101
|
Lin ML, Patel H, Remenyi J, Banerji CRS, Lai CF, Periyasamy M, Lombardo Y, Busonero C, Ottaviani S, Passey A, Quinlan PR, Purdie CA, Jordan LB, Thompson AM, Finn RS, Rueda OM, Caldas C, Gil J, Coombes RC, Fuller-Pace FV, Teschendorff AE, Buluwela L, Ali S. Expression profiling of nuclear receptors in breast cancer identifies TLX as a mediator of growth and invasion in triple-negative breast cancer. Oncotarget 2016; 6:21685-703. [PMID: 26280373 PMCID: PMC4673296 DOI: 10.18632/oncotarget.3942] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/30/2015] [Indexed: 01/23/2023] Open
Abstract
The Nuclear Receptor (NR) superfamily of transcription factors comprises 48 members, several of which have been implicated in breast cancer. Most important is estrogen receptor-α (ERα), which is a key therapeutic target. ERα action is facilitated by co-operativity with other NR and there is evidence that ERα function may be recapitulated by other NRs in ERα-negative breast cancer. In order to examine the inter-relationships between nuclear receptors, and to obtain evidence for previously unsuspected roles for any NRs, we undertook quantitative RT-PCR and bioinformatics analysis to examine their expression in breast cancer. While most NRs were expressed, bioinformatic analyses differentiated tumours into distinct prognostic groups that were validated by analyzing public microarray data sets. Although ERα and progesterone receptor were dominant in distinguishing prognostic groups, other NR strengthened these groups. Clustering analysis identified several family members with potential importance in breast cancer. Specifically, RORγ is identified as being co-expressed with ERα, whilst several NRs are preferentially expressed in ERα-negative disease, with TLX expression being prognostic in this subtype. Functional studies demonstrated the importance of TLX in regulating growth and invasion in ERα-negative breast cancer cells.
Collapse
Affiliation(s)
- Meng-Lay Lin
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Hetal Patel
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Judit Remenyi
- Division of Cancer Research, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Christopher R S Banerji
- Statistical Genomics Group, UCL Cancer Institute, University College London, London, UK.,Centre of Mathematics and Physics in Life & Experimental Sciences, University College London, London, UK
| | - Chun-Fui Lai
- Department of Surgery & Cancer, Imperial College London, London, UK
| | | | - Ylenia Lombardo
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Claudia Busonero
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Silvia Ottaviani
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Alun Passey
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Philip R Quinlan
- Dundee Cancer Centre, Clinical Research Centre, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Colin A Purdie
- Dundee Cancer Centre, Clinical Research Centre, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Lee B Jordan
- Dundee Cancer Centre, Clinical Research Centre, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Alastair M Thompson
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Oscar M Rueda
- Cancer Research UK Cambridge Institute, University of Cambridge Li Ka Shing Centre, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge Li Ka Shing Centre, Cambridge, UK
| | - Jesus Gil
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London, UK
| | | | - Frances V Fuller-Pace
- Division of Cancer Research, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Andrew E Teschendorff
- Statistical Genomics Group, UCL Cancer Institute, University College London, London, UK.,Centre of Mathematics and Physics in Life & Experimental Sciences, University College London, London, UK
| | - Laki Buluwela
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Simak Ali
- Department of Surgery & Cancer, Imperial College London, London, UK
| |
Collapse
|
102
|
Wei W, Schwaid AG, Wang X, Wang X, Chen S, Chu Q, Saghatelian A, Wan Y. Ligand Activation of ERRα by Cholesterol Mediates Statin and Bisphosphonate Effects. Cell Metab 2016; 23:479-91. [PMID: 26777690 PMCID: PMC4785078 DOI: 10.1016/j.cmet.2015.12.010] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 10/06/2015] [Accepted: 12/16/2015] [Indexed: 01/29/2023]
Abstract
Nuclear receptors (NRs) are key regulators of gene expression and physiology. Nearly half of all human NRs lack endogenous ligands including estrogen-related receptor α (ERRα). ERRα has important roles in cancer, metabolism, and skeletal homeostasis. Affinity chromatography of tissue lipidomes with the ERRα ligand-binding domain (LBD) and subsequent transcriptional assays identified cholesterol as an endogenous ERRα agonist. Perturbation of cholesterol biosynthesis or inhibition of ERRα revealed the interdependence of cholesterol and ERRα. In bone, the effects of cholesterol, statin, and bisphosphonate on osteoclastogenesis require ERRα; and consequently, cholesterol-induced bone loss or bisphosphonate osteoprotection is lost in ERRα knockout mice. Furthermore, statin induction of muscle toxicity and cholesterol suppression of macrophage cytokine secretion are impaired by loss or inhibition of ERRα. These findings reveal a key step in ERRα regulation and explain the actions of two highly prescribed drugs, statins and bisphosphonates.
Collapse
Affiliation(s)
- Wei Wei
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adam G Schwaid
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Xueqian Wang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xunde Wang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shili Chen
- Clayton Foundation Laboratories of Peptide Biology and Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Qian Chu
- Clayton Foundation Laboratories of Peptide Biology and Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Clayton Foundation Laboratories of Peptide Biology and Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Yihong Wan
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
103
|
Varešlija D, McBryan J, Fagan A, Redmond AM, Hao Y, Sims AH, Turnbull A, Dixon JM, Ó Gaora P, Hudson L, Purcell S, Hill ADK, Young LS. Adaptation to AI Therapy in Breast Cancer Can Induce Dynamic Alterations in ER Activity Resulting in Estrogen-Independent Metastatic Tumors. Clin Cancer Res 2016; 22:2765-77. [PMID: 26763249 DOI: 10.1158/1078-0432.ccr-15-1583] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/20/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Acquired resistance to aromatase inhibitor (AI) therapy is a major clinical problem in the treatment of breast cancer. The detailed mechanisms of how tumor cells develop this resistance remain unclear. Here, the adapted function of estrogen receptor (ER) to an estrogen-depleted environment following AI treatment is reported. EXPERIMENTAL DESIGN Global ER chromatin immuno-precipitation (ChIP)-seq analysis of AI-resistant cells identified steroid-independent ER target genes. Matched patient tumor samples, collected before and after AI treatment, were used to assess ER activity. RESULTS Maintained ER activity was observed in patient tumors following neoadjuvant AI therapy. Genome-wide ER-DNA-binding analysis in AI-resistant cell lines identified a subset of classic ligand-dependent ER target genes that develop steroid independence. The Kaplan-Meier analysis revealed a significant association between tumors, which fail to decrease this steroid-independent ER target gene set in response to neoadjuvant AI therapy, and poor disease-free survival and overall survival (n = 72 matched patient tumor samples, P = 0.00339 and 0.00155, respectively). The adaptive ER response to AI treatment was highlighted by the ER/AIB1 target gene, early growth response 3 (EGR3). Elevated levels of EGR3 were detected in endocrine-resistant local disease recurrent patient tumors in comparison with matched primary tissue. However, evidence from distant metastatic tumors demonstrates that the ER signaling network may undergo further adaptations with disease progression as estrogen-independent ER target gene expression is routinely lost in established metastatic tumors. CONCLUSIONS Overall, these data provide evidence of a dynamic ER response to endocrine treatment that may provide vital clues for overcoming the clinical issue of therapy resistance. Clin Cancer Res; 22(11); 2765-77. ©2016 AACR.
Collapse
Affiliation(s)
- Damir Varešlija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jean McBryan
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ailís Fagan
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Aisling M Redmond
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Andrew H Sims
- University of Edinburgh Cancer Research Centre, Carrington Crescent, Edinburgh, EH4 2XU, United Kingdom
| | - Arran Turnbull
- University of Edinburgh Cancer Research Centre, Carrington Crescent, Edinburgh, EH4 2XU, United Kingdom
| | - J M Dixon
- University of Edinburgh Cancer Research Centre, Carrington Crescent, Edinburgh, EH4 2XU, United Kingdom
| | - Peadar Ó Gaora
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Lance Hudson
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Siobhan Purcell
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Arnold D K Hill
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Leonie S Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| |
Collapse
|
104
|
Mocanu MM, Nagy P, Szöllősi J. Chemoprevention of Breast Cancer by Dietary Polyphenols. Molecules 2015; 20:22578-620. [PMID: 26694341 PMCID: PMC6332464 DOI: 10.3390/molecules201219864] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
Abstract
The review will discuss in detail the effects of polyphenols on breast cancer, including both the advantages and disadvantages of the applications of these natural compounds. First, we focus on the characterization of the main classes of polyphenols and then on in vitro and in vivo experiments carried out in breast cancer models. Since the therapeutic effects of the administration of a single type of polyphenol might be limited because of the reduced bioavailability of these drugs, investigations on combination of several polyphenols or polyphenols with conventional therapy will also be discussed. In addition, we present recent data focusing on clinical trials with polyphenols and new approaches with nanoparticles in breast cancer. Besides the clinical and translational findings this review systematically summarizes our current knowledge about the molecular mechanisms of anti-cancer effects of polyphenols, which are related to apoptosis, cell cycle regulation, plasma membrane receptors, signaling pathways and epigenetic mechanisms. At the same time the effects of polyphenols on primary tumor, metastasis and angiogenesis in breast cancer are discussed. The increasing enthusiasm regarding the combination of polyphenols and conventional therapy in breast cancer might lead to additional efforts to motivate further research in this field.
Collapse
Affiliation(s)
- Maria-Magdalena Mocanu
- Department of Biophysics, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Péter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| |
Collapse
|
105
|
TELANG NITIN. Putative cancer-initiating stem cells in cell culture models for molecular subtypes of clinical breast cancer. Oncol Lett 2015; 10:3840-3846. [PMID: 26788219 PMCID: PMC4665642 DOI: 10.3892/ol.2015.3780] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/09/2015] [Indexed: 02/07/2023] Open
Abstract
Cancer-initiating stem cells (CISC) represent a minor subpopulation of heterogeneous breast cancer. CISC are responsible for the acquired resistance to conventional chemoendocrine therapy and eventual relapse observed in patients with breast cancer. Certain molecular subtypes of clinical breast cancer that exhibit differential expression of genes coding for hormone and growth factor receptors differ in their response to conventional chemoendocrine therapy and targeted therapeutic inhibitors. Thus, the development of reliable cell culture models for CISC may provide a valuable experimental approach for the study of stem cell-targeted therapy for the treatment of breast cancer. The present study utilized optimized cell culture systems as experimental models for different molecular subtypes of clinical breast cancer, including luminal A, human epidermal growth factor receptor (HER)-2-enriched and triple negative breast cancer. Biomarker end points, including control of homeostatic growth, cancer risk and drug resistance, were quantitatively analyzed in the selected models. The results of the analyses indicated that, compared with the non-tumorigenic controls, the cell models representing the aforementioned molecular subtypes of clinical breast cancer exhibited aberrant cell cycle progression, downregulated cellular apoptosis and loss of control of homeostatic growth, as evidenced by hyperproliferation. Additionally, these models displayed persistent cancer risk, as indicated by their high incidence and frequency of anchorage-independent (AI) colony formation in vitro and their tumor development capacity in vivo. Furthermore, in the presence of maximum cytostatic drug concentrations, the drug-resistant phenotypes isolated from the parental drug-sensitive cell lines representing luminal A, HER-2-enriched and triple negative breast cancer exhibited an 11.5, 5.0 and 6.2 fold increase in cell growth, and a 5.6, 5.4 and 4.4 fold increase in the number of AI colonies, respectively, compared with the drug-sensitive controls. Collectively, the data of the present study demonstrated the presence of putative CISC in these breast cancer models.
Collapse
Affiliation(s)
- NITIN TELANG
- Cancer Prevention Research Program, Palindrome Liaisons Consultants, Montvale, NJ 07645-1559, USA
| |
Collapse
|
106
|
Transcriptomic characterization of fibrolamellar hepatocellular carcinoma. Proc Natl Acad Sci U S A 2015; 112:E5916-25. [PMID: 26489647 DOI: 10.1073/pnas.1424894112] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Fibrolamellar hepatocellular carcinoma (FLHCC) tumors all carry a deletion of ∼ 400 kb in chromosome 19, resulting in a fusion of the genes for the heat shock protein, DNAJ (Hsp40) homolog, subfamily B, member 1, DNAJB1, and the catalytic subunit of protein kinase A, PRKACA. The resulting chimeric transcript produces a fusion protein that retains kinase activity. No other recurrent genomic alterations have been identified. Here we characterize the molecular pathogenesis of FLHCC with transcriptome sequencing (RNA sequencing). Differential expression (tumor vs. adjacent normal tissue) was detected for more than 3,500 genes (log2 fold change ≥ 1, false discovery rate ≤ 0.01), many of which were distinct from those found in hepatocellular carcinoma. Expression of several known oncogenes, such as ErbB2 and Aurora Kinase A, was increased in tumor samples. These and other dysregulated genes may serve as potential targets for therapeutic intervention.
Collapse
|
107
|
Ochieng J, Nangami GN, Ogunkua O, Miousse IR, Koturbash I, Odero-Marah V, McCawley LJ, Nangia-Makker P, Ahmed N, Luqmani Y, Chen Z, Papagerakis S, Wolf GT, Dong C, Zhou BP, Brown DG, Colacci AM, Hamid RA, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Al-Temaimi R, Al-Mulla F, Bisson WH, Eltom SE. The impact of low-dose carcinogens and environmental disruptors on tissue invasion and metastasis. Carcinogenesis 2015; 36 Suppl 1:S128-59. [PMID: 26106135 DOI: 10.1093/carcin/bgv034] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.
Collapse
Affiliation(s)
- Josiah Ochieng
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Gladys N Nangami
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Olugbemiga Ogunkua
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Valerie Odero-Marah
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Lisa J McCawley
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Nuzhat Ahmed
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Yunus Luqmani
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Silvana Papagerakis
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Gregory T Wolf
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Chenfang Dong
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Binhua P Zhou
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
| | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - A Ivana Scovassi
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Hosni K Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
| | - Rabeah Al-Temaimi
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Fahd Al-Mulla
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Sakina E Eltom
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| |
Collapse
|
108
|
Wang X, Sang X, Diorio C, Lin SX, Doillon CJ. In vitro interactions between mammary fibroblasts (Hs 578Bst) and cancer epithelial cells (MCF-7) modulate aromatase, steroid sulfatase and 17β-hydroxysteroid dehydrogenases. Mol Cell Endocrinol 2015; 412:339-48. [PMID: 26044867 DOI: 10.1016/j.mce.2015.05.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 05/16/2015] [Accepted: 05/26/2015] [Indexed: 12/21/2022]
Abstract
Our objectives were to investigate the interactions between mammary cancer epithelial cells (MCF-7) and stromal cells (Hs-578Bst) at the level of the expression and inhibition of steroidogenesis enzymes by using monolayer and three dimensional co-culture models. Expressions of steroidogenesis enzymes and E2/DHT conversions in co-cultured MCF-7 and Hs-578Bst cells as well as the effects of aromatase inhibitor combined to steroid sulfatase (STS) and 17β-hydroxysteroid dehydrogenases (17βHSDs) inhibitors were evaluated. 17β-HSD type 7 was mostly modulated in MCF-7 cells whereas aromatase was mostly regulated in Hs578Bst cells thereby increasing E2 conversion and MCF-7 cell growth. A combination of inhibitors toward aromatase, STS and 17β-HSD7, was found to be the most significant treatment in decreasing E2 and elevating DHT thus inhibiting MCF-7 cell proliferation and spheroid-like cancer cell aggregation in collagen gel. The interactions between those cells modulated E2 formation in paracrine/intracrine manners by synergistically regulating aromatase, 17β-HSD7 and STS. Among tumor-associated cells, stromal fibroblasts may participate in intratumoral E2 deposition; therefore promoting breast cancer cell growth.
Collapse
Affiliation(s)
- Xiaoqiang Wang
- Research Center of the CHU de Québec, CHUL, 2705, Boulevard Laurier, Quebec City, G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, 1050, Avenue de la Médecine, Quebec City, G1V OA6, Canada
| | - Xiaoye Sang
- Research Center of the CHU de Québec, CHUL, 2705, Boulevard Laurier, Quebec City, G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, 1050, Avenue de la Médecine, Quebec City, G1V OA6, Canada
| | - Caroline Diorio
- Centre des Maladie du Sein Deschênes-Fabia, Hôpital du Saint-Sacrement, 1050, Chemin Sainte-Foy, Quebec City, G1S 4L8, Canada; Department of Social and Preventive Medicine, Faculty of Medicine, Laval University, 1050, Avenue de la Médecine, Quebec City, G1V OA6, Canada
| | - Sheng-Xiang Lin
- Research Center of the CHU de Québec, CHUL, 2705, Boulevard Laurier, Quebec City, G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, 1050, Avenue de la Médecine, Quebec City, G1V OA6, Canada
| | - Charles J Doillon
- Research Center of the CHU de Québec, CHUL, 2705, Boulevard Laurier, Quebec City, G1V 4G2, Canada; Department of Surgery, Faculty of Medicine, Laval University, 1050, Avenue de la Médecine, Quebec City, G1V OA6, Canada.
| |
Collapse
|
109
|
Thomas C, Gustafsson JÅ. Estrogen receptor mutations and functional consequences for breast cancer. Trends Endocrinol Metab 2015; 26:467-76. [PMID: 26183887 DOI: 10.1016/j.tem.2015.06.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 02/07/2023]
Abstract
A significant number of estrogen receptor α (ERα)-positive breast tumors develop resistance to endocrine therapy and recur with metastatic disease. Several mechanisms of endocrine resistance have been proposed, including genetic alterations that lead to ERs with altered protein sequence. By altering the conformation of the protein and increasing the interaction with coactivators, point mutations in ESR1, the gene encoding ERα, promote an active form of the receptor in the absence of hormone that assists tumor cells to evade hormonal treatments. Recent studies have confirmed that ESR1 point mutations frequently occur in metastatic breast tumors that are refractory to endocrine therapy, and suggest the development of novel strategies that may be more effective in controlling ER signaling and benefit patients with recurrent and metastatic disease.
Collapse
Affiliation(s)
- Christoforos Thomas
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Boulevard, Houston, TX 77204, USA.
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Boulevard, Houston, TX 77204, USA.
| |
Collapse
|
110
|
Wu VS, Kanaya N, Lo C, Mortimer J, Chen S. From bench to bedside: What do we know about hormone receptor-positive and human epidermal growth factor receptor 2-positive breast cancer? J Steroid Biochem Mol Biol 2015; 153:45-53. [PMID: 25998416 PMCID: PMC4568143 DOI: 10.1016/j.jsbmb.2015.05.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/08/2015] [Accepted: 05/12/2015] [Indexed: 12/11/2022]
Abstract
Breast cancer is a heterogeneous disease. Thanks to extensive efforts from research scientists and clinicians, treatment for breast cancer has advanced into the era of targeted medicine. With the use of several well-established biomarkers, such as hormone receptors (HRs) (i.e., estrogen receptor [ER] and progesterone receptor [PgR]) and human epidermal growth factor receptor-2 (HER2), breast cancer patients can be categorized into multiple subgroups with specific targeted treatment strategies. Although therapeutic strategies for HR-positive (HR+) HER2-negative (HER2-) breast cancer and HR-negative (HR-) HER2-positive (HER2+) breast cancer are well-defined, HR+ HER2+ breast cancer is still an overlooked subgroup without tailored therapeutic options. In this review, we have summarized the molecular characteristics, etiology, preclinical tools and therapeutic options for HR+ HER2+ breast cancer. We hope to raise the attention of both the research and the medical community on HR+ HER2+ breast cancer, and to advance patient care for this subtype of disease.
Collapse
Affiliation(s)
- Victoria Shang Wu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiao Lo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Joanne Mortimer
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Medical Center Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States.
| |
Collapse
|
111
|
Sawesi S, Carpenter JS, Jones J. Reasons for nonadherence to tamoxifen and aromatase inhibitors for the treatment of breast cancer: a literature review. Clin J Oncol Nurs 2015; 18:E50-7. [PMID: 24867124 DOI: 10.1188/14.cjon.e50-e57] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Despite improved breast cancer survival rates with the use of tamoxifen and aromatase inhibitors, patients remain at risk for cancer recurrence and mortality because of nonadherence to medication. The objective of this review was to identify factors associated with nonadherence among patients with breast cancer. Electronic databases were searched for studies, and potentially relevant studies were retrieved and assessed for eligibility. Potential factors associated with nonadherence were identified, and they included patient-related factors (e.g., patient beliefs and knowledge, fear of adverse effects, forgetfulness, smoking, age, race), therapy-related factors (e.g., duration, side effects, additional prescribed medications, treatment interfering with lifestyle), healthcare system factors (e.g., patient/provider relationships), socioeconomic factors (e.g., medication costs, burden, scheduling problems, religion, marital status), and disease-related factors (e.g., comorbidities, stage of breast cancer). Those findings highlight the need for development of interventions to promote long-term adherence in patients with breast cancer.
Collapse
Affiliation(s)
- Suhila Sawesi
- Department of BioHealth Informatics, School of Informatics, Indiana University-Purdue University in Indianapolis
| | - Janet S Carpenter
- Center for Enhancing Quality of Life in Chronic Illness, School of Nursing, Indiana University-Purdue University in Indianapolis
| | - Josette Jones
- Department of BioHealth Informatics in the School of Informatics, Indiana University-Purdue University in Indianapolis
| |
Collapse
|
112
|
Nayak SR, Harrington E, Boone D, Hartmaier R, Chen J, Pathiraja TN, Cooper KL, Fine JL, Sanfilippo J, Davidson NE, Lee AV, Dabbs D, Oesterreich S. A Role for Histone H2B Variants in Endocrine-Resistant Breast Cancer. Discov Oncol 2015; 6:214-24. [PMID: 26113056 DOI: 10.1007/s12672-015-0230-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/09/2015] [Indexed: 12/12/2022] Open
Abstract
Acquired resistance to aromatase inhibitors (AIs) remains a major clinical problem in the treatment of estrogen receptor-positive (ER+) breast cancer. We and others have previously reported widespread changes in DNA methylation using breast cancer cell line models of endocrine resistance. Here, we show that the histone variant HIST1H2BE is hypomethylated in estrogen deprivation-resistant C4-12 and long-term estrogen-deprived (LTED) cells compared with parental MCF-7 cells. As expected, this hypomethylation associates with increased expression of HIST1H2BE in C4-12 and LTED cells. Both overexpression and downregulation of HIST1H2BE caused decreased proliferation in breast cancer cell lines suggesting the need for tightly controlled expression of this histone variant. Gene expression analysis showed varied expression of HIST1H2BE in a large panel of breast cancer cell lines, without restriction to specific molecular subtypes. Analysis of HIST1H2BE messenger RNA (mRNA) expression in ER+ AI-treated breast tumors showed significantly higher expression in resistant (n = 19) compared with sensitive (n = 37) tumors (p = 0.01). Using nanostring analysis, we measured expression of all 61 histone variants in endocrine-resistant and endocrine-sensitive tumors. We found significant overexpression of 22 variant histone genes in tumors resistant to AI therapy. In silico The Cancer Genome Atlas (TCGA) analysis showed frequent amplification of the HIST1 locus. In summary, our studies show, for the first time, that overexpression of histone variants might be important in endocrine response in ER+ breast cancer, and that overexpression is at least in part mediated via epigenetic mechanisms and amplifications. Future studies addressing endocrine response should include a potential role of these currently understudied histone variants.
Collapse
Affiliation(s)
- Shweta R Nayak
- Division of Reproductive Endocrinology, Magee-Womens Hospital, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Emily Harrington
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center (WCRC), Magee-Womens Research Institute (MWRI), University of Pittsburgh Cancer Institute (UPCI), 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - David Boone
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center (WCRC), Magee-Womens Research Institute (MWRI), University of Pittsburgh Cancer Institute (UPCI), 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Ryan Hartmaier
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center (WCRC), Magee-Womens Research Institute (MWRI), University of Pittsburgh Cancer Institute (UPCI), 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Jian Chen
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center (WCRC), Magee-Womens Research Institute (MWRI), University of Pittsburgh Cancer Institute (UPCI), 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | | | | | - Jeffrey L Fine
- Department of Pathology, Magee-Womens Hospital, UPMC, Pittsburgh, PA, USA
| | - Joseph Sanfilippo
- Division of Reproductive Endocrinology, Magee-Womens Hospital, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Nancy E Davidson
- Department of Medicine, UPCI, UPMC, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, UPCI, UPMC, Pittsburgh, PA, USA
| | - Adrian V Lee
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center (WCRC), Magee-Womens Research Institute (MWRI), University of Pittsburgh Cancer Institute (UPCI), 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - David Dabbs
- Department of Pathology, Magee-Womens Hospital, UPMC, Pittsburgh, PA, USA
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center (WCRC), Magee-Womens Research Institute (MWRI), University of Pittsburgh Cancer Institute (UPCI), 204 Craft Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
113
|
Wei W, Lewis MT. Identifying and targeting tumor-initiating cells in the treatment of breast cancer. Endocr Relat Cancer 2015; 22:R135-55. [PMID: 25876646 PMCID: PMC4447610 DOI: 10.1530/erc-14-0447] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2015] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most common cancer in women (excluding skin cancer), and it is the second leading cause of cancer-related deaths. Although conventional and targeted therapies have improved survival rates, there are still considerable challenges in treating breast cancer, including treatment resistance, disease recurrence, and metastasis. Treatment resistance can be either de novo - because of traits that tumor cells possess before treatment - or acquired - because of traits that tumor cells gain in response to treatment. A recently proposed mechanism of de novo resistance invokes the existence of a specialized subset of cancer cells defined as tumor-initiating cells (TICs), or cancer stem cells (CSCs). TICs have the capacity to self-renew and to generate new tumors that consist entirely of clonally derived cell types present in the parental tumor. There are data to suggest that TICs are resistant to many conventional cancer therapies and that they can survive treatment in spite of dramatic shrinkage of the tumor. Residual TICs can then eventually regrow, which results in disease relapse. It has also been hypothesized that TIC may be responsible for metastatic disease. If these hypotheses are correct, targeting TICs may be imperative for achieving a cure. In the present review, we discuss evidence for breast TICs and their apparent resistance to conventional chemotherapy and radiotherapy as well as to various targeted therapies. We also address the potential impact of breast TIC plasticity and metastatic potential on therapeutic strategies. Finally, we describe several genes and signaling pathways that appear to be important for TIC function and may represent promising therapeutic targets.
Collapse
Affiliation(s)
- Wei Wei
- Baylor College of MedicineLester and Sue Smith Breast Center, Houston, Texas, USADepartments of Molecular and Cellular BiologyRadiologyBaylor College of Medicine, One Baylor Plaza, BCM600, Room N1210, Houston, Texas 77030, USA Baylor College of MedicineLester and Sue Smith Breast Center, Houston, Texas, USADepartments of Molecular and Cellular BiologyRadiologyBaylor College of Medicine, One Baylor Plaza, BCM600, Room N1210, Houston, Texas 77030, USA
| | - Michael T Lewis
- Baylor College of MedicineLester and Sue Smith Breast Center, Houston, Texas, USADepartments of Molecular and Cellular BiologyRadiologyBaylor College of Medicine, One Baylor Plaza, BCM600, Room N1210, Houston, Texas 77030, USA Baylor College of MedicineLester and Sue Smith Breast Center, Houston, Texas, USADepartments of Molecular and Cellular BiologyRadiologyBaylor College of Medicine, One Baylor Plaza, BCM600, Room N1210, Houston, Texas 77030, USA Baylor College of MedicineLester and Sue Smith Breast Center, Houston, Texas, USADepartments of Molecular and Cellular BiologyRadiologyBaylor College of Medicine, One Baylor Plaza, BCM600, Room N1210, Houston, Texas 77030, USA
| |
Collapse
|
114
|
Generali D, Berruti A, Cappelletti MR, Zanotti L, Brugnoli G, Forti M, Bedussi F, Vailati ME, Milani M, Strina C, Ardine M, Aguggini S, Allevi G, Ferrero G, Bertoni R, Bottini A, Harris AL, Fox SB. Effect of Primary Letrozole Treatment on Tumor Expression of mTOR and HIF-1α and Relation to Clinical Response. J Natl Cancer Inst Monogr 2015; 2015:64-6. [PMID: 26063890 DOI: 10.1093/jncimonographs/lgv018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
INTRODUCTION Recently the combination of the mammalian target of rapamycin (mTOR) inhibitor everolimus and the aromatase inhibitor exemestane has been shown to double the progression-free survival rate in advanced breast cancer. However, the effect of the interrelated pathways of hypoxia-inducible factor-1α (HIF-1α) and mTOR signaling, both of which are associated with a more aggressive breast cancer phenotype and endocrine resistance, on response in the neoadjuvant setting is unknown. We, therefore, have investigated the influence of these pathways with the aim of better defining those patients most likely to benefit from an endocrine-based therapy associated with/without mTOR inhibitors. PATIENTS AND METHODS A total of 107 women with T2-4 N0-1 and estrogen receptor-positive breast cancer were randomly assigned to 6 months of primary letrozole (2.5 mg/daily) (LET) or LET plus oral "metronomic" cyclophosphamide (50mg/daily) (LET-CYC). Phospo-mTOR and HIF-1α were evaluated in tumor specimens collected before and after treatment using a tissue microarray format. RESULTS LET-based therapy induced a downregulation of phospho-mTOR and HIF-1α expression (P = .0001 and P < .004, respectively). The reduction of HIF-1α expression observed was positively correlated with phospho-mTOR reduction (P < .03); however, no treatment interaction between the two proteins was detected. HIF-1α expression was significantly modulated by the treatment (P < .004) with a reduction both in the LET arm (45%, n = 36/80) (P = .05) and LET-CYC arm (55%, n = 44/80) (P = .04). HIF-1α reduction showed a relationship with clinical response confined in LET arm only (P < .03). CONCLUSIONS In this neoadjuvant population, LET was able to modulate the phospho-mTOR and HIF-1α pathways and may define a subpopulation of nonresponders who may be most likely to benefit from mTOR inhibitors.
Collapse
Affiliation(s)
- Daniele Generali
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF).
| | - Alfredo Berruti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Maria Rosa Cappelletti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Laura Zanotti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Giulia Brugnoli
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Michela Forti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Francesca Bedussi
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Maria Elena Vailati
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Manuela Milani
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Carla Strina
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Mara Ardine
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Sergio Aguggini
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Giovanni Allevi
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Giuseppina Ferrero
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Ramona Bertoni
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Alberto Bottini
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Adrian L Harris
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Stephen B Fox
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| |
Collapse
|
115
|
Diverse roles of SIRT1 in cancer biology and lipid metabolism. Int J Mol Sci 2015; 16:950-65. [PMID: 25569080 PMCID: PMC4307284 DOI: 10.3390/ijms16010950] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 12/24/2014] [Indexed: 12/18/2022] Open
Abstract
SIRT1, an NAD+-dependent deacetylase, has been described in the literature as a major player in the regulation of cellular stress responses. Its expression has been shown to be altered in cancer cells, and it targets both histone and non-histone proteins for deacetylation and thereby alters metabolic programs in response to diverse physiological stress. Interestingly, many of the metabolic pathways that are influenced by SIRT1 are also altered in tumor development. Not only does SIRT1 have the potential to regulate oncogenic factors, it also orchestrates many aspects of metabolism and lipid regulation and recent reports are beginning to connect these areas. SIRT1 influences pathways that provide an alternative means of deriving energy (such as fatty acid oxidation and gluconeogenesis) when a cell encounters nutritive stress, and can therefore lead to altered lipid metabolism in various pathophysiological contexts. This review helps to show the various connections between SIRT1 and major pathways in cellular metabolism and the consequence of SIRT1 deregulation on carcinogenesis and lipid metabolism.
Collapse
|
116
|
Abstract
Most recent investigations into cancer etiology have identified a key role played by epigenetics. Specifically, aberrant DNA and histone modifications which silence tumor suppressor genes or promote oncogenes have been demonstrated in multiple cancer models. While the role of epigenetics in several solid tumor cancers such as colorectal cancer are well established, there is emerging evidence that epigenetics also plays a critical role in breast and prostate cancer. In breast cancer, DNA methylation profiles have been linked to hormone receptor status and tumor progression. Similarly in prostate cancer, epigenetic patterns have been associated with androgen receptor status and response to therapy. The regulation of key receptor pathways and activities which affect clinical therapy treatment options by epigenetics renders this field high priority for elucidating mechanisms and potential targets. A new set of methylation arrays are now available to screen epigenetic changes and provide the cutting-edge tools needed to perform such investigations. The role of nutritional interventions affecting epigenetic changes particularly holds promise. Ultimately, determining the causes and outcomes from epigenetic changes will inform translational applications for utilization as biomarkers for risk and prognosis as well as candidates for therapy.
Collapse
Affiliation(s)
- Yanyuan Wu
- Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, USA
| | - Marianna Sarkissyan
- Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, USA
- Corresponding Author Contact Information: Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 East 120th Street, Los Angeles, CA 90059, USA. Tele: 323-563-4853. Fax: 323-563-4859 ;
| |
Collapse
|
117
|
Pan MH, Chiou YS, Chen LH, Ho CT. Breast cancer chemoprevention by dietary natural phenolic compounds: Specific epigenetic related molecular targets. Mol Nutr Food Res 2014; 59:21-35. [DOI: 10.1002/mnfr.201400515] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/08/2014] [Accepted: 11/03/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Min-Hsiung Pan
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
- Department of Medical Research, China Medical University Hospital; China Medical University; Taichung Taiwan
| | - Yi-Siou Chiou
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
| | - Li-Hua Chen
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University; New Brunswick; NJ USA
| |
Collapse
|
118
|
Karam M, Bièche I, Legay C, Vacher S, Auclair C, Ricort JM. Protein kinase D1 regulates ERα-positive breast cancer cell growth response to 17β-estradiol and contributes to poor prognosis in patients. J Cell Mol Med 2014; 18:2536-52. [PMID: 25287328 PMCID: PMC4302658 DOI: 10.1111/jcmm.12322] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/07/2014] [Indexed: 12/21/2022] Open
Abstract
About 70% of human breast cancers express and are dependent for growth on estrogen receptor α (ERα), and therefore are sensitive to antiestrogen therapies. However, progression to an advanced, more aggressive phenotype is associated with acquisition of resistance to antiestrogens and/or invasive potential. In this study, we highlight the role of the serine/threonine-protein kinase D1 (PKD1) in ERα-positive breast cancers. Growth of ERα-positive MCF-7 and MDA-MB-415 human breast cancer cells was assayed in adherent or anchorage-independent conditions in cells overexpressing or depleted for PKD1. PKD1 induces cell growth through both an ERα-dependent manner, by increasing ERα expression and cell sensitivity to 17β-estradiol, and an ERα-independent manner, by reducing cell dependence to estrogens and conferring partial resistance to antiestrogen ICI 182,780. PKD1 knockdown in MDA-MB-415 cells strongly reduced estrogen-dependent and independent invasion. Quantification of PKD1 mRNA levels in 38 cancerous and non-cancerous breast cell lines and in 152 ERα-positive breast tumours from patients treated with adjuvant tamoxifen showed an association between PKD1 and ERα expression in 76.3% (29/38) of the breast cell lines tested and a strong correlation between PKD1 expression and invasiveness (P < 0.0001). In tamoxifen-treated patients, tumours with high PKD1 mRNA levels (n = 77, 50.66%) were significantly associated with less metastasis-free survival than tumours with low PKD1 mRNA expression (n = 75, 49.34%; P = 0.031). Moreover, PKD1 mRNA levels are strongly positively associated with EGFR and vimentin levels (P < 0.0000001). Thus, our study defines PKD1 as a novel attractive prognostic factor and a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Manale Karam
- Laboratoire de Biologie et de Pharmacologie Appliquée, UMR 8113 CNRS, Ecole Normale Supérieure de Cachan, Cachan, France
| | | | | | | | | | | |
Collapse
|
119
|
Abstract
Around 70% of all breast cancers are estrogen receptor alpha positive and hence their development is highly dependent on estradiol. While the invention of endocrine therapies has revolusioned the treatment of the disease, resistance to therapy eventually occurs in a large number of patients. This paper seeks to illustrate and discuss the complexity and heterogeneity of the mechanisms which underlie resistance and the approaches proposed to combat them. It will also focus on the use and development of methods for predicting which patients are likely to develop resistance.
Collapse
|
120
|
Hasson SP, Rubinek T, Ryvo L, Wolf I. Endocrine resistance in breast cancer: focus on the phosphatidylinositol 3-kinase/akt/mammalian target of rapamycin signaling pathway. ACTA ACUST UNITED AC 2014; 8:248-55. [PMID: 24415977 DOI: 10.1159/000354757] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Breast cancer is the most common cancer among women. Up to 75% of breast cancers express the estrogen receptor (ER)α and/or the progesterone receptor (PR). Patients with hormone receptor-positive metastatic breast cancer are typically treated with endocrine therapy. Yet, not all patients with metastatic breast cancer respond to endocrine treatments and are considered to have primary (de novo) resistance. Furthermore, all patients who initially respond to endocrine treatment will eventually develop acquired resistance. Several mechanisms have been linked to the development of endocrine resistance, including reduced expression of ERα, altered regulation of the ER pathway, and activation of various growth factor signaling pathways, among them the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway. This pathway is involved in critical processes including cell survival, proliferation, and angiogenesis, and plays a central role in breast cancer development. Recent laboratory and clinical data implicate this pathway as mediating endocrine resistance, and agents directed against critical components of this pathway are either already approved for clinical use in breast cancer patients or are currently being tested in clinical trials. In this review, we describe the interaction between the PI3K/Akt/mTOR pathway and the ER cascade, its role in mediating endocrine resistance, and the clinical implications of this interaction.
Collapse
Affiliation(s)
- Shira Peleg Hasson
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Israel ; Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Tami Rubinek
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Israel
| | - Larysa Ryvo
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Israel
| | - Ido Wolf
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Israel ; Sackler Faculty of Medicine, Tel Aviv University, Israel
| |
Collapse
|
121
|
Kim J, Jayaprakasha GK, Patil BS. Obacunone exhibits anti-proliferative and anti-aromatase activity in vitro by inhibiting the p38 MAPK signaling pathway in MCF-7 human breast adenocarcinoma cells. Biochimie 2014; 105:36-44. [PMID: 24927687 DOI: 10.1016/j.biochi.2014.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 06/02/2014] [Indexed: 01/08/2023]
Abstract
Overexpression of the aromatase enzyme CYP19 has been implicated in the onset of estrogen-dependent breast carcinogenesis. Obacunone, a natural compound present in citrus fruits, has been demonstrated for various biological activities including anti-cancer and anti-inflammatory properties. In the present study, we have isolated obacunone and obacunone glucoside (OG) from lemon seeds, then fractionated these compounds using chromatographic techniques and characterized them by HPLC, LC-MS, and 2D NMR spectral analysis. To investigate the mechanism of anti-cancer and anti-aromatase activities of limonoids, their cytotoxic effect was tested on human breast cancer (MCF-7) and non-malignant (MCF-12F) breast cells. MTT assays confirmed that obacunone was strongly inhibited MCF-7 cell proliferation without affecting non-malignant breast cells. Treatment with obacunone increased apoptosis by up-regulating expression of the pro-apoptotic protein Bax and down-regulating the anti-apoptotic protein Bcl2, as well as inducing G1 cell cycle arrest. In addition, obacunone significantly inhibited aromatase activity in an in vitro enzyme assay. Exposure of MCF-7 breast cancer cells to obacunone down-regulated expression of inflammatory molecules including nuclear factor-kappa B (NF-κB) and cyclooxygenase-2 (COX-2). Furthermore, we found that obacunone inhibited COX-2 and NF-κB by activation of the p38 mitogen-activated protein kinase (MAPK). Finally, the uptake level of obacunone into MCF-7 cells was measured by HPLC and its structure was confirmed by LC-HR-MS. This study demonstrated that obacunone may have the potential to prevent estrogen-responsive breast cancer through inhibition of the aromatase enzyme and inflammatory pathways, as well as activation of apoptosis.
Collapse
Affiliation(s)
- Jinhee Kim
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University, College Station, TX 77845-2119, USA
| | - G K Jayaprakasha
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University, College Station, TX 77845-2119, USA.
| | - Bhimanagouda S Patil
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University, College Station, TX 77845-2119, USA.
| |
Collapse
|
122
|
Two natural products, trans-phytol and (22E)-ergosta-6,9,22-triene-3β,5α,8α-triol, inhibit the biosynthesis of estrogen in human ovarian granulosa cells by aromatase (CYP19). Toxicol Appl Pharmacol 2014; 279:23-32. [PMID: 24853760 DOI: 10.1016/j.taap.2014.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/08/2014] [Accepted: 05/13/2014] [Indexed: 10/25/2022]
Abstract
Aromatase is the only enzyme in vertebrates to catalyze the biosynthesis of estrogens. Although inhibitors of aromatase have been developed for the treatment of estrogen-dependent breast cancer, the whole-body inhibition of aromatase causes severe adverse effects. Thus, tissue-selective aromatase inhibitors are important for the treatment of estrogen-dependent cancers. In this study, 63 natural products with diverse structures were examined for their effects on estrogen biosynthesis in human ovarian granulosa-like KGN cells. Two compounds-trans-phytol (SA-20) and (22E)-ergosta-6,9,22-triene-3β,5α,8α-triol (SA-48)-were found to potently inhibit estrogen biosynthesis (IC50: 1μM and 0.5μM, respectively). Both compounds decreased aromatase mRNA and protein expression levels in KGN cells, but had no effect on the aromatase catalytic activity in aromatase-overexpressing HEK293A cells and recombinant expressed aromatase. The two compounds decreased the expression of aromatase promoter I.3/II. Neither compound affected intracellular cyclic AMP (cAMP) levels, but they inhibited the phosphorylation or protein expression of cAMP response element-binding protein (CREB). The effects of these two compounds on extracellular regulated kinase (ERK), c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinases (MAPKs), and AKT/phosphoinositide 3-kinase (PI3K) pathway were examined. Inhibition of p38 MAPK could be the mechanism underpinning the actions of these compounds. Our results suggests that natural products structurally similar to SA-20 and SA-48 may be a new source of tissue-selective aromatase modulators, and that p38 MAPK is important in the basal control of aromatase in ovarian granulosa cells. SA-20 and SA-48 warrant further investigation as new pharmaceutical tools for the prevention and treatment of estrogen-dependent cancers.
Collapse
|
123
|
Sun Y, Chung HH, Woo ARE, Lin VCL. Protein arginine methyltransferase 6 enhances ligand-dependent and -independent activity of estrogen receptor α via distinct mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2067-78. [PMID: 24742914 DOI: 10.1016/j.bbamcr.2014.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 11/19/2022]
Abstract
Recent studies reported that protein arginine methyltransferase 6 (PRMT6) enhances estrogen-induced activity of estrogen receptor α (ERα) and dysfunction of PRMT6 is associated with overall better survival for ERα-positive breast cancer patients. However, it is unclear how PRMT6 promotes ERα activity. Here we report that PRMT6 specifically interacts with ERα at its ligand-binding domain. PRMT6 also methylates ERα both in vitro and in vivo. In addition to enhancing estrogen-induced ERα activity, PRMT6 over-expression up-regulates estrogen-independent activity of ERα and PRMT6 gene silencing in MCF7 cells inhibits ligand-independent ERα activation. More interestingly, the effect of PRMT6 on the ligand-independent ERα activity does not require its methyltransferase activity. Instead, PRMT6 competes with Hsp90 for ERα binding: PRMT6 and Hsp90 bindings to ERα are mutually exclusive and PRMT6 over-expression reduces ERα interaction with Hsp90. In conclusion, PRMT6 requires its methyltransferase activity to enhance ERα's ligand-induced activity, but its effect on ligand-independent activity is likely mediated through competing with Hsp90 for binding to the C-terminal domain of ERα. PRMT6-ERα interaction would prevent ERα-Hsp90 association. Since Hsp90 and associated chaperones serve to maintain ERα conformation for ligand-binding yet functionally inactive, inhibition of ERα-Hsp90 interaction would relieve ERα from the constraint of chaperone complex.
Collapse
Affiliation(s)
- Yang Sun
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Hwa Hwa Chung
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Amanda Rui En Woo
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Valerie C-L Lin
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
124
|
Gao Q, Patani N, Dunbier AK, Ghazoui Z, Zvelebil M, Martin LA, Dowsett M. Effect of aromatase inhibition on functional gene modules in estrogen receptor-positive breast cancer and their relationship with antiproliferative response. Clin Cancer Res 2014; 20:2485-94. [PMID: 24634384 DOI: 10.1158/1078-0432.ccr-13-2602] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate potential associations between gene modules representing key biologic processes and response to aromatase inhibitors (AI) in estrogen receptor-positive (ER(+)) breast cancer. PATIENTS AND METHODS Paired gene expression and Ki67 protein expression were available from 69 postmenopausal women with ER(+) early breast cancer, at baseline and 2 weeks post-anastrozole treatment, in the presurgical setting. Functional gene modules (n = 26) were retrieved from published studies and their module scores were computed before and after elimination of proliferation-associated genes (PAG). Ki67 and module scores were assessed at baseline and 2 weeks post-anastrozole. Unsupervised clustering was used to assess associations between modules and Ki67. RESULTS Proliferation-based modules were highly correlated with Ki67 expression both pretreatment and on-treatment. At baseline with and without PAGs, Ki67 expression was significantly inversely correlated with ERG, ESR1.2, SET, and PIK3CA modules. Modules measuring estrogen signaling strongly predicted antiproliferative response to therapy with and without PAGs. Baseline expression of insulin-like growth factor-1 (IGF-I) module predicted a poor change in Ki67-implicating genes within the module as involved in de novo resistance to AIs. High expression of Immune.2.STAT1 module pretreatment predicted poor antiproliferative response to therapy. A significant association between estrogen-regulated genes modules (ESR1, ESR1-2, SET, and ERG) was evident post AI. CONCLUSIONS Multiple processes and pathways are affected by AI treatment in ER(+) breast cancer. Modules closely associated with ESR1 expression were predictive of good antiproliferative response to AIs, but modules representing immune activity and IGF-I/MAPK were predictive of poor Ki67 response, supporting their therapeutic targeting in combination with AIs.
Collapse
Affiliation(s)
- Qiong Gao
- Authors' Affiliations: Breakthrough Breast Cancer Research Centre, Institute of Cancer Research; Academic Department of Biochemistry, Royal Marsden Foundation Trust, London, United Kingdom; and Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | | | |
Collapse
|
125
|
Patani N, Martin LA. Understanding response and resistance to oestrogen deprivation in ER-positive breast cancer. Mol Cell Endocrinol 2014; 382:683-694. [PMID: 24121024 DOI: 10.1016/j.mce.2013.09.038] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/30/2013] [Accepted: 09/30/2013] [Indexed: 01/01/2023]
Abstract
Oestrogens (E) and oestrogen receptor alpha (ERα) play fundamental roles in the development and progression of more than three-quarters of breast cancers (BC). The ability to influence the natural history of BC by hormonal manipulation is well established and endocrine therapies represent the cornerstone of systemic management for women with ERα-positive disease. Endocrine agents abrogate oestrogenic signalling through distinct and incompletely overlapping mechanisms, either impeding the transcriptional activity of ERα or diminishing E-synthesis. In post-menopausal women, E-production is chiefly attributable to the enzymatic conversion of androgens in extra-gonadal tissues by the cytochrome P-450 superfamily member aromatase. Greater understanding of steroid biosynthesis has underpinned rational drug design and pharmacological development of potent and specific aromatase inhibitors (AIs). Contemporary agents induce profound E-suppression in post-menopausal women and are first-line neo-adjuvant, adjuvant and metastatic therapies, with greater efficacy and tolerability than tamoxifen. The principal qualifier for endocrine treatment, including AIs, remains ERα expression. However, it is increasingly apparent that ERα expression is not synonymous with sensitivity to treatment and insufficient to account for the considerable heterogeneity of response. Better predictive biomarkers of de novo resistance are required to improve patient selection and identify those poor-responders who may benefit from alternative or additional systemic treatment from the outset. Among patients who do respond well initially, many relapse during their clinical course and there is also an unmet need for biomarkers of acquired resistance. The majority of women who relapse on AIs continue to express functional ERα which remains a legitimate target for second-line endocrine therapy. Understanding and overcoming acquired resistance to AIs requires a greater appreciation of ERα biology and the mechanisms though which E-dependence can be subverted. In this article, we review the impact of therapeutic E-deprivation on the natural history of ERα-positive breast cancer. Consideration is given to established and emerging biomarkers and/or determinants of response and resistance to E-deprivation. In vitro and in vivo evidence of the molecular mechanisms underpinning the transition from sensitivity to resistance are reviewed in the context of current models of ERα activity and their potential translational relevance.
Collapse
Affiliation(s)
- N Patani
- Academic Department of Biochemistry, Royal Marsden Foundation Trust, London, UK; Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JB, UK
| | - L-A Martin
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JB, UK.
| |
Collapse
|
126
|
Zhang D, Wang G, Wang Y. Transcriptional regulation prediction of antiestrogen resistance in breast cancer based on RNA polymerase II binding data. BMC Bioinformatics 2014; 15 Suppl 2:S10. [PMID: 24564526 PMCID: PMC4015922 DOI: 10.1186/1471-2105-15-s2-s10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Although endocrine therapy impedes estrogen-ER signaling pathway and thus reduces breast cancer mortality, patients remain at continued risk of relapse after tamoxifen or other endocrine therapies. Understanding the mechanisms of endocrine resistance, particularly the role of transcriptional regulation is very important and necessary. Methods We propose a two-step workflow based on linear model to investigate the significant differences between MCF7 and OHT cells stimulated by 17β-estradiol (E2) respect to regulatory transcription factors (TFs) and their interactions. We additionally compared predicted regulatory TFs based on RNA polymerase II (PolII) binding quantity data and gene expression data, which were taken from MCF7/MCF7+E2 and OHT/OHT+E2 cell lines following the same analysis workflow. Enrichment analysis concerning diseases and cell functions and regulatory pattern analysis of different motifs of the same TF also were performed. Results The results showed PolII data could provide more information and predict more recognizably important regulatory TFs. Large differences in TF regulatory mode were found between two cell lines. Through verified through GO annotation, enrichment analysis and related literature regarding these TFs, we found some regulatory TFs such as AP-1, C/EBP, FoxA1, GATA1, Oct-1 and NF-κB, maintained OHT cells through molecular interactions or signaling pathways that were different from the surviving MCF7 cells. From TF regulatory interaction network, we identified E2F, E2F-1 and AP-2 as hub-TFs in MCF7 cells; whereas, in addition to E2F and E2F-1, we identified C/EBP and Oct-1 as hub-TFs in OHT cells. Notably, we found the regulatory patterns of different motifs of the same TF were very different from one another sometimes. Conclusions We inferred some regulatory TFs, such as AP-1 and NF-κB, cooperated with ER through both genomic action and non-genomic action. The TFs that were involved in both protein-protein interactions and signaling pathways could be one of the key resistant mechanisms of endocrine therapy and thus also could be new treatment targets for endocrine resistance. Our flexible workflow could be integrated into an existing analytical framework and guide biologists to further determine underlying mechanisms in human diseases.
Collapse
|
127
|
Zhang PZ, Wang F, Yang LJ, Zhang GL. Pregnane alkaloids from Sarcococca hookeriana var. digyna. Fitoterapia 2013; 89:143-8. [PMID: 23651560 DOI: 10.1016/j.fitote.2013.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 04/19/2013] [Accepted: 04/28/2013] [Indexed: 11/12/2022]
Abstract
Fourteen pregnane-type steroidal alkaloids were isolated from the ethanolic extracts of whole Sarcococca hookeriana var. digyna plants. Their structures were elucidated on the basis of spectral data. Three of them were identified as new steroidal alkaloids: (S)-20-(N,N-dimethylamino)-16α,17α-epoxy-3β-methoxy-pregn-5-ene (1), (20S)-20-(N,N-dimethylamino)-3β-tigloylamino-5α-pregn-11β-ol (2), and (20S)-2α,4β-bis(acetoxy)-20-(N,N-dimethylamino)-3β-tigloylamino-5α-pregnane (3). Some of the isolated compounds showed estrogen biosynthesis-promoting effects in human ovarian granulosa-like KGN cells. The EC50 values for the most effective compounds, vagnine B (6) and funtumafrine C (12), were 71 μM and 67 μM, respectively.
Collapse
Affiliation(s)
- Pu-zhao Zhang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, PR China
| | | | | | | |
Collapse
|
128
|
Tan PS, Haaland B, Montero AJ, Lopes G. A meta-analysis of anastrozole in combination with fulvestrant in the first line treatment of hormone receptor positive advanced breast cancer. Breast Cancer Res Treat 2013; 138:961-5. [DOI: 10.1007/s10549-013-2495-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 03/20/2013] [Indexed: 11/29/2022]
|
129
|
Phytoestrogens in menopausal supplements induce ER-dependent cell proliferation and overcome breast cancer treatment in an in vitro breast cancer model. Toxicol Appl Pharmacol 2013; 269:132-40. [PMID: 23541764 DOI: 10.1016/j.taap.2013.03.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 03/18/2013] [Indexed: 10/27/2022]
Abstract
Breast cancer treatment by the aromatase inhibitor Letrozole (LET) or Selective Estrogen Receptor Modulator Tamoxifen (TAM) can result in the onset of menopausal symptoms. Women often try to relieve these symptoms by taking menopausal supplements containing high levels of phytoestrogens. However, little is known about the potential interaction between these supplements and breast cancer treatment, especially aromatase inhibitors. In this study, interaction of phytoestrogens with the estrogen receptor alpha and TAM action was determined in an ER-reporter gene assay (BG1Luc4E2 cells) and human breast epithelial tumor cells (MCF-7). Potential interactions with aromatase activity and LET were determined in human adrenocorticocarcinoma H295R cells. We also used the previously described H295R/MCF-7 co-culture model to study interactions with steroidogenesis and tumor cell proliferation. In this model, genistein (GEN), 8-prenylnaringenin (8PN) and four commercially available menopausal supplements all induced ER-dependent tumor cell proliferation, which could not be prevented by physiologically relevant LET and 4OH-TAM concentrations. Differences in relative effect potencies between the H295R/MCF-7 co-culture model and ER-activation in BG1Luc4E2 cells, were due to the effects of the phytoestrogens on steroidogenesis. All tested supplements and GEN induced aromatase activity, while 8PN was a strong aromatase inhibitor. Steroidogenic profiles upon GEN and 8PN exposure indicated a strong inhibitory effect on steroidogenesis in H295R cells and H295R/MCF-7 co-cultures. Based on our in vitro data we suggest that menopausal supplement intake during breast cancer treatment should better be avoided, at least until more certainty regarding the safety of supplemental use in breast cancer patients can be provided.
Collapse
|
130
|
Yang L, Lu D, Guo J, Meng X, Zhang G, Wang F. Icariin from Epimedium brevicornum Maxim promotes the biosynthesis of estrogen by aromatase (CYP19). JOURNAL OF ETHNOPHARMACOLOGY 2013; 145:715-721. [PMID: 23261485 DOI: 10.1016/j.jep.2012.11.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/05/2012] [Accepted: 11/12/2012] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedium brevicornum Maxim has long been used for the treatment of osteoporosis in China and other Asian countries. However, the mechanism behind the antiosteoporotic activity of this medicinal plant is not fully understood. AIM OF THE STUDY The present study was designed to investigate the effects of five widely used antiosteoporotic medicinal plants (Epimedium brevicornum, Cuscuta chinensis, Rhizoma drynariae, Polygonum multiflorum, and Ligustrum lucidum) on the production of estrogen, and identify the bioactive compounds responsible for the estrogen biosynthesis-promoting effect. MATERIALS AND METHODS Human ovarian granulosa-like KGN cells were used to evaluate estrogen biosynthesis, and the production of 17β-estradiol was quantified by a magnetic particle-based enzyme-linked immunosorbent assay (ELISA) kit. Further, the mRNA expression of aromatase was determined by a quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR), and the protein expression of aromatase was detected by western blotting. The activity of alkaline phosphatase (ALP) in rat osteoblastic UMR-106 cells was measured using p-nitrophenyl sodium phosphate assay. RESULTS Among the 5 antiosteoporotic medicinal plants, the extract of Epimedium brevicornum was found to significantly promote estrogen biosynthesis in KGN cells. Icariin, the major compound in Epimedium brevicornum, was identified to be the active compound for the estrogen biosynthesis-promoting effect. Icariin promoted estrogen biosynthesis in KGN cells in a concentration- and time-dependant manner and enhanced the mRNA and protein expressions of aromatase, which is the only enzyme for the conversion of androgens to estrogens in vertebrates. Further study showed that icariin also promoted estrogen biosynthesis and ALP activity in osteoblastic UMR-106 cells. CONCLUSIONS These results show that the promotion of estrogen biosynthesis is a novel effect of Epimedium brevicornum, and icariin could be utilized for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Lijuan Yang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | | | | | | | | | | |
Collapse
|
131
|
Holloway KR, Barbieri A, Malyarchuk S, Saxena M, Nedeljkovic-Kurepa A, Cameron Mehl M, Wang A, Gu X, Pruitt K. SIRT1 positively regulates breast cancer associated human aromatase (CYP19A1) expression. Mol Endocrinol 2013; 27:480-90. [PMID: 23340254 DOI: 10.1210/me.2012-1347] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Breast cancer remains one of the leading causes of death in women diagnosed with cancer. In breast cancer, aberrant expression of the CYP19A1 gene, which encodes the aromatase enzyme, contributes to increased intratumoral levels of estradiol. Regardless of whether this estrogen is produced by peripheral tissues or within specific subpopulations of cells within the breast tumor, it is clear that the aromatase enzymatic activity is critical for the growth of estrogen-dependent tumors. Currently, aromatase inhibitors have proven to be highly effective in blocking the growth of estrogen-dependent forms of breast cancer. CYP19A1 transcription is tightly controlled by 10 tissue-specific promoters. In breast cancer, however, aromatase transcription is driven by multiple promoters that somehow override the tissue-specific regulation of normal tissue. Here, we explore the role that the deacetylase, sirtuin-1 (SIRT1), plays in positively regulating aromatase in breast cancer. We demonstrate that the use of cambinol and the SIRT1/2 inhibitor VII, 2 small molecule inhibitors of SIRT1 and SIRT2, as well as small molecule inhibitors and small interfering RNA specific to SIRT1, all reduce the levels of aromatase mRNA. We further demonstrate that pharmacologic inhibition causes a marked reduction in aromatase protein levels. Additionally, by chromatin immunoprecipitation, we demonstrate that SIRT1 occupies the promoter regions PI.3/PII and PI.4, and its inhibition leads to increased acetylation of estrogen-related receptorα, a transcription factor that positively regulates CYP19A1 transcription in epithelial cells. Finally, we demonstrate by immunohistochemistry that SIRT1 is significantly up-regulated in invasive ductal carcinoma relative to normal tissue adjacent to tumor, further suggesting a role of SIRT1 in breast cancer. This work uncovers a new mechanism for the regulation of aromatase and provides rationale for further investigation of how the inhibition of specific sirtuins may provide a unique strategy for inhibiting aromatase that may complement or synergize with existing therapies.
Collapse
Affiliation(s)
- Kimberly R Holloway
- Departments of Molecular and Cellular Physiology, Louisiana State University Health, Sciences Center School of Medicine in Shreveport, Shreveport, LA 71130, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Kim MJ, Park YJ, Chung KH, Oh SM. The Inhibitory Effects of the Standardized Extracts of Ginkgo biloba
on Aromatase Activity in JEG-3 Human Choriocarcinoma Cells. Phytother Res 2013; 27:1756-62. [DOI: 10.1002/ptr.4927] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Revised: 12/18/2012] [Accepted: 12/18/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Mi Jie Kim
- School of Pharmacy; Sungkyunkwan University; 300 Cheoncheon dong Jangan-gu, Suwon Kyeonggi-do 440-746 South Korea
| | - Yong Joo Park
- School of Pharmacy; Sungkyunkwan University; 300 Cheoncheon dong Jangan-gu, Suwon Kyeonggi-do 440-746 South Korea
| | - Kyu Hyuck Chung
- School of Pharmacy; Sungkyunkwan University; 300 Cheoncheon dong Jangan-gu, Suwon Kyeonggi-do 440-746 South Korea
| | - Seung Min Oh
- Hoseo Fusion Technology Laboratory; Hoseo University; 165 Sechul-ri, Asan ChungcheongNam-do 336-795 South Korea
| |
Collapse
|
133
|
Liu R, Dong JT, Chen C. Role of KLF5 in hormonal signaling and breast cancer development. VITAMINS AND HORMONES 2013; 93:213-25. [PMID: 23810009 DOI: 10.1016/b978-0-12-416673-8.00002-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Steroid hormones, including ovarian steroid hormones progesterone and estrogen and androgen, play vital roles in the development of normal mammary gland and breast cancer via their receptors. How these hormones regulate these physiological and pathological processes remains to be elucidated. Krüppel-like factor 5 (KLF5) is a transcription factor playing significant roles in breast carcinogenesis, whose expression has been shown to be regulated by hormones. In this review, the relationships among hormonal signaling, KLF5, and breast cancer are summarized and discussed.
Collapse
Affiliation(s)
- Rong Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | | | | |
Collapse
|
134
|
Poulard C, Treilleux I, Lavergne E, Bouchekioua-Bouzaghou K, Goddard-Léon S, Chabaud S, Trédan O, Corbo L, Le Romancer M. Activation of rapid oestrogen signalling in aggressive human breast cancers. EMBO Mol Med 2012; 4:1200-13. [PMID: 23065768 PMCID: PMC3494876 DOI: 10.1002/emmm.201201615] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 11/10/2022] Open
Abstract
Oestrogen receptors can mediate rapid activation of cytoplasmic signalling cascades by recruiting Src and PI3K. However, the involvement of this pathway in breast cancer remains poorly defined. We have previously shown that methylation of ERα is required for the formation of the ERα/Src/PI3K complex and that ERα is hypermethylated in a subset of breast cancers. Here, we used Proximity Ligation Assay to demonstrate that this complex is present in the cytoplasm of breast cancer cell lines as well as formalin-fixed, paraffin-embedded tumours. Of particular interest, the analysis of 175 breast tumours showed that overexpression of this complex in a subset of breast tumours correlates to the activation of the downstream effector Akt. Survival analysis revealed that high expression of this complex is an independent marker of poor prognosis and associated with reduced disease-free survival. Our data introduces the new concept that the rapid oestrogen pathway is operative in vivo. It also provides a rationale for patient stratification defined by the activation of this pathway and the identification of target therapies.
Collapse
|
135
|
Lu D, Yang L, Li Q, Gao X, Wang F, Zhang G. Egonol gentiobioside and egonol gentiotrioside from Styrax perkinsiae promote the biosynthesis of estrogen by aromatase. Eur J Pharmacol 2012; 691:275-82. [DOI: 10.1016/j.ejphar.2012.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/02/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
|
136
|
Lu DF, Yang LJ, Wang F, Zhang GL. Inhibitory effect of luteolin on estrogen biosynthesis in human ovarian granulosa cells by suppression of aromatase (CYP19). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:8411-8418. [PMID: 22838964 DOI: 10.1021/jf3022817] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Inhibition of aromatase, the key enzyme in estrogen biosynthesis, is an important strategy in the treatment of breast cancer. Several dietary flavonoids show aromatase inhibitory activity, but their tissue specificity and mechanism remain unclear. This study found that the dietary flavonoid luteolin potently inhibited estrogen biosynthesis in a dose- and time-dependent manner in KGN cells derived from human ovarian granulosa cells, the major source of estrogens in premenopausal women. Luteolin decreased aromatase mRNA and protein expression in KGN cells. Luteolin also promoted aromatase protein degradation and inhibited estrogen biosynthesis in aromatase-expressing HEK293A cells, but had no effect on recombinant expressed aromatase. Estrogen biosynthesis in KGN cells was inhibited with differing potencies by extracts of onion and bird chili and by four other dietary flavonoids: kaempferol, quercetin, myricetin, and isorhamnetin. The present study suggests that luteolin inhibits estrogen biosynthesis by decreasing aromatase expression and destabilizing aromatase protein, and it warrants further investigation as a potential treatment for estrogen-dependent cancers.
Collapse
Affiliation(s)
- Dan-feng Lu
- Chengdu Institute of Biology, Chinese Academy of Sciences , Chengdu 610041, China
| | | | | | | |
Collapse
|
137
|
Abstract
Aromatase inhibitors are well-established therapies in the neoadjuvant, adjuvant and metastatic settings for breast cancer. In adjuvant trials, this class of drugs has shown preventative properties by decreasing the rate of contralateral breast cancer. Recently, the National Cancer Institute of Canada Clinical Trials Group MAP.3 study evaluated exemestane as a breast cancer prevention agent for women with specified higher risks of developing breast cancer. We review the history of exemestane and evaluate the available evidence of its use for breast cancer prevention.
Collapse
Affiliation(s)
- Jennifer Keating Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX 77030, USA
| | | | | |
Collapse
|
138
|
Bauer M, Bryce J, Hadji P. Aromatase inhibitor-associated bone loss and its management with bisphosphonates in patients with breast cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2012; 4:91-101. [PMID: 24367197 PMCID: PMC3846762 DOI: 10.2147/bctt.s29432] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Postmenopausal women have an increased risk of osteopenia and osteoporosis due to loss of the bone-protective effects of estrogen. Disease-related processes may also contribute to the risk of bone loss in postmenopausal women with breast cancer. One of the most common and severe safety issues associated with cancer therapy for patients with breast cancer is bone loss and the associated increase in risk of fractures. This paper reviews the recent literature pertaining to aromatase inhibitor (AI)-associated bone loss, and discusses suggested management and preventative approaches that may help patients remain on therapy to derive maximum clinical benefit. A case study is presented to illustrate the discussion. We observed that AIs are in widespread use for women with hormone receptor-positive breast cancer and are now recommended as adjuvant therapy, either as primary therapy or sequential to tamoxifen, for postmenopausal women. AIs target the estrogen biosynthetic pathway and deprive tumor cells of the growth-promoting effects of estrogen, and AI therapies provide benefits to patients in terms of improved disease-free survival. However, there is a concern regarding the increased risk of bone loss with prolonged AI therapy, which can be managed in many cases with the use of bisphosphonates and other interventions (eg, calcium, vitamin D supplementation, exercise).
Collapse
Affiliation(s)
- M Bauer
- University of Marburg, Marburg, Germany
| | - J Bryce
- National Cancer Institute, Naples, Italy
| | - P Hadji
- University of Marburg, Marburg, Germany
| |
Collapse
|
139
|
Eastman BM, Jo M, Webb DL, Takimoto S, Gonias SL. A transformation in the mechanism by which the urokinase receptor signals provides a selection advantage for estrogen receptor-expressing breast cancer cells in the absence of estrogen. Cell Signal 2012; 24:1847-55. [PMID: 22617030 DOI: 10.1016/j.cellsig.2012.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/08/2012] [Accepted: 05/13/2012] [Indexed: 10/28/2022]
Abstract
Binding of urokinase-type plasminogen activator (uPA) to its receptor, uPAR, in estrogen receptor-α (ERα) expressing breast cancer cells, transiently activates ERK downstream of FAK, Src family kinases, and H-Ras. Herein, we show that when uPAR is over-expressed, in two separate ERα-positive breast cancer cell lines, ERK activation occurs autonomously of uPA and is sustained. Autonomous ERK activation by uPAR requires H-Ras and Rac1. A mutated form of uPAR, which does not bind vitronectin (uPAR-W32A), failed to induce autonomous ERK activation. Expression of human uPAR or mouse uPAR but not uPAR-W32A in MCF-7 cells provided a selection advantage when these cells were deprived of estrogen in cell culture for two weeks. Similarly, MCF-7 cells that express mouse uPAR formed xenografts in SCID mice that survived and increased in volume in the absence of estrogen supplementation, probably reflecting the pro-survival activity of phospho-ERK. Autonomous uPAR signaling to ERK was sensitive to the EGFR tyrosine kinase inhibitors, Erlotinib and Gefitinib. The transition in uPAR signaling from uPA-dependent and transient to autonomous and sustained is reminiscent of the transformation in ErbB2/HER2 signaling observed when this gene is amplified in breast cancer. uPAR over-expression may provide a pathway for escape of breast cancer cells from ERα-targeting therapeutics.
Collapse
Affiliation(s)
- Boryana M Eastman
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA 92093‐0612, USA
| | | | | | | | | |
Collapse
|
140
|
Miller WR, Larionov AA. Understanding the mechanisms of aromatase inhibitor resistance. Breast Cancer Res 2012; 14:201. [PMID: 22277572 PMCID: PMC3496110 DOI: 10.1186/bcr2931] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aromatase inhibitors (AIs) have a central role in the treatment of breast cancer; however, resistance is a major obstacle to optimal management. Evidence from endocrine, molecular and pathological measurements in clinical material taken before and after therapy with AIs and data from clinical trials in which AIs have been given as treatment either alone or in combination with other targeted agents suggest diverse causes for resistance. These include inherent tumour insensitivity to oestrogen, ineffective inhibition of aromatase, sources of oestrogenic hormones independent of aromatase, activation of signalling by non-endocrine pathways, enhanced cell survival and selection of hormone-insensitive cellular clones during treatment.
Collapse
|
141
|
Litton JK, Arun BK, Brown PH, Hortobagyi GN. Aromatase inhibitors and breast cancer prevention. Expert Opin Pharmacother 2012; 13:325-31. [DOI: 10.1517/14656566.2012.651459] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
142
|
Abstract
The nuclear estrogen receptors (ER) are the major targets for endocrine treatment of hormone-dependent breast cancers. Hormone therapy blocked endogenous estrogen activation of ER, either by competitive inhibition of endogenous estrogens (selective estrogen receptor modulators - SERM or selective estrogen receptor down regulators - SERD) or by inhibition of estrogen synthesis (aromatase inhibitors) from adrenal androgens in post-menopausal women. The efficacy of these treatments has been shown on large series of breast cancer patients. However de novo or acquired resistance to treatment occurs. The better knowledge of the mechanism of action of such treatment may help to better understand them, and also for the determinism of adverse side effects of the different class of molecules.
Collapse
|
143
|
Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM, Corbo L. Cracking the estrogen receptor's posttranslational code in breast tumors. Endocr Rev 2011; 32:597-622. [PMID: 21680538 DOI: 10.1210/er.2010-0016] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estrogen signaling pathways, because of their central role in regulating the growth and survival of breast tumor cells, have been identified as suitable and efficient targets for cancer therapies. Agents blocking estrogen activity are already widely used clinically, and many new molecules have entered clinical trials, but intrinsic or acquired resistance to treatment limits their efficacy. The basic molecular studies underlying estrogen signaling have defined the critical role of estrogen receptors (ER) in many aspects of breast tumorigenesis. However, important knowledge gaps remain about the role of posttranslational modifications (PTM) of ER in initiation and progression of breast carcinogenesis. Whereas major attention has been focused on the phosphorylation of ER, many other PTM (such as acetylation, ubiquitination, sumoylation, methylation, and palmitoylation) have been identified as events modifying ER expression and stability, subcellular localization, and sensitivity to hormonal response. This article will provide an overview of the current and emerging knowledge on ER PTM, with a particular focus on their deregulation in breast cancer. We also discuss their clinical relevance and the functional relationship between PTM. A thorough understanding of the complete picture of these modifications in ER carcinogenesis might not only open new avenues for identifying new markers for prognosis or prediction of response to endocrine therapy but also could promote the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Muriel Le Romancer
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment Cheney D, 28 rue Laennec, 69373 Lyon Cedex 08, France.
| | | | | | | | | | | |
Collapse
|
144
|
Fiszman GL, Jasnis MA. Molecular Mechanisms of Trastuzumab Resistance in HER2 Overexpressing Breast Cancer. Int J Breast Cancer 2011; 2011:352182. [PMID: 22295219 PMCID: PMC3262573 DOI: 10.4061/2011/352182] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 07/01/2011] [Indexed: 01/24/2023] Open
Abstract
The epidermal growth factor receptor 2 (HER2) is a tyrosine kinase overexpressed in nearly 20% to 25% of invasive breast cancers. Trastuzumab is a humanized monoclonal antibody that targets HER2. The majority of patients with metastatic breast cancer initially respond to trastuzumab, however, within 1 year of treatment disease progresses. Several molecular mechanisms have been described as contributing to the development of trastuzumab resistance. They could be grouped as impaired access of trastuzumab to HER2, upregulation of HER2 downstream signaling pathways, signaling of alternative pathways, and impaired immune antitumor mechanisms. However, since many of them have overlapping effects, it would be of great clinical impact to identify the principal signaling pathways involved in drug resistance. Significant efforts are being applied to find other therapeutic modalities besides trastuzumab treatment to be used alone or in combination with current modalities.
Collapse
Affiliation(s)
- Gabriel L Fiszman
- Immunobiology Department, Institute of Oncology A. H. Roffo, University of Buenos Aires, Avenida San Martín 5481, CP1417 DTB Buenos Aires, Argentina
| | | |
Collapse
|
145
|
Park J, Euhus DM, Scherer PE. Paracrine and endocrine effects of adipose tissue on cancer development and progression. Endocr Rev 2011; 32:550-70. [PMID: 21642230 PMCID: PMC3369575 DOI: 10.1210/er.2010-0030] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The past few years have provided substantial evidence for the vital role of the local tumor microenvironment for various aspects of tumor progression. With obesity and its pathophysiological sequelae still on the rise, the adipocyte is increasingly moving center stage in the context of tumor stroma-related studies. To date, we have limited insight into how the systemic metabolic changes associated with obesity and the concomitant modification of the paracrine and endocrine panel of stromal adipocyte-derived secretory products ("adipokines") influence the incidence and progression of obesity-related cancers. Here, we discuss the role of adipocyte dysfunction associated with obesity and its potential impact on cancer biology.
Collapse
Affiliation(s)
- Jiyoung Park
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, USA
| | | | | |
Collapse
|
146
|
Brufsky AM. Understanding the estrogen receptor signaling pathway: focus on current endocrine agents for breast cancer in postmenopausal women. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/s1548-5315(12)70048-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
147
|
The Role of Proteomics in the Diagnosis and Treatment of Women's Cancers: Current Trends in Technology and Future Opportunities. INTERNATIONAL JOURNAL OF PROTEOMICS 2011; 2011. [PMID: 21886869 PMCID: PMC3163496 DOI: 10.1155/2011/373584] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Technological and scientific innovations over the last decade have greatly contributed to improved diagnostics, predictive models, and prognosis among cancers affecting women. In fact, an explosion of information in these areas has almost assured future generations that outcomes in cancer will continue to improve. Herein we discuss the current status of breast, cervical, and ovarian cancers as it relates to screening, disease diagnosis, and treatment options. Among the differences in these cancers, it is striking that breast cancer has multiple predictive tests based upon tumor biomarkers and sophisticated, individualized options for prescription therapeutics while ovarian cancer lacks these tools. In addition, cervical cancer leads the way in innovative, cancer-preventative vaccines and multiple screening options to prevent disease progression. For each of these malignancies, emerging proteomic technologies based upon mass spectrometry, stable isotope labeling with amino acids, high-throughput ELISA, tissue or protein microarray techniques, and click chemistry in the pursuit of activity-based profiling can pioneer the next generation of discovery. We will discuss six of the latest techniques to understand proteomics in cancer and highlight research utilizing these techniques with the goal of improvement in the management of women's cancers.
Collapse
|
148
|
Abstract
By eliciting distinct transcriptional responses, the oestrogen receptors (ERs) ERα and ERβ exert opposite effects on cellular processes that include proliferation, apoptosis and migration and that differentially influence the development and the progression of cancer. Perturbation of ER subtype-specific expression has been detected in various types of cancer, and the differences in the expression of ERs are correlated with the clinical outcome. The changes in the bioavailability of ERs in tumours, together with their specific biological functions, promote the selective restoration of their activity as one of the major therapeutic approaches for hormone-dependent cancers.
Collapse
Affiliation(s)
- Christoforos Thomas
- Center for Nuclear Receptors and Cell Signalling, Department of Biology and Biochemistry, University of Houston, Houston 77204, Texas, USA
| | | |
Collapse
|
149
|
Dunbier AK, Martin LA, Dowsett M. New and translational perspectives of oestrogen deprivation in breast cancer. Mol Cell Endocrinol 2011; 340:137-41. [PMID: 21255630 DOI: 10.1016/j.mce.2010.12.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 12/21/2010] [Accepted: 12/22/2010] [Indexed: 10/18/2022]
Abstract
Over the last 20 years, aromatase inhibitors have been developed to become a highly effective treatment strategy for treatment of hormone receptor positive breast cancer. Despite their success, poor response and resistance limit the effectiveness of these agents in up to 50% of patients. In recent years, studies using highly sensitive hormone assays have provided insight into the source of oestrogen production for the stimulation of oestrogen receptor positive breast cancer growth, suggesting that uptake from the circulation is likely to make a significant contribution to intratumoural oestradiol. To obtain insight into how tumours become resistant to oestrogen after aromatase inhibition, long term oestrogen deprivation of cultured cells has been used to mimic acquired resistance to aromatase inhibitors. This work has aided the selection of agents to rationally combine with aromatase inhibitors to combat resistance. Molecular profiling using genome-wide approaches has shed new light on the heterogeneity of responses to oestrogen deprivation and predictors of resistance in vivo. Testing new agents and combinations in short-term pre-surgical studies using biomarkers such as Ki67 is critical for increasing the rate at which new rational combinations can be assessed for efficacy.
Collapse
|
150
|
Tolhurst RS, Thomas RS, Kyle FJ, Patel H, Periyasamy M, Photiou A, Thiruchelvam PTR, Lai CF, Al-Sabbagh M, Fisher RA, Barry S, Crnogorac-Jurcevic T, Martin LA, Dowsett M, Charles Coombes R, Kamalati T, Ali S, Buluwela L. Transient over-expression of estrogen receptor-α in breast cancer cells promotes cell survival and estrogen-independent growth. Breast Cancer Res Treat 2011; 128:357-68. [PMID: 20730598 DOI: 10.1007/s10549-010-1122-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Accepted: 08/09/2010] [Indexed: 01/30/2023]
Abstract
Estrogen receptor-α (ERα) positive breast cancer frequently responds to inhibitors of ERα activity, such as tamoxifen, and/or to aromatase inhibitors that block estrogen biosynthesis. However, many patients become resistant to these agents through mechanisms that remain unclear. Previous studies have shown that expression of ERα in ERα-negative breast cancer cell lines frequently inhibits their growth. In order to determine the consequence of ERα over-expression in ERα-positive breast cancer cells, we over-expressed ERα in the MCF-7 breast cancer cell line using adenovirus gene transduction. ERα over-expression led to ligand-independent expression of the estrogen-regulated genes pS2 and PR and growth in the absence of estrogen. Interestingly, prolonged culturing of these cells in estrogen-free conditions led to the outgrowth of cells capable of growth in cultures from ERα transduced, but not in control cultures. From these cultures a line, MLET5, was established which remained ERα-positive, but grew in an estrogen-independent manner. Moreover, MLET5 cells were inhibited by anti-estrogens showing that ERα remains important for their growth. Gene expression microarray analysis comparing MCF-7 cells with MLET5 highlighted apoptosis as a major functional grouping that is altered in MLET5 cells, such that cell survival would be favoured. This conclusion was further substantiated by the demonstration that MLET5 show resistance to etoposide-induced apoptosis. As the gene expression microarray analysis also shows that the apoptosis gene set differentially expressed in MLET5 is enriched for estrogen-regulated genes, our findings suggest that transient over-expression of ERα could lead to increased cell survival and the development of estrogen-independent growth, thereby contributing to resistance to endocrine therapies in breast cancer patients.
Collapse
Affiliation(s)
- Robert S Tolhurst
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|