151
|
Bataillon G, Fuhrmann L, Girard E, Menet E, Laé M, Capovilla M, Treilleux I, Arnould L, Penault-Llorca F, Rouzier R, Marchiò C, Bieche I, Vincent-Salomon A. High rate of PIK3CA mutations but no TP53 mutations in low-grade adenosquamous carcinoma of the breast. Histopathology 2018. [PMID: 29537649 DOI: 10.1111/his.13514] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
AIMS Low-grade adenosquamous carcinoma of the breast (LGASC) is a rare variant of metaplastic carcinoma characterised by a favourable outcome and histologically composed of glandular and squamous elements in a spindle cell background typically associated with a lymphocytic stromal reaction. Because of its rarity, the immunophenotypic and genetic profile of LGASC has not been sufficiently characterised. The aim of this study was to gain insights into the molecular and phenotypic characteristics of LGASC. METHODS AND RESULTS We reviewed the clinical and morphological features and detailed the immunohistochemical characteristics of a retrospective series of 13 LGASCs. Targeted sequencing of 50 genes was performed in 10 of 13 cases. Identified mutations were further assessed by Sanger sequencing in a validation series of 11 additional cases. All tumours showed a triple-negative immunophenotype, expressed 'basal' keratins, showed variable levels of epidermal growth factor receptor expression, and did not express androgen receptor. Sequencing analysis of the screening set of LGASCs revealed a high rate (seven of 10 cases) of PIK3CA mutations, whereas no TP53 mutations were found. All PIK3CA mutations were missense mutations located either in exon 20 (n = 6) or in exon 9 (n = 1). The global PIK3CA mutation rate, including the validation series, was 52% (11 of 21 cases). No disease recurrences were observed. [Correction added on 11 June 2018, after first online publication: The percentage of mutation rate was corrected to 52%] CONCLUSIONS: Our results indicate that LGASC of the breast is a low-grade triple-negative breast cancer that harbours a basal-like phenotype with no androgen receptor expression, and shows a high rate of PIK3CA mutations but no TP53 mutations.
Collapse
Affiliation(s)
- Guillaume Bataillon
- Institut Curie, Paris Sciences Lettres Research University, Pôle de médecine diagnostique et théranostique, Paris, France
| | - Laetitia Fuhrmann
- Institut Curie, Paris Sciences Lettres Research University, Pôle de médecine diagnostique et théranostique, Paris, France
| | - Elodie Girard
- Institut Curie, Paris Sciences Lettres Research University, Pôle de médecine diagnostique et théranostique, Paris, France
| | - Emanuelle Menet
- Institut Curie, Versailles Saint Quentin University, Pôle de médecine diagnostique et théranostique, Saint-Cloud, France
| | - Marick Laé
- Institut Curie, Paris Sciences Lettres Research University, Pôle de médecine diagnostique et théranostique, Paris, France
| | | | | | - Laurent Arnould
- Department of Pathology, Centre Georges François Leclerc, Dijon, France
| | | | - Roman Rouzier
- Institut Curie, Versailles Saint Quentin University, Surgery Department, Saint-Cloud, France
| | - Caterina Marchiò
- Institut Curie, Paris Sciences Lettres Research University, Pôle de médecine diagnostique et théranostique, Paris, France.,Department of Medical Sciences, University of Turin, Turin, Italy
| | - Ivan Bieche
- Institut Curie, Paris Sciences Lettres Research University, Pôle de médecine diagnostique et théranostique, Paris, France
| | - Anne Vincent-Salomon
- Institut Curie, Paris Sciences Lettres Research University, Pôle de médecine diagnostique et théranostique, Paris, France
| |
Collapse
|
152
|
Africander D, Storbeck KH. Steroid metabolism in breast cancer: Where are we and what are we missing? Mol Cell Endocrinol 2018; 466:86-97. [PMID: 28527781 DOI: 10.1016/j.mce.2017.05.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
It is well-known that breast cancer is hormone-dependent and that steroid hormones exert their mitogenic effects by binding to estrogen, progesterone and androgen receptors. Vital to our understanding and treatment of this malignancy, is the local metabolism of steroid hormones in breast cancer tissue. This review summarises our current knowledge on steroid producing pathways in the adrenal, ovary and breast, while focussing on the availability of specific circulating hormone precursors and steroidogenic enzymes involved in the local synthesis and metabolism of steroid hormones in the breast. Consequently, we highlight alternate pathways that may be instrumental in the etiology of breast cancer.
Collapse
Affiliation(s)
- Donita Africander
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa.
| |
Collapse
|
153
|
Augusto TV, Correia-da-Silva G, Rodrigues CMP, Teixeira N, Amaral C. Acquired resistance to aromatase inhibitors: where we stand! Endocr Relat Cancer 2018. [PMID: 29530940 DOI: 10.1530/erc-17-0425] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aromatase inhibitors (AIs) are one of the principal therapeutic approaches for estrogen receptor-positive (ER+) breast cancer in postmenopausal women. They block estrogen biosynthesis through aromatase inhibition, thus preventing tumour progression. Besides the therapeutic success of the third-generation AIs, acquired resistance may develop, leading to tumour relapse. This resistance is thought to be the result of a change in the behaviour of ER in these breast cancer cells, presumably by PI3K/AKT pathway enhancement along with alterations in other signalling pathways. Nevertheless, biological mechanisms, such as apoptosis, autophagy, cell cycle modulation and activation of androgen receptor (AR), are also implicated in acquired resistance. Moreover, clinical evidence demonstrated that there is a lack of cross-resistance among AIs, although the reason is not fully understood. Thus, there is a demand to understand the mechanisms involved in endocrine resistance to each AI, since the search for new strategies to surpass breast cancer acquired resistance is of major concern.
Collapse
Affiliation(s)
- Tiago Vieira Augusto
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Natércia Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
154
|
Liao Y, Xia X, Liu N, Cai J, Guo Z, Li Y, Jiang L, Dou QP, Tang D, Huang H, Liu J. Growth arrest and apoptosis induction in androgen receptor-positive human breast cancer cells by inhibition of USP14-mediated androgen receptor deubiquitination. Oncogene 2018; 37:1896-1910. [PMID: 29353883 PMCID: PMC5886989 DOI: 10.1038/s41388-017-0069-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/23/2017] [Accepted: 09/19/2017] [Indexed: 11/10/2022]
Abstract
It has been well known that androgen receptor (AR) is critical to prostate cancer development and progression. It has also been documented that AR is expressed in more than 60% of breast tumors, which promotes the growth of estrogen receptor-negative (ER-)/AR-positive (AR+) breast cancer cells. Thus, AR might be a potential therapeutic target for AR-positive/ER-negative breast cancer patients. Previously we reported that in prostate cancer cells proteasome-associated deubiquitinase ubiquitin-specific protease 14 (USP14) stabilized AR protein level by removing its ubiquitin chain. In the current study, we studied the USP14-AR protein interaction and cell proliferation status after USP14 reduction or inhibition in breast cancer cells, and our results support the conclusion that targeting USP14 is a novel strategy for treating AR-responsive breast cancer. We found that inhibition of USP14 accelerated the K48-ubiquitination and proteasome-mediated degradation of AR protein. Additionally, both genetic and pharmacological inhibition of USP14 significantly suppressed cell proliferation in AR-responsive breast cancer cells by blocking G0/G1 to S phase transition and inducing apoptosis. Moreover, AR overexpression inhibited USP14 inhibition-induced events, suggesting that AR deubiquitination by USP14 is critical for breast cancer growth and USP14 inhibition is a possible strategy to treat AR-positive breast cancer.
Collapse
Affiliation(s)
- Yuning Liao
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohong Xia
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianyu Cai
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiqiang Guo
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanling Li
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lili Jiang
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Q Ping Dou
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
- Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Daolin Tang
- The Third Affiliated Hospital, Center for DAMP Biology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Protein Modification and Degradation Laboratory, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hongbiao Huang
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Jinbao Liu
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
155
|
Bronte G, Rocca A, Ravaioli S, Puccetti M, Tumedei MM, Scarpi E, Andreis D, Maltoni R, Sarti S, Cecconetto L, Fedeli A, Pietri E, De Simone V, Asioli S, Amadori D, Bravaccini S. Androgen receptor in advanced breast cancer: is it useful to predict the efficacy of anti-estrogen therapy? BMC Cancer 2018; 18:348. [PMID: 29587674 PMCID: PMC5872575 DOI: 10.1186/s12885-018-4239-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/16/2018] [Indexed: 01/20/2023] Open
Abstract
Background Androgen receptor (AR) is widely expressed in breast cancer (BC) but its role in estrogen receptor (ER)-positive tumors is still controversial. The AR/ER ratio has been reported to impact prognosis and response to antiestrogen endocrine therapy (ET). Methods We assessed whether AR in primary tumors and/or matched metastases is a predictor of efficacy of first-line ET in advanced BC. Patients who had received first-line ET (2002–2011) were recruited, while those given concomitant chemotherapy or trastuzumab or pretreated with > 2 lines of chemotherapy were excluded. ER, progesterone receptor (PgR), Ki67 and AR expression were assessed by immunohistochemistry, and HER2 mainly by fluorescent in-situ hybridization. Cut-offs of 1 and 10% immunostained cells were used to categorize AR expression. Results Among 102 evaluable patients, biomarkers were assessed in primary tumors in 70 cases and in metastases in 49, with 17 patients having both determinations. The overall concordance rate between primary tumors and metastases was 64.7% (95% CI 42%-87.4%) for AR status. AR status did not affect TTP significantly, whereas PgR and Ki67 status did. AR/PgR ≥0.96 was associated with a significantly shorter TTP (HR = 1.65, 95% CI 1.05-2.61, p = 0.028). AR status in primary tumors or metastases was not associated with progressive disease (PD) as best response. In contrast, Ki67 ≥ 20% and PgR < 10% showed a statistically significant association with PD as best response. Conclusions AR expression does not appear to be useful to predict the efficacy of ET in advanced BC, whereas Ki67 and PgR exert a greater impact on its efficacy.
Collapse
Affiliation(s)
- Giuseppe Bronte
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Andrea Rocca
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy.
| | - Sara Ravaioli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | | | - Maria Maddalena Tumedei
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Emanuela Scarpi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Daniele Andreis
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Roberta Maltoni
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Samanta Sarti
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Lorenzo Cecconetto
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Anna Fedeli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Elisabetta Pietri
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Valeria De Simone
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Silvia Asioli
- Department of Pathology, Morgagni-Pierantoni Hospital, 47121, Forlì, Italy
| | - Dino Amadori
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy.
| |
Collapse
|
156
|
Traina TA, Miller K, Yardley DA, Eakle J, Schwartzberg LS, O’Shaughnessy J, Gradishar W, Schmid P, Winer E, Kelly C, Nanda R, Gucalp A, Awada A, Garcia-Estevez L, Trudeau ME, Steinberg J, Uppal H, Tudor IC, Peterson A, Cortes J. Enzalutamide for the Treatment of Androgen Receptor-Expressing Triple-Negative Breast Cancer. J Clin Oncol 2018; 36:884-890. [PMID: 29373071 PMCID: PMC5858523 DOI: 10.1200/jco.2016.71.3495] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose Studies suggest that a subset of patients with triple-negative breast cancer (TNBC) have tumors that express the androgen receptor (AR) and may benefit from an AR inhibitor. This phase II study evaluated the antitumor activity and safety of enzalutamide in patients with locally advanced or metastatic AR-positive TNBC. Patients and Methods Tumors were tested for AR with an immunohistochemistry assay optimized for breast cancer; nuclear AR staining > 0% was considered positive. Patients received enzalutamide 160 mg once per day until disease progression. The primary end point was clinical benefit rate (CBR) at 16 weeks. Secondary end points included CBR at 24 weeks, progression-free survival, and safety. End points were analyzed in all enrolled patients (the intent-to-treat [ITT] population) and in patients with one or more postbaseline assessment whose tumor expressed ≥ 10% nuclear AR (the evaluable subgroup). Results Of 118 patients enrolled, 78 were evaluable. CBR at 16 weeks was 25% (95% CI, 17% to 33%) in the ITT population and 33% (95% CI, 23% to 45%) in the evaluable subgroup. Median progression-free survival was 2.9 months (95% CI, 1.9 to 3.7 months) in the ITT population and 3.3 months (95% CI, 1.9 to 4.1 months) in the evaluable subgroup. Median overall survival was 12.7 months (95% CI, 8.5 months to not yet reached) in the ITT population and 17.6 months (95% CI, 11.6 months to not yet reached) in the evaluable subgroup. Fatigue was the only treatment-related grade 3 or higher adverse event with an incidence of > 2%. Conclusion Enzalutamide demonstrated clinical activity and was well tolerated in patients with advanced AR-positive TNBC. Adverse events related to enzalutamide were consistent with its known safety profile. This study supports additional development of enzalutamide in advanced TNBC.
Collapse
Affiliation(s)
- Tiffany A. Traina
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Kathy Miller
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Denise A. Yardley
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Janice Eakle
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Lee S. Schwartzberg
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Joyce O’Shaughnessy
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - William Gradishar
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Peter Schmid
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Eric Winer
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Catherine Kelly
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Rita Nanda
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Ayca Gucalp
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Ahmad Awada
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Laura Garcia-Estevez
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Maureen E. Trudeau
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Joyce Steinberg
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Hirdesh Uppal
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Iulia Cristina Tudor
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Amy Peterson
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| | - Javier Cortes
- Tiffany A. Traina and Ayca Gucalp, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Kathy Miller, Indiana University Simon Cancer Center, Indianapolis, IN; Denise A. Yardley, Tennessee Oncology, Nashville; Lee S. Schwartzberg, The West Clinic, Memphis, TN; Janice Eakle, Florida Cancer Specialists, Fort Myers, FL; Joyce O’Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology, Dallas, TX; William Gradishar, Northwestern University Feinberg School of Medicine; Rita Nanda, University of Chicago, Chicago; Joyce Steinberg, Astellas Pharma, Northbrook, IL; Peter Schmid, Barts Cancer Institute, Queen Mary University London, London, United Kingdom; Eric Winer, Dana-Farber Cancer Institute, Boston, MA; Catherine Kelly, All Ireland Collaborative Oncology Research Group, Dublin, Ireland; Ahmad Awada, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Laura Garcia-Estevez, Centro Integral Oncologico Clara Campal, Hospital Madrid Norte-Sanchinarro; Javier Cortes, Ramon y Cajal University Hospital, Madrid, and, Vall d’Hebron Institute of Oncology and Baselga Oncological Institute, Barcelona, Spain; Maureen E. Trudeau, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; and Hirdesh Uppal, Amy Peterson, and Iulia Cristina Tudor, Medivation, San Francisco, CA
| |
Collapse
|
157
|
Ricciardelli C, Bianco-Miotto T, Jindal S, Butler LM, Leung S, McNeil CM, O'Toole SA, Ebrahimie E, Millar EKA, Sakko AJ, Ruiz AI, Vowler SL, Huntsman DG, Birrell SN, Sutherland RL, Palmieri C, Hickey TE, Tilley WD. The Magnitude of Androgen Receptor Positivity in Breast Cancer Is Critical for Reliable Prediction of Disease Outcome. Clin Cancer Res 2018. [PMID: 29514843 DOI: 10.1158/1078-0432.ccr-17-1199] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: Consensus is lacking regarding the androgen receptor (AR) as a prognostic marker in breast cancer. The objectives of this study were to comprehensively review the literature on AR prognostication and determine optimal criteria for AR as an independent predictor of breast cancer survival.Experimental Design: AR positivity was assessed by immunostaining in two clinically validated primary breast cancer cohorts [training cohort, n = 219; validation cohort, n = 418; 77% and 79% estrogen receptor alpha (ERα) positive, respectively]. The optimal AR cut-point was determined by ROC analysis in the training cohort and applied to both cohorts.Results: AR was an independent prognostic marker of breast cancer outcome in 22 of 46 (48%) previous studies that performed multivariate analyses. Most studies used cut-points of 1% or 10% nuclear positivity. Herein, neither 1% nor 10% cut-points were robustly prognostic. ROC analysis revealed that a higher AR cut-point (78% positivity) provided optimal sensitivity and specificity to predict breast cancer survival in the training (HR, 0.41; P = 0.015) and validation (HR, 0.50; P = 0.014) cohorts. Tenfold cross-validation confirmed the robustness of this AR cut-point. Patients with ERα-positive tumors and AR positivity ≥78% had the best survival in both cohorts (P < 0.0001). Among the combined ERα-positive cases, those with comparable or higher levels of AR (AR:ERα-positivity ratio >0.87) had the best outcomes (P < 0.0001).Conclusions: This study defines an optimal AR cut-point to reliably predict breast cancer survival. Testing this cut-point in prospective cohorts is warranted for implementation of AR as a prognostic factor in the clinical management of breast cancer. Clin Cancer Res; 24(10); 2328-41. ©2018 AACR.
Collapse
Affiliation(s)
- Carmela Ricciardelli
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Tina Bianco-Miotto
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,School of Agriculture, Food and Wine, University of Adelaide, Adelaide, South Australia, Australia
| | - Shalini Jindal
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Lisa M Butler
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Samuel Leung
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Catriona M McNeil
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Sandra A O'Toole
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Esmaeil Ebrahimie
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Ewan K A Millar
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Andrew J Sakko
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Alexandra I Ruiz
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah L Vowler
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, United Kingdom
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Stephen N Birrell
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Robert L Sutherland
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Carlo Palmieri
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom.,Academic Department of Medical Oncology, Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
158
|
Imawari Y, Mimoto R, Hirooka S, Morikawa T, Takeyama H, Yoshida K. Downregulation of dual-specificity tyrosine-regulated kinase 2 promotes tumor cell proliferation and invasion by enhancing cyclin-dependent kinase 14 expression in breast cancer. Cancer Sci 2018; 109:363-372. [PMID: 29193658 PMCID: PMC5797831 DOI: 10.1111/cas.13459] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 01/16/2023] Open
Abstract
Tumor progression is the main cause of death in patients with breast cancer. Accumulating evidence suggests that dual-specificity tyrosine-regulated kinase 2 (DYRK2) functions as a tumor suppressor by regulating cell survival, differentiation, proliferation and apoptosis. However, little is known about the mechanisms of transcriptional regulation by DYRK2 in cancer progression, particularly with respect to cancer proliferation and invasion. Here, using a comprehensive expression profiling approach, we show that cyclin-dependent kinase 14 (CDK14) is a target of DYRK2. We found that reduced DYRK2 expression increases CDK14 expression, which promotes cancer cell proliferation and invasion in vitro, in addition to tumorigenicity in vivo. CDK14 and DYRK2 expression inversely correlated in human breast cancer tissues. We further identified androgen receptor (AR) as a candidate of DYRK2-dependent transcription factors regulating CDK14. Taken together, our findings suggest a mechanism by which DYRK2 controls CDK14 expression to regulate tumor cell proliferation and invasion in breast cancer. Targeting of this pathway may be a promising therapeutic strategy for treating breast cancer.
Collapse
Affiliation(s)
- Yoshimi Imawari
- Department of BiochemistryJikei University School of MedicineTokyoJapan
- Department of SurgeryJikei University School of MedicineTokyoJapan
| | - Rei Mimoto
- Department of SurgeryJikei University School of MedicineTokyoJapan
| | - Shinichi Hirooka
- Department of PathologyJikei University School of MedicineTokyoJapan
| | | | - Hiroshi Takeyama
- Department of SurgeryJikei University School of MedicineTokyoJapan
| | - Kiyotsugu Yoshida
- Department of BiochemistryJikei University School of MedicineTokyoJapan
| |
Collapse
|
159
|
Kwilas AR, Ardiani A, Gameiro SR, Richards J, Hall AB, Hodge JW. Androgen deprivation therapy sensitizes triple negative breast cancer cells to immune-mediated lysis through androgen receptor independent modulation of osteoprotegerin. Oncotarget 2018; 7:23498-511. [PMID: 27015557 PMCID: PMC5029642 DOI: 10.18632/oncotarget.8274] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/28/2016] [Indexed: 12/31/2022] Open
Abstract
Among breast cancer types, triple-negative breast cancer (TNBC) has the fewest treatment options and the lowest 5-year survival rate. Androgen receptor (AR) inhibition has displayed efficacy against breast cancer preclinically and is currently being examined clinically in AR positive TNBC patients. Androgen deprivation has been shown to induce immunogenic modulation; the alteration of tumor cell phenotype resulting in increased sensitivity to immune-mediated killing. We evaluated the ability of AR inhibition to reduce the growth and improve the immune-mediated killing of breast cancer cells with differing expression of the estrogen receptor and AR. While AR expression was required for the growth inhibitory effects of enzalutamide on breast cancer cells, both enzalutamide and abiraterone improved the sensitivity of breast cancer cells to immune-mediated lysis independent of detectable AR expression. This increase in sensitivity was linked to an increase in cell surface tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor expression as well as a significant reduction in the expression of osteoprotegerin (OPG). The reduction in OPG was further examined and found to be critical for the increase in sensitivity of AR- TNBC cells to immune-mediated killing. The data presented herein further support the use of AR inhibition therapy in the AR+ TNBC setting. These data, however, also support the consideration of AR inhibition therapy for the treatment of AR- TNBC, especially in combination with cancer immunotherapy, providing a potential novel therapeutic option for select patients.
Collapse
Affiliation(s)
- Anna R Kwilas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andressa Ardiani
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jacob Richards
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ashley B Hall
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
160
|
Christenson JL, Trepel JB, Ali HY, Lee S, Eisner JR, Baskin-Bey ES, Elias AD, Richer JK. Harnessing a Different Dependency: How to Identify and Target Androgen Receptor-Positive Versus Quadruple-Negative Breast Cancer. Discov Oncol 2018; 9:82-94. [PMID: 29340907 DOI: 10.1007/s12672-017-0314-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/07/2017] [Indexed: 12/12/2022] Open
Abstract
The androgen receptor (AR) is a promising therapeutic target for a subset of triple-negative breast cancers (TNBCs) in which AR is expressed. However, the mechanistic action of AR and the degree to which primary and metastatic tumors depend on AR, both before and after conventional treatment, remain to be defined. We discuss preclinical and clinical data for AR+ TNBC, the difficulties in monitoring AR protein levels, new methods for determining AR status, the influence of AR on "stemness" in the context of TNBC, the role of combined inhibition of sex steroid production and AR, and the role of AR in regulation of the immune system. Although the exact role of AR in subsets of TNBC is still being characterized, new therapies that target AR and the production of androgens may provide additional options for patients with TNBC for whom chemotherapy is currently the sole treatment option.
Collapse
Affiliation(s)
- Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jane B Trepel
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Sunmin Lee
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | | - Anthony D Elias
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
161
|
Omarini C, Guaitoli G, Pipitone S, Moscetti L, Cortesi L, Cascinu S, Piacentini F. Neoadjuvant treatments in triple-negative breast cancer patients: where we are now and where we are going. Cancer Manag Res 2018; 10:91-103. [PMID: 29391830 PMCID: PMC5772398 DOI: 10.2147/cmar.s146658] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Triple-negative breast cancer (TNBC) remains the poorest-prognosis breast cancer (BC) subtype. Gene expression profiling has identified at least six different triple-negative subtypes with different biology and sensitivity to therapies. The heterogeneous nature of TN tumors may justify the difficulty in treating this BC subtype. Several targeted agents have been investigated in clinical trials without demonstrating a clear survival benefit. Therefore, systemic chemotherapy remains the cornerstone of current clinical practice. Improving the knowledge of tumor biology is mandatory for patient management. In stages II and III, neoadjuvant systemic treatment is an effective option of care. The achievement of a pathological complete response represents an optimal surrogate for survival outcome as well as a test for tumor drug sensitivity. In this review, we provide a brief description of the main predictive biomarkers for tumor response to systemic treatment. Moreover, we review the treatment strategies investigated for TNBCs in neoadjuvant settings focusing on experimental drugs such as immunotherapy and poly [ADP-ribose] polymerase inhibitors that hold promise in the treatment of this aggressive disease. Therefore, the management of TNBC represents an urgent, current, unmet need in daily clinical practice. A key recommendation is to design biology-driven clinical trials wherein TNBC patients may be treated on the basis of tumor molecular profile.
Collapse
Affiliation(s)
- Claudia Omarini
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Giorgia Guaitoli
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Stefania Pipitone
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Luca Moscetti
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Laura Cortesi
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Stefano Cascinu
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Federico Piacentini
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena, Italy
| |
Collapse
|
162
|
Patnayak R, Jena A, Bhargavi D, Chowhan AK. Androgen Receptor Expression in Triple Negative Breast Cancer - Study from a Tertiary Health Care Center in South India. Indian J Med Paediatr Oncol 2018. [DOI: 10.4103/ijmpo.ijmpo_4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Abstract
Background: The treatment of breast cancer is based on a multi-modality approach. Analysis of the hormone receptor has been accepted as a standard procedure, in the routine management of breast cancer patients. Triple negative breast cancers (TNBCs) are those which are negative for expression of all three markers, i.e., estrogen receptor, progesterone receptor (PR), and human epidermal growth factor receptor. High expression of Ki-67, a proliferation index, has been associated with a worse prognosis in TNBC. TN cancers are aggressive in nature as they do not respond to routine targeted therapy. The role of the androgen receptor (AR) in breast carcinomas is important as AR has been suggested as a potential therapeutic target. We did this study to assess AR immunoreactivity in TNBCs and correlate with Ki-67 index. Materials and Methods: In this study group, there were 45 cases of TN invasive breast carcinomas. These tumors were analyzed with respect to AR and Ki-67 index. Results: Of 45 TN breast carcinomas analyzed, 42 were infiltrating duct cell carcinoma (IDCC) of not otherwise specified type. There were one medullary and two metaplastic carcinomas. The median age was 46 years. AR was positive in 20% (9/45) of cases. All the AR positive cases were an IDCC (nos). Out of the nine AR positive cases, six showed positivity for Ki-67. The statistical analysis using Pearson's Chi-squared method did not reveal any correlation between AR and Ki-67 index (P = 0.574). Conclusion: Although our study did not reveal any statistically significant correlation between AR and Ki-67 index, assessment of AR status in TNBC patients is desirable as it may help to develop a targeted therapy in future in these aggressive cancers.
Collapse
Affiliation(s)
- Rashmi Patnayak
- Department of Pathology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Amitabh Jena
- Department of Surgical Oncology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Dandumudu Bhargavi
- Department of Medical Oncology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Amit Kumar Chowhan
- Department of Pathology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| |
Collapse
|
163
|
Zhao H, Li D, Zhang B, Qi Y, Diao Y, Zhen Y, Shu X. PP2A as the Main Node of Therapeutic Strategies and Resistance Reversal in Triple-Negative Breast Cancer. Molecules 2017; 22:molecules22122277. [PMID: 29261144 PMCID: PMC6149800 DOI: 10.3390/molecules22122277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC), is defined as a type of tumor lacking the expression of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2). The ER, PR and HER2 are usually the molecular therapeutic targets for breast cancers, but they are ineffective for TNBC because of their negative expressions, so chemotherapy is currently the main treatment strategy in TNBC. However, drug resistance remains a major impediment to TNBC chemotherapeutic treatment. Recently, the protein phosphatase 2A (PP2A) has been found to regulate the phosphorylation of some substrates involved in the relevant target of TNBC, such as cell cycle control, DNA damage responses, epidermal growth factor receptor, immune modulation and cell death resistance, which may be the effective therapeutic strategies or influence drug sensitivity to TNBCs. Furthermore, PP2A has also been found that could induce ER re-expression in ER-negative breast cancer cells, and which suggests PP2A could promote the sensitivity of tamoxifen to TNBCs as a resistance reversal agent. In this review, we will summarize the potential therapeutic value of PP2A as the main node in developing targeting agents, disrupting resistance or restoring drug sensitivity in TNBC.
Collapse
Affiliation(s)
- Henan Zhao
- Department of Pathophysiology, Dalian Medical University, Dalian 116044, China.
| | - Duojiao Li
- Kamp Pharmaceutical Co. Ltd., Changsha 410008, China.
| | - Baojing Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
164
|
Yu Z, He S, Wang D, Patel HK, Miller CP, Brown JL, Hattersley G, Saeh JC. Selective Androgen Receptor Modulator RAD140 Inhibits the Growth of Androgen/Estrogen Receptor-Positive Breast Cancer Models with a Distinct Mechanism of Action. Clin Cancer Res 2017; 23:7608-7620. [PMID: 28974548 DOI: 10.1158/1078-0432.ccr-17-0670] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/14/2017] [Accepted: 09/28/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Steroidal androgens suppress androgen receptor and estrogen receptor positive (AR/ER+) breast cancer cells and were used to treat breast cancer, eliciting favorable response. The current study evaluates the activity and efficacy of the oral selective AR modulator RAD140 in in vivo and in vitro models of AR/ER+ breast cancer.Experimental Design: A series of in vitro assays were used to determine the affinity of RAD140 to 4 nuclear receptors and evaluate its tissue-selective AR activity. The efficacy and pharmacodynamics of RAD140 as monotherapy or in combination with palbociclib were evaluated in AR/ER+ breast cancer xenograft models.Results: RAD140 bound AR with high affinity and specificity and activated AR in breast cancer but not prostate cancer cells. Oral administration of RAD140 substantially inhibited the growth of AR/ER+ breast cancer patient-derived xenografts (PDX). Activation of AR and suppression of ER pathway, including the ESR1 gene, were seen with RAD140 treatment. Coadministration of RAD140 and palbociclib showed improved efficacy in the AR/ER+ PDX models. In line with efficacy, a subset of AR-repressed genes associated with DNA replication was suppressed with RAD140 treatment, an effect apparently enhanced by concurrent administration of palbociclib.Conclusions: RAD140 is a potent AR agonist in breast cancer cells with a distinct mechanism of action, including the AR-mediated repression of ESR1 It inhibits the growth of multiple AR/ER+ breast cancer PDX models as a single agent, and in combination with palbociclib. The preclinical data presented here support further clinical investigation of RAD140 in AR/ER+ breast cancer patients. Clin Cancer Res; 23(24); 7608-20. ©2017 AACR.
Collapse
Affiliation(s)
- Ziyang Yu
- Radius Health, Inc., Waltham, Massachusetts.
| | - Suqin He
- Radius Health, Inc., Waltham, Massachusetts
| | | | | | | | | | | | | |
Collapse
|
165
|
Lyons TG, Traina TA. Androgen Receptor-Targeted Therapy for Breast Cancer. CURRENT BREAST CANCER REPORTS 2017. [DOI: 10.1007/s12609-017-0261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
166
|
Targeting Androgen Receptor in Treating HER2 Positive Breast Cancer. Sci Rep 2017; 7:14584. [PMID: 29109513 PMCID: PMC5674043 DOI: 10.1038/s41598-017-14607-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 10/12/2017] [Indexed: 11/11/2022] Open
Abstract
Androgen receptor (AR) is widely expressed in different subtypes of breast cancer (BC). However, it is unclear how AR functions in HER2 positive (+) BC. Knockdown of AR with shRNAs and a new generation anti-androgen drug, Enzalutamide, were used to explore the involvement of AR on the growth of HER2 + BC cells (HCC1954 and SKBR3). AR shRNA or Enzalutamide inhibited the growth of SKBR3 cells at a similar extend compared to trastuzumab, an approved HER2 targeted drug. Combining Enzalutamide with trastuzumab further decreased the growth of HCC1954 and SKBR3 cells compared with single agent alone in vitro. Biochemical analysis revealed that inhibiting AR resulted in decreased HER2 phosphorylation and activation of Erk and Akt, without affecting the HER2 and HER3 expression. The in vivo efficacy of Enzalutamide was further tested using the HCC1954 xenograft model. Enzalutamide impaired the growth of HCC1954 tumor at a level comparable to that by trastuzumab. Enzalutamide decreased Ki67 staining and increased activated caspase3 staining compared with vehicle control in HCC1954 tumors. Our results indicate AR plays an important role in promoting the growth of HER2 + BC by cross-talking with the HER2 signaling. AR drug may be used as an alternative second line therapy for treating HER2 + BC.
Collapse
|
167
|
Kumar V, Yu J, Phan V, Tudor IC, Peterson A, Uppal H. Androgen Receptor Immunohistochemistry as a Companion Diagnostic Approach to Predict Clinical Response to Enzalutamide in Triple-Negative Breast Cancer. JCO Precis Oncol 2017; 1:1-19. [DOI: 10.1200/po.17.00075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The androgen receptor (AR) is increasingly recognized as a potential biomarker for identifying a subset of patients with possible hormonally driven triple-negative breast cancer (TNBC). However, its performance as a companion diagnostic remains elusive. Thus, we evaluated AR expression by immunohistochemistry in patients with advanced TNBC before treatment with the AR inhibitor enzalutamide. Methods We optimized and validated immunohistochemistry assays in breast and prostate cancer cell lines and tissues using two commercial AR monoclonal antibodies (SP107 and AR441). AR expression was then examined in patients with advanced TNBC enrolled in a phase II study of enzalutamide (ClinicalTrials.gov identifier: NCT01889238) on archived or fresh tissue before treatment. Association with clinical response was assessed by sensitivity, specificity, positive predictive value (PPV), drop-out rate, and survival. Results AR expression was detected in 80% and 63% of breast cancer tissue using SP107 and AR441, respectively. SP107 was selected for additional analyses because of its higher sensitivity and robustness. Total AR nuclear staining demonstrated the best accuracy in predicting clinical response (area under receiver operating characteristic curve, 0.72; P = .0001). At a threshold of 10%, 74.6% of patients were AR positive, leading to 30% PPV, 90% sensitivity, and 30% specificity. These patients showed a significantly higher median progression-free survival (hazard ratio, 0.56; 95% CI, 0.36 to 0.88; P = .011) and overall survival (hazard ratio, 0.54; 95% CI, 0.32 to 0.91; P = .019) compared with those with AR-negative (< 10%) TNBC. Conclusion At a threshold of ≥ 10% nuclear expression, the AR was associated with TNBC response to enzalutamide. However, the modest PPV may restrict its clinical application, and additional diagnostic tools may be helpful for improved patient selection.
Collapse
Affiliation(s)
| | - Jianjun Yu
- All authors: Medivation, San Francisco, CA
| | | | | | | | | |
Collapse
|
168
|
Majumder A, Singh M, Tyagi SC. Post-menopausal breast cancer: from estrogen to androgen receptor. Oncotarget 2017; 8:102739-102758. [PMID: 29254284 PMCID: PMC5731994 DOI: 10.18632/oncotarget.22156] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022] Open
Abstract
In the United States, breast cancer is the second leading cause of death among women, and even though different therapies can treat primary breast tumors, most breast cancer-related deaths (>95%) occur due to metastasis. A majority (~70%) of breast tumors are found to express estrogen receptor, and a significant portion (~90%) of ER-positive (ER+) breast tumors are also androgen receptor-positive (AR+). Although ER is known to promote tumorigenesis, the role and underlying mechanism(s) of AR in these closely knit processes remain controversial. Endocrine therapies are the most commonly used treatment for patients with ER+ breast tumors; but, ~30%-50% of initially responsive patients develop resistance to these therapies. Whereas 70%–90% of all breast tumors are AR+ and AR overexpression is correlated with endocrine resistance, but the precise molecular mechanism(s) for this association is yet to be studied. Multiple mechanisms have been proposed to show AR and ER interactions, which indicate that AR may preferentially regulate expression of a subset of ER-responsive genes and that may be responsible for breast cancer and its progression in affected patients. On the other hand, most of the ER+ breast tumors found in post-menopausal women (~80%); and they have very low 17β-estradiol and high androgen levels, but how these hormonal changes make someone more prone to cancer phenotype has long been a disputed issue. In this study, we have discussed multiple molecular mechanisms that we believe are central to the understanding of the overall contributions of AR in breast cancer and its metastasis in post-menopausal women.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville 40202, Kentucky, USA.,Department of Physiology, University of Louisville School of Medicine, Louisville 40202, Kentucky, USA
| | - Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine, Louisville 40202, Kentucky, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville 40202, Kentucky, USA
| |
Collapse
|
169
|
Ji Z, Yang L, Ruan Q. Correlation of epidermal growth factor receptor (EGFR), androgen receptor (AR) and 14-3-3 sigma expression in breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10419-10430. [PMID: 31966379 PMCID: PMC6965813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/01/2017] [Indexed: 06/10/2023]
Abstract
Epidermal growth factor receptor (EGFR), androgen receptor (AR) and 14-3-3 sigma have been reported to be implicated in breast tumorigenesis. Their correlations, however, remain elusive in this condition. In order to examine the correlation of EGFR, AR and 14-3-3 sigma in breast cancer, and analyze their relationships with molecular subtypes of breast cancer and their impacts on overall survival, we immunohistochemistrically detected EGFR, AR and 14-3-3 sigma expression in 139 cases of breast cancer. We found that EGFR expression was negatively correlated with AR (r=-0.223, P=0.008) and positively with 14-3-3 sigma expression (r=0.181, P=0.033). There were significant differences in EGFR and AR expression between different molecular subtypes (P=0.000 and P=0.000 respectively). Kaplan-Meier cumulative survival analysis showed that none of the three biomarkers had significant impacts on overall survival of breast cancer patients (P=0.315, P=0.709, P=0.789 respectively). Univariate survival analysis revealed that tumor size (P=0.044), lymph node status (P=0.006) and clinical stage (P=0.008) were significantly associated with overall survival. Multivariate analysis demonstrated that lymph node status was the only statistically significant independent prognostic factor for overall survival [P=0.006, exp (B) =1.511, CI (1.124-2.032)]. In conclusion, EGFR expression is negatively correlated with AR and positively with 14-3-3 sigma expression in breast cancer. Furthermore, there are significant differences in EGFR and AR expression between various molecular subtypes of breast cancer. Lastly, EGFR, AR and 14-3-3 sigma have no significant impacts on overall survival of breast cancer patients.
Collapse
Affiliation(s)
- Zhimin Ji
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Lili Yang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Qiurong Ruan
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
170
|
Fujii T, Reuben JM, Huo L, Espinosa Fernandez JR, Gong Y, Krupa R, Suraneni MV, Graf RP, Lee J, Greene S, Rodriguez A, Dugan L, Louw J, Lim B, Barcenas CH, Marx AN, Tripathy D, Wang Y, Landers M, Dittamore R, Ueno NT. Androgen receptor expression on circulating tumor cells in metastatic breast cancer. PLoS One 2017; 12:e0185231. [PMID: 28957377 PMCID: PMC5619732 DOI: 10.1371/journal.pone.0185231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/09/2017] [Indexed: 11/19/2022] Open
Abstract
Purpose Methods Results Conclusions
Collapse
Affiliation(s)
- Takeo Fujii
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - James M. Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jose Rodrigo Espinosa Fernandez
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yun Gong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Rachel Krupa
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Mahipal V. Suraneni
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Ryon P. Graf
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Jerry Lee
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Stephanie Greene
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Angel Rodriguez
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Lyndsey Dugan
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Jessica Louw
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Carlos H. Barcenas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Angela N. Marx
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yipeng Wang
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Mark Landers
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Ryan Dittamore
- Department of Translational Research, Epic Sciences, La Jolla, California, United States of America
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
171
|
Mina A, Yoder R, Sharma P. Targeting the androgen receptor in triple-negative breast cancer: current perspectives. Onco Targets Ther 2017; 10:4675-4685. [PMID: 29033586 PMCID: PMC5614778 DOI: 10.2147/ott.s126051] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype associated with frequent recurrence and metastasis. Unlike hormone receptor-positive subtypes, treatment of TNBC is currently limited by the lack of clinically available targeted therapies. Androgen signaling is necessary for normal breast development, and its dysregulation has been implicated in breast tumorigenesis. In recent years, gene expression studies have identified a subset of TNBC that is enriched for androgen receptor (AR) signaling. Interference with androgen signaling in TNBC is promising, and AR-inhibiting drugs have shown antitumorigenic activity in preclinical and proof of concept clinical studies. Recent advances in our understanding of androgenic signaling in TNBC, along with the identification of interacting pathways, are allowing development of the next generation of clinical trials with AR inhibitors. As novel AR-targeting agents are developed and evaluated in clinical trials, it is equally important to establish a robust set of biomarkers for identification of TNBC tumors that are most likely to respond to AR inhibition.
Collapse
Affiliation(s)
- Alain Mina
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Westwood
| | - Rachel Yoder
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Priyanka Sharma
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Westwood
| |
Collapse
|
172
|
Venema CM, Mammatas LH, Schröder CP, van Kruchten M, Apollonio G, Glaudemans AW, Bongaerts AH, Hoekstra OS, Verheul HM, Boven E, van der Vegt B, de Vries EF, de Vries EG, Boellaard R, Menke van der Houven van Oordt CW, Hospers GA. Androgen and Estrogen Receptor Imaging in Metastatic Breast Cancer Patients as a Surrogate for Tissue Biopsies. J Nucl Med 2017; 58:1906-1912. [DOI: 10.2967/jnumed.117.193649] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/24/2017] [Indexed: 11/16/2022] Open
|
173
|
Shao F, Sun H, Deng CX. Potential therapeutic targets of triple-negative breast cancer based on its intrinsic subtype. Oncotarget 2017; 8:73329-73344. [PMID: 29069872 PMCID: PMC5641215 DOI: 10.18632/oncotarget.20274] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/06/2017] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subgroup of human breast cancer, which is characterized as estrogen receptor (ER) negative, progesterone receptor (PR) negative, and human epidermal growth factor receptor 2 (HER2) negative. TNBC is the most difficult breast cancer subgroup to treat, due to its unresponsiveness to current clinical targeted therapies, high rate of recurrence, and poor prognosis. Thus, there is an urgent medical need to identify therapeutic targets and develop more effective stratified medicine for the treatment of TNBC. Here we review the potential therapeutic targets for TNBC based on its intrinsic subtype. We also review the aberrant activated signals found in different subgroups of TNBC, including androgen receptor (AR) and PI3K/AKT/mTOR, Notch, Wnt/β-catenin, Hedge-hog, and TGF-β signaling pathways, which play essential roles in multiple development stages of TNBC. The careful analysis of these signaling pathways and therapeutic targets would have significant impact on the drug development and clinical trials, leading to effective therapies for this deadly disease.
Collapse
Affiliation(s)
- Fangyuan Shao
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Heng Sun
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
174
|
Healey MA, Hirko KA, Beck AH, Collins LC, Schnitt SJ, Eliassen AH, Holmes MD, Tamimi RM, Hazra A. Assessment of Ki67 expression for breast cancer subtype classification and prognosis in the Nurses' Health Study. Breast Cancer Res Treat 2017; 166:613-622. [PMID: 28791482 DOI: 10.1007/s10549-017-4421-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022]
Abstract
PURPOSE Ki67 is a proliferation marker commonly assessed by immunohistochemistry in breast cancer, and it has been proposed as a clinical marker for subtype classification, prognosis, and prediction of therapeutic response. However, the clinical utility of Ki67 is limited by the lack of consensus on the optimal cut point for each application. METHODS We assessed Ki67 by immunohistochemistry using Definiens digital image analysis (DIA) in 2653 cases of incident invasive breast cancer diagnosed in the Nurses' Health Study from 1976 to 2006. Ki67 was scored as continuous percentage of positive tumor cells, and dichotomized at various cut points. Multivariable hazard ratios (HR) and 95% confidence intervals (CI) were calculated using Cox regression models for distant recurrence, breast cancer-specific mortality and overall mortality in relation to luminal subtypes defined with various Ki67 cut points, adjusting for breast cancer prognostic factors, clinico-pathologic features and treatment. RESULTS DIA was highly correlated with manual scoring of Ki67 (Spearman correlation ρ = 0.86). Mean Ki67 score was higher in grade-defined luminal B (12.6%), HER2-enriched (17.9%) and basal-like (20.6%) subtypes compared to luminal A (8.9%). In multivariable-adjusted models, luminal B tumors had higher breast cancer-specific mortality compared to luminal A cancer classified using various cut points for Ki67 positivity including the 14% cut point routinely reported in the literature (HR 1.38, 95% CI 1.11-1.72, p = 0.004). There was no significant difference in clinical outcomes for ER- tumors according to Ki67 positivity defined at various cut points. CONCLUSIONS Assessment of Ki67 in breast tumors by DIA was a robust and quantitative method. Results from this large prospective cohort study provide support for the clinical relevance of using Ki67 at the 14% cut point for luminal subtype classification and breast cancer prognosis.
Collapse
Affiliation(s)
- Megan A Healey
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Kelly A Hirko
- Department of Epidemiology and Biostatistics, College of Human Medicine, Traverse City Campus, Michigan State University, East Lansing, MI, USA
| | | | - Laura C Collins
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Stuart J Schnitt
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Michelle D Holmes
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Aditi Hazra
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA.
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
175
|
The Androgen Receptor Supports Tumor Progression After the Loss of Ovarian Function in a Preclinical Model of Obesity and Breast Cancer. Discov Oncol 2017; 8:269-285. [PMID: 28741260 DOI: 10.1007/s12672-017-0302-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
The androgen receptor (AR) has context-dependent roles in breast cancer growth and progression. Overall, high tumor AR levels predict a favorable patient outcome, but several studies have established a tumor promotional role for AR, particularly in supporting the growth of estrogen receptor positive (ER-positive) breast cancers after endocrine therapy. Our previous studies have demonstrated that obesity promotes mammary tumor progression after ovariectomy (OVX) in a rat model of postmenopausal breast cancer. Here, we investigated a potential role for AR in obesity-associated post-OVX mammary tumor progression following ovarian estrogen loss. In this model, we found that obese but not lean rats had nuclear localized AR in tumors that progressed 3 weeks after OVX, compared to those that regressed. AR nuclear localization is consistent with activation of AR-dependent transcription. Longer-term studies (8 weeks post-OVX) showed that AR nuclear localization and expression were maintained in tumors that had progressed, but AR expression was nearly lost in tumors that were regressing. The anti-androgen enzalutamide effectively blocked tumor progression in obese rats by promoting tumor necrosis and also prevented the formation of new tumors after OVX. Neither circulating nor mammary adipose tissue levels of the AR ligand testosterone were elevated in obese compared to lean rats; however, IL-6, which we previously reported to be higher in plasma from obese versus lean rats, sensitized breast cancer cells to low levels of testosterone. Our study demonstrates that, in the context of obesity, AR plays a role in driving ER-positive mammary tumor progression in an environment of low estrogen availability, and that circulating factors unique to the obese host, including IL-6, may influence how cancer cells respond to steroid hormones.
Collapse
|
176
|
Gordon MA, D'Amato NC, Gu H, Babbs B, Wulfkuhle J, Petricoin EF, Gallagher I, Dong T, Torkko K, Liu B, Elias A, Richer JK. Synergy between Androgen Receptor Antagonism and Inhibition of mTOR and HER2 in Breast Cancer. Mol Cancer Ther 2017; 16:1389-1400. [PMID: 28468774 PMCID: PMC5517319 DOI: 10.1158/1535-7163.mct-17-0111] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/24/2017] [Accepted: 04/18/2017] [Indexed: 12/22/2022]
Abstract
The androgen receptor (AR) is widely expressed in breast cancer, and evidence suggests dependence on AR signaling for growth and survival. AR antagonists such as enzalutamide and seviteronel have shown success in preclinical models and clinical trials of prostate cancer and are currently being evaluated in breast cancer. Reciprocal regulation between AR and the HER2/PI3K/mTOR pathway may contribute to resistance to HER2- and mTOR-targeted therapies; thus, dual inhibition of these pathways may synergistically inhibit breast cancer growth. HER2+ and triple-negative breast cancer cell lines were treated with AR antagonist plus anti-HER2 mAb trastuzumab or mTOR inhibitor everolimus. Apoptosis, cell proliferation, and drug synergy were measured in vitro Pathway component genes and proteins were measured by qRT-PCR, Western blot, and reverse phase protein array. In vivo, HER2+ breast cancer xenografts were treated with enzalutamide, everolimus, trastuzumab, and combinations of these drugs. AR antagonists inhibited proliferation of both HER2+ and TNBC cell lines. Combining AR antagonist and either everolimus or trastuzumab resulted in synergistic inhibition of proliferation. Dihydrotestosterone caused increased phosphorylation of HER2 and/or HER3 that was attenuated by AR inhibition. Everolimus caused an increase in total AR, phosphorylation of HER2 and/or HER3, and these effects were abrogated by enzalutamide. Growth of trastuzumab-resistant HER2+ xenograft tumors was inhibited by enzalutamide, and combining enzalutamide with everolimus decreased tumor viability more than either single agent. AR antagonists synergize with FDA-approved breast cancer therapies such as everolimus and trastuzumab through distinct mechanisms. Treatment combinations are effective in trastuzumab-resistant HER2+ breast cancer cells in vivoMol Cancer Ther; 16(7); 1389-400. ©2017 AACR.
Collapse
Affiliation(s)
- Michael A Gordon
- 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
| | - Haihua Gu
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Beatrice Babbs
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Isela Gallagher
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Ting Dong
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Kathleen Torkko
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Bolin Liu
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony Elias
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
177
|
Anand A, Singh KR, Kumar S, Husain N, Kushwaha JK, Sonkar AA. Androgen Receptor Expression in an Indian Breast Cancer Cohort with Relation to Molecular Subtypes and Response to Neoadjuvant Chemotherapy - a Prospective Clinical Study. Breast Care (Basel) 2017; 12:160-164. [PMID: 28785183 DOI: 10.1159/000458433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Breast cancer (BC) is associated with advanced presentation in developing countries like India due to various socio-economic factors. The presence of BC molecular subtypes such as the triple-negative (TN) subtype adds to this menace. Androgen receptor (AR) is emerging as a new biological marker. The aim of this study was to examine the prevalence of AR with relation to different BC subtypes, and its role in predicting response to neoadjuvant chemotherapy. METHODS 116 cases of invasive BC (infiltrating ductal carcinoma, not otherwise specified) were evaluated. AR expression was correlated with clinicopathological factors, established prognostic markers, BC subtypes and it ability for predicting response to neoadjuvant chemotherapy. RESULTS AR was expressed in 56% of the cases. AR expression was significantly associated with early stage (p < 0.03), low axillary burden (p < 0.04), estrogen receptor (p = 0.002), progesterone receptor (p = 0.001) expression and luminal A molecular subtype. No significant association was observed with age, tumor size and HER2/neu status. One-third of TNBC cases expressed AR. Higher AR expression corelated to good clinical response to neoadjuvant chemotherapy. CONCLUSION AR can be utilized as a predictor of response to neoadjuvant chemotherapy especially in developing countries such as India where the load of advanced disease is high.
Collapse
Affiliation(s)
- Akshay Anand
- Department of Surgery, King George's Medical University, Lucknow, India
| | - Kul R Singh
- Department of Endocrine Surgery, King George's Medical University, Lucknow, India
| | - Surender Kumar
- Department of Surgery, King George's Medical University, Lucknow, India
| | - Nuzhat Husain
- Department of Pathology, Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | | | - Abhinav A Sonkar
- Department of Surgery, King George's Medical University, Lucknow, India
| |
Collapse
|
178
|
Song W, Tang L, Xu Y, Sun Q, Yang F, Guan X. ERβ1 inhibits metastasis of androgen receptor-positive triple-negative breast cancer by suppressing ZEB1. J Exp Clin Cancer Res 2017; 36:75. [PMID: 28583190 PMCID: PMC5460479 DOI: 10.1186/s13046-017-0545-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increasing evidence has indicated an important role for estrogen receptor beta 1 (ERβ1) in breast cancer. However, the role of ERβ1 in the metastasis of androgen receptor (AR)-positive triple-negative breast cancer (TNBC) and the underlying mechanisms are still unknown. METHODS Stable ERβ1-expressing TNBC cell lines were generated for this study. We detected the abilities of cell migration and invasion by wound-healing and transwell assays and the expression of E-cadherin and N-cadherin by quantitative RT-PCR (qRT-PCR) and western blotting assays in TNBC cell lines. Chromatin immunoprecipitation (ChIP) analysis was performed to assess the effect of AR on ERβ1 promoter. Tumor metastasis was evaluated in vivo using a lung metastasis mouse model. Lastly, immunohistochemical expression of ERβ1 in TNBC tissues was analyzed and correlated with clinicopathological features. RESULTS ERβ1 suppressed the invasion and migration abilities of AR-positive TNBC cells and induced the downregulation of ZEB1. ZEB1 overexpression abrogated the increase in E-cadherin expression and the decrease in N-cadherin expression modulated by ERβ1. A lung metastasis mouse model showed that the incidence of metastasis was lower in ERβ1-expressing TNBC cells. Further, AR activation increased the anti-metastatic effect of ERβ1 in AR-positive TNBC cells, which accelerated ERβ1 transcription by functioning as a transcription factor that bound to the promoter of ERβ1. No significant change was observed in AR expression induced by ERβ1. Immunohistochemistry (IHC) analysis of TNBC clinical samples showed that ERβ1 and AR were positive in 31.7% and 23.2% of samples, respectively. ERβ1 expression was negatively correlated with ZEB1 expression and lymph node metastasis, and positively correlated with the expression of AR and E-cadherin. CONCLUSION Our findings suggested a potential role of ERβ1 in metastasis of AR-positive TNBC and provided novel insights into the mechanism of action of ERβ1 and the possible relationship between ERβ1 and AR.
Collapse
Affiliation(s)
- Wei Song
- Department of Medical Oncology, Jinling Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Lin Tang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
| | - Yumei Xu
- Department of Medical Oncology, Jinling Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Qian Sun
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
| | - Fang Yang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Southern Medical University, Guangzhou, 510515 China
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
179
|
Barton VN, Christenson JL, Gordon MA, Greene LI, Rogers TJ, Butterfield K, Babbs B, Spoelstra NS, D'Amato NC, Elias A, Richer JK. Androgen Receptor Supports an Anchorage-Independent, Cancer Stem Cell-like Population in Triple-Negative Breast Cancer. Cancer Res 2017; 77:3455-3466. [PMID: 28512248 DOI: 10.1158/0008-5472.can-16-3240] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/17/2017] [Accepted: 05/10/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Valerie N Barton
- Department of Pathology, University of Colorado Anschutz Medical Campus Aurora, Colorado
| | - Jessica L Christenson
- 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
| | - Lisa I Greene
- Department of Pathology, University of Colorado Anschutz Medical Campus Aurora, Colorado
| | - Thomas J Rogers
- Department of Pathology, University of Colorado Anschutz Medical Campus Aurora, Colorado
| | - Kiel Butterfield
- Department of Pathology, University of Colorado Anschutz Medical Campus Aurora, Colorado
| | - Beatrice Babbs
- 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
| | - Nicholas C D'Amato
- Department of Pathology, University of Colorado Anschutz Medical Campus Aurora, Colorado
| | - Anthony Elias
- Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus Aurora, Colorado.
| |
Collapse
|
180
|
Naderi A. C1orf64 is a novel androgen receptor target gene and coregulator that interacts with 14-3-3 protein in breast cancer. Oncotarget 2017; 8:57907-57933. [PMID: 28915724 PMCID: PMC5593696 DOI: 10.18632/oncotarget.17826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/11/2017] [Indexed: 12/12/2022] Open
Abstract
This study investigated the network of genes that are co-expressed with androgen receptor (AR) to discover novel AR targets in breast cancer. Bioinformatics analysis of two datasets from breast cancer cell lines resulted in the identification of an AR-gene signature constituted of 98 genes that highly correlated with AR expression. Notably, C1orf64 showed the highest positive correlation with AR across the datasets with a correlation coefficient (CC) of 0.737. In addition, C1orf64 closely correlated with AR expression in primary and metastatic breast tumors and C1orf64 expression was relatively higher in breast tumors with a lower grade and lobular histology. Furthermore, there is a functional interplay between AR and C1orf64 in breast cancer. In this process, AR activation directly represses C1orf64 transcription and C1orf64, in turn, interacts with AR as a corepressor and negatively regulates the AR-mediated induction of prolactin-induced protein (PIP) and AR reporter activity. Moreover, the corepressor effect of C1orf64 results in a reduction of AR binding to PIP promoter. The other aspect of this interplay involves a cross-talk between AR and estrogen receptor (ER) signaling in which C1orf64 silencing intensifies the AR-mediated down-regulation of ER target gene, progesterone receptor. Therefore, the repression of C1orf64 by AR provides an underlying mechanism for the AR inhibitory effects on ER signaling. To elucidate the biochemical mechanisms of C1orf64 function, this study demonstrates that C1orf64 is a phosphothreonine protein that interacts with the chaperone protein 14-3-3. In summary, C1orf64 is a novel AR coregulator and a 14-3-3 binding partner in breast cancer.
Collapse
Affiliation(s)
- Ali Naderi
- University of Hawaii Cancer Center, Cancer Biology Program, Honolulu, Hawaii 96813, USA
| |
Collapse
|
181
|
Roubaud G, Liaw BC, Oh WK, Mulholland DJ. Strategies to avoid treatment-induced lineage crisis in advanced prostate cancer. Nat Rev Clin Oncol 2017; 14:269-283. [PMID: 27874061 PMCID: PMC5567685 DOI: 10.1038/nrclinonc.2016.181] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The increasing potency of therapies that target the androgen receptor (AR) signalling axis has correlated with a rise in the proportion of patients with prostate cancer harbouring an adaptive phenotype, termed treatment-induced lineage crisis. This phenotype is characterized by features that include soft-tissue metastasis and/or resistance to standard anticancer therapies. Potent anticancer treatments might force cancer cells to evolve and develop alternative cell lineages that are resistant to primary therapies, a mechanism similar to the generation of multidrug- resistant microorganisms after continued antibiotic use. Herein, we assess the hypothesis that treatment-adapted phenotypes harbour reduced AR expression and/or activity, and acquire compensatory strategies for cell survival. We highlight the striking similarities between castration-resistant prostate cancer and triple-negative breast cancer, another poorly differentiated endocrine malignancy. Alternative treatment paradigms are needed to avoid therapy-induced resistance. Herein, we present a new clinical trial strategy designed to evaluate the potential of rapid drug cycling as an approach to delay the onset of resistance and treatment-induced lineage crisis in patients with metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Bobby C Liaw
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 1470 Madison Avenue, New York, New York 10029, USA
| | - William K Oh
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 1470 Madison Avenue, New York, New York 10029, USA
| | - David J Mulholland
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 1470 Madison Avenue, New York, New York 10029, USA
| |
Collapse
|
182
|
Schwartzberg LS, Yardley DA, Elias AD, Patel M, LoRusso P, Burris HA, Gucalp A, Peterson AC, Blaney ME, Steinberg JL, Gibbons JA, Traina TA. A Phase I/Ib Study of Enzalutamide Alone and in Combination with Endocrine Therapies in Women with Advanced Breast Cancer. Clin Cancer Res 2017; 23:4046-4054. [PMID: 28280092 DOI: 10.1158/1078-0432.ccr-16-2339] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/17/2016] [Accepted: 03/02/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Several lines of evidence support targeting the androgen signaling pathway in breast cancer. Enzalutamide is a potent inhibitor of androgen receptor signaling. Preclinical data in estrogen-expressing breast cancer models demonstrated activity of enzalutamide monotherapy and enhanced activity when combined with various endocrine therapies (ET). Enzalutamide is a strong cytochrome P450 3A4 (CYP3A4) inducer, and ETs are commonly metabolized by CYP3A4. The pharmacokinetic (PK) interactions, safety, and tolerability of enzalutamide monotherapy and in combination with ETs were assessed in this phase I/Ib study.Experimental Design: Enzalutamide monotherapy was assessed in dose-escalation and dose-expansion cohorts of patients with advanced breast cancer. Additional cohorts examined effects of enzalutamide on anastrozole, exemestane, and fulvestrant PK in patients with estrogen receptor-positive/progesterone receptor-positive (ER+/PgR+) breast cancer.Results: Enzalutamide monotherapy (n = 29) or in combination with ETs (n = 70) was generally well tolerated. Enzalutamide PK in women was similar to prior data on PK in men with prostate cancer. Enzalutamide decreased plasma exposure to anastrozole by approximately 90% and exemestane by approximately 50%. Enzalutamide did not significantly affect fulvestrant PK. Exposure of exemestane 50 mg/day given with enzalutamide was similar to exemestane 25 mg/day alone.Conclusions: These results support a 160 mg/day enzalutamide dose in women with breast cancer. Enzalutamide can be given in combination with fulvestrant without dose modifications. Exemestane should be doubled from 25 mg/day to 50 mg/day when given in combination with enzalutamide; this combination is being investigated in a randomized phase II study in patients with ER+/PgR+ breast cancer. Clin Cancer Res; 23(15); 4046-54. ©2017 AACR.
Collapse
Affiliation(s)
| | - Denise A Yardley
- Sarah Cannon Research Institute, Nashville, Tennessee.,Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Anthony D Elias
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Manish Patel
- Sarah Cannon Research Institute, Nashville, Tennessee.,Florida Cancer Specialists, Sarah Cannon Research Institute, Sarasota, Florida
| | - Patricia LoRusso
- Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Howard A Burris
- Sarah Cannon Research Institute, Nashville, Tennessee.,Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Ayca Gucalp
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Amy C Peterson
- Medivation, Inc. (Medivation, Inc., was acquired by Pfizer, Inc., in September 2016), San Francisco, California
| | - Martha E Blaney
- Medivation, Inc. (Medivation, Inc., was acquired by Pfizer, Inc., in September 2016), San Francisco, California
| | | | - Jacqueline A Gibbons
- Medivation, Inc. (Medivation, Inc., was acquired by Pfizer, Inc., in September 2016), San Francisco, California
| | - Tiffany A Traina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| |
Collapse
|
183
|
Rodgers RJ, Reid GD, Koch J, Deans R, Ledger WL, Friedlander M, Gilchrist RB, Walters KA, Abbott JA. The safety and efficacy of controlled ovarian hyperstimulation for fertility preservation in women with early breast cancer: a systematic review. Hum Reprod 2017; 32:1033-1045. [DOI: 10.1093/humrep/dex027] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/29/2017] [Indexed: 12/29/2022] Open
|
184
|
AR Signaling in Breast Cancer. Cancers (Basel) 2017; 9:cancers9030021. [PMID: 28245550 PMCID: PMC5366816 DOI: 10.3390/cancers9030021] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/13/2017] [Accepted: 02/18/2017] [Indexed: 12/31/2022] Open
Abstract
Androgen receptor (AR, a member of the steroid hormone receptor family) status has become increasingly important as both a prognostic marker and potential therapeutic target in breast cancer. AR is expressed in up to 90% of estrogen receptor (ER) positive breast cancer, and to a lesser degree, human epidermal growth factor 2 (HER2) amplified tumors. In the former, AR signaling has been correlated with a better prognosis given its inhibitory activity in estrogen dependent disease, though conversely has also been shown to increase resistance to anti-estrogen therapies such as tamoxifen. AR blockade can mitigate this resistance, and thus serves as a potential target in ER-positive breast cancer. In HER2 amplified breast cancer, studies are somewhat conflicting, though most show either no effect or are associated with poorer survival. Much of the available data on AR signaling is in triple-negative breast cancer (TNBC), which is an aggressive disease with inferior outcomes comparative to other breast cancer subtypes. At present, there are no approved targeted therapies in TNBC, making study of the AR signaling pathway compelling. Gene expression profiling studies have also identified a luminal androgen receptor (LAR) subtype that is dependent on AR signaling in TNBC. Regardless, there seems to be an association between AR expression and improved outcomes in TNBC. Despite lower pathologic complete response (pCR) rates with neoadjuvant therapy, patients with AR-expressing TNBC have been shown to have a better prognosis than those that are AR-negative. Clinical studies targeting AR have shown somewhat promising results. In this paper we review the literature on the biology of AR in breast cancer and its prognostic and predictive roles. We also present our thoughts on therapeutic strategies.
Collapse
|
185
|
Pakula H, Xiang D, Li Z. A Tale of Two Signals: AR and WNT in Development and Tumorigenesis of Prostate and Mammary Gland. Cancers (Basel) 2017; 9:E14. [PMID: 28134791 PMCID: PMC5332937 DOI: 10.3390/cancers9020014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers and among the leading causes of cancer deaths for men in industrialized countries. It has long been recognized that the prostate is an androgen-dependent organ and PCa is an androgen-dependent disease. Androgen action is mediated by the androgen receptor (AR). Androgen deprivation therapy (ADT) is the standard treatment for metastatic PCa. However, almost all advanced PCa cases progress to castration-resistant prostate cancer (CRPC) after a period of ADT. A variety of mechanisms of progression from androgen-dependent PCa to CRPC under ADT have been postulated, but it remains largely unclear as to when and how castration resistance arises within prostate tumors. In addition, AR signaling may be modulated by extracellular factors among which are the cysteine-rich glycoproteins WNTs. The WNTs are capable of signaling through several pathways, the best-characterized being the canonical WNT/β-catenin/TCF-mediated canonical pathway. Recent studies from sequencing PCa genomes revealed that CRPC cells frequently harbor mutations in major components of the WNT/β-catenin pathway. Moreover, the finding of an interaction between β-catenin and AR suggests a possible mechanism of cross talk between WNT and androgen/AR signaling pathways. In this review, we discuss the current knowledge of both AR and WNT pathways in prostate development and tumorigenesis, and their interaction during development of CRPC. We also review the possible therapeutic application of drugs that target both AR and WNT/β-catenin pathways. Finally, we extend our review of AR and WNT signaling to the mammary gland system and breast cancer. We highlight that the role of AR signaling and its interaction with WNT signaling in these two hormone-related cancer types are highly context-dependent.
Collapse
Affiliation(s)
- Hubert Pakula
- Division of Genetics, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Room 466, Boston, MA 02115, USA.
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Dongxi Xiang
- Division of Genetics, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Room 466, Boston, MA 02115, USA.
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Zhe Li
- Division of Genetics, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Room 466, Boston, MA 02115, USA.
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
186
|
AR-Signaling in Human Malignancies: Prostate Cancer and Beyond. Cancers (Basel) 2017; 9:cancers9010007. [PMID: 28085048 PMCID: PMC5295778 DOI: 10.3390/cancers9010007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
In the 1940s Charles Huggins reported remarkable palliative benefits following surgical castration in men with advanced prostate cancer, and since then the androgen receptor (AR) has remained the main therapeutic target in this disease. Over the past couple of decades, our understanding of AR-signaling biology has dramatically improved, and it has become apparent that the AR can modulate a number of other well-described oncogenic signaling pathways. Not surprisingly, mounting preclinical and epidemiologic data now supports a role for AR-signaling in promoting the growth and progression of several cancers other than prostate, and early phase clinical trials have documented preliminary signs of efficacy when AR-signaling inhibitors are used in several of these malignancies. In this article, we provide an overview of the evidence supporting the use of AR-directed therapies in prostate as well as other cancers, with an emphasis on the rationale for targeting AR-signaling across tumor types.
Collapse
|
187
|
Narayanan R, Dalton JT. Androgen Receptor: A Complex Therapeutic Target for Breast Cancer. Cancers (Basel) 2016; 8:cancers8120108. [PMID: 27918430 PMCID: PMC5187506 DOI: 10.3390/cancers8120108] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/01/2016] [Accepted: 11/23/2016] [Indexed: 12/29/2022] Open
Abstract
Molecular and histopathological profiling have classified breast cancer into multiple sub-types empowering precision treatment. Although estrogen receptor (ER) and human epidermal growth factor receptor (HER2) are the mainstay therapeutic targets in breast cancer, the androgen receptor (AR) is evolving as a molecular target for cancers that have developed resistance to conventional treatments. The high expression of AR in breast cancer and recent discovery and development of new nonsteroidal drugs targeting the AR provide a strong rationale for exploring it again as a therapeutic target in this disease. Ironically, both nonsteroidal agonists and antagonists for the AR are undergoing clinical trials, making AR a complicated target to understand in breast cancer. This review provides a detailed account of AR’s therapeutic role in breast cancer.
Collapse
Affiliation(s)
- Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - James T Dalton
- College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
188
|
Lim E, Tarulli G, Portman N, Hickey TE, Tilley WD, Palmieri C. Pushing estrogen receptor around in breast cancer. Endocr Relat Cancer 2016; 23:T227-T241. [PMID: 27729416 DOI: 10.1530/erc-16-0427] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 12/21/2022]
Abstract
The estrogen receptor-α (herein called ER) is a nuclear sex steroid receptor (SSR) that is expressed in approximately 75% of breast cancers. Therapies that modulate ER action have substantially improved the survival of patients with ER-positive breast cancer, but resistance to treatment still remains a major clinical problem. Treating resistant breast cancer requires co-targeting of ER and alternate signalling pathways that contribute to resistance to improve the efficacy and benefit of currently available treatments. Emerging data have shown that other SSRs may regulate the sites at which ER binds to DNA in ways that can powerfully suppress the oncogenic activity of ER in breast cancer. This includes the progesterone receptor (PR) that was recently shown to reprogram the ER DNA binding landscape towards genes associated with a favourable outcome. Another attractive candidate is the androgen receptor (AR), which is expressed in the majority of breast cancers and inhibits growth of the normal breast and ER-positive tumours when activated by ligand. These findings have led to the initiation of breast cancer clinical trials evaluating therapies that selectively harness the ability of SSRs to 'push' ER towards anti-tumorigenic activity. Our review will focus on the established and emerging clinical evidence for activating PR or AR in ER-positive breast cancer to inhibit the tumour growth-promoting functions of ER.
Collapse
Affiliation(s)
- Elgene Lim
- Garvan Institute of Medical Research and St Vincent's HospitalUniversity of New South Wales, Sydney, New South Wales, Australia
| | - Gerard Tarulli
- Dame Roma Mitchell Cancer Research Laboratories and Adelaide Prostate Cancer Research CentreUniversity of Adelaide, Adelaide, South Australia, Australia
| | - Neil Portman
- Garvan Institute of Medical Research and St Vincent's HospitalUniversity of New South Wales, Sydney, New South Wales, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories and Adelaide Prostate Cancer Research CentreUniversity of Adelaide, Adelaide, South Australia, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories and Adelaide Prostate Cancer Research CentreUniversity of Adelaide, Adelaide, South Australia, Australia
| | - Carlo Palmieri
- Institute of Translational MedicineUniversity of Liverpool, Clatterbridge Cancer Centre, NHS Foundation Trust, and Royal Liverpool University Hospital, Liverpool, Merseyside, UK
| |
Collapse
|
189
|
Steroid Hormone Receptor Positive Breast Cancer Patient-Derived Xenografts. Discov Oncol 2016; 8:4-15. [PMID: 27796944 DOI: 10.1007/s12672-016-0275-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/14/2016] [Indexed: 12/25/2022] Open
Abstract
The vast majority of breast cancers are positive for estrogen receptor (ER) and depend on estrogens for growth. These tumors are treated with a variety of ER-targeted endocrine therapies, although eventual resistance remains a major clinical problem. Other steroid hormone receptors such as progesterone receptor (PR) and androgen receptor (AR) are emerging as additional prospective targets in breast cancer. The fundamental mechanism of action of these steroid receptors in gene regulation has been defined mainly by several breast cancer cell lines that were established in the late 1970s. More recently, breast cancer patient-derived xenografts (PDX) have been developed by multiple groups at institutions in several countries. These new models capture the large degree of heterogeneity between patients and within tumors and promise to advance our understanding of steroid hormone receptor positive breast cancer and endocrine resistance. Unfortunately, steroid hormone receptor positive breast cancers are much more difficult than their receptor negative counterparts to establish into sustainable PDX. Herein we discuss the derivation of steroid hormone receptor positive breast cancer PDX, several pitfalls in their genesis, and their utility in preclinical and translational steroid hormone receptor research.
Collapse
|
190
|
Abstract
Clinical studies have shown that the androgen receptor (AR) is ubiquitously expressed in breast cancers and this could provide prognostic implication in the diagnosis and treatment of breast cancers. Data from Nurse’s Health Study on women with invasive breast cancer suggest that a significant number of tumors were AR-positive as defined by immunohistochemistry. In addition, the distribution of AR among different breast cancer subtypes varies significantly, and the biological reasons for this variation are not well understood. Despite strong histochemical evidence, the AR status is not applied for assessing pathological findings and disease outcome in clinical practice. AR antagonists are not currently used as therapy in breast cancer. This is in part due to conflicting results from early clinical trials with first generation of AR antagonists together with the complexity in breast cancer heterogeneity. In addition, role of AR in breast cancer is not fully understood. Here we will review the role of AR in different subtypes of breast cancers and elucidate its mechanisms. We will also discuss some recent interesting findings on the second generation of AR antagonists for treatment of breast cancer.
Collapse
Affiliation(s)
- Y Wu
- Department of Medicine, Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - J V Vadgama
- Department of Medicine, Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
191
|
Wu X, Zhan Y, Li X, Wei J, Santiago L, Daniels G, Deng F, Zhong X, Chiriboga L, Basch R, Xiong S, Dong Y, Zhang X, Lee P. Nuclear TBLR1 as an ER corepressor promotes cell proliferation, migration and invasion in breast and ovarian cancer. Am J Cancer Res 2016; 6:2351-2360. [PMID: 27822424 PMCID: PMC5088298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/06/2016] [Indexed: 06/06/2023] Open
Abstract
Estrogen receptors (ER) play important roles in the development and progression of breast and ovarian cancers. ERs mediate transcriptional regulation through interaction with cofactors and binding to response elements within the regulatory elements of target genes. Here, we examined the expression and function of TBLR1/TBL1XR1, a core component of NCoR (nuclear receptor corepressor) and SMRT (silencing mediator of retinoic acid and thyroid receptor) corepressor complexes, in breast and ovarian cancers. We found that although TBLR1 is present in both the nucleus and cytoplasm of normal and neoplastic breast and ovarian cells, it is expressed at significantly higher levels in the nucleus of malignant breast and ovarian cells compared to benign cells. TBLR1 functions as an ER corepressor to inhibit ER-mediated transcriptional activation in both breast and ovarian cell lines, but it has no effect on androgen receptor (AR) mediated transcriptional activation in these cells. Furthermore, ectopic expression of nuclear TBLR1 in breast and ovarian cancer cells stimulates cell proliferation. The increased cell proliferation by nuclear TBLR1 is through both ER-independent and ER-dependent mechanisms as evidenced by increased growth in hormone-free medium and estrogen medium, as well as reduced growth with ER knockdown by siRNA. Nuclear TBLR1 overexpression also increased migration and invasion in both breast and ovarian cancer cells. Determining the functional relationship between TBLR1 and ER may provide insights to develop novel treatment strategies and improve response to hormonal therapy in breast and ovarian cancers.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Yang Zhan
- Structural & Cellular Biology, Tulane University School of MedicineNew Orleans, LA
| | - Xin Li
- Basic Science and Craniofacial Biology, New York University College of DentistryNew York, NY
- Department of Urology, New York University School of MedicineNew York, NY
- Department of NYU Cancer Institute, New York University School of MedicineNew York, NY
| | - Jianjun Wei
- Department of Pathology, Northwestern School of MedicineChicago, IL
| | - Larion Santiago
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Garrett Daniels
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Fangming Deng
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Xuelin Zhong
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Luis Chiriboga
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Ross Basch
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Sheng Xiong
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Yan Dong
- Structural & Cellular Biology, Tulane University School of MedicineNew Orleans, LA
| | - Xinmin Zhang
- Department of Pathology, Hofstra North Shore-LIJ School of MedicineHempstead, New York, NY
| | - Peng Lee
- Department of Pathology, New York University School of MedicineNew York, NY
- Department of Urology, New York University School of MedicineNew York, NY
- Department of NYU Cancer Institute, New York University School of MedicineNew York, NY
- Department of New York Harbor Healthcare System, New York University School of MedicineNew York, NY
| |
Collapse
|
192
|
Kono M, Fujii T, Lyons GR, Huo L, Bassett R, Gong Y, Karuturi MS, Tripathy D, Ueno NT. Impact of androgen receptor expression in fluoxymesterone-treated estrogen receptor-positive metastatic breast cancer refractory to contemporary hormonal therapy. Breast Cancer Res Treat 2016; 160:101-109. [PMID: 27663436 DOI: 10.1007/s10549-016-3986-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/16/2016] [Indexed: 01/22/2023]
Abstract
PURPOSE The purpose of this study is to evaluate survival outcome in patients with hormone receptor (HR)-positive (+) metastatic breast cancer (MBC) who received fluoxymesterone after disease progression while receiving contemporary hormonal therapy, as well as the association between estrogen receptor (ER)/androgen receptor (AR) status and survival outcome in these patients. METHODS We retrospectively identified 103 patients treated with fluoxymesterone for HR + MBC from 2000 to 2014 and with at least one previous hormonal therapy in a metastatic setting. RESULTS A median of 3 (range 1-10) hormonal therapies (aromatase inhibitors, tamoxifen, and/or fulvestrant) were received before fluoxymesterone; 33 patients discontinued fluoxymesterone before progression because of physician decision or adverse events including toxicity in 14 patients. Of the remaining 70 patients, 2 (3 %) had complete response, 7 (10 %) partial response, and 21 (30 %) stable disease for at least 6 months, yielding a clinical benefit rate of 43 %. The median PFS was 3.9 months (95 % CI 3.2-5.3 months). Multivariate analysis revealed no significant association between PFS and the type or number of prior systemic treatments. All 39 patients who had archived tumor slides available for AR staining had ER + carcinoma; 10 had ≥1 % but <10 %, 18 had ≥10 %, and 11 had no AR nuclear expression. AR positivity with various cutoffs (i.e. any AR + cells, ≥1 % AR + cells, or ≥10 % AR + cells) was not significantly associated with survival outcome. CONCLUSIONS Fluoxymesterone can be considered for patients whose ER + MBC progresses on contemporary hormonal therapy, regardless of the level of AR expression.
Collapse
Affiliation(s)
- Miho Kono
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi Minamiku, Hiroshima, 7348553, Japan
| | - Takeo Fujii
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Genevieve Ray Lyons
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Roland Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yun Gong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Meghan Sri Karuturi
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA
| | - Debu Tripathy
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
193
|
D'Amato NC, Gordon MA, Babbs B, Spoelstra NS, Carson Butterfield KT, Torkko KC, Phan VT, Barton VN, Rogers TJ, Sartorius CA, Elias A, Gertz J, Jacobsen BM, Richer JK. Cooperative Dynamics of AR and ER Activity in Breast Cancer. Mol Cancer Res 2016; 14:1054-1067. [PMID: 27565181 DOI: 10.1158/1541-7786.mcr-16-0167] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 11/16/2022]
Abstract
Androgen receptor (AR) is expressed in 90% of estrogen receptor alpha-positive (ER+) breast tumors, but its role in tumor growth and progression remains controversial. Use of two anti-androgens that inhibit AR nuclear localization, enzalutamide and MJC13, revealed that AR is required for maximum ER genomic binding. Here, a novel global examination of AR chromatin binding found that estradiol induced AR binding at unique sites compared with dihydrotestosterone (DHT). Estradiol-induced AR-binding sites were enriched for estrogen response elements and had significant overlap with ER-binding sites. Furthermore, AR inhibition reduced baseline and estradiol-mediated proliferation in multiple ER+/AR+ breast cancer cell lines, and synergized with tamoxifen and fulvestrant. In vivo, enzalutamide significantly reduced viability of tamoxifen-resistant MCF7 xenograft tumors and an ER+/AR+ patient-derived model. Enzalutamide also reduced metastatic burden following cardiac injection. Finally, in a comparison of ER+/AR+ primary tumors versus patient-matched local recurrences or distant metastases, AR expression was often maintained even when ER was reduced or absent. These data provide preclinical evidence that anti-androgens that inhibit AR nuclear localization affect both AR and ER, and are effective in combination with current breast cancer therapies. In addition, single-agent efficacy may be possible in tumors resistant to traditional endocrine therapy, as clinical specimens of recurrent disease demonstrate AR expression in tumors with absent or refractory ER. IMPLICATIONS This study suggests that AR plays a previously unrecognized role in supporting E2-mediated ER activity in ER+/AR+ breast cancer cells, and that enzalutamide may be an effective therapeutic in ER+/AR+ breast cancers. Mol Cancer Res; 14(11); 1054-67. ©2016 AACR.
Collapse
Affiliation(s)
- 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
| | - Beatrice Babbs
- 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
| | | | - Kathleen C Torkko
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Valerie N Barton
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Thomas J Rogers
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Carol A Sartorius
- 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
| | - Jason Gertz
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - 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
|
194
|
Bao Y, Bertoia ML, Lenart EB, Stampfer MJ, Willett WC, Speizer FE, Chavarro JE. Origin, Methods, and Evolution of the Three Nurses' Health Studies. Am J Public Health 2016; 106:1573-81. [PMID: 27459450 DOI: 10.2105/ajph.2016.303338] [Citation(s) in RCA: 393] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We have summarized the evolution of the Nurses' Health Study (NHS), a prospective cohort study of 121 700 married registered nurses launched in 1976; NHS II, which began in 1989 and enrolled 116 430 nurses; and NHS3, which began in 2010 and has ongoing enrollment. Over 40 years, these studies have generated long-term, multidimensional data, including lifestyle- and health-related information across the life course and an extensive repository of various biological specimens. We have described the questionnaire data collection, disease follow-up methods, biorepository resources, and data management and statistical procedures. Through integrative analyses, these studies have sustained a high level of scientific productivity and substantially influenced public health recommendations. We have highlighted recent interdisciplinary research projects and discussed future directions for collaboration and innovation.
Collapse
Affiliation(s)
- Ying Bao
- Ying Bao, Meir J. Stampfer, and Frank E. Speizer are with the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Monica L. Bertoia, Elizabeth B. Lenart, Walter C. Willett, and Jorge E. Chavarro are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Monica L Bertoia
- Ying Bao, Meir J. Stampfer, and Frank E. Speizer are with the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Monica L. Bertoia, Elizabeth B. Lenart, Walter C. Willett, and Jorge E. Chavarro are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Elizabeth B Lenart
- Ying Bao, Meir J. Stampfer, and Frank E. Speizer are with the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Monica L. Bertoia, Elizabeth B. Lenart, Walter C. Willett, and Jorge E. Chavarro are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Meir J Stampfer
- Ying Bao, Meir J. Stampfer, and Frank E. Speizer are with the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Monica L. Bertoia, Elizabeth B. Lenart, Walter C. Willett, and Jorge E. Chavarro are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Walter C Willett
- Ying Bao, Meir J. Stampfer, and Frank E. Speizer are with the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Monica L. Bertoia, Elizabeth B. Lenart, Walter C. Willett, and Jorge E. Chavarro are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Frank E Speizer
- Ying Bao, Meir J. Stampfer, and Frank E. Speizer are with the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Monica L. Bertoia, Elizabeth B. Lenart, Walter C. Willett, and Jorge E. Chavarro are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Jorge E Chavarro
- Ying Bao, Meir J. Stampfer, and Frank E. Speizer are with the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Monica L. Bertoia, Elizabeth B. Lenart, Walter C. Willett, and Jorge E. Chavarro are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| |
Collapse
|
195
|
Barton VN, Gordon MA, Richer JK, Elias A. Anti-androgen therapy in triple-negative breast cancer. Ther Adv Med Oncol 2016; 8:305-8. [PMID: 27482289 PMCID: PMC4952024 DOI: 10.1177/1758834016646735] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Valerie N. Barton
- Department of Pathology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael A. Gordon
- Department of Pathology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer K. Richer
- Department of Pathology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Anthony Elias
- Division of Medical Oncology, University of Colorado School of Medicine, Anschutz Medical Campus, ACP 5310, MS 8117 1665 Aurora Court, Aurora, CO 80045, USA
| |
Collapse
|
196
|
Christgen M, Steinemann D, Kühnle E, Länger F, Gluz O, Harbeck N, Kreipe H. Lobular breast cancer: Clinical, molecular and morphological characteristics. Pathol Res Pract 2016; 212:583-97. [DOI: 10.1016/j.prp.2016.05.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/11/2016] [Accepted: 05/04/2016] [Indexed: 01/20/2023]
|
197
|
Potential role of targeted therapies in the treatment of triple-negative breast cancer. Anticancer Drugs 2016; 27:147-55. [PMID: 26682525 DOI: 10.1097/cad.0000000000000328] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Breast cancer is the most common cancer type that affects women and is the major cause of morbidity and mortality. Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype and accounts for 10-20% of all breast cancer cases. TNBC is commonly characterized by the absence of estrogen, progesterone, and the Her2/neu receptor and is usually diagnosed by immunohistochemistry. Mutations in the BRCA1 gene, as well as overexpression of oncogenic kinases, such as human epidermal growth factor receptor 2, vascular endothelial growth factor-A, insulin-like growth factor-1 (IGF-1)/IGF-1 receptor, and transforming growth factor-β1, have been found to be correlated with a higher risk of metastasis and poor overall survival in TNBC patients. The current review briefly discusses the various treatment options including chemotherapeutics and targeted therapies that are available currently for the therapy of TNBC patients and highlights their comparative benefits and disadvantages for clinical application.
Collapse
|
198
|
Hidradenoma Papilliferum With Oncocytic Metaplasia: A Histopathological and Immunohistochemical Study. Am J Dermatopathol 2016; 38:444-7. [PMID: 27097337 DOI: 10.1097/dad.0000000000000599] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hidradenoma papilliferum is a benign cutaneous adnexal neoplasm, commonly occurring in the vulva and perianal region of adult women. It has characteristic histopathological features composed of anastomosing and branching tubules, lined by columnar cells, and a basal layer of myoepithelial cells. A 39-year-old woman was evaluated for 2 asymptomatic labial masses. The histopathological examination revealed a Bartholin's cyst and a hidradenoma papilliferum. The latter contains a distinct area of oncocytic/oxyphilic metaplasia. Immunohistochemical stains revealed positive staining for gross cystic disease fluid protein (GCDFP)-15 and androgen receptor. GATA-3, a protein expressed in sweat glands, highlights a similar positive staining pattern with weaker staining in areas of oncocytic metaplasia. P63 highlighted the myoepithelial differentiation. In situ hybridization for Human Papilloma Virus 6, 11, 16, and 18 was negative. P53 was negative and Ki-67 was low, confirming its benign nature. Oncocytes are enlarged epithelial cells with voluminous eosinophilic granular cytoplasm resulting from staining of nonribosomal cytoplasmic components. Few reports documented it in hidradenoma papilliferum. Our case demonstrated a florid distinct appearance of this metaplasia. The immunoprofiles of this oncocytic metaplasia such as p53 negativity and positivity for androgen receptor and GCDFP-15 demonstrates similarity to apocrine metaplasia in the breast. The authors' case demonstrates the benign nature of oncocytic metaplasia and supports the common origin of oncocytic cells and columnar cells in hidradenoma papilliferum.
Collapse
|
199
|
Caiazza F, Murray A, Madden SF, Synnott NC, Ryan EJ, O'Donovan N, Crown J, Duffy MJ. Preclinical evaluation of the AR inhibitor enzalutamide in triple-negative breast cancer cells. Endocr Relat Cancer 2016; 23:323-34. [PMID: 26932782 DOI: 10.1530/erc-16-0068] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/01/2016] [Indexed: 12/22/2022]
Abstract
The androgen receptor (AR) is present in approximately 80% of invasive breast cancer patients and in up to 30% of patients with triple-negative breast cancer (TNBC). Therefore, our aim was to investigate the targeting of AR as a possible hormonal approach to the treatment of TNBC. Analysis of 2091 patients revealed an association between AR expression and poor overall survival, selectively in patients with the basal subtype of breast cancer, the vast majority of which are TNBC. IC50 values for the second-generation anti-androgen enzalutamide across 11 breast cancer cell lines varied from 4 µM to >50 µM. The activity of enzalutamide was similar in TN and non-TN cell lines but was dependent on the presence of AR. Enzalutamide reduced clonogenic potential and cell growth in a 3D matrix in AR-positive cells. In addition, enzalutamide also inhibited cell migration and invasion in an AR-dependent manner. Enzalutamide appeared to mediate these processes through down-regulation of the transcription factors AP-1 and SP-1. The first-generation anti-androgen flutamide similarly blocked cell growth, migration and invasion. AR-positive TNBC cells clustered separately from AR-negative cells based on an androgen-related gene expression signature, independently of TNBC subtype. We conclude that targeting of the AR with drugs such as enzalutamide may provide an alternative treatment strategy for patients with AR-positive TNBC.
Collapse
Affiliation(s)
- Francesco Caiazza
- School of MedicineUniversity College Dublin, Ireland Centre for Colorectal DiseaseSt. Vincent's University Hospital, Dublin, Ireland
| | - Alyson Murray
- School of MedicineUniversity College Dublin, Ireland
| | - Stephen F Madden
- Population Health SciencesDepartment of Psychology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Elizabeth J Ryan
- School of MedicineUniversity College Dublin, Ireland Centre for Colorectal DiseaseSt. Vincent's University Hospital, Dublin, Ireland
| | - Norma O'Donovan
- National Institute for Cellular Biotechnology (NICB)Dublin City University, Dublin, Ireland
| | - John Crown
- School of MedicineUniversity College Dublin, Ireland Department of Medical OncologySt. Vincent's University Hospital, Dublin, Ireland
| | - Michael J Duffy
- School of MedicineUniversity College Dublin, Ireland UCD Clinical Research CentreSt. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
200
|
Prevalence and predictors of androgen receptor and programmed death-ligand 1 in BRCA1-associated and sporadic triple-negative breast cancer. NPJ Breast Cancer 2016; 2:16002. [PMID: 28721372 PMCID: PMC5515333 DOI: 10.1038/npjbcancer.2016.2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/28/2015] [Indexed: 12/13/2022] Open
Abstract
Background: Triple-negative breast cancers comprise 15% of breast cancers and are more common in women with BRCA1 mutations. Although most have basal gene expression signatures, others resemble luminal tumors with expression of androgen receptor-related genes and some express the immunoinhibitory protein programmed death-ligand 1 (PD-L1). Given the availability of androgen receptor-targeted and immune therapies for triple-negative breast cancers, determining predictors of these biomarkers is important. Aims: To determine the prevalence and predictors of androgen receptor and PD-L1 expression in BRCA1-associated and sporadic triple-negative breast cancer. Methods: We studied 197 triple-negative breast cancers: 78 (39.6%) from BRCA1 mutation carriers and 119 (60.4%) from noncarriers. Tumor pathology was reviewed and tissue microarray sections were immunostained for androgen receptor and PD-L1. Results: Androgen receptor expression was seen in 18% of tumors and was significantly less common in tumors from BRCA1 mutation carriers than noncarriers (9.2 vs. 23.7%; P=0.01). Twenty-six percent of cancers expressed PD-L1 with no significant difference in frequency between carriers and noncarriers. Factors predicting androgen receptor expression were lower histologic grade (odds ratio (OR) 4.6; 95% confidence interval (CI) 1.1–19.7), older age at diagnosis (OR 1.3; 95% CI 1.03–1.7) and PD-L1 expression (OR 2.6; 95% CI 1.1–6.1). PD-L1 expression was significantly more common in cancers with lymphocytic infiltrates (OR, 3.3; 95% CI 1.1–10.4) and androgen receptor expression (OR, 3.2; 95% CI 1.4–7.5), and less common in tumors with lymphovascular invasion (OR 0.41; 95% CI 0.18–0.92). Conclusions: These results identify predictors for androgen receptor and PD-L1 expression among triple-negative breast cancers that may lead to better treatment selection and participation in clinical trials.
Collapse
|