Androgen receptor expression on circulating tumor cells in metastatic breast cancer

Takeo Fujii, James M Reuben, Lei Huo, Jose Rodrigo Espinosa Fernandez, Yun Gong, Rachel Krupa, Mahipal V Suraneni, Ryon P Graf, Jerry Lee, Stephanie Greene, Angel Rodriguez, Lyndsey Dugan, Jessica Louw, Bora Lim, Carlos H Barcenas, Angela N Marx, Debu Tripathy, Yipeng Wang, Mark Landers, Ryan Dittamore, Naoto T Ueno, Takeo Fujii, James M Reuben, Lei Huo, Jose Rodrigo Espinosa Fernandez, Yun Gong, Rachel Krupa, Mahipal V Suraneni, Ryon P Graf, Jerry Lee, Stephanie Greene, Angel Rodriguez, Lyndsey Dugan, Jessica Louw, Bora Lim, Carlos H Barcenas, Angela N Marx, Debu Tripathy, Yipeng Wang, Mark Landers, Ryan Dittamore, Naoto T Ueno

Abstract

Purpose: Androgen receptor (AR) is frequently detected in breast cancers, and AR-targeted therapies are showing activity in AR-positive (AR+) breast cancer. However, the role of AR in breast cancers is still not fully elucidated and the biology of AR in breast cancer remains incompletely understood. Circulating tumor cells (CTCs) can serve as prognostic and diagnostic tools, prompting us to measure AR protein expression and conduct genomic analyses on CTCs in patients with metastatic breast cancer.

Methods: Blood samples from patients with metastatic breast cancer were deposited on glass slides, subjected to nuclear staining with DAPI, and reacted with fluorescent-labeled antibodies to detect CD45, cytokeratin (CK), and biomarkers of interest (AR, estrogen receptor [ER], and HER2) on all nucleated cells. The stained slides were scanned and enumerated by non-enrichment-based non-biased approach independent of cell surface epithelial cell adhesion molecule (EpCAM) using the Epic Sciences CTC platform. Data were analyzed using established digital pathology algorithms.

Results: Of 68 patients, 51 (75%) had at least 1 CTC, and 49 of these 51 (96%) had hormone-receptor-positive (HR+)/HER2-negative primary tumors. AR was expressed in CK+ CTCs in 10 patients. Of these 10 patients, 3 also had ER expression in CK+ CTCs. Single cell genomic analysis of 78 CTCs from 1 of these 3 patients identified three distinct copy number patterns. AR+ cells had a lower frequency of chromosomal changes than ER+ and HER2+ cells.

Conclusions: CTC enumeration and analysis using no enrichment or selection provides a non-biased approach to detect AR expression and chromosomal aberrations in CTCs in patients with metastatic breast cancer. The heterogeneity of intrapatient AR expression in CTCs leads to the new hypothesis that patients with AR+ CTCs have heterogeneous disease with multiple drivers. Further studies are warranted to investigate the clinical applicability of AR+ CTCs and their heterogeneity.

Conflict of interest statement

Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: NTU has a legal agreement with Epic Sciences for using their technology for analysis. Epic Sciences paid salary only for Rachel Krupa, Mahipal V. Suraneni, Ryon P. Graf, Jerry Lee, Stephanie Greene, Angel Rodriguez, Lyndsey Dugan, Jessica Louw, Yipeng Wang, Mark Landers and Ryan Dittamore at Epic Sciences. No one from The University of Texas MD Anderson Cancer Center has received direct financial support from Epic Sciences. This affiliation with Epic Sciences does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. AR expression by IHC staining.
Fig 1. AR expression by IHC staining.
Representative images of AR-positive (A) and AR-negative (B) tumors. Arrows: tumors; arrowhead, normal breast epithelium. Original magnifications, x100.
Fig 2. Representative images of CTC subtypes…
Fig 2. Representative images of CTC subtypes identified by the Epic Sciences CTC platform.
Representative fluorescence microscopy images of subtypes of CTCs identified by the Epic Sciences CTC platform. Blood samples from patients with metastatic breast cancer were deposited on glass slides and stained with a cocktail of DAPI and antibodies against CK, CD45, and AR. After staining, CTCs were detected using a digital pathology algorithm and classified into CTC subtypes on the basis of marker expression profile into CK+ CTCs, CK- CTCs, CTC clusters, and apoptotic CTCs. The top panel shows an AR+CK+ CTC with AR expression localized in the nucleus.
Fig 3. Prevalence of AR+ and ER+…
Fig 3. Prevalence of AR+ and ER+ CTCs in metastatic breast cancer samples.
(A) Dot plot depicting AR expression in CTCs identified in patients with metastatic breast cancer with respect to the threshold for AR positivity (indicated by dotted line). Each dot represents a single CTC, and the color indicates the subtype, defined as CK+, CK-, CK+ cluster, or apoptotic (Apop). AR+ CTCs were detected in 10 of 68 patients tested. The 2 patients with only apoptotic AR+ CTCs were excluded. Interpatient heterogeneity was observed in levels of AR expression and subtypes of CTCs. N-term, N-terminal region. (B) Dot plot depicting ER expression in CTCs in the 10 patients with AR+ CTCs with respect to the threshold for ER positivity (indicated by dotted line). Each dot represents a single CTC, and the color indicates the subtype, defined as CK+, CK-, CK+ cluster, or apoptotic (Apop). ER+ CTCs were identified in 3 of 10 patients tested.
Fig 4. Single-cell CNV analysis of CTCs.
Fig 4. Single-cell CNV analysis of CTCs.
Seventy eight CTCs with various biomarker (AR, ER and HER2) positive and negative and 1 white blood cell (germline control) detected in the sample from patient 11 were sequenced and analyzed for the presence of CNVs. (A) Characteristics of CTCs sequenced for CNV analysis according to AR, ER, and HER2 expression. (B) Representative examples of the 3 different CNV patterns identified in patient #11. The bottom figure is the CNV profile of the WBC. (C) Incidences of the 3 CNV patterns in CTCs according to AR, ER, and HER2 expression.

References

    1. Heemers HV, Tindall DJ. Androgen receptor (AR) coregulators: a diversity of functions converging on and regulating the AR transcriptional complex. Endocrine reviews. 2007;28(7):778–808. Epub 2007/10/18. doi: .
    1. Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer. 2001;1(1):34–45. Epub 2002/03/20. doi: .
    1. Reyes EE, VanderWeele DJ, Isikbay M, Duggan R, Campanile A, Stadler WM, et al. Quantitative characterization of androgen receptor protein expression and cellular localization in circulating tumor cells from patients with metastatic castration-resistant prostate cancer. Journal of translational medicine. 2014;12:313 Epub 2014/11/27. doi: ; PubMed Central PMCID: PMCPmc4252013.
    1. Lonergan PE, Tindall DJ. Androgen receptor signaling in prostate cancer development and progression. Journal of carcinogenesis. 2011;10:20 Epub 2011/09/03. doi: ; PubMed Central PMCID: PMCPmc3162670.
    1. Traish AM, Morgentaler A. Epidermal growth factor receptor expression escapes androgen regulation in prostate cancer: a potential molecular switch for tumour growth. British journal of cancer. 2009;101(12):1949–56. doi: ; PubMed Central PMCID: PMC2795439.
    1. Abreu-Martin MT, Chari A, Palladino AA, Craft NA, Sawyers CL. Mitogen-activated protein kinase kinase kinase 1 activates androgen receptor-dependent transcription and apoptosis in prostate cancer. Molecular and cellular biology. 1999;19(7):5143–54. ; PubMed Central PMCID: PMC84357.
    1. Sarker D, Reid AH, Yap TA, de Bono JS. Targeting the PI3K/AKT pathway for the treatment of prostate cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2009;15(15):4799–805. Epub 2009/07/30. doi: .
    1. Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Harris JM, et al. Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer. Cancer research. 2009;69(15):6131–40. Epub 2009/07/30. doi: .
    1. Gucalp A, Tolaney S, Isakoff SJ, Ingle JN, Liu MC, Carey LA, et al. Phase II trial of bicalutamide in patients with androgen receptor-positive, estrogen receptor-negative metastatic Breast Cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2013;19(19):5505–12. Epub 2013/08/24. doi: ; PubMed Central PMCID: PMCPMC4086643.
    1. Traina T, Traina, KM., Denise AY., Joyce O'Shaughnessy, JC., Ahmad A., Catherine MK., Maureen ET., Peter S., Luca Gianni, LG., Rita N., Foluso OA., Stephen C., Joyce LS., Martha EB., Iulia CT., Hirdesh Uppal, ACP., Clifford AH., editor Results from a phase 2 study of enzalutamide (ENZA), an androgen receptor (AR) inhibitor, in advanced AR+ triple-negative breast cancer (TNBC). 2015 ASCO annual meeting; 2014; Chicago, IL.
    1. Chang C, Lee SO, Yeh S, Chang TM. Androgen receptor (AR) differential roles in hormone-related tumors including prostate, bladder, kidney, lung, breast and liver. Oncogene. 2014;33(25):3225–34. doi: .
    1. Hu R, Dawood S, Holmes MD, Collins LC, Schnitt SJ, Cole K, et al. Androgen receptor expression and breast cancer survival in postmenopausal women. Clinical cancer research: an official journal of the American Association for Cancer Research. 2011;17(7):1867–74. doi: ; PubMed Central PMCID: PMC3076683.
    1. Collins LC, Cole KS, Marotti JD, Hu R, Schnitt SJ, Tamimi RM. Androgen receptor expression in breast cancer in relation to molecular phenotype: results from the Nurses' Health Study. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc. 2011;24(7):924–31. doi: ; PubMed Central PMCID: PMC3128675.
    1. Guiu S, Charon-Barra C, Vernerey D, Fumoleau P, Campone M, Spielmann M, et al. Coexpression of androgen receptor and FOXA1 in nonmetastatic triple-negative breast cancer: ancillary study from PACS08 trial. Future oncology (London, England). 2015;11(16):2283–97. Epub 2015/08/12. doi: .
    1. Elebro K, Borgquist S, Simonsson M, Markkula A, Jirstrom K, Ingvar C, et al. Combined Androgen and Estrogen Receptor Status in Breast Cancer: Treatment Prediction and Prognosis in a Population-Based Prospective Cohort. Clinical cancer research: an official journal of the American Association for Cancer Research. 2015;21(16):3640–50. doi: .
    1. De Amicis F, Thirugnansampanthan J, Cui Y, Selever J, Beyer A, Parra I, et al. Androgen receptor overexpression induces tamoxifen resistance in human breast cancer cells. Breast cancer research and treatment. 2010;121(1):1–11. doi: ; PubMed Central PMCID: PMC2995248.
    1. Takagi K, Miki Y, Nagasaki S, Hirakawa H, Onodera Y, Akahira J, et al. Increased intratumoral androgens in human breast carcinoma following aromatase inhibitor exemestane treatment. Endocrine-related cancer. 2010;17(2):415–30. doi: .
    1. Vera-Badillo FE, Templeton AJ, de Gouveia P, Diaz-Padilla I, Bedard PL, Al-Mubarak M, et al. Androgen receptor expression and outcomes in early breast cancer: a systematic review and meta-analysis. Journal of the National Cancer Institute. 2014;106(1):djt319 Epub 2013/11/26. doi: .
    1. Tsang JY, Ni YB, Chan SK, Shao MM, Law BK, Tan PH, et al. Androgen receptor expression shows distinctive significance in ER positive and negative breast cancers. Annals of surgical oncology. 2014;21(7):2218–28. Epub 2014/03/19. doi: .
    1. Hilborn E, Gacic J, Fornander T, Nordenskjold B, Stal O, Jansson A. Androgen receptor expression predicts beneficial tamoxifen response in oestrogen receptor-alpha-negative breast cancer. British journal of cancer. 2016;114(3):248–55. Epub 2016/01/08. doi: ; PubMed Central PMCID: PMCPMC4742586.
    1. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. The Journal of clinical investigation. 2011;121(7):2750–67. doi: ; PubMed Central PMCID: PMC3127435.
    1. Masuda H, Baggerly KA, Wang Y, Zhang Y, Gonzalez-Angulo AM, Meric-Bernstam F, et al. Differential response to neoadjuvant chemotherapy among 7 triple-negative breast cancer molecular subtypes. Clinical cancer research: an official journal of the American Association for Cancer Research. 2013;19(19):5533–40. doi: ; PubMed Central PMCID: PMC3813597.
    1. Parker JS, Peterson AC, Tudor IC, Hoffman J, H. U. A novel biomarker to predict sensitivity to enzalutamide (ENZA) in TNBC. 2015 ASCO Annual Meeting. Chicago, IL.
    1. Kono M, Fujii T, Lim B, Karuturi MS, Tripathy D, Ueno NT. Androgen Receptor Function and Androgen Receptor-Targeted Therapies in Breast Cancer: A Review. JAMA oncology. 2017. Epub 2017/03/17. doi: .
    1. Giuliano M, Giordano A, Jackson S, Hess KR, De Giorgi U, Mego M, et al. Circulating tumor cells as prognostic and predictive markers in metastatic breast cancer patients receiving first-line systemic treatment. Breast Cancer Res. 2011;13(3):R67 doi:
    1. de Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2008;14(19):6302–9. doi: .
    1. Budd GT, Cristofanilli M, Ellis MJ, Stopeck A, Borden E, Miller MC, et al. Circulating tumor cells versus imaging—predicting overall survival in metastatic breast cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2006;12(21):6403–9. doi: .
    1. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. The New England journal of medicine. 2004;351(8):781–91. doi: .
    1. Pantel K, Riethdorf S. Pathology: are circulating tumor cells predictive of overall survival? Nature Reviews Clinical Oncology. 2009;6(4):190–1. doi:
    1. Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2008;26(19):3213–21. doi: .
    1. Smerage JB, Barlow WE, Hortobagyi GN, Winer EP, Leyland-Jones B, Srkalovic G, et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2014;32(31):3483–9. Epub 2014/06/04. doi: ; PubMed Central PMCID: PMCPMC4209100.
    1. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clinical cancer research: an official journal of the American Association for Cancer Research. 2004;10(20):6897–904. doi: .
    1. Pantel K, Alix-Panabieres C, Riethdorf S. Cancer micrometastases. Nature reviews Clinical oncology. 2009;6(6):339–51. Epub 2009/04/29. doi: .
    1. Lianidou ES, Mavroudis D, Georgoulias V. Clinical challenges in the molecular characterization of circulating tumour cells in breast cancer. British journal of cancer. 2013;108(12):2426–32. Epub 2013/06/13. doi: ; PubMed Central PMCID: PMCPmc3694246.
    1. Plaks V, Koopman CD, Werb Z. Circulating Tumor Cells. Science (New York, NY). 2013;341(6151):1186–8. doi:
    1. Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, Collura CV, et al. Detection of mutations in EGFR in circulating lung-cancer cells. New England Journal of Medicine. 2008;359(4):366–77. doi:
    1. Markou A, Farkona S, Schiza C, Efstathiou T, Kounelis S, Malamos N, et al. PIK3CA mutational status in circulating tumor cells can change during disease recurrence or progression in patients with breast cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2014;20(22):5823–34. Epub 2014/11/16. doi: .
    1. Pestrin M, Salvianti F, Galardi F, De Luca F, Turner N, Malorni L, et al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Molecular oncology. 2015;9(4):749–57. Epub 2014/12/30. doi: .
    1. Fehm T, Muller V, Aktas B, Janni W, Schneeweiss A, Stickeler E, et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: a prospective, multicenter trial. Breast cancer research and treatment. 2010;124(2):403–12. doi: .
    1. Scher HI, Lu D, Schreiber NA, Louw J, Graf RP, Vargas HA, et al. Association of AR-V7 on Circulating Tumor Cells as a Treatment-Specific Biomarker With Outcomes and Survival in Castration-Resistant Prostate Cancer. JAMA oncology. 2016;2(11):1441–9. Epub 2016/06/05. doi: ; PubMed Central PMCID: PMCPMC5206761.
    1. Lu D, Graf RP, Harvey M, Madan RA, Heery C, Marte J, et al., editors. Detection and Characterization of Circulating Tumor Cells (CTCs) from Frozen Peripheral Blood Mononuclear Cells. American Association for Cancer Research (AACR) Annual Meeting 2015; 2015; Philadelphia, PA.
    1. Werner SL, Graf RP, Landers ML, Valenta DT, Schroeder M, Greene SB, et al. Analytical Validation and Capabilities of the Epic CTC Platform: Enrichment-Free Circulating Tumour Cell Detection and Characterization. Journal of Circulating Biomarkers. 2015;4(3).
    1. Punnoose EA, Ferraldeschi R, Szafer-Glusman E, Tucker EK, Mohan S, Flohr P, et al. PTEN loss in circulating tumour cells correlates with PTEN loss in fresh tumour tissue from castration-resistant prostate cancer patients. British journal of cancer. 2015;113(8):1225–33. doi:
    1. Beltran H, Jendrisak A, Landers M, Mosquera JM, Kossai M, Louw J, et al. The Initial Detection and Partial Characterization of Circulating Tumor Cells in Neuroendocrine Prostate Cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2015. Epub 2015/12/17. doi: .
    1. Marrinucci D, Bethel K, Bruce RH, Curry DN, Hsieh B, Humphrey M, et al. Case study of the morphologic variation of circulating tumor cells. Human pathology. 2007;38(3):514–9. doi:
    1. Marrinucci D, Bethel K, Kolatkar A, Luttgen MS, Malchiodi M, Baehring F, et al. Fluid biopsy in patients with metastatic prostate, pancreatic and breast cancers. Physical biology. 2012;9(1):016003 doi:
    1. Marrinucci D, Bethel K, Lazar D, Fisher J, Huynh E, Clark P, et al. Cytomorphology of circulating colorectal tumor cells:a small case series. Journal of oncology. 2010;2010:861341 Epub 2010/01/30. doi: ; PubMed Central PMCID: PMCPMC2810476.
    1. Greene SB, Dago AE, Leitz LJ, Wang Y, Lee J, Werner SL, et al. Chromosomal Instability Estimation Based on Next Generation Sequencing and Single Cell Genome Wide Copy Number Variation Analysis. PLoS One. 2016;11(11):e0165089 Epub 2016/11/17. doi: .
    1. Anantharaman A, Friedlander T, Lu D, Krupa R, Premasekharan G, Hough J, et al. Programmed death-ligand 1 (PD-L1) characterization of circulating tumor cells (CTCs) in muscle invasive and metastatic bladder cancer patients. BMC cancer. 2016;16(1):744 doi:
    1. Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2013;31(31):3997–4013. Epub 2013/10/09. doi: .
    1. McDaniel AS, Ferraldeschi R, Krupa R, Landers M, Graf R, Louw J, et al. Phenotypic diversity of circulating tumour cells in patients with metastatic castration‐resistant prostate cancer. BJU international. 2016.
    1. Graf RP, Wang Y, Schreiber N, McLaughlin B, Greene S, Rodriguez A, et al. Abstract 1740: Phenotypic, genomic, and clinical associations of Circulating Tumor Cells (CTCs) lacking epithelial biomarkers in metastatic Castration Resistant Prostate Cancer (mCRPC). Cancer research. 2017;77(13 Supplement):1740–. doi:
    1. Fehm T, Müller V, Aktas B, Janni W, Schneeweiss A, Stickeler E, et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: a prospective, multicenter trial. Breast cancer research and treatment. 2010;124(2):403–12. doi:
    1. Turner-Ivey B, Guest ST, Irish JC, Kappler CS, Garrett-Mayer E, Wilson RC, et al. KAT6A, a chromatin modifier from the 8p11-p12 amplicon is a candidate oncogene in luminal breast cancer. Neoplasia. 2014;16(8):644–55. doi:
    1. Keilty D, Buchanan M, Ntapolias K, Aleynikova O, Tu D, Li X, et al. RSF1 and Not Cyclin D1 Gene Amplification May Predict Lack of Benefit from Adjuvant Tamoxifen in High-Risk Pre-Menopausal Women in the MA. 12 Randomized Clinical Trial. PloS one. 2013;8(12):e81740 doi:
    1. Scher H. I., Graf R., Louw J., Jendrisak A., Johnson A., Green S., et al., editors. Single CTC Characterization Identifies Phenotypic and Genomic Heterogeneity as a Mechanism of Resistance to AR Signaling Directed Therapies (AR Tx) in mCRPC Patient. ASCO 2016 Genitourinary Cancers Symposium; 2016; San Francisco, CA.

Source: PubMed

Подписаться