201
|
Jongen L, Paridaens R, Floris G, Wildiers H, Neven P. Androgen deprivation by adrenal suppression using low-dose hydrocortisone for the treatment of breast carcinoma with apocrine features: a case report illustrating this new paradigm. Breast Cancer Res Treat 2016; 155:603-7. [PMID: 26868122 DOI: 10.1007/s10549-016-3708-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 02/05/2016] [Indexed: 12/11/2022]
Abstract
We report on a postmenopausal patient with a secondary metastatic apocrine breast cancer successfully treated with low-dose hydrocortisone only following several lines of chemotherapy. The tumor cells in the primary and metastatic lesion exhibited a 'triple-negative' status (estrogen receptor (ER)-, progesterone receptor (PR)-, and human epidermal growth factor receptor 2 (HER2)-negative); the androgen receptor (AR) was strongly expressed. Twenty milligrams of hydrocortisone, a low substitution dose known to suppress adrenal steroid production, twice daily led to a clinical benefit lasting for one year, with symptom control, radiologically stable disease, and steady decrease in CA15.3. Our observation demonstrates that an AR-expressing apocrine breast cancer may respond to androgen deprivation, as an ER-positive breast cancer may benefit from estrogen deprivation. It highlights the importance of further research targeting the AR pathway in apocrine carcinoma, for which androgens represent the sole (known) steroid hormone stimulating tumor growth. Future clinical trials should not only focus on AR inhibitors like enzalutamide, but also on ablative modalities like low-dose hydrocortisone aiming at medical adrenalectomy. This method of androgen deprivation is largely available, cheap, and nearly devoid of toxicity.
Collapse
Affiliation(s)
- Lynn Jongen
- Department of Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium.
| | - Robert Paridaens
- Department of Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
- Department of General Medical Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| | - Giuseppe Floris
- Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| | - Hans Wildiers
- Department of Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
- Department of General Medical Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Gynecology and Obstetrics, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
202
|
Bosch A, Li Z, Bergamaschi A, Ellis H, Toska E, Prat A, Tao JJ, Spratt DE, Viola-Villegas NT, Castel P, Minuesa G, Morse N, Rodón J, Ibrahim Y, Cortes J, Perez-Garcia J, Galvan P, Grueso J, Guzman M, Katzenellenbogen JA, Kharas M, Lewis JS, Dickler M, Serra V, Rosen N, Chandarlapaty S, Scaltriti M, Baselga J. PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor-positive breast cancer. Sci Transl Med 2016; 7:283ra51. [PMID: 25877889 DOI: 10.1126/scitranslmed.aaa4442] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Activating mutations of PIK3CA are the most frequent genomic alterations in estrogen receptor (ER)-positive breast tumors, and selective phosphatidylinositol 3-kinase α (PI3Kα) inhibitors are in clinical development. The activity of these agents, however, is not homogeneous, and only a fraction of patients bearing PIK3CA-mutant ER-positive tumors benefit from single-agent administration. Searching for mechanisms of resistance, we observed that suppression of PI3K signaling results in induction of ER-dependent transcriptional activity, as demonstrated by changes in expression of genes containing ER-binding sites and increased occupancy by the ER of promoter regions of up-regulated genes. Furthermore, expression of ESR1 mRNA and ER protein were also increased upon PI3K inhibition. These changes in gene expression were confirmed in vivo in xenografts and patient-derived models and in tumors from patients undergoing treatment with the PI3Kα inhibitor BYL719. The observed effects on transcription were enhanced by the addition of estradiol and suppressed by the anti-ER therapies fulvestrant and tamoxifen. Fulvestrant markedly sensitized ER-positive tumors to PI3Kα inhibition, resulting in major tumor regressions in vivo. We propose that increased ER transcriptional activity may be a reactive mechanism that limits the activity of PI3K inhibitors and that combined PI3K and ER inhibition is a rational approach to target these tumors.
Collapse
Affiliation(s)
- Ana Bosch
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA
| | - Zhiqiang Li
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA
| | - Anna Bergamaschi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 524 Burrill Hall, Urbana, IL 61801, USA
| | - Haley Ellis
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA
| | - Eneda Toska
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA
| | - Aleix Prat
- Translational Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain. Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Hospital Clinic Barcelona, C/Rosselló 149-153, Barcelona 08035, Spain
| | - Jessica J Tao
- Massachusetts General Hospital Cancer Center and Harvard Medical School, 425 13th Street, Charlestown, MA 02129, USA
| | - Daniel E Spratt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Pau Castel
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA
| | - Gerard Minuesa
- Molecular Pharmacology and Chemistry Program and Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Natasha Morse
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA
| | - Jordi Rodón
- Department of Medical Oncology, VHIO, Barcelona 08035, Spain. Universitat Autònoma de Barcelona, Plaza Cívica, Campus UAB, 08193 Bellaterra, Spain
| | - Yasir Ibrahim
- Experimental Therapeutics Group, VHIO, Barcelona 08035, Spain
| | - Javier Cortes
- Department of Medical Oncology, VHIO, Barcelona 08035, Spain
| | | | - Patricia Galvan
- Translational Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Judit Grueso
- Experimental Therapeutics Group, VHIO, Barcelona 08035, Spain
| | - Marta Guzman
- Experimental Therapeutics Group, VHIO, Barcelona 08035, Spain
| | | | - Michael Kharas
- Molecular Pharmacology and Chemistry Program and Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. Molecular Pharmacology and Chemistry Program and Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maura Dickler
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Violeta Serra
- Experimental Therapeutics Group, VHIO, Barcelona 08035, Spain
| | - Neal Rosen
- Molecular Pharmacology and Chemistry Program and Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA. Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. Weill Cornell Medical College, New York, NY 10065, USA.
| | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA.
| | - José Baselga
- Human Oncology and Pathogenesis Program and Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY 10065, USA. Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
203
|
Hilborn E, Gacic J, Fornander T, Nordenskjöld B, Stål O, Jansson A. Androgen receptor expression predicts beneficial tamoxifen response in oestrogen receptor-α-negative breast cancer. Br J Cancer 2016; 114:248-55. [PMID: 26742006 PMCID: PMC4742586 DOI: 10.1038/bjc.2015.464] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/09/2015] [Accepted: 11/26/2015] [Indexed: 12/29/2022] Open
Abstract
Background: Although the androgen receptor (AR) is frequently expressed in breast cancer, its relevance in the disease is not fully understood. In addition, the relevance of AR in determining tamoxifen treatment efficiency requires evaluation. Purpose: To investigate the tamoxifen predictive relevance of the AR protein expression in breast cancer. Methods Patients were randomised to tamoxifen 40 mg daily for 2 or 5 years or to no endocrine treatment. Mean follow-up was 15 years. Hazard ratios were calculated with recurrence-free survival as end point. Results: In patients with oestrogen receptor (ER)-negative tumours, expression of AR predicted decreased recurrence rate with tamoxifen (hazard ratio (HR)=0.34; 95% confidence interval (CI)=0.14–0.81; P=0.015), whereas the opposite was seen in the AR− group (HR=2.92; 95% CI=1.16–7.31; P=0.022). Interaction test was significant P<0.001. Patients with triple-negative and AR+ tumours benefitted from tamoxifen treatment (HR=0.12; 95% CI=0.014–0.95 P=0.044), whereas patients with AR− tumours had worse outcome when treated with tamoxifen (HR=3.98; 95% CI=1.32–12.03; P=0.014). Interaction test was significant P=0.003. Patients with ER+ tumours showed benefit from tamoxifen treatment regardless of AR expression. Conclusions: AR can predict tamoxifen treatment benefit in patients with ER− tumours and triple-negative breast cancer.
Collapse
Affiliation(s)
- Erik Hilborn
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| | - Jelena Gacic
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| | - Tommy Fornander
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Bo Nordenskjöld
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| | - Olle Stål
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden.,Department of Oncology, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Agneta Jansson
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| |
Collapse
|
204
|
Abstract
Systems-wide profiling of breast cancer has almost always entailed RNA and DNA analysis by microarray and sequencing techniques. Marked developments in proteomic technologies now enable very deep profiling of clinical samples, with high identification and quantification accuracy. We analysed 40 oestrogen receptor positive (luminal), Her2 positive and triple negative breast tumours and reached a quantitative depth of >10,000 proteins. These proteomic profiles identified functional differences between breast cancer subtypes, related to energy metabolism, cell growth, mRNA translation and cell–cell communication. Furthermore, we derived a signature of 19 proteins, which differ between the breast cancer subtypes, through support vector machine (SVM)-based classification and feature selection. Remarkably, only three proteins of the signature were associated with gene copy number variations and eleven were also reflected on the mRNA level. These breast cancer features revealed by our work provide novel insights that may ultimately translate to development of subtype-specific therapeutics. Breast cancers have been extensively studied at the genomic and transcriptomic levels in the hope of tailoring therapeutic regimens. Here the authors generate deep coverage proteomes from several clinical breast cancer samples, and use machine learning techniques to uncover biological processes altered in specific cancer subtypes.
Collapse
|
205
|
Wairagu PM, Phan ANH, Kim MK, Han J, Kim HW, Choi JW, Kim KW, Cha SK, Park KH, Jeong Y. Insulin priming effect on estradiol-induced breast cancer metabolism and growth. Cancer Biol Ther 2016; 16:484-92. [PMID: 25701261 DOI: 10.1080/15384047.2015.1016660] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Diabetes is a risk factor for breast cancer development and is associated with poor prognosis for breast cancer patients. However, the molecular and biochemical mechanisms underlying the association between diabetes and breast cancer have not been fully elucidated. Here, we investigated estradiol response in MCF-7 breast cancer cells with or without chronic exposure to insulin. We found that insulin priming is necessary and specific for estradiol-induced cancer cell growth, and induces anaplerotic shunting of glucose into macromolecule biosynthesis in the estradiol treated cells. Treatment with ERK or Akt specific inhibitors, U0126 or LY294002, respectively, suppressed estradiol-induced growth. Interestingly, molecular analysis revealed that estradiol treatment markedly increases expression of cyclin A and B, and decreases p21 and p27 in the insulin-primed cells. In addition, estradiol treatment activated metabolic genes in pentose phosphate (PPP) and serine biosynthesis pathways in the insulin-primed cells while insulin priming decreased metabolic gene expression associated with glucose catabolism in the breast cancer cells. Finally, we found that anti-diabetic drug metformin and AMPK ligand AICAR, but not thiazolidinediones (TZDs), specifically suppress the estradiol-induced cellular growth in the insulin-primed cells. These findings suggest that estrogen receptor (ER) activation under chronic hyperinsulinemic condition increases breast cancer growth through the modulation of cell cycle and apoptotic factors and nutrient metabolism, and further provide a mechanistic evidence for the clinical benefit of metformin use for ER-positive breast cancer patients with diabetes.
Collapse
Key Words
- AR, androgen receptor
- CKI, cyclin dependent-kinase inhibitor
- DHT, dihydrotestosterone
- ER, estrogen receptor
- G6PD, glucose-6-phosphodehydrogenase
- GLUT1, glucose transporter 1
- HER2, human epidermal growth factor receptor 2
- IGF-1, insulin-like growth factor 1
- LDHA, lactate dehydrogenase A
- MCF-7
- PDK1, pyruvate dehydrogenase kinase 1
- PFK, phosphofructokinase
- PFKFB3, 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphate 3
- PGD, 6-phosphogluconate dehydrogenase
- PHGDH, phosphoglycerate dehydrogenase
- PKM, pyruvate kinase M
- PPAR γ, peroxisome proliferator-activated receptor gamma
- PPP, pentose phosphate pathway
- PR, progesterone receptor
- PSPH, phosphoserine phosphatase
- RPE, ribulose-5-phosphate-3-epimerase
- RPIA, ribulose-5-phosphate isomerase A
- SHMT, serine hydroxymethyltransferase
- TALDO1, transaldolase 1
- TKT, transketolase
- TZDs, thiazolidinediones
- breast cancer
- diabetes
- estradiol
- estrogen receptor
- iInsulin priming
- metformin
Collapse
Affiliation(s)
- Peninah M Wairagu
- a Department of Biochemistry ; Wonju College of Medicine ; Yonsei University ; Wonju , Gangwon-do , Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Hickey TE, Irvine CM, Dvinge H, Tarulli GA, Hanson AR, Ryan NK, Pickering MA, Birrell SN, Hu DG, Mackenzie PI, Russell R, Caldas C, Raj GV, Dehm SM, Plymate SR, Bradley RK, Tilley WD, Selth LA. Expression of androgen receptor splice variants in clinical breast cancers. Oncotarget 2015; 6:44728-44. [PMID: 26554309 PMCID: PMC4792588 DOI: 10.18632/oncotarget.6296] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/26/2015] [Indexed: 02/06/2023] Open
Abstract
The importance of androgen receptor (AR) signaling is increasingly being recognized in breast cancer, which has elicited clinical trials aimed at assessing the efficacy of androgen deprivation therapy (ADT) for metastatic disease. In prostate cancer, resistance to ADT is frequently associated with the emergence of androgen-independent splice variants of the AR (AR variants, AR-Vs) that lack the LBD and are constitutively active. Women with breast cancer may be prone to a similar phenomenon. Herein, we show that in addition to the prototypical transcript, the AR gene produces a diverse range of AR-V transcripts in primary breast tumors. The most frequently and highly expressed variant was AR-V7 (exons 1/2/3/CE3), which was detectable at the mRNA level in > 50% of all breast cancers and at the protein level in a subset of ERα-negative tumors. Functionally, AR-V7 is a constitutively active and ADT-resistant transcription factor that promotes growth and regulates a transcriptional program distinct from AR in ERα-negative breast cancer cells. Importantly, we provide ex vivo evidence that AR-V7 is upregulated by the AR antagonist enzalutamide in primary breast tumors. These findings have implications for treatment response in the ongoing clinical trials of ADT in breast cancer.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Antineoplastic Agents, Hormonal/pharmacology
- Benzamides
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Databases, Genetic
- Drug Resistance, Neoplasm
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- HEK293 Cells
- Humans
- MCF-7 Cells
- Nitriles
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/pharmacology
- Protein Isoforms
- RNA Interference
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Androgen/drug effects
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Signal Transduction
- Time Factors
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- Theresa E. Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
| | - Connie M. Irvine
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
| | - Heidi Dvinge
- Computational Biology Program, Public Health Sciences Division, Seattle, WA 98109, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Gerard A. Tarulli
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
| | - Adrienne R. Hanson
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
| | - Natalie K. Ryan
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
| | - Marie A. Pickering
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
| | - Stephen N. Birrell
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
| | - Dong Gui Hu
- Department of Clinical Pharmacology, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Peter I. Mackenzie
- Department of Clinical Pharmacology, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Roslin Russell
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Ganesh V. Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55905, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55905, USA
| | - Stephen R. Plymate
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA 98109, USA
| | - Robert K. Bradley
- Computational Biology Program, Public Health Sciences Division, Seattle, WA 98109, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Wayne D. Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
- Freemasons Foundation Centre for Men's Health, School of Medicine, The University of Adelaide, SA 5005, Australia
| | - Luke A. Selth
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, SA 5005, Australia
- Freemasons Foundation Centre for Men's Health, School of Medicine, The University of Adelaide, SA 5005, Australia
| |
Collapse
|
207
|
Heat shock protein 27 and gross cystic disease fluid protein 15 play critical roles in molecular apocrine breast cancer. Tumour Biol 2015; 37:8027-36. [PMID: 26711786 DOI: 10.1007/s13277-015-4712-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/20/2015] [Indexed: 10/22/2022] Open
Abstract
Molecular apocrine breast cancer (MABC) has a distinct hormonal profile, being estrogen receptor (ER) and progesterone receptor (PR) negative but androgen receptor (AR) positive. The clinical significance of MABC and its relative variables have not been absolutely clarified and remain to be determined. Five hundred cases of invasive breast carcinoma were randomly selected in this study, including 158 MABC cases and 342 nonMABC cases. Expression of ER, PR, epidermal growth factor receptor 2 (HER2), Ki67, AR, gross cystic disease fluid protein 15 (GCDFP15), and heat shock protein 27 (HSP27) were analyzed by immunohistochemistry. Differences of continuous variables between MABC and nonMABC subgroups were evaluated by the chi-square test. The Kaplan-Meier method was performed to evaluate disease-free survival (DFS) and overall survival (OS). The MABC subgroup had higher histological grade, bigger tumor size, more lymph node metastasis, and higher pTNM stage than the nonMABC subgroup (P < 0.05), and patients with MABC had poorer prognosis than those of the nonMABC subgroup (P < 0.05). Both GCDFP15 and HSP27 were expressed differently in the MABC and nonMABC subgroups (P < 0.05). Furthermore, in the MABC subgroup, positive HSP27 expression indicated higher risk of recurrence (P < 0.05) and positive GCDFP15 expression was also a poor marker for patient outcome (P < 0.05). MABC patients with HSP27 and GCDFP15 co-expression had worse outcome (P < 0.05). Our data suggested that MABC had a high risk of recurrence. Positive expression of both GCDFP15 and HSP27 were correlated with MABC malignancy. Targeting AR and HSP27 at the same time might offer a useful strategy to MABC.
Collapse
|
208
|
Jézéquel P, Sharif Z, Lasla H, Gouraud W, Guérin-Charbonnel C, Campion L, Chrétien S, Campone M. Gene-expression signature functional annotation of breast cancer tumours in function of age. BMC Med Genomics 2015; 8:80. [PMID: 26597277 PMCID: PMC4657228 DOI: 10.1186/s12920-015-0153-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/17/2015] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Breast cancer biological characteristics change as age advances. Today, there is a lack of knowledge regarding age-specific molecular alterations that characterize breast tumours, notably in elderly patients. The vast majority of studies that aimed at exploring breast cancer in function of age are based on clinico-pathological data. Gene-expression signatures (GES), which in some ways capture biological information in a non-reductionist manner, represent powerful tools able to explore tumour heterogeneity. METHODS Twenty-five GES were used for functional annotation of breast tumours in function of age: five for molecular subtyping, seven for immune response, three for metabolism, seven for critical pathways in cancer and three for prognosis. Affymetrix® genomics datasets were exclusively used to avoid cross-platform normalization issues. Available corresponding clinico-pathological data were also retrieved and analysed. RESULTS Fifteen publicly available datasets were pooled for a total of 2378 breast cancer patients (whole cohort), out of whom 1413 were of Caucasian origin. Three age groups were defined: ≤ 40 years (AG1), > 40 to < 70 years (AG2) and ≥ 70 years (AG3). We confirmed that age influenced the incidence of molecular subtypes. We found a significant growing incidence of luminal B and a decreasing kinetics for basal-like in function of age. We showed that AG3 luminal B tumours were less aggressive than AG1 luminal B tumours based on different GES (iron metabolism, mitochondrial oxidative phosphorylation and reactive stroma), recurrence score prognostic GES and histological grade (SBR). Contrary to tumours of young patients, tumours of elderly patients concentrated favourable GES scores: high oestrogen receptor and mitochondrial oxidative phosphorylation, low proliferation, basal-like, glycolysis, chromosomal instability and iron metabolism, and low GES prognostic scores (van't Veer 70-GES, genomic grade index and recurrence score). CONCLUSIONS Functional annotation of breast tumours by means of 25 GES demonstrated a decreasing aggressiveness of breast tumours in function of age. This strategy, which can be strengthened by increasing the number of representative GES to gain more insight into biological systems involved in this disease, provides a framework to develop rational therapeutic strategies in function of age.
Collapse
Affiliation(s)
- Pascal Jézéquel
- Bioinfomics unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France. .,Cancer Genomic Unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France. .,INSERM U892, IRT-UN, 8 quai Moncousu, Nantes Cedex, 44007, France. .,Biopatholgy department, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France.
| | - Zein Sharif
- Départemental de Vendée - site de Montaigu, Polyvalent medicine service, Centre Hospitalier, 54, rue Saint Jacques, BP 259, Montaigu, 85602, France.
| | - Hamza Lasla
- Bioinfomics unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France.
| | - Wilfried Gouraud
- Bioinfomics unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France. .,Cancer Genomic Unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France. .,INSERM U892, IRT-UN, 8 quai Moncousu, Nantes Cedex, 44007, France.
| | - Catherine Guérin-Charbonnel
- Bioinfomics unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France. .,Cancer Genomic Unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France. .,INSERM U892, IRT-UN, 8 quai Moncousu, Nantes Cedex, 44007, France.
| | - Loïc Campion
- INSERM U892, IRT-UN, 8 quai Moncousu, Nantes Cedex, 44007, France. .,Biostatistics unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France.
| | - Stéphane Chrétien
- Mathematics laboratory, UMR CNRS 6623 et Université de Franche Comté, 16 route de Gray, Besançon Cedex, 25030, France.
| | - Mario Campone
- Bioinfomics unit, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France. .,INSERM U892, IRT-UN, 8 quai Moncousu, Nantes Cedex, 44007, France. .,Medical oncology service, Integrated Centre for Oncology - René Gauducheau, Bd J. Monod, Nantes, Saint Herblain Cedex, 44805, France.
| |
Collapse
|
209
|
Prolactin-Induced Protein regulates cell adhesion in breast cancer. Biochem Biophys Res Commun 2015; 468:850-6. [PMID: 26585492 DOI: 10.1016/j.bbrc.2015.11.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
Prolactin-Induced Prolactin (PIP) is widely expressed in breast cancer and has key cellular functions in this disease that include promoting invasion and cell cycle progression. Notably, we have recently identified a strong association between PIP-binding partners and a number of cell functions that are involved in cell adhesion. Therefore in this study, we investigated the effect of PIP on the regulation of cell adhesion using PIP-silencing in breast cancer cell lines T-47D, BT-474, and MFM-223. Our findings suggest that PIP expression is necessary for cell adhesion in a process that shows variation in the pattern of PIP regulation of cell-matrix and cell-cell adhesions based on the types of adhesion surface and breast cancer cell line. In this respect, we observed that PIP-silencing markedly reduced cell adhesion to uncoated plates in all three cell lines. In addition, in T-47D and MFM-223 cells fibronectin matrix induced baseline adhesion and reversed the PIP-silencing mediated reduction of cell adhesion. However, in BT-474 cells we did not observe an induction of baseline adhesion by fibronectin and PIP-silencing led to a marked reduction in cell adhesion to both uncoated and fibronectin-coated plates. Furthermore, we observed a significant reduction in cell-cell adhesion of BT-474 cell line following PIP-silencing. To explain an underlying mechanism for PIP regulation of cell adhesion, we found that PIP expression is necessary for the formation of α-actinin/actin-rich podosomes at the adhesion-sites of breast cancer cells. In summary, this study suggests that PIP expression regulates the process of cell adhesion in breast cancer.
Collapse
|
210
|
Lau WH, Pandey V, Kong X, Wang XN, Wu Z, Zhu T, Lobie PE. Trefoil Factor-3 (TFF3) Stimulates De Novo Angiogenesis in Mammary Carcinoma both Directly and Indirectly via IL-8/CXCR2. PLoS One 2015; 10:e0141947. [PMID: 26559818 PMCID: PMC4641663 DOI: 10.1371/journal.pone.0141947] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/13/2015] [Indexed: 12/19/2022] Open
Abstract
Mammary carcinoma cells produce pro-angiogenic factors to stimulate angiogenesis and tumor growth. Trefoil factor-3 (TFF3) is an oncogene secreted from mammary carcinoma cells and associated with poor prognosis. Herein, we demonstrate that TFF3 produced in mammary carcinoma cells functions as a promoter of tumor angiogenesis. Forced expression of TFF3 in mammary carcinoma cells promoted proliferation, survival, invasion and in vitro tubule formation of human umbilical vein endothelial cells (HUVEC). MCF7-TFF3 cells with forced expression of TFF3 generated tumors with enhanced microvessel density as compared to tumors formed by vector control cells. Depletion of TFF3 in mammary carcinoma cells by siRNA concordantly decreased the angiogenic behavior of HUVEC. Forced expression of TFF3 in mammary carcinoma cells stimulated IL-8 transcription and subsequently enhanced IL-8 expression in both mammary carcinoma cells and HUVEC. Depletion of IL-8 in mammary carcinoma cells with forced expression of TFF3, or antibody inhibition of IL-8, partially abrogated mammary carcinoma cell TFF3-stimulated HUVEC angiogenic behavior in vitro, as did inhibition of the IL-8 receptor, CXCR2. Depletion of STAT3 by siRNA in MCF-7 cells with forced expression of TFF3 partially diminished the angiogenic capability of TFF3 on stimulation of cellular processes of HUVEC. Exogenous recombinant hTFF3 also directly promoted the angiogenic behavior of HUVEC. Hence, TFF3 is a potent angiogenic factor and functions as a promoter of de novo angiogenesis in mammary carcinoma, which may co-coordinate with the growth promoting and metastatic actions of TFF3 in mammary carcinoma to enhance tumor progression.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Blotting, Western
- Breast Neoplasms/blood supply
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cells, Cultured
- Coculture Techniques
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Interleukin-8/genetics
- Interleukin-8/metabolism
- MCF-7 Cells
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Fluorescence
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Peptides/genetics
- Peptides/metabolism
- RNA Interference
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Transplantation, Heterologous
- Trefoil Factor-3
Collapse
Affiliation(s)
- Wai-Hoe Lau
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Vijay Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xiangjun Kong
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiao-Nan Wang
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
- Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - ZhengSheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
- * E-mail: (PEL); (TZ)
| | - Peter E Lobie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- National University Cancer Institute of Singapore, National Health System, National University of Singapore, Singapore, Singapore
- * E-mail: (PEL); (TZ)
| |
Collapse
|
211
|
Takeshita T, Yamamoto Y, Yamamoto-Ibusuki M, Inao T, Sueta A, Fujiwara S, Omoto Y, Iwase H. Prognostic role of PIK3CA mutations of cell-free DNA in early-stage triple negative breast cancer. Cancer Sci 2015; 106:1582-9. [PMID: 26353837 PMCID: PMC4714688 DOI: 10.1111/cas.12813] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 12/14/2022] Open
Abstract
PIK3CA is an oncogene that encodes the p110α component of phosphatidylinositol 3‐kinase (PI3K); it is the second most frequently mutated gene following the TP53 gene. In the clinical setting, PIK3CA mutations may have favorable prognostic value for hormone receptor‐positive breast cancer patients and, during the past few years, PIK3CA mutations of cell‐free DNA (cfDNA) have attracted attention as a potential noninvasive biomarker of cancer. However, there are few reports on the clinical implications of PIK3CA mutations for TNBC patients. We investigated the PIK3CA major mutation status of cfDNA as a noninvasive biomarker of cancer using droplet digital polymerase chain reaction (ddPCR), which has high level sensitivity and specificity for cancer mutation, in early‐stage 49 triple negative breast cancer (TNBC) patients. A total of 12 (24.4%) of 49 patients had PIK3CA mutations of cfDNA. In a median follow up of 54.4 months, the presence of PIK3CA mutations of cfDNA had significant impacts on relapse‐free survival (RFS; P = 0.0072) and breast cancer‐specific survival (BCSS; P = 0.016), according to the log‐lank test. In a Cox proportional hazards model, the presence of PIK3CA mutations of cfDNA had significant prognostic value in the univariate and multivariate analysis. Additionally, the presence of PIK3CA mutations of cfDNA was significantly correlated with positive androgen receptor phosphorylated form depending on PI3K signaling pathway (pAR) which is independent favorable prognostic factors of TNBC. We demonstrated that the presence of PIK3CA major mutations of cfDNA could be a discriminatory predictor of RFS and BCSS in early‐stage TNBC patients and it was associated with PI3K pathway‐dependent AR phosphorylation. We demonstrated the presence of PIK3CA major mutations of cfDNA could be discriminatory predictor of RFS and BCSS in early‐stage TNBC patients which may be associated with PI3K pathway dependent AR phosphorylation.
Collapse
Affiliation(s)
- Takashi Takeshita
- Department of Breast and Endocrine surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yutaka Yamamoto
- Department of Molecular-Targeting Therapy for Breast Cancer, Kumamoto University Hospital, Kumamoto, Japan
| | - Mutsuko Yamamoto-Ibusuki
- Department of Breast and Endocrine surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Toko Inao
- Department of Breast and Endocrine surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Aiko Sueta
- Department of Molecular-Targeting Therapy for Breast Cancer, Kumamoto University Hospital, Kumamoto, Japan
| | - Saori Fujiwara
- Department of Breast and Endocrine surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yoko Omoto
- Department of Breast and Endocrine surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Department of Endocrinological and Breast Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirotaka Iwase
- Department of Breast and Endocrine surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
212
|
Malinen M, Toropainen S, Jääskeläinen T, Sahu B, Jänne OA, Palvimo JJ. Androgen receptor- and PIAS1-regulated gene programs in molecular apocrine breast cancer cells. Mol Cell Endocrinol 2015. [PMID: 26219822 DOI: 10.1016/j.mce.2015.07.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
We have analyzed androgen receptor (AR) chromatin binding sites (ARBs) and androgen-regulated transcriptome in estrogen receptor negative molecular apocrine breast cancer cells. These analyses revealed that 42% of ARBs and 39% androgen-regulated transcripts in MDA-MB453 cells have counterparts in VCaP prostate cancer cells. Pathway analyses showed a similar enrichment of molecular and cellular functions among AR targets in both breast and prostate cancer cells, with cellular growth and proliferation being among the most enriched functions. Silencing of the coregulator SUMO ligase PIAS1 in MDA-MB453 cells influenced AR function in a target-selective fashion. An anti-apoptotic effect of the silencing suggests involvement of the PIAS1 in the regulation of cell death and survival pathways. In sum, apocrine breast cancer and prostate cancer cells share a core AR cistrome and target gene signature linked to cancer cell growth, and PIAS1 plays a similar coregulatory role for AR in both cancer cell types.
Collapse
Affiliation(s)
- Marjo Malinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Sari Toropainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Tiina Jääskeläinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland; Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
| | - Biswajyoti Sahu
- Institute of Biomedicine, Physiology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Research Programs Unit, Genome-Scale Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Olli A Jänne
- Institute of Biomedicine, Physiology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
213
|
Wang T, Chen M, Zhao H. Estimating DNA methylation levels by joint modeling of multiple methylation profiles from microarray data. Biometrics 2015; 72:354-63. [PMID: 26433612 DOI: 10.1111/biom.12422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 06/01/2015] [Accepted: 08/01/2015] [Indexed: 12/29/2022]
Abstract
DNA methylation studies have been revolutionized by the recent development of high throughput array-based platforms. Most of the existing methods analyze microarray methylation data on a probe-by-probe basis, ignoring probe-specific effects and correlations among methylation levels at neighboring genomic locations. These methods can potentially miss functionally relevant findings associated with genomic regions. In this article, we propose a statistical model that allows us to pool information on the same probe across multiple samples to estimate the probe affinity effect, and to borrow strength from the neighboring probe sites to better estimate the methylation values. Using a simulation study, we demonstrate that our method can provide accurate model-based estimates. We further use the proposed method to develop a new procedure for detecting differentially methylated regions, and compare it with a state-of-the-art approach via a data application.
Collapse
Affiliation(s)
- Tao Wang
- Department of Biostatistics, Yale University, New Haven, Connecticut, 06520, U.S.A
| | - Mengjie Chen
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, 27599, U.S.A
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, New Haven, Connecticut, 06520, U.S.A
| |
Collapse
|
214
|
Barton VN, D'Amato NC, Gordon MA, Christenson JL, Elias A, Richer JK. Androgen Receptor Biology in Triple Negative Breast Cancer: a Case for Classification as AR+ or Quadruple Negative Disease. Discov Oncol 2015. [PMID: 26201402 DOI: 10.1007/s12672-015-0232-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype that lacks estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2) amplification. Due to the absence of these receptors, TNBC does not respond to traditional endocrine or HER2-targeted therapies that improve patient prognosis in other breast cancer subtypes. TNBC has a poor prognosis, and currently, there are no effective targeted therapies. Some TNBC tumors express androgen receptor (AR) and may benefit from AR-targeted therapies. Here, we review the literature on AR in TNBC and propose that TNBC be further sub-classified as either AR+ TNBC or quadruple negative breast cancer since targeting AR may represent a viable therapeutic option for a subset of TNBC.
Collapse
Affiliation(s)
- Valerie N Barton
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Nicholas C D'Amato
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Michael A Gordon
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Anthony Elias
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA.
| |
Collapse
|
215
|
Gromov P, Espinoza JA, Gromova I. Molecular and diagnostic features of apocrine breast lesions. Expert Rev Mol Diagn 2015; 15:1011-22. [DOI: 10.1586/14737159.2015.1057125] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
216
|
Bonsang-Kitzis H, Sadacca B, Hamy-Petit AS, Moarii M, Pinheiro A, Laurent C, Reyal F. Biological network-driven gene selection identifies a stromal immune module as a key determinant of triple-negative breast carcinoma prognosis. Oncoimmunology 2015; 5:e1061176. [PMID: 26942074 DOI: 10.1080/2162402x.2015.1061176] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/02/2015] [Accepted: 06/08/2015] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous group of aggressive breast cancers for which no targeted treatment is available. Robust tools for TNBC classification are required, to improve the prediction of prognosis and to develop novel therapeutic interventions. We analyzed 3,247 primary human breast cancer samples from 21 publicly available datasets, using a five-step method: (1) selection of TNBC samples by bimodal filtering on ER-HER2 and PR, (2) normalization of the selected TNBC samples, (3) selection of the most variant genes, (4) identification of gene clusters and biological gene selection within gene clusters on the basis of String© database connections and gene-expression correlations, (5) summarization of each gene cluster in a metagene. We then assessed the ability of these metagenes to predict prognosis, on an external public dataset (METABRIC). Our analysis of gene expression (GE) in 557 TNBCs from 21 public datasets identified a six-metagene signature (167 genes) in which the metagenes were enriched in different gene ontologies. The gene clusters were named as follows: Immunity1, Immunity2, Proliferation/DNA damage, AR-like, Matrix/Invasion1 and Matrix2 clusters respectively. This signature was particularly robust for the identification of TNBC subtypes across many datasets (n = 1,125 samples), despite technology differences (Affymetrix© A, Plus2 and Illumina©). Weak Immunity two metagene expression was associated with a poor prognosis (disease-specific survival; HR = 2.68 [1.59-4.52], p = 0.0002). The six-metagene signature (167 genes) was validated over 1,125 TNBC samples. The Immunity two metagene had strong prognostic value. These findings open up interesting possibilities for the development of new therapeutic interventions.
Collapse
Affiliation(s)
- H Bonsang-Kitzis
- Residual Tumor & Response to Treatment Laboratory; RT2Lab; Translational Research Department; Institut Curie; Paris, France; U932 Immunity and Cancer; INSERM; Institut Curie; Paris, France; Department of Surgery; Institut Curie; Paris, France
| | - B Sadacca
- Residual Tumor & Response to Treatment Laboratory; RT2Lab; Translational Research Department; Institut Curie; Paris, France; U932 Immunity and Cancer; INSERM; Institut Curie; Paris, France; Laboratoire de Mathématiques et Modélisation d'Evry, Université d'Évry Val d'Essonne; UMR CNRS 8071, ENSIIE, USC INRA, France
| | - A S Hamy-Petit
- Residual Tumor & Response to Treatment Laboratory; RT2Lab; Translational Research Department; Institut Curie; Paris, France; U932 Immunity and Cancer; INSERM; Institut Curie; Paris, France
| | - M Moarii
- Mines Paristech; PSL-Research University; CBIO-Centre for Computational Biology; Mines ParisTech; Fontainebleau, France; U900, INSERM; Institut Curie; Paris, France
| | - A Pinheiro
- Residual Tumor & Response to Treatment Laboratory; RT2Lab; Translational Research Department; Institut Curie; Paris, France; U932 Immunity and Cancer; INSERM; Institut Curie; Paris, France
| | - C Laurent
- Residual Tumor & Response to Treatment Laboratory; RT2Lab; Translational Research Department; Institut Curie; Paris, France; U932 Immunity and Cancer; INSERM; Institut Curie; Paris, France
| | - F Reyal
- Residual Tumor & Response to Treatment Laboratory; RT2Lab; Translational Research Department; Institut Curie; Paris, France; U932 Immunity and Cancer; INSERM; Institut Curie; Paris, France; Department of Surgery; Institut Curie; Paris, France
| |
Collapse
|
217
|
Vranic S, Marchiò C, Castellano I, Botta C, Scalzo MS, Bender RP, Payan-Gomez C, di Cantogno LV, Gugliotta P, Tondat F, di Celle PF, Mariani S, Gatalica Z, Sapino A. Immunohistochemical and molecular profiling of histologically defined apocrine carcinomas of the breast. Hum Pathol 2015. [PMID: 26208846 DOI: 10.1016/j.humpath.2015.05.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the marked improvement in the understanding of molecular mechanisms and classification of apocrine carcinoma, little is known about its specific molecular genetic alterations and potentially targetable biomarkers. In this study, we explored immunohistochemical and molecular genetic characteristics of 37 invasive apocrine carcinomas using immunohistochemistry (IHC), fluorescent in situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA), and next-generation sequencing (NGS) assays. IHC revealed frequent E-cadherin expression (89%), moderate (16%) proliferation activity [Ki-67, phosphohistone H3], infrequent (~10%) expression of basal cell markers [CK5/6, CK14, p63, caveolin-1], loss of PTEN (83%), and overexpression of HER2 (32%), EGFR (41%), cyclin D1 (50%), and MUC-1 (88%). MLPA assay revealed gene copy gains of MYC, CCND1, ZNF703, CDH1, and TRAF4 in 50% or greater of the apocrine carcinomas, whereas gene copy losses frequently affected BRCA2 (75%), ADAM9 (54%), and BRCA1 (46%). HER2 gain, detected by MLPA in 38% of the cases, was in excellent concordance with HER2 results obtained by IHC/FISH (κ = 0.915, P < .001). TOP2A gain was observed in one case, while five cases (21%) exhibited TOP2A loss. Unsupervised hierarchical cluster analysis revealed two distinct clusters: HER2-positive and HER2-negative (P = .03 and .04, respectively). NGS assay revealed mutations of the TP53 (2 of 7, 29%), BRAF/KRAS (2 of 7, 29%), and PI3KCA/PTEN genes (7 of 7, 100%). We conclude that morphologically defined apocrine carcinomas exhibit complex molecular genetic alterations that are consistent with the "luminal-complex" phenotype. Some of the identified molecular targets are promising biomarkers; however, functional studies are needed to prove these observations.
Collapse
Affiliation(s)
- Semir Vranic
- Department of Medical Sciences, University of Turin, Turin 10126, Italy; Department of Pathology, Clinical Center, University of Sarajevo, Sarajevo 71000, Bosnia and Herzegovina.
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
| | | | - Cristina Botta
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
| | | | | | - Cesar Payan-Gomez
- Medical Genetics Center, Department of Cell Biology and Genetics, Center of Biomedical Genetics, Erasmus MC, Rotterdam 3015, the Netherlands; Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá 110010, Colombia
| | | | | | - Fabrizio Tondat
- Center for Experimental Research and Medical Studies, San Giovanni Battista Hospital, Turin 10100, Italy
| | - Paola Francia di Celle
- Center for Experimental Research and Medical Studies, San Giovanni Battista Hospital, Turin 10100, Italy
| | - Sara Mariani
- Center for Experimental Research and Medical Studies, San Giovanni Battista Hospital, Turin 10100, Italy
| | | | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin 10126, Italy.
| |
Collapse
|
218
|
Proverbs-Singh T, Feldman JL, Morris MJ, Autio KA, Traina TA. Targeting the androgen receptor in prostate and breast cancer: several new agents in development. Endocr Relat Cancer 2015; 22:R87-R106. [PMID: 25722318 PMCID: PMC4714354 DOI: 10.1530/erc-14-0543] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2015] [Indexed: 12/29/2022]
Abstract
Prostate cancer (PCa) and breast cancer (BCa) share similarities as hormone-sensitive cancers with a wide heterogeneity of both phenotype and biology. The androgen receptor (AR) is a hormone receptor involved in both benign and malignant processes. Targeting androgen synthesis and the AR pathway has been and remains central to PCa therapy. Recently, there has been increased interest in the role of the AR in BCa development and growth, with results indicating AR co-expression with estrogen, progesterone, and human epidermal growth factor receptors, across all intrinsic subtypes of BCa. Targeting the AR axis is an evolving field with novel therapies in development which may ultimately be applicable to both tumor types. In this review, we offer an overview of available agents which target the AR axis in both PCa and BCa and provide insights into the novel drugs in development for targeting this signaling pathway.
Collapse
Affiliation(s)
- Tracy Proverbs-Singh
- Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA
| | - Jarett L Feldman
- Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA
| | - Michael J Morris
- Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA
| | - Karen A Autio
- Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA
| | - Tiffany A Traina
- Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA Breast Medicine ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, New York 10065, USAGenitourinary Oncology ServiceDepartment of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USAWeill Cornell Medical College1300 York Avenue, New York, New York 10065, USA
| |
Collapse
|
219
|
Abstract
INTRODUCTION Triple negative breast cancer (TNBC) is a heterogeneous disease associated with a high risk of recurrence, and therapeutic options are currently limited to cytotoxic therapy. Germ-line mutations may occur in up to 20% of unselected patients with TNBC, which may serve as a biomarker identifying which patients may have tumors that are particularly sensitive to platinums and/or inhibitors of poly(ADP-ribose)polymerase. A substantial proportion of patients with TNBCs not associated with germ-line BRCA mutations may have tumors that are ‘BRCA-like’, rendering those individuals potential candidates for similar strategies. AREAS COVERED The purpose of this review is to highlight the current standard and experimental treatment strategies. EXPERT OPINION Recent research that has illuminated the molecular heterogeneity of the disease rationalizes its diverse biological behavior and differential response to chemotherapy. Modern technology platforms provide molecular signatures that can be mined for therapeatic interventions. Target pathways that are commonly dysregulated in cancer cells control cellular processes such as apoptosis, proliferation, angiogenesis, DNA repair, cell cycle progression, immune modulation and invasion, and metastasis. Novel trial design and re-defined endpoints as surrogates to clinical outcome have been introduced to expedite the development of breakthrough therapies to treat high-risk early-stage breast cancer.
Collapse
Affiliation(s)
- Eleni Andreopoulou
- Associate Professor of Medicine, Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, 1695 Eastchester Rd Bronx, NY 10461 USA
| | - Sarah J Schweber
- Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, Bronx, NY, USA
| | - Joseph A Sparano
- Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, Bronx, NY, USA
| | - Hayley M McDaid
- Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, Bronx, NY, USA
| |
Collapse
|
220
|
Targeting thyroid hormone receptor beta in triple-negative breast cancer. Breast Cancer Res Treat 2015; 150:535-45. [PMID: 25820519 DOI: 10.1007/s10549-015-3354-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/22/2022]
Abstract
The purpose of this study was to discover novel nuclear receptor targets in triple-negative breast cancer. Expression microarray, Western blot, qRT-PCR analyses, MTT growth assay, soft agar anchorage-independent growth assay, TRE reporter transactivation assay, and statistical analysis were performed in this study. We performed microarray analysis using 227 triple-negative breast tumors, and clustered the tumors into five groups according to their nuclear receptor expression. Thyroid hormone receptor beta (TRβ) was one of the most differentially expressed nuclear receptors in group 5 compared to other groups. TRβ low expressing patients were associated with poor outcome. We evaluated the role of TRβ in triple-negative breast cancer cell lines representing group 5 tumors. Knockdown of TRβ increased soft agar colony and reduced sensitivity to docetaxel and doxorubicin treatment. Docetaxel or doxorubicin long-term cultured cell lines also expressed decreased TRβ protein. Microarray analysis revealed cAMP/PKA signaling was the only KEGG pathways upregulated in TRβ knockdown cells. Inhibitors of cAMP or PKA, in combination with doxorubicin further enhanced cell apoptosis and restored sensitivity to chemotherapy. TRβ-specific agonists enhanced TRβ expression, and further sensitized cells to both docetaxel and doxorubicin. Sensitization was mediated by increased apoptosis with elevated cleaved PARP and caspase 3. TRβ represents a novel nuclear receptor target in triple-negative breast cancer; low TRβ levels were associated with enhanced resistance to both docetaxel and doxorubicin treatment. TRβ-specific agonists enhance chemosensitivity to these two agents. Mechanistically enhanced cAMP/PKA signaling was associated with TRβ's effects on response to chemotherapy.
Collapse
|
221
|
Mehta J, Asthana S, Mandal CC, Saxena S. A molecular analysis provides novel insights into androgen receptor signalling in breast cancer. PLoS One 2015; 10:e0120622. [PMID: 25781993 PMCID: PMC4364071 DOI: 10.1371/journal.pone.0120622] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 02/05/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Androgen Receptor (AR) is an essential transcription factor for the development of secondary sex characteristics, spermatogenesis and carcinogenesis. Recently AR has been implicated in the development and progression of breast and prostate cancers. Although some of the functions of the AR are known but the mechanistic details of these divergent processes are still not clear. Therefore understanding the regulatory mechanisms of the functioning of the AR in ER-/AR+ breast cancer will provide many novel targets for the purpose of therapeutic intervention. METHODS/RESULTS Using bioinformatics tools, we have identified 75 AR targets having prominent roles in cell cycle, apoptosis and metabolism. Herein, we validated 10 genes as AR targets by studying the regulation of these genes in MDA-MB-453 cell line on stimulation by androgens like 5α-dihydrotestosterone (DHT), using RT-qPCR and ChIP assay. It was observed that all the identified genes involved in cell cycle except MAD1L1 were found to be up regulated whereas expression of apoptosis related genes was decreased in response to DHT treatment. We performed an exhaustive, rigid-body docking between individual ARE and DNA binding domain (DBD) of the AR protein and it was found that novel residues K567, K588, K591 and R592 are involved in the process of DNA binding. To verify these specific DNA-protein interactions electrostatic energy term calculations for each residue was determined using the linearized Poisson-Boltzmann equation. Our experimental data showed that treatment of breast cancer cells with DHT promotes cell proliferation and decreases apoptosis. It was observed that bicalutamide treatment was able to reverse the effect of DHT. CONCLUSION Taken together, our results provide new insights into the mechanism by which AR promotes breast cancer progression. Moreover our work proposes to use bicalutamide along with taxanes as novel therapy for the treatment of TNBCs, which are positive for downstream AR signalling.
Collapse
Affiliation(s)
- Jatin Mehta
- National Institute of Pathology, ICMR, Safdarjang Hospital, New Delhi, India
| | - Shailendra Asthana
- National Institute of Pathology, ICMR, Safdarjang Hospital, New Delhi, India
| | | | - Sunita Saxena
- National Institute of Pathology, ICMR, Safdarjang Hospital, New Delhi, India
- * E-mail:
| |
Collapse
|
222
|
O'Reilly EA, Gubbins L, Sharma S, Tully R, Guang MHZ, Weiner-Gorzel K, McCaffrey J, Harrison M, Furlong F, Kell M, McCann A. The fate of chemoresistance in triple negative breast cancer (TNBC). BBA CLINICAL 2015; 3:257-75. [PMID: 26676166 PMCID: PMC4661576 DOI: 10.1016/j.bbacli.2015.03.003] [Citation(s) in RCA: 272] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Treatment options for women presenting with triple negative breast cancer (TNBC) are limited due to the lack of a therapeutic target and as a result, are managed with standard chemotherapy such as paclitaxel (Taxol®). Following chemotherapy, the ideal tumour response is apoptotic cell death. Post-chemotherapy, cells can maintain viability by undergoing viable cellular responses such as cellular senescence, generating secretomes which can directly enhance the malignant phenotype. SCOPE OF REVIEW How tumour cells retain viability in response to chemotherapeutic engagement is discussed. In addition we discuss the implications of this retained tumour cell viability in the context of the development of recurrent and metastatic TNBC disease. Current adjuvant and neo-adjuvant treatments available and the novel potential therapies that are being researched are also reviewed. MAJOR CONCLUSIONS Cellular senescence and cytoprotective autophagy are potential mechanisms of chemoresistance in TNBC. These two non-apoptotic outcomes in response to chemotherapy are inextricably linked and are neglected outcomes of investigation in the chemotherapeutic arena. Cellular fate assessments may therefore have the potential to predict TNBC patient outcome. GENERAL SIGNIFICANCE Focusing on the fact that cancer cells can bypass the desired cellular apoptotic response to chemotherapy through cellular senescence and cytoprotective autophagy will highlight the importance of targeting non-apoptotic survival pathways to enhance chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Elma A O'Reilly
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Luke Gubbins
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Shiva Sharma
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Riona Tully
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Matthew Ho Zhing Guang
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Karolina Weiner-Gorzel
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - John McCaffrey
- Department of Oncology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Michele Harrison
- Department of Pathology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Fiona Furlong
- School of Pharmacy, Queens University Belfast, Belfast BT7 1NN, UK
| | - Malcolm Kell
- Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Amanda McCann
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| |
Collapse
|
223
|
mRNA profiling reveals determinants of trastuzumab efficiency in HER2-positive breast cancer. PLoS One 2015; 10:e0117818. [PMID: 25710561 PMCID: PMC4339844 DOI: 10.1371/journal.pone.0117818] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 12/30/2014] [Indexed: 12/30/2022] Open
Abstract
Intrinsic and acquired resistance to the monoclonal antibody drug trastuzumab is a major problem in the treatment of HER2-positive breast cancer. A deeper understanding of the underlying mechanisms could help to develop new agents. Our intention was to detect genes and single nucleotide polymorphisms (SNPs) affecting trastuzumab efficiency in cell culture. Three HER2-positive breast cancer cell lines with different resistance phenotypes were analyzed. We chose BT474 as model of trastuzumab sensitivity, HCC1954 as model of intrinsic resistance, and BTR50, derived from BT474, as model of acquired resistance. Based on RNA-Seq data, we performed differential expression analyses on these cell lines with and without trastuzumab treatment. Differentially expressed genes between the resistant cell lines and BT474 are expected to contribute to resistance. Differentially expressed genes between untreated and trastuzumab treated BT474 are expected to contribute to drug efficacy. To exclude false positives from the candidate gene set, we removed genes that were also differentially expressed between untreated and trastuzumab treated BTR50. We further searched for SNPs in the untreated cell lines which could contribute to trastuzumab resistance. The analysis resulted in 54 differentially expressed candidate genes that might be connected to trastuzumab efficiency. 90% of 40 selected candidates were validated by RT-qPCR. ALPP, CALCOCO1, CAV1, CYP1A2 and IGFBP3 were significantly higher expressed in the trastuzumab treated than in the untreated BT474 cell line. GDF15, IL8, LCN2, PTGS2 and 20 other genes were significantly higher expressed in HCC1954 than in BT474, while NCAM2, COLEC12, AFF3, TFF3, NRCAM, GREB1 and TFF1 were significantly lower expressed. Additionally, we inferred SNPs in HCC1954 for CAV1, PTGS2, IL8 and IGFBP3. The latter also had a variation in BTR50. 20% of the validated subset have already been mentioned in literature. For half of them we called and analyzed SNPs. These results contribute to a better understanding of trastuzumab action and resistance mechanisms.
Collapse
|
224
|
Barton VN, D'Amato NC, Gordon MA, Lind HT, Spoelstra NS, Babbs BL, Heinz RE, Elias A, Jedlicka P, Jacobsen BM, Richer JK. Multiple molecular subtypes of triple-negative breast cancer critically rely on androgen receptor and respond to enzalutamide in vivo. Mol Cancer Ther 2015; 14:769-78. [PMID: 25713333 DOI: 10.1158/1535-7163.mct-14-0926] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/11/2015] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC) has the lowest 5-year survival rate of invasive breast carcinomas, and currently there are no approved targeted therapies for this aggressive form of the disease. The androgen receptor (AR) is expressed in up to one third of TNBC and we find that all AR(+) TNBC primary tumors tested display nuclear localization of AR, indicative of transcriptionally active receptors. While AR is most abundant in the "luminal AR (LAR)" molecular subtype of TNBC, here, for the first time, we use both the new-generation anti-androgen enzalutamide and AR knockdown to demonstrate that the other non-LAR molecular subtypes of TNBC are critically dependent on AR protein. Indeed, AR inhibition significantly reduces baseline proliferation, anchorage-independent growth, migration, and invasion and increases apoptosis in four TNBC lines (SUM159PT, HCC1806, BT549, and MDA-MB-231), representing three non-LAR TNBC molecular subtypes (mesenchymal-like, mesenchymal stem-like, and basal-like 2). In vivo, enzalutamide significantly decreases viability of SUM159PT and HCC1806 xenografts. Furthermore, mechanistic analysis reveals that AR activation upregulates secretion of the EGFR ligand amphiregulin (AREG), an effect abrogated by enzalutamide in vitro and in vivo. Exogenous AREG partially rescues the effects of AR knockdown on proliferation, migration, and invasion, demonstrating that upregulation of AREG is one mechanism by which AR influences tumorigenicity. Together, our findings indicate that non-LAR subtypes of TNBC are AR dependent and, moreover, that enzalutamide is a promising targeted therapy for multiple molecular subtypes of AR(+) TNBC.
Collapse
Affiliation(s)
- Valerie N Barton
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicholas C D'Amato
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael A Gordon
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hanne T Lind
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Beatrice L Babbs
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard E Heinz
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony Elias
- Department of Medicine, Division of Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Britta M Jacobsen
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
225
|
Ng CKY, Schultheis AM, Bidard FC, Weigelt B, Reis-Filho JS. Breast cancer genomics from microarrays to massively parallel sequencing: paradigms and new insights. J Natl Cancer Inst 2015; 107:djv015. [PMID: 25713166 DOI: 10.1093/jnci/djv015] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rapid advancements in massively parallel sequencing methods have enabled the analysis of breast cancer genomes at an unprecedented resolution, which have revealed the remarkable heterogeneity of the disease. As a result, we now accept that despite originating in the breast, estrogen receptor (ER)-positive and ER-negative breast cancers are completely different diseases at the molecular level. It has become apparent that there are very few highly recurrently mutated genes such as TP53, PIK3CA, and GATA3, that no two breast cancers display an identical repertoire of somatic genetic alterations at base-pair resolution and that there might not be a single highly recurrently mutated gene that defines each of the "intrinsic" subtypes of breast cancer (ie, basal-like, HER2-enriched, luminal A, and luminal B). Breast cancer heterogeneity, however, extends beyond the diversity between tumors. There is burgeoning evidence to demonstrate that at least some primary breast cancers are composed of multiple, genetically diverse clones at diagnosis and that metastatic lesions may differ in their repertoire of somatic genetic alterations when compared with their respective primary tumors. Several biological phenomena may shape the reported intratumor genetic heterogeneity observed in breast cancers, including the different mutational processes and multiple types of genomic instability. Harnessing the emerging concepts of the diversity of breast cancer genomes and the phenomenon of intratumor genetic heterogeneity will be essential for the development of optimal methods for diagnosis, disease monitoring, and the matching of patients to the drugs that would benefit them the most.
Collapse
Affiliation(s)
- Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF)
| | - Anne M Schultheis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF)
| | - Francois-Clement Bidard
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF)
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF).
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF).
| |
Collapse
|
226
|
Murray JI, West NR, Murphy LC, Watson PH. Intratumoural inflammation and endocrine resistance in breast cancer. Endocr Relat Cancer 2015; 22:R51-67. [PMID: 25404688 DOI: 10.1530/erc-14-0096] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is becoming clear that inflammation-associated mechanisms can affect progression of breast cancer and modulate responses to treatment. Estrogen receptor alpha (ERα (ESR1)) is the principal biomarker and therapeutic target for endocrine therapies in breast cancer. Over 70% of patients are ESR1-positive at diagnosis and are candidates for endocrine therapy. However, ESR1-positive tumours can become resistant to endocrine therapy. Multiple mechanisms of endocrine resistance have been proposed, including suppression of ESR1. This review discusses the relationship between intratumoural inflammation and endocrine resistance with a particular focus on inflammation-mediated suppression of ESR1.
Collapse
Affiliation(s)
- Jill I Murray
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Nathan R West
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Leigh C Murphy
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Peter H Watson
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
227
|
Prolactin-induced protein is required for cell cycle progression in breast cancer. Neoplasia 2015; 16:329-42.e1-14. [PMID: 24862759 DOI: 10.1016/j.neo.2014.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/06/2014] [Accepted: 03/24/2014] [Indexed: 11/21/2022] Open
Abstract
Prolactin-induced protein (PIP) is expressed in the majority of breast cancers and is used for the diagnostic evaluation of this disease as a characteristic biomarker; however, the molecular mechanisms of PIP function in breast cancer have remained largely unknown. In this study, we carried out a comprehensive investigation of PIP function using PIP silencing in a broad group of breast cancer cell lines, analysis of expression microarray data, proteomic analysis using mass spectrometry, and biomarker studies on breast tumors. We demonstrated that PIP is required for the progression through G1 phase, mitosis, and cytokinesis in luminal A, luminal B, and molecular apocrine breast cancer cells. In addition, PIP expression is associated with a transcriptional signature enriched with cell cycle genes and regulates key genes in this process including cyclin D1, cyclin B1, BUB1, and forkhead box M1 (FOXM1). It is notable that defects in mitotic transition and cytokinesis following PIP silencing are accompanied by an increase in aneuploidy of breast cancer cells. Importantly, we have identified novel PIP-binding partners in breast cancer and shown that PIP binds to β-tubulin and is necessary for microtubule polymerization. Furthermore, PIP interacts with actin-binding proteins including Arp2/3 and is needed for inside-out activation of integrin-β1 mediated through talin. This study suggests that PIP is required for cell cycle progression in breast cancer and provides a rationale for exploring PIP inhibition as a therapeutic approach in breast cancer that can potentially target microtubule polymerization.
Collapse
|
228
|
Naderi A. Prolactin-induced protein in breast cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 846:189-200. [PMID: 25472539 DOI: 10.1007/978-3-319-12114-7_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Prolactin-induced protein (PIP) is a 17-kDa single polypeptide chain that is secreted by a number of normal apocrine cells, such as milk, saliva, and seminal fluid. PIP is widely expressed in breast cancer and is commonly used as a diagnostic biomarker for the histopathological diagnosis of this disease. Expression of PIP in breast cancer is regulated by androgen and prolactin through a number of transcription factors and signaling cross-talks, including STAT5, Runx2, and CREB1. Notably, PIP is induced by a positive feedback loop between androgen receptor (AR) and extracellular signal-regulated kinase (ERK). The available data indicate that PIP has an aspartyl protease activity that can degrade fibronectin. Importantly, PIP is necessary for outside-in activation of integrin-β1 signaling pathway and regulation of key downstream signaling targets of this pathway such as interaction of integrin-β1 with integrin-linked kinase 1 (ILK1) and ErbB2. Furthermore, the importance of PIP in cell proliferation has been demonstrated by the fact that purified PIP promotes growth of breast cancer cells and PIP expression is necessary for the proliferation of T-47D and MDA-MB-453 cell lines. In addition to cell proliferation, PIP mediates invasion of breast cancer cells in a process that partially depends on the degradation of fibronectin by this protein. Therefore, PIP is a breast cancer-related protein that is expressed in a majority of breast tumors and has a significant function in the biology of this disease.
Collapse
Affiliation(s)
- Ali Naderi
- Holden Comprehensive Cancer Center, University of Iowa, 3202 MERF, 375 Newton Road, 52242, Iowa City, IA, USA,
| |
Collapse
|
229
|
Bae J, Samur M, Munshi A, Hideshima T, Keskin D, Kimmelman A, Lee AH, Dranoff G, Anderson KC, Munshi NC. Heteroclitic XBP1 peptides evoke tumor-specific memory cytotoxic T lymphocytes against breast cancer, colon cancer, and pancreatic cancer cells. Oncoimmunology 2014; 3:e970914. [PMID: 25941601 DOI: 10.4161/21624011.2014.970914] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022] Open
Abstract
XBP1 is a critical transcriptional activator of the unfolded protein response (UPR), which increases tumor cell survival under prolonged endoplasmic reticulum (ER) stress and hypoxic conditions.This study was designed to evaluate the immunogenicity of heteroclitic XBP1 unspliced (US)184-192 (YISPWILAV) and heteroclictic XBP1 spliced (SP)367-375 (YLFPQLISV) HLA-A2 peptides, and to characterize the specific activities of XBP1 peptides-specific cytotoxic T lymphocytes (XBP1-CTL) against breast cancer, colon cancer, and pancreatic cancer cells.The XBP1-CTL had upregulated expression of critical T cell markers and displayed HLA-A2-restricted and antigen-specific activities against breast cancer, colon cancer and pancreatic cancer cells. XBP1-CTL were enriched withCD45RO+ memory CTL, which showed high expression of critical T cell markers (CD28, ICOS, CD69, CD40L), cell proliferation and antitumor activities as compared to CD45RO- non-memory CTL. The effector memory (EM: CD45RO+CCR7-) subset had the highest level of cell proliferation while the central memory (CM: CD45RO+CCR7+) subset demonstrated enhanced functional activities (CD107a degranulation, IFNγ/IL-2 production) upon recognition of the respective tumor cells. Furthermore, both the EM and CM XBP1-CTL subsets expressed high levels of Th1 transcription regulators Tbet and Eomes. The highest frequencies of IFNγ or granzyme B producing cells were detected within CM XBP1-CTL subset that were either Tbet+ or Eomes+ in responding to the tumor cells.These results demonstrate the immunotherapeutic potential of a cocktail of immunogenic HLA-A2 specific heteroclitic XBP1 US184-192 and heteroclictic XBP1 SP367-375 peptides to induce CD3+CD8+ CTL enriched for CM and EM cells with specific antitumor activities against a variety of solid tumors.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Mehmet Samur
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Aditya Munshi
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Teru Hideshima
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Derin Keskin
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Alec Kimmelman
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Ann-Hwee Lee
- Weill Cornell Medical College ; New York, NY USA
| | - Glen Dranoff
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Kenneth C Anderson
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA
| | - Nikhil C Munshi
- Dana-Farber Cancer Institute ; Boston, MA USA ; Harvard Medical School ; Boston, MA USA ; VA Boston Healthcare System ; Boston, MA USA
| |
Collapse
|
230
|
Gromov P, Espinoza JA, Talman ML, Honma N, Kroman N, Wielenga VT, Moreira JMA, Gromova I. FABP7 and HMGCS2 are novel protein markers for apocrine differentiation categorizing apocrine carcinoma of the breast. PLoS One 2014; 9:e112024. [PMID: 25389781 PMCID: PMC4229141 DOI: 10.1371/journal.pone.0112024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/09/2014] [Indexed: 02/01/2023] Open
Abstract
Apocrine carcinoma of the breast is a distinctive malignancy with unique morphological and molecular features, generally characterized by being negative for estrogen and progesterone receptors, and thus not electable for endocrine therapy. Despite the fact that they are morphologically distinct from other breast lesions, no standard molecular criteria are currently available for their diagnosis. Using gel-based proteomics in combination with mass spectrometry and immunohistochemistry we have identified two novel markers, HMGCS2 and FABP7 that categorize the entire breast apocrine differentiation spectrum from benign metaplasia and cysts to invasive stages. Expression of HMGCS2 and FABP7 is strongly associated with apocrine differentiation; their expression is retained by most invasive apocrine carcinomas (IAC) showing positive immunoreactivity in 100% and 78% of apocrine carcinomas, respectively, as compared to non-apocrine tumors (16.7% and 6.8%). The nuclear localization of FABP7 in tumor cells was shown to be associated with more aggressive stages of apocrine carcinomas. In addition, when added to the panel of apocrine biomarkers previously reported by our group: 15-PGDH, HMGCR and ACSM1, together they provide a signature that may represent a golden molecular standard for defining the apocrine phenotype in the breast. Moreover, we show that combining HMGCS2 to the steroidal profile (HMGCS2+/Androgen Receptor (AR)+/Estrogen Receptor(ER)-/Progesteron Receptor (PR)- identifies IACs with a greater sensitivity (79%) as compared with the steroidal profile (AR+/ER-/PR-) alone (54%). We have also presented a detailed immunohistochemical analysis of breast apocrine lesions with a panel of antibodies against proteins which correspond to 10 genes selected from published transcriptomic signatures that currently characterize molecular apocrine subtype and shown that except for melanophilin that is overexpressed in benign apocrine lesions, these proteins were not specific for morphological apocrine differentiation in breast.
Collapse
Affiliation(s)
- Pavel Gromov
- Danish Cancer Society Research Center, Genome Integrity Unit, Copenhagen, Denmark
- * E-mail:
| | - Jaime A. Espinoza
- Department of Pathology, Center for Investigation in Translational Oncology (CITO), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maj-Lis Talman
- Department of Pathology, the Centre of Diagnostic Investigations, Copenhagen University Hospital, Copenhagen, Denmark
| | - Naoko Honma
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Niels Kroman
- Department of Breast Surgery, Copenhagen University Hospital, Copenhagen, Denmark
| | - Vera Timmermans Wielenga
- Department of Pathology, the Centre of Diagnostic Investigations, Copenhagen University Hospital, Copenhagen, Denmark
| | - José M. A. Moreira
- Section of Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irina Gromova
- Danish Cancer Society Research Center, Genome Integrity Unit, Copenhagen, Denmark
| |
Collapse
|
231
|
Abstract
Androgens were at one time a therapeutic mainstay in the treatment of advanced breast cancer. Despite comparable efficacy, SERMs and aromatase inhibitors eventually became the therapies of choice due to in part to preferred side-effect profiles. Molecular characterization of breast tumors has revealed an abundance of androgen receptor expression but the choice of an appropriate androgen receptor ligand (agonist or antagonist) has been confounded by multiple conflicting reports concerning the role of the receptor in the disease. Modern clinical efforts have almost exclusively utilized antagonists. However, the recent clinical development of selective androgen receptor modulators with greatly improved side-effect profiles has renewed interest in androgen agonist therapy for advanced breast cancer.
Collapse
|
232
|
Mancini P, Angeloni A, Risi E, Orsi E, Mezi S. Standard of care and promising new agents for triple negative metastatic breast cancer. Cancers (Basel) 2014; 6:2187-223. [PMID: 25347122 PMCID: PMC4276962 DOI: 10.3390/cancers6042187] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/05/2014] [Accepted: 09/26/2014] [Indexed: 12/11/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a cluster of heterogeneous diseases, all of them sharing the lack of expression of estrogen and progesterone receptors and HER2 protein. They are characterized by different biological, molecular and clinical features, including a poor prognosis despite the increased sensitivity to the current cytotoxic therapies. Several studies have identified important molecular features which enable further subdivision of this type of tumor. We are drawing from genomics, transcription and translation analysis at different levels, to improve our knowledge of the molecular alterations along the pathways which are activated during carcinogenesis and tumor progression. How this information should be used for the rational selection of therapy is an ongoing challenge and the subject of numerous research studies in progress. Currently, the vascular endothelial growth factor (VEGF), poly (ADP-ribose) polymerase (PARP), HSP90 and Aurora inhibitors are most used as targeting agents in metastatic setting clinical trials. In this paper we will review the current knowledge about the genetic subtypes of TNBC and their different responses to conventional therapeutic strategies, as well as to some new promising molecular target agents, aimed to achieve more tailored therapies.
Collapse
Affiliation(s)
- Patrizia Mancini
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Antonio Angeloni
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Emanuela Risi
- Department of Radiology, Oncology and Human Pathology, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Errico Orsi
- Department of Surgical Science, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Silvia Mezi
- Department of Radiology, Oncology and Human Pathology, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| |
Collapse
|
233
|
Hu DG, Meech R, McKinnon RA, Mackenzie PI. Transcriptional regulation of human UDP-glucuronosyltransferase genes. Drug Metab Rev 2014; 46:421-58. [PMID: 25336387 DOI: 10.3109/03602532.2014.973037] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucuronidation is an important metabolic pathway for many small endogenous and exogenous lipophilic compounds, including bilirubin, steroid hormones, bile acids, carcinogens and therapeutic drugs. Glucuronidation is primarily catalyzed by the UDP-glucuronosyltransferase (UGT) 1A and two subfamilies, including nine functional UGT1A enzymes (1A1, 1A3-1A10) and 10 functional UGT2 enzymes (2A1, 2A2, 2A3, 2B4, 2B7, 2B10, 2B11, 2B15, 2B17 and 2B28). Most UGTs are expressed in the liver and this expression relates to the major role of hepatic glucuronidation in systemic clearance of toxic lipophilic compounds. Hepatic glucuronidation activity protects the body from chemical insults and governs the therapeutic efficacy of drugs that are inactivated by UGTs. UGT mRNAs have also been detected in over 20 extrahepatic tissues with a unique complement of UGT mRNAs seen in almost every tissue. This extrahepatic glucuronidation activity helps to maintain homeostasis and hence regulates biological activity of endogenous molecules that are primarily inactivated by UGTs. Deciphering the molecular mechanisms underlying tissue-specific UGT expression has been the subject of a large number of studies over the last two decades. These studies have shown that the constitutive and inducible expression of UGTs is primarily regulated by tissue-specific and ligand-activated transcription factors (TFs) via their binding to cis-regulatory elements (CREs) in UGT promoters and enhancers. This review first briefly summarizes published UGT gene transcriptional studies and the experimental models and tools utilized in these studies, and then describes in detail the TFs and their respective CREs that have been identified in the promoters and/or enhancers of individual UGT genes.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre , Bedford Park, SA , Australia
| | | | | | | |
Collapse
|
234
|
Coagulation factor VII is regulated by androgen receptor in breast cancer. Exp Cell Res 2014; 331:239-250. [PMID: 25447311 DOI: 10.1016/j.yexcr.2014.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 11/22/2022]
Abstract
Androgen receptor (AR) is widely expressed in breast cancer; however, there is limited information on the key molecular functions and gene targets of AR in this disease. In this study, gene expression data from a cohort of 52 breast cancer cell lines was analyzed to identify a network of AR co-expressed genes. A total of 300 genes, which were significantly enriched for cell cycle and metabolic functions, showed absolute correlation coefficients (|CC|) of more than 0.5 with AR expression across the dataset. In this network, a subset of 35 "AR-signature" genes were highly co-expressed with AR (|CC|>0.6) that included transcriptional regulators PATZ1, NFATC4, and SPDEF. Furthermore, gene encoding coagulation factor VII (F7) demonstrated the closest expression pattern with AR (CC=0.716) in the dataset and factor VII protein expression was significantly associated to that of AR in a cohort of 209 breast tumors. Moreover, functional studies demonstrated that AR activation results in the induction of factor VII expression at both transcript and protein levels and AR directly binds to a proximal region of F7 promoter in breast cancer cells. Importantly, AR activation in breast cancer cells induced endogenous factor VII activity to convert factor X to Xa in conjunction with tissue factor. In summary, F7 is a novel AR target gene and AR activation regulates the ectopic expression and activity of factor VII in breast cancer cells. These findings have functional implications in the pathobiology of thromboembolic events and regulation of factor VII/tissue factor signaling in breast cancer.
Collapse
|
235
|
Pandey V, Wu ZS, Zhang M, Li R, Zhang J, Zhu T, Lobie PE. Trefoil factor 3 promotes metastatic seeding and predicts poor survival outcome of patients with mammary carcinoma. Breast Cancer Res 2014; 16:429. [PMID: 25266665 PMCID: PMC4303111 DOI: 10.1186/s13058-014-0429-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 08/15/2014] [Indexed: 12/14/2022] Open
Abstract
Introduction Recurrence or early metastasis remains the predominant cause of mortality in patients with estrogen receptor positive (ER+) mammary carcinoma (MC). However, the molecular mechanisms underlying the initial progression of ER+ MC to metastasis remains poorly understood. Trefoil factor 3 (TFF3) is an estrogen-responsive oncogene in MC. Herein, we provide evidence for a functional role of TFF3 in metastatic progression of ER+ MC. Methods The association of TFF3 expression with clinicopathological parameters and survival outcome in a cohort of MC patients was assessed by immunohistochemistry. The expression of TFF3 in MCF7 and T47D cells was modulated by forced expression or siRNA-mediated depletion of TFF3. mRNA and protein levels were determined using qPCR and western blot. The functional effect of modulation of TFF3 expression in MC cells was determined in vitro and in vivo. Mechanistic analyses were performed using reporter constructs, modulation of signal transducer and activator of transcription 3 (STAT3) expression, and pharmacological inhibitors against c-SRC and STAT3 activity. Results TFF3 protein expression was positively associated with larger tumour size, lymph node metastasis, higher stage, and poor survival outcome. Forced expression of TFF3 in ER+ MC cells stimulated colony scattering, cell adhesion to a Collagen I-coated matrix, colony formation on a Collagen I- or Matrigel-coated matrix, endothelial cell adhesion, and transmigration through an endothelial cell barrier. In vivo, forced expression of TFF3 in MCF7 cells stimulated the formation of metastatic nodules in animal lungs. TFF3 regulation of the mRNA levels of epithelial, mesenchymal, and metastatic-related genes in ER+ MC cells were consistent with the altered cell behaviour. Forced expression of TFF3 in ER+ MC cells stimulated phosphorylation of c-SRC that subsequently increased STAT3 activity, which lead to the downregulation of E-cadherin. siRNA-mediated depletion of TFF3 reduced the invasiveness of ER+ MC cells. Conclusions TFF3 expression predicts metastasis and poor survival outcome of patients with MC and functionally stimulates cellular invasion and metastasis of ER+ MC cells. Adjuvant functional inhibition of TFF3 may therefore be considered to ameliorate outcome of ER+ MC patients. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0429-3) contains supplementary material, which is available to authorized users.
Collapse
|
236
|
Combined conjugated esterified estrogen plus methyltestosterone supplementation and risk of breast cancer in postmenopausal women. Maturitas 2014; 79:70-6. [DOI: 10.1016/j.maturitas.2014.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 11/18/2022]
|
237
|
Shike M, Doane AS, Russo L, Cabal R, Reis-Filho JS, Gerald W, Cody H, Khanin R, Bromberg J, Norton L. The effects of soy supplementation on gene expression in breast cancer: a randomized placebo-controlled study. J Natl Cancer Inst 2014; 106:dju189. [PMID: 25190728 PMCID: PMC4817128 DOI: 10.1093/jnci/dju189] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/22/2014] [Accepted: 05/28/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND There are conflicting reports on the impact of soy on breast carcinogenesis. This study examines the effects of soy supplementation on breast cancer-related genes and pathways. METHODS Women (n = 140) with early-stage breast cancer were randomly assigned to soy protein supplementation (n = 70) or placebo (n = 70) for 7 to 30 days, from diagnosis until surgery. Adherence was determined by plasma isoflavones: genistein and daidzein. Gene expression changes were evaluated by NanoString in pre- and posttreatment tumor tissue. Genome-wide expression analysis was performed on posttreatment tissue. Proliferation (Ki67) and apoptosis (Cas3) were assessed by immunohistochemistry. RESULTS Plasma isoflavones rose in the soy group (two-sided Wilcoxon rank-sum test, P < .001) and did not change in the placebo group. In paired analysis of pre- and posttreatment samples, 21 genes (out of 202) showed altered expression (two-sided Student's t-test, P < .05). Several genes including FANCC and UGT2A1 revealed different magnitude and direction of expression changes between the two groups (two-sided Student's t-test, P < .05). A high-genistein signature consisting of 126 differentially expressed genes was identified from microarray analysis of tumors. This signature was characterized by overexpression (>2-fold) of cell cycle transcripts, including those that promote cell proliferation, such as FGFR2, E2F5, BUB1, CCNB2, MYBL2, CDK1, and CDC20 (P < .01). Soy intake did not result in statistically significant changes in Ki67 or Cas3. CONCLUSIONS Gene expression associated with soy intake and high plasma genistein defines a signature characterized by overexpression of FGFR2 and genes that drive cell cycle and proliferation pathways. These findings raise the concerns that in a subset of women soy could adversely affect gene expression in breast cancer.
Collapse
MESH Headings
- Adult
- Aged
- Apoptosis/drug effects
- Biomarkers/blood
- Breast Neoplasms/blood
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/blood
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Caspase 3/metabolism
- Cell Proliferation/drug effects
- Dietary Supplements/adverse effects
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Genistein/blood
- Humans
- Immunohistochemistry
- Isoflavones/blood
- Ki-67 Antigen/metabolism
- Middle Aged
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Soybean Proteins/administration & dosage
- Soybean Proteins/adverse effects
- Tissue Array Analysis
- Up-Regulation
Collapse
Affiliation(s)
- Moshe Shike
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN).
| | - Ashley S Doane
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Lianne Russo
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Rafael Cabal
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Jorge S Reis-Filho
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - William Gerald
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Hiram Cody
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Raya Khanin
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Jacqueline Bromberg
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Larry Norton
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| |
Collapse
|
238
|
Choi JE, Kang SH, Lee SJ, Bae YK. Androgen receptor expression predicts decreased survival in early stage triple-negative breast cancer. Ann Surg Oncol 2014; 22:82-9. [PMID: 25145503 DOI: 10.1245/s10434-014-3984-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND A subset of triple-negative breast cancer (TNBC) has been reported to express androgen receptor (AR); however, the clinical significance of AR expression in TNBC is unclear. METHODS We examined immunohistochemical expression of AR in a large cohort of TNBC cases and correlated its expression with clinicopathologic features and clinical outcome. RESULTS AR expression was found in 17.7% (87/492) of TNBCs. Positive expression of AR showed significant correlation with older age (p < 0.001), apocrine histology (p = 0.001), and lower histologic grade (p < 0.001). AR was a poor prognostic marker for overall survival (OS) in univariate (p = 0.026) and multivariate (p = 0.008) analyses. In the lymph node-negative (n = 316) subgroup, AR expression was a significant predictor of worse OS and disease-free survival (DFS) in both univariate (p = 0.028 and 0.011) and multivariate (p = 0.024 and 0.01, respectively) analyses. AR expression also was a prognostic factor in pT1 subgroup (OS, p = 0.007; DFS, p = 0.01); however, its prognostic value was not observed in TNBC patients with lymph node metastasis or tumor size larger than pT1. CONCLUSIONS AR-expressing TNBCs represent a distinct breast cancer subgroup with adverse clinical outcome and AR blockade could be a potential endocrine therapy for these TNBC patients. Evaluation of AR status may provide additional information on prognosis and treatment in patients with TNBC.
Collapse
Affiliation(s)
- Jung Eun Choi
- Department of Surgery, Yeungnam University College of Medicine, Daegu, South Korea
| | | | | | | |
Collapse
|
239
|
Abstract
While it has been known for decades that androgen hormones influence normal breast development and breast carcinogenesis, the underlying mechanisms have only been recently elucidated. To date, most studies have focused on androgen action in breast cancer cell lines, yet these studies represent artificial systems that often do not faithfully replicate/recapitulate the cellular, molecular and hormonal environments of breast tumours in vivo. It is critical to have a better understanding of how androgens act in the normal mammary gland as well as in in vivo systems that maintain a relevant tumour microenvironment to gain insights into the role of androgens in the modulation of breast cancer development. This in turn will facilitate application of androgen-modulation therapy in breast cancer. This is particularly relevant as current clinical trials focus on inhibiting androgen action as breast cancer therapy but, depending on the steroid receptor profile of the tumour, certain individuals may be better served by selectively stimulating androgen action. Androgen receptor (AR) protein is primarily expressed by the hormone-sensing compartment of normal breast epithelium, commonly referred to as oestrogen receptor alpha (ERa (ESR1))-positive breast epithelial cells, which also express progesterone receptors (PRs) and prolactin receptors and exert powerful developmental influences on adjacent breast epithelial cells. Recent lineage-tracing studies, particularly those focussed on NOTCH signalling, and genetic analysis of cancer risk in the normal breast highlight how signalling via the hormone-sensing compartment can influence normal breast development and breast cancer susceptibility. This provides an impetus to focus on the relationship between androgens, AR and NOTCH signalling and the crosstalk between ERa and PR signalling in the hormone-sensing component of breast epithelium in order to unravel the mechanisms behind the ability of androgens to modulate breast cancer initiation and growth.
Collapse
Affiliation(s)
- Gerard A Tarulli
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Lisa M Butler
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
240
|
McNamara KM, Moore NL, Hickey TE, Sasano H, Tilley WD. Complexities of androgen receptor signalling in breast cancer. Endocr Relat Cancer 2014; 21:T161-81. [PMID: 24951107 DOI: 10.1530/erc-14-0243] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
While the clinical benefit of androgen-based therapeutics in breast cancer has been known since the 1940s, we have only recently begun to fully understand the mechanisms of androgen action in breast cancer. Androgen signalling pathways can have either beneficial or deleterious effects in breast cancer depending on the breast cancer subtype and intracellular context. This review discusses our current knowledge of androgen signalling in breast cancer, including the relationship between serum androgens and breast cancer risk, the prognostic significance of androgen receptor (AR) expression in different breast cancer subtypes and the downstream molecular pathways mediating androgen action in breast cancer cells. Intracrine androgen metabolism has also been discussed and proposed as a potential mechanism that may explain some of the reported differences regarding dichotomous androgen actions in breast cancers. A better understanding of AR signalling in this disease is critical given the current resurgence in interest in utilising contemporary AR-directed therapies for breast cancer and the need for biomarkers that will accurately predict clinical response.
Collapse
Affiliation(s)
- Keely M McNamara
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Nicole L Moore
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Theresa E Hickey
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Hironobu Sasano
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Wayne D Tilley
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| |
Collapse
|
241
|
Dvorkin-Gheva A, Hassell JA. Identification of a novel luminal molecular subtype of breast cancer. PLoS One 2014; 9:e103514. [PMID: 25076125 PMCID: PMC4116208 DOI: 10.1371/journal.pone.0103514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/03/2014] [Indexed: 12/31/2022] Open
Abstract
The molecular classification of human breast tumors has afforded insights into subtype specific biological processes, patient prognosis and response to therapies. However, using current methods roughly one quarter of breast tumors cannot be classified into one or another molecular subtype. To explore the possibility that the unclassifiable samples might comprise one or more novel subtypes we employed a collection of publically available breast tumor datasets with accompanying clinical information to assemble 1,593 transcript profiles: 25% of these samples could not be assigned to one of the current molecular subtypes of breast cancer. All of the unclassifiable samples could be grouped into a new molecular subtype, which we termed “luminal-like”. We also identified the luminal-like subtype in an independent collection of tumor samples (NKI295). We found that patients harboring tumors of the luminal-like subtype have a better prognosis than those with basal-like breast cancer, a similar prognosis to those with ERBB2+, luminal B or claudin-low tumors, but a worse prognosis than patients with luminal A or normal-like breast tumors. Our findings suggest the occurrence of another molecular subtype of breast cancer that accounts for the vast majority of previously unclassifiable breast tumors.
Collapse
Affiliation(s)
- Anna Dvorkin-Gheva
- Centre for Functional Genomics, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - John A. Hassell
- Centre for Functional Genomics, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
242
|
Darb-Esfahani S, von Minckwitz G, Denkert C, Ataseven B, Högel B, Mehta K, Kaltenecker G, Rüdiger T, Pfitzner B, Kittel K, Fiedler B, Baumann K, Moll R, Dietel M, Eidtmann H, Thomssen C, Loibl S. Gross cystic disease fluid protein 15 (GCDFP-15) expression in breast cancer subtypes. BMC Cancer 2014; 14:546. [PMID: 25070172 PMCID: PMC4122770 DOI: 10.1186/1471-2407-14-546] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 07/16/2014] [Indexed: 11/12/2022] Open
Abstract
Background Gross cystic disease fluid protein 15 (GCDFP-15), which is regulated by the androgen receptor (AR), is a diagnostic marker for mammary differentiation in histopathology. We determined the expression of GCDFP-15 in breast cancer subtypes, its potential prognostic and predictive value, as well as its relationship to AR expression. Methods 602 pre-therapeutic breast cancer core biopsies from the phase III randomized neoadjuvant GeparTrio trial (NCT00544765) were investigated for GCDFP-15 expression by immunohistochemistry. Expression data were correlated with disease-free (DFS) and overall survival (OS) time as well as pathological complete response (pCR) to neoadjuvant chemotherapy. Results 239 tumors (39.7%) were GCDFP-15 positive. GCDFP-15 expression was positively linked to hormone receptor (HR) and HER2 positive tumor type, while most triple negative carcinomas were negative (p < 0.0001). GCDFP-15 was also strongly correlated to AR expression (p 0.001), and to the so-called molecular apocrine subtype (HR-/AR+, p < 0.0001). Higher rates of GCDFP-15 positivity were seen in tumors of lower grade (<0.0001) and negative nodal status (p = 0.008). GCDFP-15 positive tumors tended to have a more favourable prognosis than GCDFP-15 negative tumors (DFS (p = 0.052) and OS (p = 0.044)), which was not independent from other factors in multivariate analysis. GCDFP-15 expression was not linked to pCR. Histological apocrine differentiation was frequent in molecular apocrine carcinomas (60.7%), and was associated with GCDFP-15 within this group (p = 0.039). Conclusions GCDFP-15 expression is higher in tumors with favorable prognostic features. GCDFP-15 expression is further a frequent feature of AR positive tumors and the molecular apocrine subtype. It might have reduced sensitivity as a diagnostic marker for mammary differentiation in triple negative tumors as compared to HR or HER2 positive tumor types. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-546) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silvia Darb-Esfahani
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Frietze S, O'Geen H, Littlepage LE, Simion C, Sweeney CA, Farnham PJ, Krig SR. Global analysis of ZNF217 chromatin occupancy in the breast cancer cell genome reveals an association with ERalpha. BMC Genomics 2014; 15:520. [PMID: 24962896 PMCID: PMC4082627 DOI: 10.1186/1471-2164-15-520] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 06/18/2014] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The ZNF217 gene, encoding a C2H2 zinc finger protein, is located at 20q13 and found amplified and overexpressed in greater than 20% of breast tumors. Current studies indicate ZNF217 drives tumorigenesis, yet the regulatory mechanisms of ZNF217 are largely unknown. Because ZNF217 associates with chromatin modifying enzymes, we postulate that ZNF217 functions to regulate specific gene signaling networks. Here, we present a large-scale functional genomic analysis of ZNF217, which provides insights into the regulatory role of ZNF217 in MCF7 breast cancer cells. RESULTS ChIP-seq analysis reveals that the majority of ZNF217 binding sites are located at distal regulatory regions associated with the chromatin marks H3K27ac and H3K4me1. Analysis of ChIP-seq transcription factor binding sites shows clustering of ZNF217 with FOXA1, GATA3 and ERalpha binding sites, supported by the enrichment of corresponding motifs for the ERalpha-associated cis-regulatory sequences. ERalpha expression highly correlates with ZNF217 in lysates from breast tumors (n = 15), and ERalpha co-precipitates ZNF217 and its binding partner CtBP2 from nuclear extracts. Transcriptome profiling following ZNF217 depletion identifies differentially expressed genes co-bound by ZNF217 and ERalpha; gene ontology suggests a role for ZNF217-ERalpha in expression programs associated with ER+ breast cancer studies found in the Molecular Signature Database. Data-mining of expression data from breast cancer patients correlates ZNF217 with reduced overall survival. CONCLUSIONS Our genome-wide ZNF217 data suggests a functional role for ZNF217 at ERalpha target genes. Future studies will investigate whether ZNF217 expression contributes to aberrant ERalpha regulatory events in ER+ breast cancer and hormone resistance.
Collapse
Affiliation(s)
- Seth Frietze
- School of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA.
| | | | | | | | | | | | | |
Collapse
|
244
|
Lim E, Ni M, Cao S, Hazra A, Tamimi RM, Brown M. Importance of Breast Cancer Subtype in the Development of Androgen Receptor Directed Therapy. CURRENT BREAST CANCER REPORTS 2014; 6:71-78. [PMID: 24860642 PMCID: PMC4026357 DOI: 10.1007/s12609-014-0140-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The androgen receptor (AR) has re-emerged as a potential therapeutic target in breast cancer. This stems from recent progress made in preclinical models, that have recognized important differences in the effect of AR expression on patient outcomes among different breast cancer subtypes. In parallel, the clinical development of new generations of AR directed therapies for prostate cancer has begun to mature. The availability of these new agents has translated into new trials to treat breast cancer. It is critical that studies of the effect of AR expression and signaling in breast cancer be context and subtype specific in order to successfully target AR signalling as a therapeutic strategy for breast cancer. We will review developments in preclinical studies, and recent clinical trials targeting AR in breast cancer.
Collapse
|
245
|
Abstract
Breast cancer (BC) is traditionally viewed as an oestrogen-dependent disease in which the androgen receptor (AR) is inhibitory, counteracting the oncogenic activity of oestrogen receptor α (ERα (ESR1)). Most probably as a result of this crosstalk, the AR has prognostic value in ER-positive disease, with AR positivity reported to correlate with a better prognosis. Activation of the AR pathway has been previously used as a therapeutic strategy to treat BC, but its usage declined following the introduction of the anti-oestrogen tamoxifen. More recently, it has been demonstrated that a subset of triple-negative BCs (molecular apocrine) are dependent upon androgen signalling for growth and therapies that inhibit androgen signalling, currently used for the treatment of prostate cancer, e.g. the antiandrogen bicalutamide and the CYP17 inhibitor abiraterone acetate are undergoing clinical trials to investigate their efficacy in this BC subtype. This review summarises the current knowledge of AR activity in BC.
Collapse
Affiliation(s)
- F M Fioretti
- Androgen Signalling LaboratoryDepartment of Surgery and Cancer, Imperial College London, London W12 0NN, UKMolecular OncologySchool of Biological Sciences, University of Essex, Colchester, Essex CO4 3SQ, UK
| | - A Sita-Lumsden
- Androgen Signalling LaboratoryDepartment of Surgery and Cancer, Imperial College London, London W12 0NN, UKMolecular OncologySchool of Biological Sciences, University of Essex, Colchester, Essex CO4 3SQ, UK
| | - C L Bevan
- Androgen Signalling LaboratoryDepartment of Surgery and Cancer, Imperial College London, London W12 0NN, UKMolecular OncologySchool of Biological Sciences, University of Essex, Colchester, Essex CO4 3SQ, UK
| | - G N Brooke
- Androgen Signalling LaboratoryDepartment of Surgery and Cancer, Imperial College London, London W12 0NN, UKMolecular OncologySchool of Biological Sciences, University of Essex, Colchester, Essex CO4 3SQ, UKAndrogen Signalling LaboratoryDepartment of Surgery and Cancer, Imperial College London, London W12 0NN, UKMolecular OncologySchool of Biological Sciences, University of Essex, Colchester, Essex CO4 3SQ, UK
| |
Collapse
|
246
|
McNamara KM, Yoda T, Nurani AM, Shibahara Y, Miki Y, Wang L, Nakamura Y, Suzuki K, Yang Y, Abe E, Hirakawa H, Suzuki T, Nemoto N, Miyashita M, Tamaki K, Ishida T, Brown KA, Ohuchi N, Sasano H. Androgenic pathways in the progression of triple-negative breast carcinoma: a comparison between aggressive and non-aggressive subtypes. Breast Cancer Res Treat 2014; 145:281-93. [PMID: 24715382 DOI: 10.1007/s10549-014-2942-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/27/2014] [Indexed: 12/21/2022]
Abstract
One of the active intracellular pathways/networks in triple-negative breast carcinoma (TNBC) is that of the androgen receptor (AR). In this study, we examined AR and androgen-metabolising enzyme immunoreactivity in subcategories of TNBC to further elucidate the roles of androgenic pathways in TNBC. We utilised formalin-fixed paraffin-embedded breast cancer samples from ductal carcinoma in situ (DCIS) and invasive ductal carcinoma patient cohorts. We then used immunohistochemistry to classify these samples into basal-like and non-basal samples and to assess interactions between AR, androgen-metabolising enzymes and proliferation. To further substantiate our hypothesis and provide mechanistic insights, we also looked at the expression and regulation of these factors in publically available microarray data and in a panel of TNBC AR-positive cell lines. DCIS was associated with higher levels of AR and enzymes (p < 0.02), although a similar difference was not noticed in basal and non-basal samples. AR and enzymes were correlated in all states. In TNBC cell lines (MDA-MD-453, MFM-223 and SUM185-PE), we found that DHT treatment up-regulated 5αR1 and 17βHSD5 suggesting a mechanistic explanation for the correlations observed in the histological samples. Publicly available microarray data in TNBC cases suggested similar patterns to those observed in histological samples. In the majority of settings, including publically available microarray data, an inverse association between AR and proliferation was detected. These findings suggest that decreases in AR and androgen-metabolising enzymes may be involved in the increased biological aggressiveness in TNBC development.
Collapse
MESH Headings
- 3-Hydroxysteroid Dehydrogenases/genetics
- 3-Hydroxysteroid Dehydrogenases/metabolism
- Aldo-Keto Reductase Family 1 Member C3
- Androgens/metabolism
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Line, Tumor
- Cell Proliferation
- Cholestenone 5 alpha-Reductase/genetics
- Cholestenone 5 alpha-Reductase/metabolism
- Dihydrotestosterone/pharmacology
- ErbB Receptors/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hydroxyprostaglandin Dehydrogenases/genetics
- Hydroxyprostaglandin Dehydrogenases/metabolism
- Keratin-5/metabolism
- Keratin-6/metabolism
- Receptors, Androgen/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
Collapse
Affiliation(s)
- Keely M McNamara
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Lakis S, Kotoula V, Eleftheraki AG, Batistatou A, Bobos M, Koletsa T, Timotheadou E, Chrisafi S, Pentheroudakis G, Koutras A, Zagouri F, Linardou H, Fountzilas G. The androgen receptor as a surrogate marker for molecular apocrine breast cancer subtyping. Breast 2014; 23:234-43. [PMID: 24703723 DOI: 10.1016/j.breast.2014.02.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 01/30/2014] [Accepted: 02/28/2014] [Indexed: 01/02/2023] Open
Abstract
The Androgen Receptor (AR) is a potential prognostic marker and therapeutic target in breast cancer. We evaluated AR protein expression in high-risk breast cancer treated in the adjuvant setting. Tumors were subtyped into luminal (ER+/PgR±/AR±), molecular apocrine (MAC, [ER-/PgR-/AR+]) and hormone receptor negative carcinomas (HR-negative, [ER-/PgR-/AR-]). Subtyping was evaluated with respect to prognosis and to taxane therapy. High histologic grade (p < 0.001) and increased proliferation (p = 0.001) more often appeared in MAC and HR-negative than in luminal tumors. Patients with MAC had outcome comparable to the luminal group, while patients with HR-negative disease had increased risk for relapse and death. MAC outcome was favorable upon taxane-containing treatment; this remained significant upon multivariate analysis for overall survival (HR 0.31, 95%CI 0.13-0.74, interaction p = 0.035) and as a trend for time to relapse (p = 0.15). In conclusion, AR-related subtyping of breast cancer may be prognostic and serve for selecting optimal treatment combinations.
Collapse
Affiliation(s)
- Sotiris Lakis
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece.
| | - Vassiliki Kotoula
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece; Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | | | - Anna Batistatou
- Department of Pathology, Ioannina University Hospital, Ioannina, Greece
| | - Mattheos Bobos
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Triantafyllia Koletsa
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Eleni Timotheadou
- Department of Medical Oncology, "Papageorgiou" Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Sofia Chrisafi
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | | | - Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, "Alexandra" Hospital, University of Athens School of Medicine, Athens, Greece
| | - Helena Linardou
- First Department of Medical Oncology, "Metropolitan" Hospital, Piraeus, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece; Department of Medical Oncology, "Papageorgiou" Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| |
Collapse
|
248
|
Schmadeka R, Harmon BE, Singh M. Triple-negative breast carcinoma: current and emerging concepts. Am J Clin Pathol 2014; 141:462-77. [PMID: 24619745 DOI: 10.1309/ajcpqn8gz8silkgn] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES Triple-negative breast cancer is regarded as an aggressive disease that affects a young patient population and for which effective targeted therapy is not yet available. METHODS Intense efforts have been made to gain a better understanding of this heterogeneous group of tumors from the histologic to the genomic and molecular levels. RESULTS Progress has been made, including the ability to subtype these tumors and the discovery of biomarkers toward which current therapeutic efforts are focused. Many novel targets under exploration have the potential to affect the clinical course of this disease. CONCLUSIONS This article reviews the current concepts regarding the clinicopathologic features of triple-negative breast carcinoma, its histologic subtypes, molecular classification, the prognostic and therapeutic potential of biomarkers, and emerging targeted therapies.
Collapse
Affiliation(s)
- Robert Schmadeka
- Department of Pathology, Stony Brook University School of Medicine, State University of New York at Stony Brook, Stony Brook, NY
| | - Bryan E. Harmon
- Department of Pathology, Stony Brook University School of Medicine, State University of New York at Stony Brook, Stony Brook, NY
| | - Meenakshi Singh
- Department of Pathology, Stony Brook University School of Medicine, State University of New York at Stony Brook, Stony Brook, NY
| |
Collapse
|
249
|
Tsang JYS, Ni YB, Chan SK, Shao MM, Law BKB, Tan PH, Tse GM. Androgen receptor expression shows distinctive significance in ER positive and negative breast cancers. Ann Surg Oncol 2014; 21:2218-28. [PMID: 24639191 DOI: 10.1245/s10434-014-3629-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND Androgen receptor (AR), a nuclear steroid hormone receptor, is differentially expressed in breast cancer subgroups with distinct clinical implications. METHODS To investigate the clinical significance of AR in breast cancers more precisely, the expression of AR in a large cohort of breast cancer was correlated with clinicopathological features, biomarker expression, and patients' survival according to different molecular groupings in this study. RESULTS Higher AR expression was found in ER+ (57.8 %) than in ER- (24.7 %) cancers. In the ER+ cancers, AR expression was associated with favorable clinicopathological features, including lower grade (p < .001), lower pT stage (p < .001), and positivity for PR (p < .001). It was an independent prognostic factor for longer disease-free survival, mainly in the HER2+ luminal B cancers (hazard ratio [HR] = 0.251, 95 % CI 0.065-0.972, p = .045). In ER- cancers, AR expression was associated with features distinct from basal-like breast cancer, and such features were found in molecular apocrine (MA) cancers. AR correlated with presence of extensive in situ component (p = .006) and apocrine phenotype (p < .001), HER2 (p = .026), and EGFR (p = .048), but negatively with c-kit (p = .041), CK5/6 (p < .001), CK14 (p = .002), and αB-crystallin (p = .038). However, AR expression was found only in 37.8 % of immunohistochemically defined MA. Of note, AR-MA appeared to have a trend of worse overall survival than AR+MA. CONCLUSIONS AR expression was different in ER+ and ER- cancers and had different clinical implications. AR alone may not be a good marker for MA subtype. Its expression in MA may have substantial prognostic implication and as such warrants further validation.
Collapse
Affiliation(s)
- Julia Y S Tsang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
250
|
Thike AA, Yong-Zheng Chong L, Cheok PY, Li HH, Wai-Cheong Yip G, Huat Bay B, Tse GMK, Iqbal J, Tan PH. Loss of androgen receptor expression predicts early recurrence in triple-negative and basal-like breast cancer. Mod Pathol 2014; 27:352-60. [PMID: 23929266 DOI: 10.1038/modpathol.2013.145] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/20/2013] [Accepted: 06/21/2013] [Indexed: 01/24/2023]
Abstract
Treatment of triple-negative invasive breast cancers, defined by the absence of estrogen and progesterone receptors and c-erbB2 expression, remains challenging. Androgen receptor, a member of the nuclear receptor superfamily that is involved in signaling pathways regulating cell proliferation, has been implicated in breast tumorigenesis. We immunohistochemically examined the expression of androgen receptor, basal markers (CK14, 34βE12) and EGFR in 699 triple-negative invasive breast cancers in tissue microarrays using the streptavidin-biotin method, and correlated the findings with clinical outcome. Positive androgen receptor expression was defined as staining of 1% or more of tumor cell nuclei. Survival outcomes were estimated with the Kaplan-Meier method and compared between groups with log-rank statistics. Cox proportional hazards models were used to determine the effect of androgen receptor on survival outcomes. Immunohistochemical positivity was observed in 38% of tumors, with the proportion of stained tumor cells ranging from 1 to 95% (mean 29%, median 10%). Androgen receptor expression was inversely associated with histologic grade and mitotic score. CK14, 34βE12 and EGFR confirmed 85% of cases to be basal-like, without significant association of basal-like phenotype with androgen receptor expression. Disease-free survival was significantly better in androgen receptor-positive triple-negative breast cancer, with a trend for improved overall survival. Decreased recurrence likelihood in both triple-negative and basal-like tumors (hazard ratio, 0.704; 95% confidence intervals, 0.498-0.994; P=0.0464; and hazard ratio, 0.675; 95% confidence intervals, 0.468-0.974; P=0.0355, respectively) was noted within 5 years of diagnosis but not thereafter. Our study suggests that loss of androgen receptor in triple-negative breast cancers augurs a worse prognosis, including those with basal-like features. More work in elucidating its relationship with mechanisms of progression, as well as trials of targeted treatment for androgen receptor-expressing triple-negative tumors, needs to be performed.
Collapse
Affiliation(s)
- Aye Aye Thike
- 1] Department of Pathology, Singapore General Hospital, Singapore, Singapore [2] Department of Clinical Research, Singapore General Hospital, Singapore, Singapore [3] Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore [4] Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Luke Yong-Zheng Chong
- 1] Department of Pathology, Singapore General Hospital, Singapore, Singapore [2] Department of Clinical Research, Singapore General Hospital, Singapore, Singapore [3] Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore [4] Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Poh Yian Cheok
- Department of Clinical Research, Singapore General Hospital, Singapore, Singapore
| | - Hui Hua Li
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - George Wai-Cheong Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Boon Huat Bay
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Gary Man-Kit Tse
- 1] Department of Pathology, Singapore General Hospital, Singapore, Singapore [2] Department of Clinical Research, Singapore General Hospital, Singapore, Singapore [3] Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore [4] Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Jabed Iqbal
- 1] Department of Pathology, Singapore General Hospital, Singapore, Singapore [2] Department of Clinical Research, Singapore General Hospital, Singapore, Singapore [3] Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore [4] Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Puay Hoon Tan
- 1] Department of Pathology, Singapore General Hospital, Singapore, Singapore [2] Department of Clinical Research, Singapore General Hospital, Singapore, Singapore [3] Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore [4] Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|