A novel method for downstream characterization of breast cancer circulating tumor cells following CellSearch isolation

Henrik Frithiof, Charlotte Welinder, Anna-Maria Larsson, Lisa Rydén, Kristina Aaltonen, Henrik Frithiof, Charlotte Welinder, Anna-Maria Larsson, Lisa Rydén, Kristina Aaltonen

Abstract

Background: Enumeration of circulating tumor cells (CTCs) obtained from minimally invasive blood samples has been well established as a valuable monitoring tool in metastatic and early breast cancer, as well as in several other cancer types. The gold standard technology for detecting CTCs in blood against a backdrop of millions of leukocytes is the FDA-approved CellSearch system (Janssen Diagnostics), which relies on EpCAM-based immunomagnetic separation. Secondary characterization of these cells could enable treatment selection based on specific targets in these cells, as well as providing a real time window into the metastatic process and offering unique insights into tumor heterogeneity. The objective of this study was to develop a method for downstream characterization of CTCs following isolation with the CellSearch system.

Methods: An in vitro CTC model system focusing on clinically useful treatment predictive biomarkers in breast cancer, specifically the estrogen receptor α (ERα) and the human epidermal growth factor receptor 2 (HER2), was established using healthy donor blood spiked with breast cancer cell lines MCF7 (ERα(+)/HER2(-)) and SKBr3 (ERα(-)/HER2(+)). Following CTC isolation by CellSearch, the captured CTCs were further enriched and fixed on a microscope slide using the in-house-developed CTC-DropMount technique.

Results: The recovery rate of CTCs after CellSearch Profile analysis and CTC-DropMount was 87%. A selective and consistent triple-immunostaining protocol was optimized. Cells positive for DAPI, cytokeratin (CK) 8, 18 and 19, but negative for the leukocyte-specific marker CD45, were classified as CTCs and subsequently analyzed for ERα and HER2 expression. The method was verified in breast cancer patient samples, thus demonstrating its clinical relevance.

Conclusions: Our results show that it is possible to ascertain the status of important predictive biomarkers expressed in breast cancer CTCs using the newly developed CTC-DropMount technique. Downstream characterization of multiple biomarkers using a standard fluorescence microscope demonstrates that important clinical and biological information may be obtained from a single patient blood sample following either CellSearch epithelial or profile analyses.

Trial registration: Clinical Trials NCT01322893.

Figures

Figure 1
Figure 1
Overview of the method. Enriched CTCs were collected after CellSearch analysis using the Profile or Epithelial cell kit. The solution containing CTCs and leukocytes was placed in a magnetic tray. Following incubation, the non-adherent solvent was removed and the ferrofluid-attached cells were re-suspended in a smaller volume of PBS, thus permitting further enrichment. This solution was dropped and fixed on a glass slide before subsequent staining according to protocols 1 or 2. Visualization is possible with a fluorescence or bright-field microscope, depending on the staining method applied. (Drawing of magnetic stand reprinted with permission from IFI CLAIMS Patent Services.)
Figure 2
Figure 2
CD45 staining. Secondary staining of cell line cells spiked into healthy donor blood, from left to right: DAPI counterstain (fluorescent blue), cytokeratins 8, 18, and 19 (CK) stained with Phycoerythrin (red), CD45 stained with AlexaFluor 647 (yellow), and a composite of all channels. The two juxtaposed CTCs (CK-positive) stained negative for CD45, while the leukocytes (white arrows) simultaneously stained positive for CD45 and negative for CK, illustrating methodological selectivity.
Figure 3
Figure 3
ERα staining of MCF7 and SKBr3 cells. Selective ERα staining demonstrated in MCF7 (ERα+) and SKBr3 (ERα−) cells. From left to right: DAPI counterstain (fluorescent blue), cytokeratins 8, 18, and 19 (CK) stained with Phycoerythrin (red), estrogen receptor (ERα) stained with AlexaFluor 488 (green), and a composite of all channels. MCF7 showed positive nuclear staining in AlexaFluor 488 indicating positive ERα expression, while SKBr3 was negative.
Figure 4
Figure 4
HER2-staining of MCF7 and SKBr3. Selective HER2 staining demonstrated in MCF7 (HER2−) and SKBr3 (HER2+) cells. First row: MCF7, from left to right: DAPI counterstain (fluorescent blue), HER2 stained with Liquid Permanent Red (red), and a composite of all channels. Second row: SKBr3, in the corresponding channels. Positive membrane staining was visible in SKBr3 cells only. Additionally, assessment of HER2 staining was also possible using bright-field microscopy, as demonstrated in the lower two rows (third row: MCF7, and fourth row: SKBr3).
Figure 5
Figure 5
Immunostaining of metastatic breast cancer patient blood samples. Representative images of positive ERα and HER2 staining in clinical samples. Row A, from left to right: DAPI counterstain (fluorescent blue), cytokeratins 8, 18, and 19 (CK) stained with Phycoerythrin (red), estrogen receptor (ERα) stained with AlexaFluor 488 (green), and a composite of all channels. This patient sample (no. 4, see Table 3) was collected prior to initiation of therapy, illustrating two clustered ERα+ CTCs, adjacent to a solitary leukocyte located in the lower left corner. This patient was diagnosed with an ERα+ metastasis. Row B, from left to right: DAPI counterstain (fluorescent blue), HER2 stained with Liquid Permanent Red (red). This patient sample (no. 1, see Table 3) was obtained following 6 months of chemotherapy, illustrating HER2+ CTCs identified by combination of fluorescence and bright-field microscopy. This patient was diagnosed with a HER2− primary tumor and HER2− metastasis.

References

    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. Clin Cancer Res. 2008;14(19):6302–9. doi: 10.1158/1078-0432.CCR-08-0872.
    1. Cohen SJ, Punt CJA, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, et al. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann Oncol. 2009;20(7):1223–9. doi: 10.1093/annonc/mdn786.
    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. N Engl J Med. 2004;351(8):781–91. doi: 10.1056/NEJMoa040766.
    1. Bidard F-C, Peeters DJ, Fehm T, Nolé F, Gisbert-Criado R, Mavroudis D, 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(4):406–14. doi: 10.1016/S1470-2045(14)70069-5.
    1. Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013;24(9):2206–23. doi: 10.1093/annonc/mdt303.
    1. Nadji M, Gomez-Fernandez C, Ganjei-Azar P, Morales AR. Immunohistochemistry of estrogen and progesterone receptors reconsidered: experience with 5,993 breast cancers. Am J Clin Pathol. 2005;123(1):21–7. doi: 10.1309/4WV79N2GHJ3X1841.
    1. Davies C, Godwin J, Gray R, Clarke M, Cutter D, Darby S, et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet. 2011;378(9793):771–84. doi: 10.1016/S0140-6736(11)60993-8.
    1. Moy I, Lin Z, Rademaker AW, Reierstad S, Khan SA, Bulun SE. Expression of estrogen-related gene markers in breast cancer tissue predicts aromatase inhibitor responsiveness. PLoS One. 2013;8(11) doi: 10.1371/journal.pone.0077543.
    1. Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer. Annu Rev Med. 2011;62:233–47. doi: 10.1146/annurev-med-070909-182917.
    1. Lower EE, Glass EL, Bradley DA, Blau R, Heffelfinger S. Impact of metastatic estrogen receptor and progesterone receptor status on survival. Breast Cancer Res Treat. 2005;90(1):65–70. doi: 10.1007/s10549-004-2756-z.
    1. Aitken SJ, Thomas JS, Langdon SP, Harrison DJ, Faratian D. Quantitative analysis of changes in ER, PR and HER2 expression in primary breast cancer and paired nodal metastases. Ann Oncol. 2010;21(6):1254–61. doi: 10.1093/annonc/mdp427.
    1. Gomez-Fernandez C, Daneshbod Y, Nassiri M, Milikowski C, Alvarez C, Nadji M. Immunohistochemically determined estrogen receptor phenotype remains stable in recurrent and metastatic breast cancer. Am J Clin Pathol. 2008;130(6):879–82. doi: 10.1309/AJCPD1AO3YSYQYNW.
    1. Iguchi C, Nio Y, Itakura M. Heterogeneic expression of estrogen receptor between the primary tumor and the corresponding involved lymph nodes in patients with node-positive breast cancer and its implications in patient outcome. J Surg Oncol. 2003;83(2):85–93. doi: 10.1002/jso.10243.
    1. Simmons C, Miller N, Geddie W, Gianfelice D, Oldfield M, Dranitsaris G, et al. Does confirmatory tumor biopsy alter the management of breast cancer patients with distant metastases? Ann Oncol. 2009;20(9):1499–504. doi: 10.1093/annonc/mdp028.
    1. Fehm T, Hoffmann O, Aktas B, Becker S, Solomayer EF, Wallwiener D, et al. Detection and characterization of circulating tumor cells in blood of primary breast cancer patients by RT-PCR and comparison to status of bone marrow disseminated cells. Breast Cancer Res. 2009;11(4):R59. doi: 10.1186/bcr2349.
    1. Aktas B, Muller V, Tewes M, Zeitz J, Kasimir-Bauer S, Loehberg CR, et al. Comparison of estrogen and progesterone receptor status of circulating tumor cells and the primary tumor in metastatic breast cancer patients. Gynecol Oncol. 2011;122(2):356–60. doi: 10.1016/j.ygyno.2011.04.039.
    1. Bock C, Rack B, Kuhn C, Hofmann S, Finkenzeller C, Jager B, et al. Heterogeneity of ERalpha and ErbB2 status in cell lines and circulating tumor cells of metastatic breast cancer patients. Transl Oncol. 2012;5(6):475–85. doi: 10.1593/tlo.12310.
    1. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244(4905):707–12. doi: 10.1126/science.2470152.
    1. Sjogren S, Inganas M, Lindgren A, Holmberg L, Bergh J. Prognostic and predictive value of c-erbB-2 overexpression in primary breast cancer, alone and in combination with other prognostic markers. J Clin Oncol. 1998;16(2):462–9.
    1. Baselga J. Herceptin alone or in combination with chemotherapy in the treatment of HER2-positive metastatic breast cancer: pivotal trials. Oncology. 2001;61(Suppl. 2):14–21. doi: 10.1159/000055397.
    1. Davoli A, Hocevar BA, Brown TL. Progression and treatment of HER2-positive breast cancer. Cancer Chemother Pharmacol. 2010;65(4):611–23. doi: 10.1007/s00280-009-1208-1.
    1. Lindstrom LS, Karlsson E, Wilking UM, Johansson U, Hartman J, Lidbrink EK, et al. Clinically used breast cancer markers such as estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are unstable throughout tumor progression. J Clin Oncol. 2012;30(21):2601–8. doi: 10.1200/JCO.2011.37.2482.
    1. Zidan J, Dashkovsky I, Stayerman C, Basher W, Cozacov C, Hadary A. Comparison of HER-2 overexpression in primary breast cancer and metastatic sites and its effect on biological targeting therapy of metastatic disease. Br J Cancer. 2005;93(5):552–6. doi: 10.1038/sj.bjc.6602738.
    1. Gancberg D, Di Leo A, Cardoso F, Rouas G, Pedrocchi M, Paesmans M, et al. Comparison of HER-2 status between primary breast cancer and corresponding distant metastatic sites. Ann Oncol. 2002;13(7):1036–43. doi: 10.1093/annonc/mdf252.
    1. Regitnig P, Schippinger W, Lindbauer M, Samonigg H, Lax SF. Change of HER-2/neu status in a subset of distant metastases from breast carcinomas. J Pathol. 2004;203(4):918–26. doi: 10.1002/path.1592.
    1. Meng S, Tripathy D, Shete S, Ashfaq R, Saboorian H, Haley B, et al. uPAR and HER-2 gene status in individual breast cancer cells from blood and tissues. Proc Natl Acad Sci U S A. 2006;103(46):17361–5. doi: 10.1073/pnas.0608113103.
    1. Pestrin M, Bessi S, Galardi F, Truglia M, Biggeri A, Biagioni C, et al. Correlation of HER2 status between primary tumors and corresponding circulating tumor cells in advanced breast cancer patients. Breast Cancer Res Treat. 2009;118(3):523–30. doi: 10.1007/s10549-009-0461-7.
    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 Res Treat. 2010;124(2):403–12. doi: 10.1007/s10549-010-1163-x.
    1. Liu Y, Liu Q, Wang T, Bian L, Zhang S, Hu H, et al. Circulating tumor cells in HER2-positive metastatic breast cancer patients: a valuable prognostic and predictive biomarker. BMC Cancer. 2013;13:202. doi: 10.1186/1471-2407-13-202.
    1. Meng S, Tripathy D, Shete S, Ashfaq R, Haley B, Perkins S, et al. HER-2 gene amplification can be acquired as breast cancer progresses. Proc Natl Acad Sci U S A. 2004;101(25):9393–8. doi: 10.1073/pnas.0402993101.
    1. Bidard FC, Fehm T, Ignatiadis M, Smerage JB, Alix-Panabieres C, Janni W, et al. Clinical application of circulating tumor cells in breast cancer: overview of the current interventional trials. Cancer Metastasis Rev. 2013;32(1–2):179–88. doi: 10.1007/s10555-012-9398-0.
    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. Phys Biol. 2012;9(1):016003. doi: 10.1088/1478-3975/9/1/016003.
    1. Babayan A, Hannemann J, Spotter J, Muller V, Pantel K, Joosse SA. Heterogeneity of estrogen receptor expression in circulating tumor cells from metastatic breast cancer patients. PLoS One. 2013;8(9) doi: 10.1371/journal.pone.0075038.
    1. Speel EJ, Schutte B, Wiegant J, Ramaekers FC, Hopman AH. A novel fluorescence detection method for in situ hybridization, based on the alkaline phosphatase-fast red reaction. J Histochem Cytochem. 1992;40(9):1299–308. doi: 10.1177/40.9.1506667.
    1. Van der Auwera I, Peeters D, Benoy IH, Elst HJ, Van Laere SJ, Prove A, et al. Circulating tumour cell detection: a direct comparison between the cell search system, the AdnaTest and CK-19/mammaglobin RT-PCR in patients with metastatic breast cancer. Br J Cancer. 2010;102(2):276–84. doi: 10.1038/sj.bjc.6605472.
    1. Lowes LE, Allan AL. Recent advances in the molecular characterization of circulating tumor cells. Cancers (Basel) 2014;6(1):595–624. doi: 10.3390/cancers6010595.
    1. Swennenhuis JF, Tibbe AGJ, Levink R, Sipkema RCJ, Terstappen LWMM. Characterization of circulating tumor cells by fluorescence in situ hybridization. Cytometry Part A. 2009;75A(6):520–7. doi: 10.1002/cyto.a.20718.
    1. Ignatiadis M, Rothe F, Chaboteaux C, Durbecq V, Rouas G, Criscitiello C, et al. HER2-positive circulating tumor cells in breast cancer. PLoS One. 2011;6(1) doi: 10.1371/journal.pone.0015624.
    1. Riethdorf S, Muller V, Zhang L, Rau T, Loibl S, Komor M, et al. Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin Cancer Res. 2010;16(9):2634–45. doi: 10.1158/1078-0432.CCR-09-2042.
    1. Paoletti C, Muniz MC, Thomas DG, Griffith KA, Kidwell KM, Tokudome N et al. Development of circulating tumor cell-endocrine therapy index in patients with hormone receptor positive breast cancer. Clin Cancer Res. 2014. Nov 7, e pub ahead of print doi: 10.1158/1078-0432.CCR-14-2781.
    1. Heitzer E, Auer M, Gasch C, Pichler M, Ulz P, Hoffmann EM, et al. Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing. Cancer Res. 2013;7:7.
    1. Punnoose EA, Atwal SK, Spoerke JM, Savage H, Pandita A, Yeh RF, et al. Molecular biomarker analyses using circulating tumor cells. PLoS One. 2010;5(9) doi: 10.1371/journal.pone.0012517.
    1. Yusa A, Toneri M, Masuda T, Ito S, Yamamoto S, Okochi M, et al. Development of a new rapid isolation device for circulating tumor cells (CTCs) using 3D palladium filter and its application for genetic analysis. PLoS One. 2014;9(2) doi: 10.1371/journal.pone.0088821.
    1. Riethdorf S, Fritsche H, Müller V, Rau T, Schindlbeck C, Rack B, et al. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the cell search system. Clin Cancer Res. 2007;13(3):920–8. doi: 10.1158/1078-0432.CCR-06-1695.
    1. Murlidhar V, Zeinali M, Grabauskiene S, Ghannad-Rezaie M, Wicha MS, Simeone DM et al. A radial flow microfluidic device for ultra-high-throughput affinity-based isolation of circulating tumor cells. Small. 2014.
    1. Lee A, Park J, Lim M, Sunkara V, Kim SY, Kim GH et al. All-in-one centrifugal microfluidic device for size-selective circulating tumor cell isolation with high purity. Anal Chem. 2014.
    1. Hillig T, Nygaard AB, Nekiunaite L, Klingelhofer J, Soletormos G. In vitro validation of an ultra-sensitive scanning fluorescence microscope for analysis of circulating tumor cells. Apmis. 2014;122(6):545–51. doi: 10.1111/apm.12183.
    1. Zhao M, Schiro PG, Kuo JS, Koehler KM, Sabath DE, Popov V, et al. An automated high-throughput counting method for screening circulating tumor cells in peripheral blood. Anal Chem. 2013;85(4):2465–71. doi: 10.1021/ac400193b.

Source: PubMed

3
订阅