A Multi-Center Clinical Study to Harvest and Characterize Circulating Tumor Cells from Patients with Metastatic Breast Cancer Using the Parsortix® PC1 System

Evan N Cohen, Gitanjali Jayachandran, Richard G Moore, Massimo Cristofanilli, Julie E Lang, Joseph D Khoury, Michael F Press, Kyu Kwang Kim, Negar Khazan, Qiang Zhang, Youbin Zhang, Pushpinder Kaur, Roberta Guzman, Michael C Miller, James M Reuben, Naoto T Ueno, Evan N Cohen, Gitanjali Jayachandran, Richard G Moore, Massimo Cristofanilli, Julie E Lang, Joseph D Khoury, Michael F Press, Kyu Kwang Kim, Negar Khazan, Qiang Zhang, Youbin Zhang, Pushpinder Kaur, Roberta Guzman, Michael C Miller, James M Reuben, Naoto T Ueno

Abstract

Circulating tumor cells (CTCs) captured from the blood of cancer patients may serve as a surrogate source of tumor material that can be obtained via a venipuncture (also known as a liquid biopsy) and used to better understand tumor characteristics. However, the only FDA-cleared CTC assay has been limited to the enumeration of surface marker-defined cells and not further characterization of the CTCs. In this study, we tested the ability of a semi-automated device capable of capturing and harvesting CTCs from peripheral blood based on cell size and deformability, agnostic of cell-surface markers (the Parsortix® PC1 System), to yield CTCs for evaluation by downstream techniques commonly available in clinical laboratories. The data generated from this study were used to support a De Novo request (DEN200062) for the classification of this device, which the FDA recently granted. As part of a multicenter clinical trial, peripheral blood samples from 216 patients with metastatic breast cancer (MBC) and 205 healthy volunteers were subjected to CTC enrichment. A board-certified pathologist enumerated the CTCs from each participant by cytologic evaluation of Wright-Giemsa-stained slides. As proof of principle, cells harvested from a concurrent parallel sample provided by each participant were evaluated using one of three additional evaluation techniques: molecular profiling by qRT-PCR, RNA sequencing, or cytogenetic analysis of HER2 amplification by FISH. The study demonstrated that the Parsortix® PC1 System can effectively capture and harvest CTCs from the peripheral blood of MBC patients and that the harvested cells can be evaluated using orthogonal methodologies such as gene expression and/or Fluorescence In Situ Hybridization (FISH).

Keywords: biomarkers; biopsy; blood; breast neoplasms/pathology; circulating; circulating tumor cells; circulating/pathology; liquid biopsy; neoplasms/diagnosis; neoplastic cells; tumor.

Conflict of interest statement

The authors ENC, GJ, KKK, NK, QZ, YZ, PK and RG declare no conflict of interest. RGM reports consulting fees from Fujirebio Diagnostics; MC reports AstraZeneca (Consulting Fees (e.g., advisory boards), Contracted Research) Celcuity (Consulting Fees (e.g., advisory boards)) Eli Lilly (Consulting Fees (e.g., advisory boards), Contracted Research, Fees for Non-CMEServices Received Directly from Commercial Interest or their Agents (e.g., speakers’ bureaus)) Ellipses (Consulting Fees (e.g., advisory boards)) Foundation Medicine (Fees for Non-CME Services Received Directly from Commercial Interest or their Agents (e.g., speakers’ bureaus)) Guardant (Fees for Non-CME Services Received Directly from Commercial Interest or their Agents (e.g., speakers’ bureaus)) Menarini (Consulting Fees (e.g., advisory boards)) Olaris (Consulting Fees (e.g., advisory boards)) Pfizer (Contracted Research, Fees for Non-CME Services Received Directly from Commercial Interest or their Agents (e.g., speakers’ bureaus)) Semonix (Consulting Fees (e.g., advisory boards)); JEL reports grant from ANGLE (Contracted Research); JDK reports Angle, plc (Consulting Fees (e.g., advisory boards), Contracted Research); MFP reports AstraZeneca (Consulting Fees (e.g., advisory boards)) Biocartis SA (Consulting Fees (e.g., advisory boards)) CEPHEID (Consulting Fees (e.g., advisory boards)) Eli Lilly & Company (Consulting Fees (e.g., advisory boards)) Lilly USA, LLC (Consulting Fees (e.g., advisory boards)) Merck & Co. (Consulting Fees (e.g., advisory boards)) Puma Biotechnology (Consulting Fees (e.g., advisory boards)) TORL BIOTHERAPEUTICS LLC (Ownership Interest (stocks, stock options, patent or other intellectual property or other ownership interest excluding diversified mutual funds)) Zymeworks Inc. (Consulting Fees (e.g., advisory boards)); MCM reports ANGLE plc (Full-time employee, Ownership Interest (stocks, stock options, patent or other intellectual property or other ownership interest excluding diversified mutual funds)); JMR reports Angle plc (Consulting Fees (e.g., advisory boards), Contracted Research); NTU reports Angle plc (Contracted Research) AnHeart Therapeutics Inc. (Consulting Fees (e.g., advisory boards)) AstraZeneca Pharmaceutical (Consulting Fees (e.g., advisory boards)) Carisma Therapeutics, Inc. (Consulting Fees (e.g., advisory boards), Contracted Research) CARNA Biosciences, Inc. (Consulting Fees (e.g., advisory boards)) ChemDiv, Inc. (Consulting Fees (e.g., advisory boards), Contracted Research) Chugai Pharmaceutical (Consulting Fees (e.g., advisory boards)) Daiichi Sankyo, Inc. (Consulting Fees (e.g., advisory boards), Contracted Research) DualityBio (Consulting Fees (e.g., advisory boards), Contracted Research) DynaMed (Consulting Fees (e.g., advisory boards)) Eisai Medical Research Inc (Consulting Fees (e.g., advisory boards), Contracted Research) Epic Science (Contracted Research) Gilead Sciences, Inc. (Consulting Fees (e.g., advisory boards), Contracted Research) Kechow Pharma (Consulting Fees (e.g., advisory boards)) Kirilys Therapeutics, Inc. (Consulting Fees (e.g., advisory boards)) Kyowa Hakko Kirin Co., Ltd. (Speaker) LARVOL (Consulting Fees (e.g., advisory boards)) Merck Co. (Contracted Research) OBI Pharma Inc. (Contracted Research) OncoCyte Co. (Consulting Fees (e.g., advisory boards)) Oncolys BioPharma Inc. (Contracted Research) Ourotech, Inc., DBA Pear Bio (Royalty) Peptilogics, Inc. (Consulting Fees (e.g., advisory boards)) Pfizer Inc. (Consulting Fees (e.g., advisory boards), Contracted Research) Phoenix Molecular Designs (Consulting Fees (e.g., advisory boards)) Preferred Medicine, Inc. (Consulting Fees (e.g., advisory boards), Contracted Research) Rakuten Medical, Inc. (Consulting Fees (e.g., advisory boards)) Sumitomo Dainippon Pharma, Inc. (Contracted Research) Sysmex Co. Ltd. (Consulting Fees (e.g., advisory boards)) Takeda Pharmaceuticals, Ltd. (Consulting Fees (e.g., advisory boards)) Unitech Medical, Inc. (Consulting Fees (e.g., advisory boards)). MCM, a full-time employee of ANGLE North America, Inc., a subsidiary of ANGLE plc, and was involved in the design of the study, the analysis and interpretation of the data, the decision to publish the results, and in the writing of the manuscript. The information and results reported in the manuscript, as well as their interpretations, are based on generally accepted scientific principals and methods and as such were not inappropriately influenced by the commercial interest of the funder (ANGLE Europe Limited).

Figures

Figure 1
Figure 1
Study enrollment, eligibility, and evaluation. HV, healthy volunteer; MBC, metastatic breast cancer; Cyto, cytology evaluation; qPCR, quantitative polymerase chain reaction; FISH, fluorescence in situ hybridization.
Figure 2
Figure 2
(a) Flow diagram for cytopathology evaluation in 207 eligible MBC patients. (b) Representative images of cells classified as CTCs (red arrows) from MBC patients that were harvested by the Parsortix PC1 system and deposited onto cytology slides by cytocentrifugation (images not to same scale) and Wright-Giemsa stained. (c) CTC numbers from the review of evaluable Wright-Giemsa-stained cytology slides. CTC, circulating tumor cell; HV, healthy volunteer; MBC, metastatic breast cancer. * p < 0.05, *** p < 0.001.
Figure 3
Figure 3
(a) Flow diagram for qPCR evaluation in eligible HV and MBC patients. (b) CTC count correlates with total CTC-related gene expression (40-Sum of KRT19, EPCAM, ERBB2, TWIST, and SNAI2 Ct values). Heat map and scatter plots of CTC gene expression show correlation of gene expression with the number of CTCs observed (c) Sum CTC related genes expression = 0.3701 ∗ CTC count + 6.771; R square = 0.3405, slope significantly different from zero (p < 0.0001). HV, healthy volunteer; MBC, metastatic breast cancer; qPCR, quantitative polymerase chain reaction. NS = p ≥0.05, * p < 0.05, ** p < 0.01, *** p < 0.001.
Figure 4
Figure 4
(a) Sum of CTC-related gene expression (40-Sum of EPCAM, KRT19, ERBB2, SNAI2, and TWIST Ct values) in HV, newly diagnosed MBC, and MBC with recurrence or progression. (b) ERBB2 expression in CTC-enriched samples by tissue HER2 status. (c) SNAI2 expression in CTC-enriched samples by tissue HER2 status. CTC, circulating tumor cell; HV, healthy volunteer; MBC, metastatic breast cancer.
Figure 5
Figure 5
(a) Flow diagram for RNA-seq evaluation in eligible HV and MBC patients. (b) Sum of TPM values for all genes differentially expressed (p < 0.001) between Parsortix PC1 harvests obtained from HV comparators and MBC patients. (c) Genes from the KEGG Cancer Pathway were differentially expressed between the HV and MBC harvests (p < 0.05) (d). Net TPM score of these 20 genes for HV and MBC samples. CTC, circulating tumor cell; HV, healthy volunteer; MBC, metastatic breast cancer.
Figure 6
Figure 6
(a) Flow diagram for HER2 FISH evaluation in eligible MBC patients. (b) Example images of HER2 FISH-stained CTCs from MBC patients that were harvested by the Parsortix PC1 system and deposited onto cytology slides (images not to scale). CTC, circulating tumor cell; HV, healthy volunteer; MBC, metastatic breast cancer; FISH, fluorescence in situ hybridization.

References

    1. Hong B., Zu Y. Detecting circulating tumor cells: Current challenges and new trends. Theranostics. 2013;3:377–394. doi: 10.7150/thno.5195.
    1. Cristofanilli M., Hayes D.F., Budd G.T., Ellis M.J., Stopeck A., Reuben J.M., Doyle G.V., Matera J., Allard W.J., Miller M.C., et al. Circulating tumor cells: A novel prognostic factor for newly diagnosed metastatic breast cancer. J. Clin. Oncol. 2005;23:1420–1430. doi: 10.1200/JCO.2005.08.140.
    1. Dawood S., Broglio K., Valero V., Reuben J., Handy B., Islam R., Jackson S., Hortobagyi G.N., Fritsche H., Cristofanilli M. Circulating tumor cells in metastatic breast cancer: From prognostic stratification to modification of the staging system? Cancer. 2008;113:2422–2430. doi: 10.1002/cncr.23852.
    1. Hayes D.F., Cristofanilli M., Budd G.T., Ellis M.J., Stopeck A., Miller M.C., Matera J., Allard W.J., Doyle G.V., Terstappen L.W. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin. Cancer Res. 2006;12:4218–4224. doi: 10.1158/1078-0432.CCR-05-2821.
    1. Budd G.T., Cristofanilli M., Ellis M.J., Stopeck A., Borden E., Miller M.C., Matera J., Repollet M., Doyle G.V., Terstappen L.W., et al. Circulating tumor cells versus imaging--predicting overall survival in metastatic breast cancer. Clin. Cancer Res. 2006;12:6403–6409. doi: 10.1158/1078-0432.CCR-05-1769.
    1. Liu M.C., Shields P.G., Warren R.D., Cohen P., Wilkinson M., Ottaviano Y.L., Rao S.B., Eng-Wong J., Seillier-Moiseiwitsch F., Noone A.M., et al. Circulating tumor cells: A useful predictor of treatment efficacy in metastatic breast cancer. J. Clin. Oncol. 2009;27:5153–5159. doi: 10.1200/JCO.2008.20.6664.
    1. Hayes D.F., Smerage J. Is there a role for circulating tumor cells in the management of breast cancer? Clin. Cancer Res. 2008;14:3646–3650. doi: 10.1158/1078-0432.CCR-07-4481.
    1. Pantel K., Alix-Panabieres C. Real-time liquid biopsy in cancer patients: Fact or fiction? Cancer Res. 2013;73:6384–6388. doi: 10.1158/0008-5472.CAN-13-2030.
    1. Alunni-Fabbroni M., Sandri M.T. Circulating tumour cells in clinical practice: Methods of detection and possible characterization. Methods. 2010;50:289–297. doi: 10.1016/j.ymeth.2010.01.027.
    1. Allard W.J., Matera J., Miller M.C., Repollet M., Connelly M.C., Rao C., Tibbe A.G., Uhr J.W., Terstappen L.W. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin. Cancer Res. 2004;10:6897–6904. doi: 10.1158/1078-0432.CCR-04-0378.
    1. Cristofanilli M., Budd G.T., Ellis M.J., Stopeck A., Matera J., Miller M.C., Reuben J.M., Doyle G.V., Allard W.J., Terstappen L.W., et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004;351:781–791. doi: 10.1056/NEJMoa040766.
    1. Zhang Z., Wuethrich A., Wang J., Korbie D., Lin L.L., Trau M. Dynamic Monitoring of EMT in CTCs as an Indicator of Cancer Metastasis. Anal. Chem. 2021;93:16787–16795. doi: 10.1021/acs.analchem.1c03167.
    1. Sankpal N.V., Fleming T.P., Sharma P.K., Wiedner H.J., Gillanders W.E. A double-negative feedback loop between EpCAM and ERK contributes to the regulation of epithelial-mesenchymal transition in cancer. Oncogene. 2017;36:3706–3717. doi: 10.1038/onc.2016.504.
    1. Yang J., Mani S.A., Donaher J.L., Ramaswamy S., Itzykson R.A., Come C., Savagner P., Gitelman I., Richardson A., Weinberg R.A. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117:927–939. doi: 10.1016/j.cell.2004.06.006.
    1. Mani S.A., Guo W., Liao M.J., Eaton E.N., Ayyanan A., Zhou A.Y., Brooks M., Reinhard F., Zhang C.C., Shipitsin M., et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–715. doi: 10.1016/j.cell.2008.03.027.
    1. Hollier B.G., Evans K., Mani S.A. The epithelial-to-mesenchymal transition and cancer stem cells: A coalition against cancer therapies. J. Mammary Gland. Biol. Neoplasia. 2009;14:29–43. doi: 10.1007/s10911-009-9110-3.
    1. Chudziak J., Burt D.J., Mohan S., Rothwell D.G., Mesquita B., Antonello J., Dalby S., Ayub M., Priest L., Carter L., et al. Clinical evaluation of a novel microfluidic device for epitope-independent enrichment of circulating tumour cells in patients with small cell lung cancer. Analyst. 2016;141:669–678. doi: 10.1039/C5AN02156A.
    1. Yap K., Cohen E.N., Reuben J.M., Khoury J.D. Circulating Tumor Cells: State-of-the-art Update on Technologies and Clinical Applications. Curr. Hematol. Malig. Rep. 2019;14:353–357. doi: 10.1007/s11899-019-00531-x.
    1. Hvichia G.E., Parveen Z., Wagner C., Janning M., Quidde J., Stein A., Muller V., Loges S., Neves R.P., Stoecklein N.H., et al. A novel microfluidic platform for size and deformability based separation and the subsequent molecular characterization of viable circulating tumor cells. Int. J. Cancer. 2016;138:2894–2904. doi: 10.1002/ijc.30007.
    1. Lampignano R., Yang L., Neumann M.H.D., Franken A., Fehm T., Niederacher D., Neubauer H. A Novel Workflow to Enrich and Isolate Patient-Matched EpCAM(high) and EpCAM(low/negative) CTCs Enables the Comparative Characterization of the PIK3CA Status in Metastatic Breast Cancer. Int. J. Mol. Sci. 2017;18:1885. doi: 10.3390/ijms18091885.
    1. Miller M.C., Robinson P.S., Wagner C., O’Shannessy D.J. The Parsortix Cell Separation System-A versatile liquid biopsy platform. Cytom. Part A. 2018;93:1234–1239. doi: 10.1002/cyto.a.23571.
    1. Gorges T.M., Kuske A., Rock K., Mauermann O., Muller V., Peine S., Verpoort K., Novosadova V., Kubista M., Riethdorf S., et al. Accession of Tumor Heterogeneity by Multiplex Transcriptome Profiling of Single Circulating Tumor Cells. Clin. Chem. 2016;62:1504–1515. doi: 10.1373/clinchem.2016.260299.
    1. Xu L., Mao X., Guo T., Chan P.Y., Shaw G., Hines J., Stankiewicz E., Wang Y., Oliver R.T.D., Ahmad A.S., et al. The Novel Association of Circulating Tumor Cells and Circulating Megakaryocytes with Prostate Cancer Prognosis. Clin. Cancer Res. 2017;23:5112–5122. doi: 10.1158/1078-0432.CCR-16-3081.
    1. Maertens Y., Humberg V., Erlmeier F., Steffens S., Steinestel J., Bogemann M., Schrader A.J., Bernemann C. Comparison of isolation platforms for detection of circulating renal cell carcinoma cells. Oncotarget. 2017;8:87710–87717. doi: 10.18632/oncotarget.21197.
    1. Obermayr E., Maritschnegg E., Agreiter C., Pecha N., Speiser P., Helmy-Bader S., Danzinger S., Krainer M., Singer C., Zeillinger R. Efficient leukocyte depletion by a novel microfluidic platform enables the molecular detection and characterization of circulating tumor cells. Oncotarget. 2018;9:812–823. doi: 10.18632/oncotarget.22549.
    1. Harris P.A., Taylor R., Thielke R., Payne J., Gonzalez N., Conde J.G. Research electronic data capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. J. Biomed. Inform. 2009;42:377–381. doi: 10.1016/j.jbi.2008.08.010.
    1. Harris P.A., Taylor R., Minor B.L., Elliott V., Fernandez M., O’Neal L., McLeod L., Delacqua G., Delacqua F., Kirby J., et al. The REDCap consortium: Building an international community of software platform partners. J. Biomed. Inform. 2019;95:103208. doi: 10.1016/j.jbi.2019.103208.
    1. Lang J.E., Ring A., Porras T., Kaur P., Forte V.A., Mineyev N., Tripathy D., Press M.F., Campo D. RNA-Seq of Circulating Tumor Cells in Stage II-III Breast Cancer. Ann. Surg. Oncol. 2018;25:2261–2270. doi: 10.1245/s10434-018-6540-4.
    1. Kaur P., Campo D., Porras T.B., Ring A., Lu J., Chairez Y., Su Y., Kang I., Lang J.E. A Pilot Study for the Feasibility of Exome-Sequencing in Circulating Tumor Cells Versus Single Metastatic Biopsies in Breast Cancer. Int. J. Mol. Sci. 2020;21:4826. doi: 10.3390/ijms21144826.
    1. Patro R., Duggal G., Love M.I., Irizarry R.A., Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods. 2017;14:417–419. doi: 10.1038/nmeth.4197.
    1. Press M.F., Villalobos I., Santiago A., Guzman R., Cervantes M., Gasparyan A., Campeau A., Ma Y., Tsao-Wei D.D., Groshen S. Assessing the New American Society of Clinical Oncology/College of American Pathologists Guidelines for HER2 Testing by Fluorescence In Situ Hybridization: Experience of an Academic Consultation Practice. Arch. Pathol. Lab. Med. 2016;140:1250–1258. doi: 10.5858/arpa.2016-0009-OA.
    1. Press M.F., Sauter G., Buyse M., Fourmanoir H., Quinaux E., Tsao-Wei D.D., Eiermann W., Robert N., Pienkowski T., Crown J., et al. HER2 Gene Amplification Testing by Fluorescent In Situ Hybridization (FISH): Comparison of the ASCO-College of American Pathologists Guidelines With FISH Scores Used for Enrollment in Breast Cancer International Research Group Clinical Trials. J. Clin. Oncol. 2016;34:3518–3528. doi: 10.1200/JCO.2016.66.6693.
    1. Press M.F., Seoane J.A., Curtis C., Quinaux E., Guzman R., Sauter G., Eiermann W., Mackey J.R., Robert N., Pienkowski T., et al. Assessment of ERBB2/HER2 Status in HER2-Equivocal Breast Cancers by FISH and 2013/2014 ASCO-CAP Guidelines. JAMA Oncol. 2019;5:366–375. doi: 10.1001/jamaoncol.2018.6012.
    1. Press M.F., Sauter G., Buyse M., Bernstein L., Guzman R., Santiago A., Villalobos I.E., Eiermann W., Pienkowski T., Martin M., et al. Alteration of topoisomerase II-alpha gene in human breast cancer: Association with responsiveness to anthracycline-based chemotherapy. J. Clin. Oncol. 2011;29:859–867. doi: 10.1200/JCO.2009.27.5644.
    1. Howlader N., Altekruse S.F., Li C.I., Chen V.W., Clarke C.A., Ries L.A., Cronin K.A. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J. Natl. Cancer Inst. 2014;106 doi: 10.1093/jnci/dju055.
    1. Bidard F.C., Peeters D.J., Fehm T., Nole F., Gisbert-Criado R., Mavroudis D., Grisanti S., Generali D., Garcia-Saenz J.A., Stebbing J., et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: A pooled analysis of individual patient data. Lancet Oncol. 2014;15:406–414. doi: 10.1016/S1470-2045(14)70069-5.
    1. Sun Y.F., Guo W., Xu Y., Shi Y.H., Gong Z.J., Ji Y., Du M., Zhang X., Hu B., Huang A., et al. Circulating Tumor Cells from Different Vascular Sites Exhibit Spatial Heterogeneity in Epithelial and Mesenchymal Composition and Distinct Clinical Significance in Hepatocellular Carcinoma. Clin. Cancer Res. 2018;24:547–559. doi: 10.1158/1078-0432.CCR-17-1063.
    1. Terai M., Mu Z., Eschelman D.J., Gonsalves C.F., Kageyama K., Chervoneva I., Orloff M., Weight R., Mastrangelo M.J., Cristofanilli M., et al. Arterial Blood, Rather Than Venous Blood, is a Better Source for Circulating Melanoma Cells. EBioMedicine. 2015;2:1821–1826. doi: 10.1016/j.ebiom.2015.09.019.
    1. Jesenko T., Modic Z., Kuhar C.G., Cemazar M., Matkovic U., Miceska S., Varl J., Kuhar A., Kloboves-Prevodnik V. Morphological features of breast cancer circulating tumor cells in blood after physical and biological type of isolation. Radiol. Oncol. 2021;55:292–304. doi: 10.2478/raon-2021-0033.
    1. Cohen E.N., Jayachandran G., Hardy M.R., Venkata Subramanian A.M., Meng X., Reuben J.M. Antigen-agnostic microfluidics-based circulating tumor cell enrichment and downstream molecular characterization. PLoS ONE. 2020;15:e0241123. doi: 10.1371/journal.pone.0241123.
    1. Ingthorsson S., Andersen K., Hilmarsdottir B., Maelandsmo G.M., Magnusson M.K., Gudjonsson T. HER2 induced EMT and tumorigenicity in breast epithelial progenitor cells is inhibited by coexpression of EGFR. Oncogene. 2016;35:4244–4255. doi: 10.1038/onc.2015.489.
    1. Gupta P., Srivastava S.K. HER2 mediated de novo production of TGFbeta leads to SNAIL driven epithelial-to-mesenchymal transition and metastasis of breast cancer. Mol. Oncol. 2014;8:1532–1547. doi: 10.1016/j.molonc.2014.06.006.
    1. Jenndahl L.E., Isakson P., Baeckstrom D. c-erbB2-induced epithelial-mesenchymal transition in mammary epithelial cells is suppressed by cell-cell contact and initiated prior to E-cadherin downregulation. Int. J. Oncol. 2005;27:439–448. doi: 10.3892/ijo.27.2.439.
    1. D’Souza B., Berdichevsky F., Kyprianou N., Taylor-Papadimitriou J. Collagen-induced morphogenesis and expression of the alpha 2-integrin subunit is inhibited in c-erbB2-transfected human mammary epithelial cells. Oncogene. 1993;8:1797–1806.
    1. D’Souza B., Taylor-Papadimitriou J. Overexpression of ERBB2 in human mammary epithelial cells signals inhibition of transcription of the E-cadherin gene. Proc. Natl. Acad. Sci. USA. 1994;91:7202–7206. doi: 10.1073/pnas.91.15.7202.
    1. Khoury H., Dankort D.L., Sadekova S., Naujokas M.A., Muller W.J., Park M. Distinct tyrosine autophosphorylation sites mediate induction of epithelial mesenchymal like transition by an activated ErbB-2/Neu receptor. Oncogene. 2001;20:788–799. doi: 10.1038/sj.onc.1204166.
    1. Giordano A., Gao H., Anfossi S., Cohen E., Mego M., Lee B.N., Tin S., De Laurentiis M., Parker C.A., Alvarez R.H., et al. Epithelial-mesenchymal transition and stem cell markers in patients with HER2-positive metastatic breast cancer. Mol. Cancer Ther. 2012;11:2526–2534. doi: 10.1158/1535-7163.MCT-12-0460.
    1. Lang J.E., Mosalpuria K., Cristofanilli M., Krishnamurthy S., Reuben J., Singh B., Bedrosian I., Meric-Bernstam F., Lucci A. HER2 status predicts the presence of circulating tumor cells in patients with operable breast cancer. Breast Cancer Res. Treat. 2009;113:501–507. doi: 10.1007/s10549-008-9951-2.

Source: PubMed

3
Tilaa