete. In this article, we review recent findings showing how the immune system plays dual host-protective and tumor-promoting roles in breast cancer initiation and progression. We then discuss estrogen receptor α (ERα)-dependent and ERα-independent mechanisms that shield breast cancers from immunosurveillance and enable breast cancer cells to evade immune cell induced apoptosis and produce an immunosuppressive tumor microenvironment. Finally, we discuss protumorigenic inflammation that is induced during tumor progression and therapy, and how inflammation promotes more aggressive phenotypes in ERα positive breast cancers.
Collapse
Affiliation(s)
- Xinguo Jiang
- Department of Medicine, VA Palo Alto Health Care System/Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - David J Shapiro
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
24
|
Stone A, Cowley MJ, Valdes-Mora F, McCloy RA, Sergio CM, Gallego-Ortega D, Caldon CE, Ormandy CJ, Biankin AV, Gee JMW, Nicholson RI, Print CG, Clark SJ, Musgrove EA. BCL-2 hypermethylation is a potential biomarker of sensitivity to antimitotic chemotherapy in endocrine-resistant breast cancer. Mol Cancer Ther 2013; 12:1874-85. [PMID: 23861345 DOI: 10.1158/1535-7163.mct-13-0012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Overexpression of the antiapoptotic factor BCL-2 is a frequent feature of malignant disease and is commonly associated with poor prognosis and resistance to conventional chemotherapy. In breast cancer, however, high BCL-2 expression is associated with favorable prognosis, estrogen receptor (ER) positivity, and low tumor grade, whereas low expression is included in several molecular signatures associated with resistance to endocrine therapy. In the present study, we correlate BCL-2 expression and DNA methylation profiles in human breast cancer and in multiple cell models of acquired endocrine resistance to determine whether BCL-2 hypermethylation could provide a useful biomarker of response to cytotoxic therapy. In human disease, diminished expression of BCL-2 was associated with hypermethylation of the second exon, in a region that overlapped a CpG island and an ER-binding site. Hypermethylation of this region, which occurred in 10% of primary tumors, provided a stronger predictor of patient survival (P = 0.019) when compared with gene expression (n = 522). In multiple cell models of acquired endocrine resistance, BCL-2 expression was significantly reduced in parallel with increased DNA methylation of the exon 2 region. The reduction of BCL-2 expression in endocrine-resistant cells lowered their apoptotic threshold to antimitotic agents: nocodazole, paclitaxel, and the PLK1 inhibitor BI2536. This phenomenon could be reversed with ectopic expression of BCL-2, and rescued with the BCL-2 inhibitor ABT-737. Collectively, these data imply that BCL-2 hypermethylation provides a robust biomarker of response to current and next-generation cytotoxic agents in endocrine-resistant breast cancer, which may prove beneficial in directing therapeutic strategy for patients with nonresectable, metastatic disease.
Collapse
Affiliation(s)
- Andrew Stone
- Corresponding Author: Andrew Stone, Garvan Institute of Medical Research, L9 TKCC, 370 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kanwal S, Fardini Y, Pagesy P, N’Tumba-Byn T, Pierre-Eugène C, Masson E, Hampe C, Issad T. O-GlcNAcylation-inducing treatments inhibit estrogen receptor α expression and confer resistance to 4-OH-tamoxifen in human breast cancer-derived MCF-7 cells. PLoS One 2013; 8:e69150. [PMID: 23935944 PMCID: PMC3730543 DOI: 10.1371/journal.pone.0069150] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 06/05/2013] [Indexed: 01/08/2023] Open
Abstract
O-GlcNAcylation (addition of N-acetyl-glucosamine on serine or threonine
residues) is a post-translational modification that regulates stability,
activity or localization of cytosolic and nuclear proteins. O-linked
N-acetylgluocosmaine transferase (OGT) uses UDP-GlcNAc, produced in the
hexosamine biosynthetic pathway to O-GlcNacylate proteins. Removal of O-GlcNAc
from proteins is catalyzed by the β-N-Acetylglucosaminidase (OGA). Recent
evidences suggest that O-GlcNAcylation may affect the growth of cancer cells.
However, the consequences of O-GlcNAcylation on anti-cancer therapy have not
been evaluated. In this work, we studied the effects of O-GlcNAcylation on
tamoxifen-induced cell death in the breast cancer-derived MCF-7 cells.
Treatments that increase O-GlcNAcylation (PUGNAc and/or glucosoamine) protected
MCF-7 cells from death induced by tamoxifen. In contrast, inhibition of OGT
expression by siRNA potentiated the effect of tamoxifen on cell death. Since the
PI-3 kinase/Akt pathway is a major regulator of cell survival, we used BRET to
evaluate the effect of PUGNAc+glucosamine on PIP3 production. We
observed that these treatments stimulated PIP3 production in MCF-7
cells. This effect was associated with an increase in Akt phosphorylation.
However, the PI-3 kinase inhibitor LY294002, which abolished the effect of
PUGNAc+glucosamine on Akt phosphorylation, did not impair the protective effects
of PUGNAc+glucosamine against tamoxifen-induced cell death. These results
suggest that the protective effects of O-GlcNAcylation are independent of the
PI-3 kinase/Akt pathway. As tamoxifen sensitivity depends on the estrogen
receptor (ERα) expression level, we evaluated the effect of PUGNAc+glucosamine
on the expression of this receptor. We observed that O-GlcNAcylation-inducing
treatment significantly reduced the expression of ERα mRNA and protein,
suggesting a potential mechanism for the decreased tamoxifen sensitivity induced
by these treatments. Therefore, our results suggest that inhibition of
O-GlcNAcylation may constitute an interesting approach to improve the
sensitivity of breast cancer to anti-estrogen therapy.
Collapse
Affiliation(s)
- Shahzina Kanwal
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
| | - Yann Fardini
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
| | - Patrick Pagesy
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
| | - Thierry N’Tumba-Byn
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
| | - Cécile Pierre-Eugène
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
| | - Elodie Masson
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
| | - Cornelia Hampe
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
| | - Tarik Issad
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris,
France
- INSERM, U1016, Paris, France
- * E-mail:
| |
Collapse
|
26
|
Ayala G, Morello M, Frolov A, You S, Li R, Rosati F, Bartolucci G, Danza G, Adam RM, Thompson TC, Lisanti MP, Freeman MR, Vizio DD. Loss of caveolin-1 in prostate cancer stroma correlates with reduced relapse-free survival and is functionally relevant to tumour progression. J Pathol 2013; 231:77-87. [PMID: 23729330 DOI: 10.1002/path.4217] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 05/24/2013] [Accepted: 05/27/2013] [Indexed: 12/14/2022]
Abstract
Levels of caveolin-1 (Cav-1) in tumour epithelial cells increase during prostate cancer progression. Conversely, Cav-1 expression in the stroma can decline in advanced and metastatic prostate cancer. In a large cohort of 724 prostate cancers, we observed significantly decreased levels of stromal Cav-1 in concordance with increased Gleason score (p = 0.012). Importantly, reduced expression of Cav-1 in the stroma correlated with reduced relapse-free survival (p = 0.009), suggesting a role for stromal Cav-1 in inhibiting advanced disease. Silencing of Cav-1 by shRNA in WPMY-1 prostate fibroblasts resulted in up-regulation of Akt phosphorylation, and significantly altered expression of genes involved in angiogenesis, invasion, and metastasis, including a > 2.5-fold increase in TGF-β1 and γ-synuclein (SNCG) gene expression. Moreover, silencing of Cav-1 induced migration of prostate cancer cells when stromal cells were used as attractants. Pharmacological inhibition of Akt caused down-regulation of TGF-β1 and SNCG, suggesting that loss of Cav-1 in the stroma can influence Akt-mediated signalling in the tumour microenvironment. Cav-1-depleted stromal cells exhibited increased levels of intracellular cholesterol, a precursor for androgen biosynthesis, steroidogenic enzymes, and testosterone. These findings suggest that loss of Cav-1 in the tumour microenvironment contributes to the metastatic behaviour of tumour cells by a mechanism that involves up-regulation of TGF-β1 and SNCG through Akt activation. They also suggest that intracrine production of androgens, a process relevant to castration resistance, may occur in the stroma.
Collapse
Affiliation(s)
- Gustavo Ayala
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | - Matteo Morello
- Cancer Biology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,The Urological Diseases Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anna Frolov
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | - Sungyong You
- Cancer Biology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rile Li
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | - Fabiana Rosati
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Gianluca Bartolucci
- Department of Pharmaceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Giovanna Danza
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Rosalyn M Adam
- The Urological Diseases Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy C Thompson
- Department of Genitourinary Medical Oncology, Unit 18-3, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P Lisanti
- Breast Oncology and Institute of Cancer Sciences, Paterson Institute of Cancer Research, The University of Manchester, Manchester, UK
| | - Michael R Freeman
- Cancer Biology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,The Urological Diseases Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Departments of Surgery and Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Dolores Di Vizio
- Cancer Biology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,The Urological Diseases Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
The HER2 amplicon in breast cancer: Topoisomerase IIA and beyond. Biochim Biophys Acta Rev Cancer 2013; 1836:146-57. [PMID: 23628726 DOI: 10.1016/j.bbcan.2013.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/17/2013] [Accepted: 04/19/2013] [Indexed: 12/20/2022]
Abstract
HER2 gene amplification is observed in about 15% of breast cancers. The subgroup of HER2-positive breast cancers appears to be heterogeneous and presents complex patterns of gene amplification at the locus on chromosome 17q12-21. The molecular variations within the chromosome 17q amplicon and their clinical implications remain largely unknown. Besides the well-known TOP2A gene encoding Topoisomerase IIA, other genes might also be amplified and could play functional roles in breast cancer development and progression. This review will focus on the current knowledge concerning the HER2 amplicon heterogeneity, its clinical and biological impact and the pitfalls associated with the evaluation of gene amplifications at this locus, with particular attention to TOP2A and the link between TOP2A and anthracycline benefit. In addition it will discuss the clinical and biological implications of the amplification of ten other genes at this locus (MED1, STARD3, GRB7, THRA, RARA, IGFPB4, CCR7, KRT20, KRT19 and GAST) in breast cancer.
Collapse
|
28
|
Lyng MB, Lænkholm AV, Tan Q, Vach W, Gravgaard KH, Knoop A, Ditzel HJ. Gene expression signatures that predict outcome of tamoxifen-treated estrogen receptor-positive, high-risk, primary breast cancer patients: a DBCG study. PLoS One 2013; 8:e54078. [PMID: 23342080 PMCID: PMC3546921 DOI: 10.1371/journal.pone.0054078] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 12/06/2012] [Indexed: 12/28/2022] Open
Abstract
Background Tamoxifen significantly improves outcome for estrogen receptor-positive (ER+) breast cancer, but the 15-year recurrence rate remains 30%. The aim of this study was to identify gene profiles that accurately predicted the outcome of ER+ breast cancer patients who received adjuvant Tamoxifen mono-therapy. Methodology/Principal Findings Post-menopausal breast cancer patients diagnosed no later than 2002, being ER+ as defined by >1% IHC staining and having a frozen tumor sample with >50% tumor content were included. Tumor samples from 108 patients treated with adjuvant Tamoxifen were analyzed for the expression of 59 genes using quantitative-PCR. End-point was clinically verified recurrence to distant organs or ipsilateral breast. Gene profiles were identified using a model building procedure based on conditional logistic regression and leave-one-out cross-validation, followed by a non-parametric bootstrap (1000x re-sampling). The optimal profiles were further examined in 5 previously-reported datasets containing similar patient populations that were either treated with Tamoxifen or left untreated (n = 623). Three gene signatures were identified, the strongest being a 2-gene combination of BCL2-CDKN1A, exhibiting an accuracy of 75% for prediction of outcome. Independent examination using 4 previously-reported microarray datasets of Tamoxifen-treated patient samples (n = 503) confirmed the potential of BCL2-CDKN1A. The predictive value was further determined by comparing the ability of the genes to predict recurrence in an additional, previously-published, cohort consisting of Tamoxifen-treated (n = 58, p = 0.015) and untreated patients (n = 62, p = 0.25). Conclusions/Significance A novel gene expression signature predictive of outcome of Tamoxifen-treated patients was identified. The validation suggests that BCL2-CDKN1A exhibit promising predictive potential.
Collapse
Affiliation(s)
- Maria B. Lyng
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- * E-mail: (MBL); (HJD)
| | - Anne-Vibeke Lænkholm
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Pathology, Slagelse Hospital, Slagelse, Denmark
| | - Qihua Tan
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Werner Vach
- Institute of Medical Biometry and Medical Informatics, University Medical Center Freiburg, Freiburg, Germany
| | - Karina H. Gravgaard
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ann Knoop
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Henrik J. Ditzel
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- * E-mail: (MBL); (HJD)
| |
Collapse
|
29
|
Zhang L, Gong C, Lau SLY, Yang N, Wong OGW, Cheung ANY, Tsang JWH, Chan KYK, Khoo US. SpliceArray Profiling of Breast Cancer Reveals a Novel Variant of NCOR2/SMRT That Is Associated with Tamoxifen Resistance and Control of ERα Transcriptional Activity. Cancer Res 2012; 73:246-55. [PMID: 23117886 DOI: 10.1158/0008-5472.can-12-2241] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Luduo Zhang
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Network insights on oxaliplatin anti-cancer mechanisms. Clin Transl Med 2012; 1:26. [PMID: 23369220 PMCID: PMC3560997 DOI: 10.1186/2001-1326-1-26] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 06/22/2012] [Indexed: 12/25/2022] Open
Abstract
Oxaliplatin has been a crucial component of combination therapies since admission into the clinic causing modest gains in survival across multiple malignancies. However, oxaliplatin functions in a non-targeted manner, posing a difficulty in ascertaining precise efficacy mechanisms. While previously thought to only affect DNA repair mechanisms, Platinum-protein adducts (Pt-Protein) far outnumber Pt-DNA adducts leaving a big part of oxaliplatin function unknown. Through preliminary network modeling of high throughput data, this article critically reviews the efficacy of oxaliplatin as well as proposes a better model for enhanced efficacy based on a network approach. In our study, not only oxaliplatin’s function in interrupting DNA-replication was confirmed, but also its role in initiating or intensifying tumorigenesis pathways was uncovered. From our data we present a novel picture of competing signaling networks that collectively provide a plausible explanation of chemotherapeutic resistance, cancer stem cell survival, as well as invasiveness and metastases. Here we highlight oxaliplatin signaling networks, their significance and the clinical implications of these interactions that verifies the importance of network modeling in rational drug design.
Collapse
|
31
|
GREB1 functions as a growth promoter and is modulated by IL6/STAT3 in breast cancer. PLoS One 2012; 7:e46410. [PMID: 23056300 PMCID: PMC3463574 DOI: 10.1371/journal.pone.0046410] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 08/29/2012] [Indexed: 11/19/2022] Open
Abstract
Background Growth Regulation by Estrogen in Breast cancer (GREB1) was an estrogen receptor (ER) target gene, and GREB1 expression inversely correlated with HER2 status, possibly as a surrogate marker for ER status and a predictor for tamoxifen resistance in breast cancer patients. In the present study, we examine the function and regulation of GREB1 in breast cancer, with the goal to develop GREB1 as a biomarker in breast cancer with de novo and acquired tamoxifen resistance. Methods We overexpressed GREB1 using adenovirus containing the full length GREB1 cDNA (Ad-GREB1) in breast cancer cell lines. The soft agar assay was used as a measure of anchorage independent growth. The effects of GREB1 on cell proliferation in MCF-7 cells transduced with Ad-GREB1 were also measured by the me olic activity using AlamarBlue assay. We tested whether there was interaction between STAT3 and ER, which could repress GREB1 expression by immunoprecipitation assay. The effects of IL-6/JAK/STAT3 cascade activation on estrogen-induced GREB1 promoter activity were determined by luciferase assay and those on gene expression were measured by real time reverse transcription polymerase chain reaction (qRT-PCR). Results We found that the ability of breast cancer cells to grow in soft agar is enhanced following GREB1 transfection. In MCF-7 cells transduced with Ad-GREB1 or transfected with siRNA GREB1, the metabolic activity was increased or completely abolished, suggesting that GREB1 may function as a growth promoter in breast cancer. E2 treatment increased GREB1 promoter luciferase activity. IL-6 inhibited E2-induced GREB1 transcription activity and GREB1 mRNA expression. Constitutively expressing active STAT3 construct (STAT3-C) dramatically decreased GREB1 transcription. Conclusions These data indicate that overexpression of GREB1 promotes cell proliferation and increases the clonogenic ability in breast cancer cells. Moreover, Il6/STAT3 modulates estrogen-induced GREB1 transcriptional activity in breast cancer cells.
Collapse
|
32
|
Emerging roles for the pro-oncogenic anterior gradient-2 in cancer development. Oncogene 2012; 32:2499-509. [PMID: 22945652 DOI: 10.1038/onc.2012.346] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Clinical studies have defined the core 'genetic blueprint' of a cancer cell, but this information does not necessarily predict the cancer phenotype. Signalling hubs that mediate such phenotype have been identified largely using OMICS platforms that measure dynamic molecular changes within the cancer cell landscape. The pro-oncogenic protein anterior gradient 2 (AGR2) is a case in point; AGR2 has been shown using a range of expression platforms to be involved in asthma, inflammatory bowel disease, cell transformation, cancer drug resistance and metastatic growth. AGR2 protein is also highly overexpressed in a diverse range of human cancers and can be secreted and detected in extracellular fluids, thus representing a compelling pro-oncogenic signalling intermediate in human cancer. AGR2 belongs to the protein disulphide isomerase family with all the key features of an endoplasmic reticulum-resident protein-this gives clues into how it might function as an oncoprotein through the regulation of protein folding, maturation and secretion that can drive metastatic cell growth. In this review, we will describe the known aspects of AGR2 molecular biology, including gene structure and regulation, emerging protein interaction networks and how its subcellular localization mediates its biological functions. We will finally review the cases of AGR2 expression in human cancers, the pathophysiological consequences of AGR2 overexpression, its potential role as a tumour biomarker that predicts the response to therapy and how the AGR2 pathway might form the basis for drug discovery programmes aimed at targeting protein folding/maturation pathways that mediate secretion and metastasis.
Collapse
|
33
|
Gene expression profiling to dissect the complexity of cancer biology: Pitfalls and promise. Semin Cancer Biol 2012; 22:250-60. [DOI: 10.1016/j.semcancer.2012.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 12/15/2022]
|
34
|
Vendrell JA, Thollet A, Nguyen NT, Ghayad SE, Vinot S, Bièche I, Grisard E, Josserand V, Coll JL, Roux P, Corbo L, Treilleux I, Rimokh R, Cohen PA. ZNF217 Is a Marker of Poor Prognosis in Breast Cancer That Drives Epithelial–Mesenchymal Transition and Invasion. Cancer Res 2012; 72:3593-606. [DOI: 10.1158/0008-5472.can-11-3095] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
35
|
Development of multigene expression signature maps at the protein level from digitized immunohistochemistry slides. PLoS One 2012; 7:e33520. [PMID: 22438942 PMCID: PMC3305321 DOI: 10.1371/journal.pone.0033520] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 02/15/2012] [Indexed: 12/03/2022] Open
Abstract
Molecular classification of diseases based on multigene expression signatures is increasingly used for diagnosis, prognosis, and prediction of response to therapy. Immunohistochemistry (IHC) is an optimal method for validating expression signatures obtained using high-throughput genomics techniques since IHC allows a pathologist to examine gene expression at the protein level within the context of histologically interpretable tissue sections. Additionally, validated IHC assays may be readily implemented as clinical tests since IHC is performed on routinely processed clinical tissue samples. However, methods have not been available for automated n-gene expression profiling at the protein level using IHC data. We have developed methods to compute expression level maps (signature maps) of multiple genes from IHC data digitized on a commercial whole slide imaging system. Areas of cancer for these expression level maps are defined by a pathologist on adjacent, co-registered H&E slides, allowing assessment of IHC statistics and heterogeneity within the diseased tissue. This novel way of representing multiple IHC assays as signature maps will allow the development of n-gene expression profiling databases in three dimensions throughout virtual whole organ reconstructions.
Collapse
|
36
|
A five-gene model predicts clinical outcome in ER+/PR+, early-stage breast cancers treated with adjuvant tamoxifen. Discov Oncol 2012; 2:261-71. [PMID: 21826535 DOI: 10.1007/s12672-011-0080-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Primary breast carcinomas expressing both estrogen and progesterone receptors are most likely to respond to tamoxifen therapy, especially in patients with early-stage lesions. However, certain patients exhibit clinicopathologic features suggesting good prognosis relapse within 10 years, justifying a search for biomarkers identifying patients at risk for recurrence. Nine candidate genes associated with estrogen signaling were selected from microarray studies and combined with those for conventional biomarkers (ESR1, PGR, ERBB2). Expression of this 12-gene subset was analyzed by RT-qPCR in frozen tissue specimens from 60 early-stage, estrogen receptor (ER)+/progestin receptor (PR)+ breast cancers from patients treated with adjuvant tamoxifen. A multivariate model was created by Cox regression using a training data set and applied to an independent validation set. A five-gene model was developed from the training set (n = 36) that exhibited significant correlations with both relapse-free and overall survival. Applying this model to Kaplan-Meier regression, patients were separated into low-risk (100% relapse-free at 150 months) and high-risk (60% relapse-free at 150 months) groups (P = 0.03). When this model was applied to the validation set (n = 24), similar risk stratification was achieved for both relapse-free and overall survival (P = 0.01 and 0.04, respectively). We developed a five-gene model composed of PgR, BCL2, ERBB4 JM-a, RERG, and CD34 that identified early-stage, ER+/PR+ breast cancers in patients treated with tamoxifen that relapsed, although they exhibited clinicopathologic features suggesting good prognosis. Within this multivariate model, increased expression of PgR, ERBB4 JM-a, RERG, and CD34 was associated with increased survival, while increased expression of BCL2 was associated with decreased survival.
Collapse
|
37
|
Becker MA, Hou X, Harrington SC, Weroha SJ, Gonzalez SE, Jacob KA, Carboni JM, Gottardis MM, Haluska P. IGFBP ratio confers resistance to IGF targeting and correlates with increased invasion and poor outcome in breast tumors. Clin Cancer Res 2012; 18:1808-17. [PMID: 22287600 DOI: 10.1158/1078-0432.ccr-11-1806] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE To improve the significance of insulin-like growth factor-binding protein 5 (IGFBP-5) as a prognostic and potentially predictive marker in patients with breast cancer. EXPERIMENTAL DESIGN Increased IGFBP-5 expression was identified in MCF-7 cells resistant (MCF-7R4) to the IGF-1R/insulin receptor (InsR) inhibitor BMS-536924 and its role examined by targeted knockdown and overexpression in multiple experimental models. Protein expression of IGFBP-5 was measured by immunohistochemistry in a cohort of 76 patients with breast cancer to examine correlative associations with invasive tumor fraction and outcome. The use of a combined IGFBP-5/IGFBP-4 (BPR) expression ratio was applied to predict anti-IGF-1R/InsR response in a panel of breast cancer lines and outcome in multiple breast tumor cohorts. RESULTS IGFBP-5 knockdown decreased BMS-536924 resistance in MCF-7R4 cells, whereas IGFBP-5 overexpression in MCF-7 cells conferred resistance. When compared with pathologically normal reduction mammoplasty tissue, IGFBP-5 expression levels were upregulated in both invasive and histologically normal adjacent breast cancer tissue. In both univariate and multivariate modeling, metastasis-free survival, recurrence free survival (RFS), and overall survival (OS) were significantly associated with high IGFBP-5 expression. Prognostic power of IGFBP-5 was further increased with the addition of IGFBP-4 where tumors were ranked based upon IGFBP-5/IGFBP-4 expression ratio (BPR). Multiple breast cancer cohorts confirm that BPR (high vs. low) was a strong predictor of RFS and OS. CONCLUSION IGFBP-5 expression is a marker of poor outcome in patients with breast cancer. An IGFBP-5/IGFBP-4 expression ratio may serve as a surrogate biomarker of IGF pathway activation and predict sensitivity to anti-IGF-1R targeting.
Collapse
Affiliation(s)
- Marc A Becker
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Evidence for biological effects of metformin in operable breast cancer: a pre-operative, window-of-opportunity, randomized trial. Breast Cancer Res Treat 2011; 128:783-94. [PMID: 21655990 DOI: 10.1007/s10549-011-1612-1] [Citation(s) in RCA: 231] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 05/24/2011] [Indexed: 10/18/2022]
Abstract
Metformin may reduce the incidence of breast cancer and enhance response to neoadjuvant chemotherapy in diabetic women. This trial examined the effects of metformin on Ki67 and gene expression in primary breast cancer. Non-diabetic women with operable invasive breast cancer received pre-operative metformin. A pilot cohort of eight patients had core biopsy of the cancer at presentation, a week later (without treatment; internal control), then following metformin 500-mg o.d. for 1 week increased to 1-g b.d. for a further week continued to surgery. A further 47 patients had core biopsy at diagnosis were randomized to metformin (the same dose regimen) or no drug, and 2 weeks later had core biopsy at surgery. Ki67 immunohistochemistry, transcriptome analysis on formalin-fixed paraffin-embedded cores and serum insulin determination were performed blinded to treatment. Seven patients (7/32, 21.9%) receiving metformin withdrew because of gastrointestinal upset. The mean percentage of cells staining for Ki67 fell significantly following metformin treatment in both the pilot cohort (P = 0.041, paired t-test) and in the metformin arm (P = 0.027, Wilcoxon rank test) but was unchanged in the internal control or metformin control arms. Messenger RNA expression was significantly downregulated by metformin for PDE3B (phosphodiesterase 3B, cGMP-inhibited; a critical regulator of cAMP levels that affect activation of AMP-activated protein kinase, AMPK), confirmed by immunohistochemistry, SSR3, TP53 and CCDC14. By ingenuity pathway analysis, the tumour necrosis factor receptor 1 (TNFR1) signaling pathway was most affected by metformin: TGFB and MEKK were upregulated and cdc42 downregulated; mTOR and AMPK pathways were also affected. Gene set analysis additionally revealed that p53, BRCA1 and cell cycle pathways also had reduced expression following metformin. Mean serum insulin remained stable in patients receiving metformin but rose in control patients. This trial presents biomarker evidence for anti-proliferative effects of metformin in women with breast cancer and provides support for therapeutic trials of metformin.
Collapse
|
39
|
Yerushalmi R, Gelmon KA, Leung S, Gao D, Cheang M, Pollak M, Turashvili G, Gilks BC, Kennecke H. Insulin-like growth factor receptor (IGF-1R) in breast cancer subtypes. Breast Cancer Res Treat 2011; 132:131-42. [DOI: 10.1007/s10549-011-1529-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 04/16/2011] [Indexed: 01/22/2023]
|
40
|
Arendt LM, Rugowski DE, Grafwallner-Huseth TA, Garcia-Barchino MJ, Rui H, Schuler LA. Prolactin-induced mouse mammary carcinomas model estrogen resistant luminal breast cancer. Breast Cancer Res 2011; 13:R11. [PMID: 21276249 PMCID: PMC3109579 DOI: 10.1186/bcr2819] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Revised: 11/30/2010] [Accepted: 01/28/2011] [Indexed: 02/07/2023] Open
Abstract
Introduction Tumors that express estrogen receptor alpha (ERα+) comprise 75% of breast cancers in women. While treatments directed against this receptor have successfully lowered mortality rates, many primary tumors initially or later exhibit resistance. The paucity of murine models of this "luminal" tumor subtype has hindered studies of factors that promote their pathogenesis and modulate responsiveness to estrogen-directed therapeutics. Since epidemiologic studies closely link prolactin and the development of ERα+ tumors in women, we examined characteristics of the aggressive ERα+ and ERα- carcinomas which develop in response to mammary prolactin in a murine transgenic model (neu-related lipocalin- prolactin (NRL-PRL)). To evaluate their relationship to clinical tumors, we determined phenotypic relationships among these carcinomas, other murine models of breast cancer, and features of luminal tumors in women. Methods We examined a panel of prolactin-induced tumors for characteristics relevant to clinical tumors: histotype, ERα/progesterone receptor (PR) expression and estrogen responsiveness, Activating Protein 1 (AP-1) components, and phosphorylation of signal transducer and activator of transcription 5 (Stat5), extracellular signal regulated kinase (ERK) 1/2 and AKT. We compared levels of transcripts in the ERα-associated "luminal" signature that defines this subtype of tumors in women and transcripts enriched in various mammary epithelial lineages to other well-studied genetically modified murine models of breast cancer. Finally, we used microarray analyses to compare prolactin-induced ERα+ and ERα- tumors, and examined responsiveness to estrogen and the anti-estrogen, Faslodex, in vivo. Results Prolactin-induced carcinomas were markedly diverse with respect to histotype, ERα/PR expression, and activated signaling cascades. They constituted a heterogeneous, but distinct group of murine mammary tumors, with molecular features of the luminal subtype of human breast cancer. In contrast to morphologically normal and hyperplastic structures in NRL-PRL females, carcinomas were insensitive to ERα-mediated signals. These tumors were distinct from mouse mammary tumor virus (MMTV)-neu tumors, and contained elevated transcripts for factors associated with luminal/alveolar expansion and differentiation, suggesting that they arose from physiologic targets of prolactin. These features were shared by ERα+ and ERα- tumors, suggesting a common origin, although the former exhibited transcript profiles reflecting greater differentiation. Conclusions Our studies demonstrate that prolactin can promote diverse carcinomas in mice, many of which resemble luminal breast cancers, providing a novel experimental model to examine the pathogenesis, progression and treatment responsiveness of this tumor subtype.
Collapse
Affiliation(s)
- Lisa M Arendt
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Dr., Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
41
|
Thompson AM, Johnson A, Quinlan P, Hillman G, Fontecha M, Bray SE, Purdie CA, Jordan LB, Ferraldeschi R, Latif A, Hadfield KD, Clarke RB, Ashcroft L, Evans DG, Howell A, Nikoloff M, Lawrence J, Newman WG. Comprehensive CYP2D6 genotype and adherence affect outcome in breast cancer patients treated with tamoxifen monotherapy. Breast Cancer Res Treat 2010; 125:279-87. [PMID: 20809362 DOI: 10.1007/s10549-010-1139-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 08/18/2010] [Indexed: 12/15/2022]
Abstract
The association between CYP2D6 genotype and outcome in breast cancer patients treated with adjuvant tamoxifen remains controversial. We assessed the influence of comprehensive versus limited CYP2D6 genotype in the context of tamoxifen adherence and co-medication in a large cohort of 618 patients. Genotyping of 33 CYP2D6 alleles used two archival cohorts from tamoxifen-treated women with invasive breast cancer (Dundee, n = 391; Manchester, n = 227). Estimates for recurrence-free survival (RFS) were calculated based on inferred CYP2D6 phenotypes using Kaplan-Meier and Cox proportional hazard models, adjusted for nodal status and tumour size. Patients with at least one reduced function CYP2D6 allele (60%) or no functional alleles (6%) had a non-significant trend for worse RFS: hazard ratio (HR) 1.52 (CI 0.98-2.36, P = 0.06). For post-menopausal women on tamoxifen monotherapy, the HR for recurrence in patients with reduced functional alleles was 1.96 (CI 1.05-3.66, P = 0.036). However, RFS analysis limited to four common CYP2D6 allelic variants was no longer significant (P = 0.39). The effect of CYP2D6 genotype was increased by adjusting for adherence to tamoxifen therapy, but not significantly changed when adjusted for co-administration of potent inhibitors of CYP2D6. Comprehensive genotyping of CYP2D6 and adherence to tamoxifen therapy may be useful to identify breast cancer patients most likely to benefit from adjuvant tamoxifen.
Collapse
Affiliation(s)
- Alastair M Thompson
- Department of Surgery and Molecular Oncology, University of Dundee, Dundee DD1 9SY, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Taylor KJ, Sims AH, Liang L, Faratian D, Muir M, Walker G, Kuske B, Dixon JM, Cameron DA, Harrison DJ, Langdon SP. Dynamic changes in gene expression in vivo predict prognosis of tamoxifen-treated patients with breast cancer. Breast Cancer Res 2010; 12:R39. [PMID: 20569502 PMCID: PMC2917034 DOI: 10.1186/bcr2593] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 01/02/2010] [Accepted: 06/22/2010] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Tamoxifen is the most widely prescribed anti-estrogen treatment for patients with estrogen receptor (ER)-positive breast cancer. However, there is still a need for biomarkers that reliably predict endocrine sensitivity in breast cancers and these may well be expressed in a dynamic manner. METHODS In this study we assessed gene expression changes at multiple time points (days 1, 2, 4, 7, 14) after tamoxifen treatment in the ER-positive ZR-75-1 xenograft model that displays significant changes in apoptosis, proliferation and angiogenesis within 2 days of therapy. RESULTS Hierarchical clustering identified six time-related gene expression patterns, which separated into three groups: two with early/transient responses, two with continuous/late responses and two with variable response patterns. The early/transient response represented reductions in many genes that are involved in cell cycle and proliferation (e.g. BUB1B, CCNA2, CDKN3, MKI67, UBE2C), whereas the continuous/late changed genes represented the more classical estrogen response genes (e.g. TFF1, TFF3, IGFBP5). Genes and the proteins they encode were confirmed to have similar temporal patterns of expression in vitro and in vivo and correlated with reduction in tumour volume in primary breast cancer. The profiles of genes that were most differentially expressed on days 2, 4 and 7 following treatment were able to predict prognosis, whereas those most changed on days 1 and 14 were not, in four tamoxifen treated datasets representing a total of 404 patients. CONCLUSIONS Both early/transient/proliferation response genes and continuous/late/estrogen-response genes are able to predict prognosis of primary breast tumours in a dynamic manner. Temporal expression of therapy-response genes is clearly an important factor in characterising the response to endocrine therapy in breast tumours which has significant implications for the timing of biopsies in neoadjuvant biomarker studies.
Collapse
Affiliation(s)
- Karen J Taylor
- CRUK Cancer Research Centre and Academic Breast Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wagoner MP, Gunsalus KTW, Schoenike B, Richardson AL, Friedl A, Roopra A. The transcription factor REST is lost in aggressive breast cancer. PLoS Genet 2010; 6:e1000979. [PMID: 20548947 PMCID: PMC2883591 DOI: 10.1371/journal.pgen.1000979] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 05/07/2010] [Indexed: 01/24/2023] Open
Abstract
The function of the tumor suppressor RE1 silencing transcription factor (REST) is lost in colon and small cell lung cancers and is known to induce anchorage-independent growth in human mammary epithelial cells. However, nothing is currently known about the role of this tumor suppressor in breast cancer. Here, we test the hypothesis that loss of REST function plays a role in breast cancer. To assay breast tumors for REST function, we developed a 24-gene signature composed of direct targets of the transcriptional repressor. Using the 24- gene signature, we identified a previously undefined RESTless breast tumor subtype. Using gene set enrichment analysis, we confirmed the aberrant expression of REST target genes in the REST–less tumors, including neuronal gene targets of REST that are normally not expressed outside the nervous system. Examination of REST mRNA identified a truncated splice variant of REST present in the REST–less tumor population, but not other tumors. Histological analysis of 182 outcome-associated breast tumor tissues also identified a subpopulation of tumors that lack full-length, functional REST and over-express the neuroendocrine marker and REST target gene Chromogranin A. Importantly, patients whose tumors were found to be REST–less using either the 24-gene signature or histology had significantly poorer prognosis and were more than twice as likely to undergo disease recurrence within the first 3 years after diagnosis. We show here that REST function is lost in breast cancer, at least in part via an alternative splicing mechanism. Patients with REST–less breast cancer undergo significantly more early disease recurrence than those with fully functional REST, regardless of estrogen receptor or HER2 status. Importantly, REST status may serve as a predictor of poor prognosis, helping to untangle the heterogeneity inherent in disease course and response to treatment. Additionally, the alternative splicing observed in REST–less breast cancer is an attractive therapeutic target. Breast cancer is a heterogeneous disease, with highly variable disease outcomes and responses to treatment for otherwise indistinguishable tumors. Understanding this heterogeneity holds the key to better determining disease prognosis and tailoring treatments to the tumors for which they will be most efficacious. Some of the most successful work dissecting the differences between histologically identical tumors with differing disease outcomes has come from profiling the array of protein-coding transcripts present in every tumor and dividing the breast cancer profiles into multiple subtypes of varying aggressiveness. Importantly, these profiles are now being used in the clinic to predict disease outcome and plan treatment. Using a similar molecular-profiling strategy, we have identified a previously unrecognized subset of breast cancers in which the tumor suppressor gene REST is lost, which display a highly aggressive disease course. Intriguingly, we have traced the loss of the tumor suppressor to the presence of a variant of the REST protein normally found in the brain following seizures, which represents a unique and attractive therapeutic target. Additionally, the gene signature used to identify REST–less tumors shows no overlap with the profiles currently used in the clinic to assess tumor aggressiveness and may be an important new diagnostic tool.
Collapse
Affiliation(s)
- Matthew P. Wagoner
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kearney T. W. Gunsalus
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Barry Schoenike
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andrea L. Richardson
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Andreas Friedl
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Avtar Roopra
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
44
|
Hens J, Dann P, Hiremath M, Pan TC, Chodosh L, Wysolmerski J. Analysis of gene expression in PTHrP-/- mammary buds supports a role for BMP signaling and MMP2 in the initiation of ductal morphogenesis. Dev Dyn 2010; 238:2713-24. [PMID: 19795511 DOI: 10.1002/dvdy.22097] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Parathyroid hormone-related protein (PTHrP) acts on the mammary mesenchyme and is required for proper embryonic mammary development. In order to understand PTHrP's effects on mesenchymal cells, we profiled gene expression in WT and PTHrP(-/-) mammary buds, and in WT and K14-PTHrP ventral skin at E15.5. By cross-referencing the differences in gene expression between these groups, we identified 35 genes potentially regulated by PTHrP in the mammary mesenchyme, including 6 genes known to be involved in BMP signaling. One of these genes was MMP2. We demonstrated that PTHrP and BMP4 regulate MMP2 gene expression and MMP2 activity in mesenchymal cells. Using mammary bud cultures, we demonstrated that MMP2 acts downstream of PTHrP to stimulate ductal outgrowth. Future studies on the functional role of other genes on this list should expand our knowledge of how PTHrP signaling triggers the onset of ductal outgrowth from the embryonic mammary buds.
Collapse
Affiliation(s)
- Julie Hens
- Department of Biology, St. Bonaventure University, St. Bonaventure, New York, USA
| | | | | | | | | | | |
Collapse
|
45
|
Ghayad SE, Vendrell JA, Ben Larbi S, Dumontet C, Bieche I, Cohen PA. Endocrine resistance associated with activated ErbB system in breast cancer cells is reversed by inhibiting MAPK or PI3K/Akt signaling pathways. Int J Cancer 2010; 126:545-62. [PMID: 19609946 DOI: 10.1002/ijc.24750] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endocrine therapy resistance is one of the main challenges in the treatment of estrogen receptor positive (ER+) breast cancer patients. This study showed that two ER+ human breast carcinoma cell lines derived from MCF-7 (MVLN cells) that have acquired under OH-Tamoxifen selection two distinct phenotypes of endocrine resistance both displayed constitutive activation of the PI3K/Akt and MAPK pathways. Aberrant expression and activation of the ErbB system (phospho-EGFR, phospho-ErbB2, phospho-ErbB3, over-expression of ErbB4 and over-expression of several ErbB ligands) were also observed in the two resistant cell lines, suggesting the existence of an autocrine loop leading to constitutive activation of MAPK and PI3K/Akt survival pathways. The recent clinical use of specific signal transduction inhibitors is one of the most promising therapeutic approaches in breast cancers. The MEK inhibitor PD98059 and the PI3K inhibitor LY294002 were both able to enhance the cytostatic effect of OH-Tamoxifen or fulvestrant on MVLN sensitive cells. In the two resistant cell lines, inhibition of the MAPK or the PI3K/Akt pathways associated with endocrine therapy was sufficient to reverse OH-Tamoxifen or fulvestrant resistance. Investigating the effect of a combination of both inhibitors on the reversion of OH-Tamoxifen and fulvestrant resistance in the two resistant cell lines suggested that, in clinical practice, a strategy combining the two inhibitors would be the best approach to target the different endocrine resistance phenotypes possibly present in a tumor. In conclusion, the combination of MAPK and PI3K inhibitors represents a promising strategy to overcome endocrine therapy resistance in ER+ breast cancer patients.
Collapse
|
46
|
Abstract
Endocrine therapies targeting oestrogen action (anti-oestrogens, such as tamoxifen, and aromatase inhibitors) decrease mortality from breast cancer, but their efficacy is limited by intrinsic and acquired therapeutic resistance. Candidate molecular biomarkers and gene expression signatures of tamoxifen response emphasize the importance of deregulation of proliferation and survival signalling in endocrine resistance. However, definition of the specific genetic lesions and molecular processes that determine clinical endocrine resistance is incomplete. The development of large-scale computational and genetic approaches offers the promise of identifying the mediators of endocrine resistance that may be exploited as potential therapeutic targets and biomarkers of response in the clinic.
Collapse
Affiliation(s)
- Elizabeth A Musgrove
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia.
| | | |
Collapse
|
47
|
Sims AH, Bartlett JMS. Approaches towards expression profiling the response to treatment. Breast Cancer Res 2008; 10:115. [PMID: 19144210 PMCID: PMC2656889 DOI: 10.1186/bcr2196] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Over the past 8 years there has been a wealth of breast cancer gene expression studies. The majority of these studies have focused upon characterising a tumour at presentation, before treatment, rather than looking at the effects of treatment on the tumour. More recently, a number of groups have moved from predicting prognosis based upon long-term follow-up to alternative approaches of using expression profiling to measure the effect of treatment on breast tumours and potentially predict response to therapy using either post-treatment samples or both pre-treatment and post-treatment samples. Whilst this provides great potential to further our understanding of the mode of action of treatments and to more accurately select which patients will benefit from a particular treatment, serious issues of experimental design must be considered.
Collapse
|