T. Holbro, R. R. Beerli, F. Maurer, M. Koziczak, C. F. Barbas-3rd et al., The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation, Proc Natl Acad Sci U S A, vol.100, pp.8933-8941, 2003.

R. Naidu, M. Yadav, S. Nair, and M. K. Kutty, Expression of c-erbB3 protein in primary breast carcinomas, Br J Cancer, vol.78, pp.1385-90, 1998.

V. Pawlowski, F. Revillion, M. Hebbar, L. Hornez, and J. P. Peyrat, Prognostic value of the type I growth factor receptors in a large series of human primary breast cancers quantified with a real-time reverse transcription-polymerase chain reaction assay, Clin Cancer Res, vol.6, pp.4217-4242, 2000.

Z. Suo, H. S. Berner, B. Risberg, M. G. Karlsson, and J. M. Nesland, Estrogen receptoralpha and C-ERBB-4 expression in breast carcinomas, Virchows Arch, vol.439, pp.62-71, 2001.

Z. Suo, B. Risberg, M. G. Kalsson, K. Willman, A. Tierens et al., EGFR family expression in breast carcinomas. c-erbB-2 and c-erbB-4 receptors have different effects on survival, J Pathol, vol.196, pp.17-25, 2002.

S. M. Tovey, C. J. Witton, J. M. Bartlett, P. D. Stanton, J. R. Reeves et al., Outcome and human epidermal growth factor receptor (HER) 1-4 status in invasive breast carcinomas with proliferation indices evaluated by bromodeoxyuridine labelling, Breast Cancer Res, vol.6, pp.246-51, 2004.

C. J. Witton, J. R. Reeves, J. J. Going, T. G. Cooke, and J. M. Bartlett, Expression of the HER1-4 family of receptor tyrosine kinases in breast cancer, J Pathol, vol.200, pp.290-297, 2003.

D. J. Slamon, B. Leyland-jones, S. Shak, H. Fuchs, V. Paton et al., Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2, N Engl J Med, vol.344, pp.783-92, 2001.

C. A. Hudis, Trastuzumab -mechanism of action and use in clinical practice, N Engl J Med, vol.357, pp.39-51, 2007.

M. J. Piccart-gebhart, M. Procter, B. Leyland-jones, A. Goldhirsch, M. Untch et al., Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer, N Engl J Med, vol.353, pp.1659-72, 2005.

I. Smith, M. Procter, R. D. Gelber, S. Guillaume, A. Feyereislova et al., 2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial, Lancet, vol.369, pp.29-36, 2007.

C. E. Geyer, J. Forster, D. Lindquist, S. Chan, C. G. Romieu et al., Lapatinib plus capecitabine for HER2-positive advanced breast cancer, N Engl J Med, vol.355, pp.2733-2776, 2006.

H. L. Gomez, D. C. Doval, M. A. Chavez, P. C. Ang, Z. Aziz et al., Efficacy and safety of lapatinib as first-line therapy for ErbB2-amplified locally advanced or metastatic breast cancer, J Clin Oncol, vol.26, pp.2999-3005, 2008.

D. Leo, A. Gomez, H. L. Aziz, Z. Zvirbule, Z. Bines et al., Phase III, double-blind, randomized study comparing lapatinib plus paclitaxel with placebo plus paclitaxel as first-line treatment for metastatic breast cancer, J Clin Oncol, vol.26, pp.5544-52, 2008.

J. Baselga, I. Bradbury, H. Eidtmann, D. Cosimo, S. De-azambuja et al., Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial, Lancet, vol.379, pp.633-673, 2012.

J. Baselga, J. Cortes, S. B. Kim, S. A. Im, R. Hegg et al., Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer, N Engl J Med, vol.366, pp.109-128, 2012.

L. Gianni, T. Pienkowski, Y. H. Im, L. Roman, L. M. Tseng et al., Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial, Lancet Oncol, vol.13, pp.25-32, 2012.

T. O. Nielsen, F. D. Hsu, K. Jensen, M. Cheang, G. Karaca et al., Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma, Clin Cancer Res, vol.10, pp.5367-74, 2004.

K. A. Hoadley, V. J. Weigman, C. Fan, L. R. Sawyer, X. He et al., EGFR associated expression profiles vary with breast tumor subtype, BMC Genomics, vol.8, p.258, 2007.

J. Baselga, P. Gomez, R. Greil, S. Braga, M. A. Climent et al., Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer, J Clin Oncol, vol.31, pp.2586-92, 2013.

L. A. Carey, H. S. Rugo, P. K. Marcom, E. L. Mayer, F. J. Esteva et al., TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer, J Clin Oncol, vol.30, pp.2615-2638, 2012.

J. O'shaughnessy, D. J. Weckstein, S. J. Vukelja, K. Mcintyre, L. Krekow et al., Preliminary results of a randomized phase II study of weekly irinotecan/carboplatin with or without cetuximab in patients with metastatic breast cancer, Breast Cancer Res Treat, vol.106, p.32, 2007.

J. M. Nabholtz, C. Abrial, M. A. Mouret-reynier, M. M. Dauplat, B. Weber et al., Multicentric neoadjuvant phase II study of panitumumab combined with an anthracycline/taxane-based chemotherapy in operable triple-negative breast cancer: identification of biologically defined signatures predicting treatment impact, Ann Oncol, vol.25, pp.1570-1577, 2014.

E. A. Perez, V. J. Suman, N. E. Davidson, S. Martino, P. A. Kaufman et al., HER2 testing by local, central, and reference laboratories in specimens from the North Central Cancer Treatment Group N9831 intergroup adjuvant trial, J Clin Oncol, vol.24, pp.3032-3040, 2006.

A. E. Mccullough, P. Dell'orto, M. M. Reinholz, R. D. Gelber, A. C. Dueck et al., Central pathology laboratory review of HER2 and ER in early breast cancer: an ALTTO trial, Breast Cancer Res Treat, vol.143, pp.485-92, 2014.

E. A. Perez, M. F. Press, A. C. Dueck, R. B. Jenkins, C. Kim et al., Immunohistochemistry and fluorescence in situ hybridization assessment of HER2 in clinical trials of adjuvant therapy for breast cancer (NCCTG N9831, BCIRG 006, and BCIRG 005), Breast Cancer Res Treat, vol.138, pp.99-108, 2013.

A. C. Wolff, M. E. Hammond, D. G. Hicks, M. Dowsett, L. M. Mcshane 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, Arch Pathol Lab Med, vol.138, pp.241-56, 2014.

C. R. Taylor and R. M. Levenson, Quantification of immunohistochemistry -issues concerning methods, utility and semiquantitative assessment II, Histopathology, vol.49, pp.411-435, 2006.

A. C. Wolff, M. E. Hammond, J. N. Schwartz, K. L. Hagerty, D. C. Allred et al., American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer, J Clin Oncol, vol.25, pp.118-163, 2007.

K. B. Engel and H. M. Moore, Effects of preanalytical variables on the detection of proteins by immunohistochemistry in formalin-fixed, paraffin-embedded tissue, Arch Pathol Lab Med, vol.135, pp.537-580, 2011.

T. Khoury, Delay to formalin fixation alters morphology and immunohistochemistry for breast carcinoma, Appl Immunohistochem Mol Morphol, vol.20, pp.531-573, 2012.

B. P. Portier, Z. Wang, E. Downs-kelly, J. J. Rowe, D. Patil et al., Delay to formalin fixation 'cold ischemia time': effect on ERBB2 detection by in-situ hybridization and immunohistochemistry, Mod Pathol, vol.26, pp.1-9, 2013.

R. L. Camp, G. G. Chung, and D. L. Rimm, Automated subcellular localization and quantification of protein expression in tissue microarrays, Nat Med, vol.8, pp.1323-1330, 2002.

M. D. Gustavson, B. Bourke-martin, D. Reilly, M. Cregger, C. Williams et al., Standardization of HER2 immunohistochemistry in breast cancer by automated quantitative analysis, Arch Pathol Laboratory Med, vol.133, pp.1413-1422, 2009.

A. Mccabe, M. Dolled-filhart, R. L. Camp, and D. L. Rimm, Automated quantitative analysis (AQUA) of in situ protein expression, antibody concentration, and prognosis, J Natl Cancer Inst, vol.97, pp.1808-1823, 2005.

R. Colomer, A. Llombart-cussac, A. Lluch, A. Barnadas, B. Ojeda et al., Biweekly paclitaxel plus gemcitabine in advanced breast cancer: phase II trial and predictive value of HER2 extracellular domain, Ann Oncol, vol.15, pp.201-207, 2004.

W. J. Kostler, G. G. Steger, A. Soleiman, B. Schwab, C. F. Singer et al., Monitoring of serum Her-2/neu predicts histopathological response to neoadjuvant trastuzumab-based therapy for breast cancer, Anticancer Res, vol.24, issue.2C, pp.1127-1157, 2004.

S. Lennon, C. Barton, L. Banken, L. Gianni, M. Marty et al., Utility of serum HER2 extracellular domain assessment in clinical decision making: pooled analysis of four trials of trastuzumab in metastatic breast cancer, J Clin Oncol, vol.27, pp.1685-93, 2009.

W. Huang, M. Reinholz, J. Weidler, Y. L. Paquet, A. Whitcomb et al., Comparison of central HER2 testing with quantitative total HER2 expression and HER2 homodimer measurements using a novel proximity-based assay, Am J Clin Pathol, vol.134, pp.303-314, 2010.

Y. Shi, W. Huang, Y. Tan, J. X. Dua, R. Penuel et al., A novel proximity assay for the detection of proteins and protein complexes: quantitation of HER1 and HER2 total protein expression and homodimerization in formalin-fixed, paraffin-embedded cell lines and breast cancer tissue, Diagn Mol Pathol, vol.18, pp.11-21, 2009.

L. Defazio-eli, K. Strommen, T. Dao-pick, G. Parry, L. Goodman et al., Quantitative assays for the measurement of HER1-HER2 heterodimerization and phosphorylation in cell lines and breast tumors: applications for diagnostics and targeted drug mechanism of action, Breast Cancer Res, vol.13, p.44, 2011.

R. Dua, J. Zhang, P. Nhonthachit, E. Penuel, C. Petropoulos et al., EGFR over-expression and activation in high HER2, ER negative breast cancer cell line induces trastuzumab resistance, Breast Cancer Res Treat, vol.122, pp.685-97, 2010.

J. Sperinde, J. X. Banerjee, J. Penuel, E. Saha, A. Diedrich et al., Quantitation of p95HER2 in paraffin sections by using a p95-specific antibody and correlation with outcome in a cohort of trastuzumab-treated breast cancer patients, Clin Cancer Res, vol.16, pp.4226-4261, 2010.

A. Mukherjee, Y. Badal, X. T. Nguyen, J. Miller, A. Chenna et al., Profiling the HER3/PI3K pathway in breast tumors using proximity-directed assays identifies correlations between protein complexes and phosphoproteins, PLoS One, vol.6, p.16443, 2011.

M. R. Arkin and J. A. Wells, Small-molecule inhibitors of protein-protein interactions: progressing towards the dream, Nat Rev Drug Discov, vol.3, pp.301-318, 2004.

C. Tovar, J. Rosinski, Z. Filipovic, B. Higgins, K. Kolinsky et al., From the Cover: Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy, Proc Natl Acad Sci, vol.103, pp.1888-93, 2006.

L. T. Vassilev, B. T. Vu, B. Graves, D. Carvajal, F. Podlaski et al., In vivo activation of the p53 pathway by small-molecule antagonists of MDM2, Science, vol.303, pp.844-852, 2004.

D. B. Solit and N. Rosen, Resistance to BRAF inhibition in melanomas, N Engl J Med, vol.364, pp.772-776, 2011.

H. Y. Chuang, E. Lee, Y. T. Liu, D. Lee, and T. Ideker, Network-based classification of breast cancer metastasis, Mol Syst Biol, vol.3, p.140, 2007.

I. W. Taylor, R. Linding, D. Warde-farley, Y. Liu, C. Pesquita et al., Dynamic modularity in protein interaction networks predicts breast cancer outcome, Nat Biotechnol, vol.27, pp.199-204, 2009.

M. Peter, S. M. Ameer-beg, M. K. Hughes, M. D. Keppler, S. Prag et al., Multiphoton-FLIM quantification of the EGFP-mRFP1 FRET pair for localization of membrane receptor-kinase interactions, Biophys J, vol.88, pp.1224-1261, 2005.

P. R. Barber, S. M. Ameer-beg, J. Gilbey, L. M. Carlin, M. Keppler et al., Multiphoton time-domain fluorescence lifetime imaging microscopy: practical application to protein-protein interactions using global analysis, J R Soc Interface, vol.6, pp.93-105, 2008.

M. T. Kelleher, G. Fruhwirth, G. Patel, E. Ofo, F. Festy et al., The potential of optical proteomic technologies to individualize prognosis and guide rational treatment for cancer patients, Target Oncol, vol.4, pp.235-52, 2009.

G. O. Fruhwirth, L. P. Fernandes, G. Weitsman, G. Patel, M. Kelleher et al., How Forster resonance energy transfer imaging improves the understanding of protein interaction networks in cancer biology, Chemphyschem, vol.12, pp.442-61, 2011.

N. Anilkumar, M. Parsons, R. Monk, T. Ng, and J. C. Adams, Interaction of fascin and protein kinase Calpha: a novel intersection in cell adhesion and motility, EMBO J, vol.22, pp.5390-402, 2003.

J. W. Legg, C. A. Lewis, M. Parsons, T. Ng, and C. M. Isacke, A novel PKC-regulated mechanism controls CD44 ezrin association and directional cell motility, Nat Cell Biol, vol.4, pp.399-407, 2002.

T. Ng, M. Parsons, W. E. Hughes, J. Monypenny, D. Zicha et al., Ezrin is a downstream effector of trafficking PKC-integrin complexes involved in the control of cell motility, EMBO J, vol.20, pp.2723-2764, 2001.

T. Ng, A. Squire, G. Hansra, F. Bornancin, C. Prevostel et al., Imaging protein kinase Calpha activation in cells, Science, vol.283, pp.2085-2094, 1999.

M. Parsons, M. D. Keppler, A. Kline, A. Messent, M. J. Humphries et al., Site-directed perturbation of protein kinase C-integrin interaction blocks carcinoma cell chemotaxis, Mol Cell Biol, vol.22, pp.5897-911, 2002.

M. Parsons, J. Monypenny, S. M. Ameer-beg, T. H. Millard, L. M. Machesky et al., Spatially distinct binding of Cdc42 to PAK1 and N-WASP in breast carcinoma cells, Mol Cell Biol, vol.25, pp.1680-95, 2005.

S. Prag, M. Parsons, M. D. Keppler, S. M. Ameer-beg, P. Barber et al., Activated ezrin promotes cell migration through recruitment of the GEF Dbl to lipid rafts and preferential downstream activation of Cdc42, Mol Biol Cell, vol.18, pp.2935-2983, 2007.

S. Ganesan, S. M. Ameer-beg, T. T. Ng, B. Vojnovic, and F. S. Wouters, A dark yellow fluorescent protein (YFP)-based Resonance Energy-Accepting Chromoprotein (REACh) for Forster resonance energy transfer with GFP, Proc Natl Acad Sci, vol.103, pp.4089-94, 2006.

S. Dadke, S. Cotteret, S. C. Yip, Z. M. Jaffer, F. Haj et al., Regulation of protein tyrosine phosphatase 1B by sumoylation, Nat Cell Biol, vol.9, pp.80-85, 2007.

K. Makrogianneli, L. M. Carlin, M. D. Keppler, D. R. Matthews, E. Ofo et al., Integrating receptor signal inputs that influence small Rho GTPase activation dynamics at the immunological synapse, Mol Cell Biol, vol.29, pp.2997-3006, 2009.

J. R. Morris, C. Boutell, M. Keppler, R. Densham, D. Weekes et al., The SUMO modification pathway is involved in the BRCA1 response to genotoxic stress, Nature, vol.462, pp.886-90, 2009.

L. M. Carlin, R. Evans, H. Milewicz, L. Fernandes, D. R. Matthews et al., A targeted siRNA screen identifies regulators of Cdc42 activity at the natural killer cell immunological synapse, Sci Signal, vol.4, p.81, 2011.

M. Keese, R. J. Magdeburg, T. Herzog, T. Hasenberg, M. Offterdinger et al., Imaging epidermal growth factor receptor phosphorylation in human colorectal cancer cells and human tissues, J Biol Chem, vol.280, pp.27826-27857, 2005.

A. Kong, P. Leboucher, R. Leek, V. Calleja, S. Winter et al., Prognostic value of an activation state marker for epidermal growth factor receptor in tissue microarrays of head and neck cancer, Cancer Res, vol.66, pp.2834-2877, 2006.

M. Parsons and T. Ng, Intracellular coupling of adhesion receptors: molecular proximity measurements, Methods Cell Biol, vol.69, pp.261-78, 2002.

P. R. Barber, I. D. Tullis, G. P. Pierce, R. G. Newman, J. Prentice et al., The Gray Institute 'open' high-content, fluorescence lifetime microscopes, J Microsc, vol.251, pp.154-67, 2013.

G. Weitsman, K. Lawler, M. Kelleher, J. Barrett, P. R. Barber et al., Imaging tumour heterogeneity of the consequence of a PKC-substrate interaction in breast cancer patients, Biochem Soc Trans, 2014.

M. Y. Berezin and S. Achilefu, Fluorescence lifetime measurements and biological imaging, Chem Rev, vol.110, pp.2641-84, 2010.

P. R. Barber, I. Tullis, M. I. Rowley, C. D. Martins, G. Weitsman et al., The Gray Institute open microscopes applied to radiobiology and protein interaction studies, Three-Dimensional and Multidimensional Microscopy: Image Acquisition and Processing XXI, vol.8949, 2014.

J. Tao, P. Castel, N. Radosevic-robin, M. Elkabets, N. Auricchio et al., Blockade of EGFR and HER3 enhances PI3K/Akt anti-tumor activity in triple negative breast cancer, Sci Signal, vol.7, p.29, 2014.

T. Kiuchi, E. Ortiz-zapater, J. Monypenny, D. R. Matthews, L. K. Nguyen et al., The ErbB4 CYT2 variant protects EGFR from ligand-induced degradation to enhance cancer cell motility, Sci Signal, vol.7, p.78, 2014.

R. Aebersold and M. Mann, Mass spectrometry-based proteomics, Nature, vol.422, pp.198-207, 2003.

B. Domon and R. Aebersold, Mass spectrometry and protein analysis, Science, vol.312, pp.212-219, 2006.

T. A. Addona, S. E. Abbatiello, B. Schilling, S. J. Skates, D. R. Mani et al., Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma, Nat Biotechnol, vol.27, pp.633-674, 2009.

T. Nilsson, M. Mann, R. Aebersold, J. R. Yates-3rd, A. Bairoch et al., Mass spectrometry in high-throughput proteomics: ready for the big time, Nat Methods, vol.7, pp.681-686, 2010.

P. A. Rudnick, K. R. Clauser, L. E. Kilpatrick, D. V. Tchekhovskoi, P. Neta et al., Performance metrics for liquid chromatography-tandem mass spectrometry systems in proteomics analyses, Mol Cell Proteomics, vol.9, pp.225-266, 2010.

H. Keshishian, T. Addona, M. Burgess, E. Kuhn, and S. A. Carr, Quantitative, multiplexed assays for low abundance proteins in plasma by targeted mass spectrometry and stable isotope dilution, Mol Cell Proteomics, vol.6, pp.2212-2241, 2007.

N. W. Bateman, M. Sun, R. Bhargava, B. L. Hood, M. M. Darfler et al., Differential proteomic analysis of late-stage and recurrent breast cancer from formalin-fixed paraffin-embedded tissues, J Proteome Res, vol.10, pp.1323-1355, 2011.

D. A. Prieto, B. L. Hood, M. M. Darfler, T. G. Guiel, D. A. Lucas et al., Liquid Tissue: proteomic profiling of formalin-fixed tissues, Biotechniques, pp.32-37, 2005.

B. L. Hood, M. M. Darfler, T. G. Guiel, B. Furusato, D. A. Lucas et al., Proteomic analysis of formalin-fixed prostate cancer tissue, Mol Cell Proteomics, vol.4, pp.1741-53, 2005.

T. Hembrough, S. Thyparambil, W. L. Liao, M. M. Darfler, J. Abdo et al., Application of selected reaction monitoring for multiplex quantification of clinically validated biomarkers in formalin-fixed, paraffin-embedded tumor tissue, J Mol Diagn, vol.15, pp.454-65, 2013.

S. K. Huang, M. M. Darfler, M. B. Nicholl, J. You, K. G. Bemis et al., LC/MSbased quantitative proteomic analysis of paraffin-embedded archival melanomas reveals potential proteomic biomarkers associated with metastasis, PLoS One, vol.4, p.4430, 2009.

N. W. Bateman, M. Sun, B. L. Hood, M. S. Flint, and T. P. Conrads, Defining central themes in breast cancer biology by differential proteomics: conserved regulation of cell spreading and focal adhesion kinase, J Proteome Res, vol.9, pp.5311-5335, 2010.

W. Cheung, M. M. Darfler, H. Alvarez, B. L. Hood, T. P. Conrads et al., Application of a global proteomic approach to archival precursor lesions: deleted in malignant brain tumors 1 and tissue transglutaminase 2 are upregulated in pancreatic cancer precursors, Pancreatology, vol.8, pp.608-624, 2008.

L. V. Desouza, O. Krakovska, M. M. Darfler, D. B. Krizman, A. D. Romaschin et al., mTRAQ-based quantification of potential endometrial carcinoma biomarkers from archived formalin-fixed paraffin-embedded tissues, Proteomics, vol.10, pp.3108-3124, 2010.

T. Hembrough, S. Thyparambil, W. L. Liao, M. M. Darfler, J. Abdo et al., Selected reaction monitoring (SRM) analysis of epidermal growth factor receptor (EGFR) in formalin fixed tumor tissue, Clin Proteomics, vol.9, p.5, 2012.

B. N. Patel, N. Sharma, M. Sanyal, and P. S. Shrivastav, High throughput and sensitive determination of trazodone and its primary metabolite, m-chlorophenylpiperazine, in human plasma by liquid chromatographytandem mass spectrometry, J Chromatogr B Analyt Technol Biomed Life Sci, vol.871, pp.44-54, 2008.

B. Spurrier, S. Ramalingam, and S. Nishizuka, Reverse-phase protein lysate microarrays for cell signaling analysis, Nat Protocols, vol.3, pp.1796-808, 2008.

P. Kim, X. Liu, T. Lee, L. Liu, R. Barham et al., Highly sensitive proximity mediated immunoassay reveals HER2 status conversion in the circulating tumor cells of metastatic breast cancer patients, Proc Natl Acad Sci U S A, vol.9, p.75, 2011.

L. Arnould, Y. Denoux, G. Macgrogan, F. Penault-llorca, M. Fiche et al., Agreement between chromogenic in situ hybridisation (CISH) and FISH in the determination of HER2 status in breast cancer, Br J Cancer, vol.88, pp.1587-91, 2003.

J. M. Bartlett, F. M. Campbell, M. Ibrahim, P. Wencyk, I. Ellis et al., Chromogenic in situ hybridization: a multicenter study comparing silver in situ hybridization with FISH, Am J Clin Pathol, vol.132, pp.514-534, 2009.

G. D. Francis, M. A. Jones, G. F. Beadle, and S. R. Stein, Bright-field in situ hybridization for HER2 gene amplification in breast cancer using tissue microarrays: correlation between chromogenic (CISH) and automated silver-enhanced (SISH) methods with patient outcome, Diagn Mol Pathol, vol.18, pp.88-95, 2009.

W. M. Hanna and K. Kwok, Chromogenic in-situ hybridization: a viable alternative to fluorescence in-situ hybridization in the HER2 testing algorithm, Modern Pathol, vol.19, pp.481-488, 2006.

A. Lebeau, D. Deimling, C. Kaltz, A. Sendelhofert, A. Iff et al., Her-2/ neu analysis in archival tissue samples of human breast cancer: comparison of immunohistochemistry and fluorescence in situ hybridization, J Clin Oncol, vol.19, pp.354-63, 2001.

G. Pauletti, S. Dandekar, H. Rong, L. Ramos, H. Peng et al., Assessment of methods for tissue-based detection of the HER-2/neu alteration in human breast cancer: a direct comparison of fluorescence in situ hybridization and immunohistochemistry, J Clin Oncol, vol.18, pp.3651-64, 2000.

A. Brugmann, G. Lelkaitis, S. Nielsen, K. G. Jensen, and V. Jensen, Testing HER2 in breast cancer: a comparative study on BRISH, FISH, and IHC, Appl Immunohistochem Mol Morphol, vol.19, pp.203-214, 2011.

W. M. Hanna, J. Ruschoff, M. Bilous, R. A. Coudry, M. Dowsett et al., HER2 in situ hybridization in breast cancer: clinical implications of polysomy 17 and genetic heterogeneity, Mod Pathol, vol.27, pp.4-18, 2014.

C. B. Moelans, R. A. De-weger, and P. J. Van-diest, Absence of chromosome 17 polysomy in breast cancer: analysis by CEP17 chromogenic in situ hybridization and multiplex ligation-dependent probe amplification, Breast Cancer Res Treat, vol.120, pp.1-7, 2010.

I. T. Yeh, M. A. Martin, R. S. Robetorye, A. R. Bolla, C. Mccaskill et al., Clinical validation of an array CGH test for HER2 status in breast cancer reveals that polysomy 17 is a rare event, Modern Pathol, vol.22, pp.1169-75, 2009.

C. Franchet, T. Filleron, A. Cayre, E. Mounie, F. Penault-llorca et al., Instant-quality fluorescence in-situ hybridization as a new tool for HER2 testing in breast cancer: a comparative study, Histopathology, vol.64, pp.274-83, 2014.

J. P. Schouten, C. J. Mcelgunn, R. Waaijer, D. Zwijnenburg, F. Diepvens et al., Relative quantification of 40 nucleic acid sequences by multiplex ligationdependent probe amplification, Nucleic Acids Res, vol.30, p.57, 2002.

E. Moerland, R. L. Van-hezik, T. C. Van-der-aa, M. W. Van-beek, and A. J. Van-den-brule, Detection of HER2 amplification in breast carcinomas: comparison of multiplex ligation-dependent probe amplification (MLPA) and fluorescence in situ hybridization (FISH) combined with automated spot counting, Cell Oncol, vol.28, pp.151-160, 2006.

C. B. Moelans, R. A. De-weger, C. Ezendam, and P. J. Van-diest, HER-2/neu amplification testing in breast cancer by multiplex ligation-dependent probe amplification: influence of manual-and laser microdissection, BMC Cancer, vol.9, p.4, 2009.

C. B. Moelans, R. A. De-weger, M. T. Van-blokland, E. Van-der-wall, and P. J. Van-diest, Simultaneous detection of TOP2A and HER2 gene amplification by multiplex ligation-dependent probe amplification in breast cancer, Modern Pathol, vol.23, pp.62-70, 2010.

C. C. Kuijpers, C. B. Moelans, H. J. Van-slooten, A. Horstman, J. W. Hinrichs et al., Added value of HER-2 amplification testing by multiplex ligation-dependent probe amplification in invasive breast cancer, PLoS One, vol.8, p.82018, 2013.

C. Jackisch and M. Untch, Systemic therapy for women with ErbB2-positive breast cancer: new options, new challenges, Breast Care (Basel), vol.5, issue.s1, pp.1-2, 2010.

F. L. Baehner, N. Achacoso, T. Maddala, S. Shak, Q. Jr et al., Human epidermal growth factor receptor 2 assessment in a case-control study: comparison of fluorescence in situ hybridization and quantitative reverse transcription polymerase chain reaction performed by central laboratories, J Clin Oncol, vol.28, pp.4300-4306, 2010.

E. Perez, S. Butler, A. Dueck, F. Baehner, D. Cherbavaz et al., The relationship between quantitative HER2 gene expression by the 21-gene RT-PCR assay and adjuvant trastuzumab (H) benefit in NCCTG (Alliance) N9831, J Clin Oncol, vol.31, p.520, 2013.

D. J. Dabbs, M. E. Klein, S. K. Mohsin, R. R. Tubbs, Y. Shuai et al., High falsenegative rate of HER2 quantitative reverse transcription polymerase chain reaction of the Oncotype DX test: an independent quality assurance study, J Clin Oncol, vol.29, pp.4279-85, 2011.

R. A. Ach, A. Floore, B. Curry, V. Lazar, A. M. Glas et al., Robust interlaboratory reproducibility of a gene expression signature measurement consistent with the needs of a new generation of diagnostic tools, BMC Genomics, vol.8, p.148, 2007.

A. M. Glas, A. Floore, L. J. Delahaye, A. T. Witteveen, R. C. Pover et al., Converting a breast cancer microarray signature into a high-throughput diagnostic test, BMC Genomics, vol.7, p.278, 2006.

P. Roepman, H. M. Horlings, O. Krijgsman, M. Kok, J. M. Bueno-de-mesquita et al., Microarray-based determination of estrogen receptor, progesterone receptor, and HER2 receptor status in breast cancer, Clin Cancer Res, vol.15, pp.7003-7014, 2009.

A. K. Majidzadeh, R. Esmaeili, and N. Abdoli, TFRC and ACTB as the best reference genes to quantify Urokinase Plasminogen Activator in breast cancer, BMC Res Notes, vol.4, p.215, 2011.

J. S. Parker, M. Mullins, M. C. Cheang, S. Leung, D. Voduc et al., Supervised risk predictor of breast cancer based on intrinsic subtypes, J Clin Oncol, vol.27, pp.1160-1167, 2009.

T. O. Nielsen, J. S. Parker, S. Leung, D. Voduc, M. Ebbert et al., A comparison of PAM50 intrinsic subtyping with immunohistochemistry and clinical prognostic factors in tamoxifen-treated estrogen receptor-positive breast cancer, Clin Cancer Res, vol.16, pp.5222-5254, 2010.

M. Dowsett, I. Sestak, E. Lopez-knowles, K. Sidhu, A. K. Dunbier et al., Comparison of PAM50 risk of recurrence score with oncotype DX and IHC4 for predicting risk of distant recurrence after endocrine therapy, J Clin Oncol, vol.31, pp.2783-90, 2013.

M. Filipits, T. O. Nielsen, M. Rudas, R. Greil, H. Stoger et al., The PAM50 risk-of-recurrence score predicts risk for late distant recurrence after endocrine therapy in postmenopausal women with endocrine-responsive early breast cancer, Clin Cancer Res, vol.20, pp.1298-305, 2014.

M. Gnant, M. Filipits, R. Greil, H. Stoeger, M. Rudas et al., Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: using the PAM50 Risk of Recurrence score in 1478 postmenopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone, Ann Oncol, vol.25, pp.339-384, 2014.

G. K. Geiss, R. E. Bumgarner, B. Birditt, T. Dahl, N. Dowidar et al., Direct multiplexed measurement of gene expression with color-coded probe pairs, Nat Biotechnol, vol.26, pp.317-342, 2008.

P. P. Reis, L. Waldron, R. S. Goswami, W. Xu, Y. Xuan et al., mRNA transcript quantification in archival samples using multiplexed, color-coded probes, BMC Biotechnol, vol.11, p.46, 2011.

R. R. Bastien, A. Rodriguez-lescure, M. T. Ebbert, A. Prat, B. Munarriz et al., PAM50 breast cancer subtyping by RT-qPCR and concordance with standard clinical molecular markers, BMC Med Genomics, vol.5, p.44, 2012.

M. C. Cheang, K. D. Voduc, D. Tu, S. Jiang, S. Leung et al., Responsiveness of intrinsic subtypes to adjuvant anthracycline substitution in the NCIC.CTG MA.5 randomized trial, Clin Cancer Res, vol.18, pp.2402-2414, 2012.

P. Benohr, V. Henkel, R. Speer, U. Vogel, K. Sotlar et al., Her-2/neu expression in breast cancer -a comparison of different diagnostic methods, Anticancer Res, vol.25, issue.3B, pp.1895-900, 2005.

L. M. Gjerdrum, B. S. Sorensen, E. Kjeldsen, F. B. Sorensen, E. Nexo et al., Real-time quantitative PCR of microdissected paraffin-embedded breast carcinoma: an alternative method for HER-2/neu analysis, J Mol Diagn, vol.6, pp.42-51, 2004.

C. Christopherson, M. Chang, D. A. Eberhard, J. J. Sninsky, S. M. Anderson et al., Comparison of immunohistochemistry (IHC) and quantitative RT-PCR: ER, PR, and HER2 receptor status, J Clin Oncol, vol.30, p.47, 2012.

F. Elloumi, Z. Hu, Y. Li, J. S. Parker, M. L. Gulley et al., Systematic bias in genomic classification due to contaminating non-neoplastic tissue in breast tumor samples, BMC Med Genomics, vol.4, p.54, 2011.

M. Toi, J. Sperinde, W. Huang, S. Saji, J. Winslow et al., Differential survival following trastuzumab treatment based on quantitative HER2 expression and HER2 homodimers in a clinic-based cohort of patients with metastatic breast cancer, BMC Cancer, vol.10, p.56, 2010.

H. Cheng, Y. L. Bai, W. Sikov, N. Sinclair, V. Bossuyt et al., Quantitative measurements of HER2 and phospho-HER2 expression: correlation with pathologic response to neoadjuvant chemotherapy and trastuzumab, BMC Cancer, vol.14, p.326, 2014.

F. Montemurro, A. Prat, V. Rossi, G. Valabrega, J. Sperinde et al., Potential biomarkers of long-term benefit from single-agent trastuzumab or lapatinib in HER2-positive metastatic breast cancer, Mol Oncol, vol.8, pp.20-26, 2014.

R. Duchnowska, W. Biernat, B. Szostakiewicz, J. Sperinde, F. Piette et al., Correlation between quantitative HER-2 protein expression and risk for brain metastases in HER-2+ advanced breast cancer patients receiving trastuzumab-containing therapy, Oncologist, vol.17, pp.26-35, 2012.

A. Lipton, W. J. Kostler, K. Leitzel, S. M. Ali, J. Sperinde et al., Quantitative HER2 protein levels predict outcome in fluorescence in situ hybridization-positive patients with metastatic breast cancer treated with trastuzumab, Cancer, vol.116, pp.5168-78, 2010.

P. Nuciforo, S. Thyparambil, A. Garrido-castro, V. Peg, L. Prudkin et al., Correlation of high levels of HER2 measured by multiplex mass spectrometry with increased overall survival in patients treated with anti-HER2-based therapy, J Clin Oncol, vol.32, p.649, 2014.

J. Christiansen, N. Barakat, D. Murphy, D. Rimm, B. Dabbas et al., Her2 expression measured by AQUA analysis on BCIRG-005 and BCIRG-006 predicts the benefit of Herceptin therapy, Cancer Res, vol.72, pp.2-3, 2012.

S. Guiu, M. Gauthier, B. Coudert, F. Bonnetain, L. Favier et al., Pathological complete response and survival according to the level of HER-2 amplification after trastuzumab-based neoadjuvant therapy for breast cancer, Br J Cancer, vol.103, pp.1335-1377, 2010.

E. A. Perez, M. M. Reinholz, D. W. Hillman, K. S. Tenner, M. J. Schroeder et al., HER2 and chromosome 17 effect on patient outcome in the N9831 adjuvant trastuzumab trial, J Clin Oncol, vol.28, pp.4307-4322, 2010.

M. F. Press, R. S. Finn, D. Cameron, D. Leo, A. Geyer et al., HER-2 gene amplification, HER-2 and epidermal growth factor receptor mRNA and protein expression, and lapatinib efficacy in women with metastatic breast cancer, Clin Cancer Res, vol.14, pp.7861-70, 2008.

M. Bates, J. Sperinde, W. J. Kostler, S. M. Ali, K. Leitzel et al., Identification of a subpopulation of metastatic breast cancer patients with very high HER2 expression levels and possible resistance to trastuzumab, Ann Oncol, vol.22, pp.2014-2034, 2011.

H. Joensuu, J. Sperinde, M. Leinonen, W. Huang, J. Weidler et al., Very high quantitative tumor HER2 content and outcome in early breast cancer, Ann Oncol, vol.22, pp.2007-2020, 2011.

G. Gullo, D. Bettio, V. Torri, G. Masci, P. Salvini et al., Level of HER2/neu gene amplification as a predictive factor of response to trastuzumab-based therapy in patients with HER2-positive metastatic breast cancer, Invest New Drugs, vol.27, pp.179-83, 2009.

M. Dowsett, M. Procter, W. Mccaskill-stevens, E. De-azambuja, U. Dafni et al., Disease-free survival according to degree of HER2 amplification for patients treated with adjuvant chemotherapy with or without 1 year of trastuzumab: the HERA Trial, J Clin Oncol, vol.27, pp.2962-2971, 2009.

S. Paik, C. Kim, and N. Wolmark, HER2 status and benefit from adjuvant trastuzumab in breast cancer, N Engl J Med, vol.358, pp.1409-1420, 2008.

E. H. Romond, E. A. Perez, J. Bryant, V. J. Suman, C. E. Geyer et al., Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer, N Engl J Med, vol.353, pp.1673-84, 2005.

K. L. Pogue-geile, C. Kim, J. H. Jeong, N. Tanaka, H. Bandos et al., Predicting Degree of Benefit From Adjuvant Trastuzumab in NSABP Trial B-31, J Natl Cancer Inst, vol.105, pp.1782-1790, 2013.

M. Scaltriti, P. Nuciforo, I. Bradbury, J. Sperinde, D. Agbor-tarh et al., High HER2 expression correlates with response to the combination of lapatinib and trastuzumab, Clin Cancer Res, vol.21, pp.569-76, 2015.

M. Scaltriti, C. Verma, M. Guzman, J. Jimenez, J. L. Parra et al., Lapatinib, a HER2 tyrosine kinase inhibitor, induces stabilization and accumulation of HER2 and potentiates trastuzumab-dependent cell cytotoxicity, Oncogene, vol.28, pp.803-817, 2009.

N. Jura, Y. Shan, X. Cao, D. E. Shaw, and J. Kuriyan, Structural analysis of the catalytically inactive kinase domain of the human EGF receptor 3, Proc Natl Acad Sci, vol.106, pp.21608-21621, 2009.

K. Aertgeerts, R. Skene, J. Yano, B. C. Sang, H. Zou et al., Structural analysis of the mechanism of inhibition and allosteric activation of the kinase domain of HER2 protein, J Biol Chem, vol.286, pp.18756-65, 2011.

J. T. Garrett, M. G. Olivares, C. Rinehart, N. D. Granja-ingram, V. Sanchez et al., Transcriptional and posttranslational up-regulation of HER3 (ErbB3) compensates for inhibition of the HER2 tyrosine kinase, Proc Natl Acad Sci U S A, vol.108, pp.5021-5027, 2011.

H. Cheng, K. Ballman, M. Vassilakopoulou, A. C. Dueck, M. M. Reinholz et al., EGFR expression is associated with decreased benefit from trastuzumab in the NCCTG N9831 (Alliance) trial, Br J Cancer, vol.111, pp.1065-71, 2014.

E. A. Mittendorf, Y. Wu, M. Scaltriti, F. Meric-bernstam, K. K. Hunt et al., Loss of HER2 amplification following trastuzumab-based neoadjuvant systemic therapy and survival outcomes, Clin Cancer Res, vol.15, pp.7381-7389, 2009.

M. Piccart-gebhart, A. Holmes, J. Baselga, D. Azambuja, E. Dueck et al.,

, N063D) comparing one year of anti-HER2 therapy with lapatinib alone (L), trastuzumab alone (T), their sequence (T?L), or their combination (T+L) in the adjuvant treatment of HER2-positive early breast cancer (EBC), J Clin Oncol, vol.32, p.2, 2014.

L. Carey, E. Winer, G. Viale, D. Cameron, and L. Gianni, Triple-negative breast cancer: disease entity or title of convenience?, Nat Rev Clin Oncol, vol.7, pp.683-92, 2010.