220
Views
9
CrossRef citations to date
0
Altmetric
Review

How may targeted proteomics complement genomic data in breast cancer?

, , , , , , & show all
Pages 43-54 | Received 04 Jul 2016, Accepted 01 Nov 2016, Published online: 15 Nov 2016

References

  • Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.
  • Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–752.
  • Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98:10869–10874.
  • Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A. 2003;100:8418–8423.
  • Kobold DC, Fulton RS, McLellan MD, et al. Comprehensive molecular portraits of human breast tumours. Nature. 2012; 490:61–70.
  • Marty M, Cognetti F, Maraninchi D, et al. Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: the M77001 study group. J Clin Oncol. 2005;23:4265–4274.
  • Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792.
  • Geyer CE, Forster J, Lindquist D, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733–2743.
  • Baselga J, Cortes J, Kim SB, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366:109–119.
  • Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783–1791.
  • Andre F, O’Regan R, Ozguroglu M, et al. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2014;15:580–591.
  • Emens LA. Breast cancer immunobiology driving immunotherapy: vaccines and immune checkpoint blockade. Expert Rev Anticancer Ther. 2012;12:1597–1611.
  • Gandhi L, Bahleda R, Tolaney SM, et al. Phase I study of neratinib in combination with temsirolimus in patients with human epidermal growth factor receptor 2-dependent and other solid tumors. J Clin Oncol. 2014;32:68–75.
  • Turner NC, Ro J, Andre F, et al. Palbociclib in hormone-receptor-positive advanced breast cancer. N Engl J Med. 2015;373:209–219.
  • Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–2465.
  • Bendell JC, Rodon J, Burris HA, et al. Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2012;30:282–290.
  • Cheang MC, Chia SK, Voduc D, et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst. 2009;101:736–750.
  • Goldhirsch A, Wood WC, Coates AS, et al. Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol. 2011;22:1736–1747.
  • Akbani R, Ng PK, Werner HM, et al. A pan-cancer proteomic perspective on the cancer Genome Atlas. Nat Commun. 2014;5:3887.
  • Lawrence RT, Perez EM, Hernandez D, et al. The proteomic landscape of triple-negative breast cancer. Cell Rep. 2015;11:630–644.
  • Moghaddas Gholami A, Hahne H, Wu Z, et al. Global proteome analysis of the NCI-60 cell line panel. Cell Rep. 2013;4:609–620.
  • Tyers M, Mann M. From genomics to proteomics. Nature. 2003;422:193–197.
  • Dittrich J, Becker S, Hecht M, et al. Sample preparation strategies for targeted proteomics via proteotypic peptides in human blood using liquid chromatography tandem mass spectrometry. Proteomics Clin Appl. 2015;9:5–16.
  • Gillette MA, Carr SA. Quantitative analysis of peptides and proteins in biomedicine by targeted mass spectrometry. Nat Methods. 2013;10:28–34.
  • Burnette WN. “Western blotting”: electrophoretic transfer of proteins from sodium dodecyl sulfate–polyacrylamide gels to unmodified nitrocellulose and radiographic detection with antibody and radioiodinated protein A. Anal Biochem. 1981;112:195–203.
  • Aebersold R, Burlingame AL. Bradshaw RA Western blots versus selected reaction monitoring assays: time to turn the tables? Mol Cell Proteomics. 2013;12:2381–2382.
  • Engvall E, Perlmann P. Enzyme-linked immunosorbent assay (ELISA). Quantitative assay of immunoglobulin G. Immunochemistry. 1971;8:871–874.
  • Duffy MJ. Urokinase plasminogen activator and its inhibitor, PAI-1, as prognostic markers in breast cancer: from pilot to level 1 evidence studies. Clin Chem. 2002;48:1194–1197.
  • Look MP, van Putten WL, Duffy MJ, et al. Pooled analysis of prognostic impact of urokinase-type plasminogen activator and its inhibitor PAI-1 in 8377 breast cancer patients. J Natl Cancer Inst. 2002;94:116–128.
  • Harbeck N, Schmitt M, Meisner C, et al. Ten-year analysis of the prospective multicentre Chemo-N0 trial validates American Society of Clinical Oncology (ASCO)-recommended biomarkers uPA and PAI-1 for therapy decision making in node-negative breast cancer patients. Eur J Cancer. 2013;49:1825–1835.
  • Lang DS, Heilenkotter U, Schumm W, et al. Optimized immunohistochemistry in combination with image analysis: a reliable alternative to quantitative ELISA determination of uPA and PAI-1 for routine risk group discrimination in breast cancer. Breast. 2013;22:736–743.
  • Matos LL, Trufelli DC, de Matos MG, et al. Immunohistochemistry as an important tool in biomarkers detection and clinical practice. Biomark Insights. 2010;5:9–20.
  • Allred DC. Should immunohistochemical examination replace biochemical hormone receptor assays in breast cancer? Am J Clin Pathol. 1993;99:1–3.
  • Mohsin SK, Weiss H, Havighurst T, et al. Progesterone receptor by immunohistochemistry and clinical outcome in breast cancer: a validation study. Mod Pathol. 2004;17:1545–1554.
  • Rhodes A, Jasani B, Balaton AJ, et al. Frequency of oestrogen and progesterone receptor positivity by immunohistochemical analysis in 7016 breast carcinomas: correlation with patient age, assay sensitivity, threshold value, and mammographic screening. J Clin Pathol. 2000;53:688–696.
  • Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–712.
  • Perez EA, Suman VJ, Davidson NE, 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. 2006;24:3032–3038.
  • Wolff AC, Hammond ME, Schwartz JN, 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. 2007;25:118–145.
  • Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a combined estrogen receptor, progesterone receptor, Ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and comparison with the Genomic Health recurrence score in early breast cancer. J Clin Oncol. 2011;29:4273–4278.
  • Paweletz CP, Charboneau L, Bichsel VE, et al. Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front. Oncogene. 2001;20:1981–1989.
  • Charboneau L, Tory H, Chen T, et al. Utility of reverse phase protein arrays: applications to signalling pathways and human body arrays. Brief Funct Genomic Proteomic. 2002;1:305–315.
  • Akbani R, Becker KF, Carragher N, et al. Realizing the promise of reverse phase protein arrays for clinical, translational, and basic research: a workshop report: the RPPA (Reverse Phase Protein Array) society. Mol Cell Proteomics. 2014;13:1625–1643.
  • Boellner S, Becker KF. Reverse phase protein arrays-quantitative assessment of multiple biomarkers in biopsies for clinical use. Microarrays (Basel). 2015;4:98–114.
  • Becker KF, Schott C, Hipp S, et al. Quantitative protein analysis from formalin-fixed tissues: implications for translational clinical research and nanoscale molecular diagnosis. J Pathol. 2007;211:370–378.
  • Berg D, Langer R, Tran K, et al. Protein microarray-based comparison of HER2, estrogen receptor, and progesterone receptor status in core biopsies and surgical specimens from FFPE breast cancer tissues. Appl Immunohistochem Mol Morphol. 2011;19:300–305.
  • Berg D, Wolff C, Malinowsky K, et al. Profiling signalling pathways in formalin-fixed and paraffin-embedded breast cancer tissues reveals cross-talk between EGFR, HER2, HER3 and uPAR. J Cell Physiol. 2012;227:204–212.
  • Cicenas J, Urban P, Kung W, et al. Phosphorylation of tyrosine 1248-ERBB2 measured by chemiluminescence-linked immunoassay is an independent predictor of poor prognosis in primary breast cancer patients. Eur J Cancer. 2006;42:636–645.
  • Frogne T, Laenkholm AV, Lyng MB, et al. Determination of HER2 phosphorylation at tyrosine 1221/1222 improves prediction of poor survival for breast cancer patients with hormone receptor-positive tumors. Breast Cancer Res. 2009;11:R11.
  • Wulfkuhle JD, Berg D, Wolff C, et al. Molecular analysis of HER2 signaling in human breast cancer by functional protein pathway activation mapping. Clin Cancer Res. 2012;18:6426–6435.
  • Malinowsky K, Raychaudhuri M, Buchner T, et al. Common protein biomarkers assessed by reverse phase protein arrays show considerable intratumoral heterogeneity in breast cancer tissues. PLoS One. 2012;7:e40285.
  • Hennessy BT, Lu Y, Gonzalez-Angulo AM, et al. A Technical assessment of the utility of reverse phase protein arrays for the study of the functional proteome in non-microdissected human breast cancers. Clin Proteomics. 2010;6:129–151.
  • Hennessy BT, Gonzalez-Angulo AM, Stemke-Hale K, et al. Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res. 2009;69:4116–4124.
  • Knuefermann C, Lu Y, Liu B, et al. HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells. Oncogene. 2003;22:3205–3212.
  • Mondesire WH, Jian W, Zhang H, et al. Targeting mammalian target of rapamycin synergistically enhances chemotherapy-induced cytotoxicity in breast cancer cells. Clin Cancer Res. 2004;10:7031–7042.
  • Stassi G, Garofalo M, Zerilli M, et al. PED mediates AKT-dependent chemoresistance in human breast cancer cells. Cancer Res. 2005;65:6668–6675.
  • Aichler M, Walch A. MALDI Imaging mass spectrometry: current frontiers and perspectives in pathology research and practice. Lab Invest. 2015;95:422–431.
  • Caprioli RM, Farmer TB, Gile J. Molecular imaging of biological samples: localization of peptides and proteins using MALDI-TOF MS. Anal Chem. 1997;69:4751–4760.
  • Stoeckli M, Chaurand P, Hallahan DE, et al. Imaging mass spectrometry: a new technology for the analysis of protein expression in mammalian tissues. Nat Med. 2001;7:493–496.
  • Gemoll T, Roblick UJ, Habermann JK. MALDI mass spectrometry imaging in oncology (Review). Mol Med Rep. 2011;4:1045–1051.
  • Pineau C. MALDI imaging mass spectrometry: applications, limitations and potential. Farm Animal Proteomics. 2013;34–35.
  • Rauser S, Marquardt C, Balluff B, et al. Classification of HER2 receptor status in breast cancer tissues by MALDI imaging mass spectrometry. J Proteome Res. 2010;9:1854–1863.
  • Reyzer ML, Caldwell RL, Dugger TC, et al. Early changes in protein expression detected by mass spectrometry predict tumor response to molecular therapeutics. Cancer Res. 2004;64:9093–9100.
  • Winkelmann I, Walch A, Grüner BM, et al. FAST-SRM: a new single reaction monitoring method for fast, targeted drug distribution testing in tissue sections. Bruker Daltonics application note MT-103. 2010.
  • Domon B, Aebersold R. Options and considerations when selecting a quantitative proteomics strategy. Nat Biotechnol. 2010;28:710–721.
  • Deutsch EW. Lam H, Aebersold R. PeptideAtlas: a resource for target selection for emerging targeted proteomics workflows. EMBO Rep. 2008;9:429–434.
  • Addona TA, Abbatiello SE, Schilling B, et al. Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma. Nat Biotechnol. 2009;27:633–641.
  • Muraoka S, Kume H, Watanabe S, et al. Strategy for SRM-based verification of biomarker candidates discovered by iTRAQ method in limited breast cancer tissue samples. J Proteome Res. 2012;11:4201–4210.
  • Liu NQ, Dekker LJ, Stingl C, et al. Quantitative proteomic analysis of microdissected breast cancer tissues: comparison of label-free and SILAC-based quantification with shotgun, directed, and targeted MS approaches. J Proteome Res. 2013;12:4627–4641.
  • Ong SE, Blagoev B, Kratchmarova I, et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics. 2002;1:376–386.
  • Hanke S, Besir H, Oesterhelt D, et al. Absolute SILAC for accurate quantitation of proteins in complex mixtures down to the attomole level. J Proteome Res. 2008;7:1118–1130.
  • Schoenherr RM, Whiteaker JR, Zhao L, et al. Multiplexed quantification of estrogen receptor and HER2/Neu in tissue and cell lysates by peptide immunoaffinity enrichment mass spectrometry. Proteomics. 2012;12:1253–1260.
  • Sjostrom M, Ossola R, Breslin T, et al. A Combined Shotgun and Targeted Mass Spectrometry Strategy for Breast Cancer Biomarker Discovery. J Proteome Res. 2015;14(7):2807-2818.
  • Steiner C, Tille JC, Lamerz J, et al. Quantification of HER2 by targeted mass spectrometry in Formalin-Fixed Paraffin-Embedded (FFPE) breast cancer tissues. Mol Cell Proteomics. 2015;14:2786–2799.
  • Nuciforo P, Thyparambil S, Aura C, et al. High HER2 protein levels correlate with increased survival in breast cancer patients treated with anti-HER2 therapy. Mol Oncol. 2016;10:138–147.
  • Trinidad JC, Specht CG, Thalhammer A, et al. Comprehensive identification of phosphorylation sites in postsynaptic density preparations. Mol Cell Proteomics. 2006;5:914–922.
  • Munton RP, Tweedie-Cullen R, Livingstone-Zatchej M, et al. Qualitative and quantitative analyses of protein phosphorylation in naive and stimulated mouse synaptosomal preparations. Mol Cell Proteomics. 2007;6:283–293.
  • Steen H, Jebanathirajah JA, Rush J, et al. Phosphorylation analysis by mass spectrometry: myths, facts, and the consequences for qualitative and quantitative measurements. Mol Cell Proteomics. 2006;5:172–181.
  • Picotti P, Aebersold R. Selected reaction monitoring-based proteomics: workflows, potential, pitfalls and future directions. Nat Methods. 2012;9:555–566.
  • Domanski D, Murphy LC, Borchers CH. Assay development for the determination of phosphorylation stoichiometry using multiple reaction monitoring methods with and without phosphatase treatment: application to breast cancer signaling pathways. Anal Chem. 2010;82:5610–5620.
  • Britton DJ, Scott GK, Schilling B, et al. A novel serine phosphorylation site detected in the N-terminal domain of estrogen receptor isolated from human breast cancer cells. J Am Soc Mass Spectrom. 2008;19:729–740.
  • Lesur A, Ancheva L, Kim YJ, et al. Screening protein isoforms predictive for cancer using immunoaffinity capture and fast LC-MS in PRM mode. Proteomics Clin Appl. 2015;9:695–705.
  • Law KP, Lim YP. Recent advances in mass spectrometry: data independent analysis and hyper reaction monitoring. Expert Rev Proteomics. 2013;10:551–566.
  • Schubert OT, Gillet LC, Collins BC, et al. Building high-quality assay libraries for targeted analysis of SWATH MS data. Nat Protoc. 2015;10:426–441.
  • Schilling B, Rardin MJ, MacLean BX, et al. Platform-independent and label-free quantitation of proteomic data using MS1 extracted ion chromatograms in skyline: application to protein acetylation and phosphorylation. Mol Cell Proteomics. 2012;11:202–214.
  • Held JM, Schilling B, D’Souza AK, et al. Label-free quantitation and mapping of the ErbB2 tumor receptor by multiple protease digestion with data-dependent (MS1) and data-independent (MS2) acquisitions. Int J Proteomics. 2013;2013:1–11.
  • Doerr A. Mass spectrometry-based targeted proteomics. Nat Methods. 2013;10:23.
  • Gallien S, Duriez E, Crone C, et al. Targeted proteomic quantification on quadrupole-orbitrap mass spectrometer. Mol Cell Proteomics. 2012;11:1709–1723.
  • Peterson AC, Russell JD, Bailey DJ, et al. Parallel reaction monitoring for high resolution and high mass accuracy quantitative, targeted proteomics. Mol Cell Proteomics. 2012;11:1475–1488.
  • Gallien S, Domon B. Detection and quantification of proteins in clinical samples using high resolution mass spectrometry. Methods. 2015;81:15–23.
  • Andre F, Bachelot T, Commo F, et al. Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol. 2014;15:267–274.
  • S Le Tourneau C, Delord JP, Goncalves A, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–1334.
  • Hembrough T, Thyparambil S, Liao WL, et al. Application of selected reaction monitoring for multiplex quantification of clinically validated biomarkers in formalin-fixed, paraffin-embedded tumor tissue. J Mol Diagn. 2013;15:454–465.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.