Circulating Tumor Cell Composition in Renal Cell Carcinoma

Ivonne Nel, Thomas C Gauler, Kira Bublitz, Lazaros Lazaridis, André Goergens, Bernd Giebel, Martin Schuler, Andreas-Claudius Hoffmann, Ivonne Nel, Thomas C Gauler, Kira Bublitz, Lazaros Lazaridis, André Goergens, Bernd Giebel, Martin Schuler, Andreas-Claudius Hoffmann

Abstract

Purpose: Due to their minimal-invasive yet potentially current character circulating tumor cells (CTC) might be useful as a "liquid biopsy" in solid tumors. However, successful application in metastatic renal cell carcinoma (mRCC) has been very limited so far. High plasticity and heterogeneity of CTC morphology challenges currently available enrichment and detection techniques with EpCAM as the usual surface marker being underrepresented in mRCC. We recently described a method that enables us to identify and characterize non-hematopoietic cells in the peripheral blood stream with varying characteristics and define CTC subgroups that distinctly associate to clinical parameters. With this pilot study we wanted to scrutinize feasibility of this approach and its potential usage in clinical studies.

Experimental design: Peripheral blood was drawn from 14 consecutive mRCC patients at the West German Cancer Center and CTC profiles were analyzed by Multi-Parameter Immunofluorescence Microscopy (MPIM). Additionally angiogenesis-related genes were measured by quantitative RT-PCR analysis.

Results: We detected CTC with epithelial, mesenchymal, stem cell-like or mixed-cell characteristics at different time-points during anti-angiogenic therapy. The presence and quantity of N-cadherin-positive or CD133-positive CTC was associated with inferior PFS. There was an inverse correlation between high expression of HIF1A, VEGFA, VEGFR and FGFR and the presence of N-cadherin-positive and CD133-positive CTC.

Conclusions: Patients with mRCC exhibit distinct CTC profiles that may implicate differences in therapeutic outcome. Prospective evaluation of phenotypic and genetic CTC profiling as prognostic and predictive biomarker in mRCC is warranted.

Trial registration: ClinicalTrials.gov NCT01731158 NCT01521715.

Conflict of interest statement

Competing Interests: Dr. Nel, shared first author, declares that she has no significant competing financial, professional or personal interests that might have influenced the performance or presentation of the work described in this manuscript. The lab work was completed and the manuscript was written while she was employed at M.O.R.E.—Molecular Oncology Risk-Profile Evaluation, Department of Medical Oncology, West German Cancer Center, University Duisburg-Essen. Final changes, submission and revision of the manuscript was processed during her employment at ABA GmbH & Co. KG, where she is currently working on a co-operative project. In accordance with her ethical obligation as a researcher, she is reporting that she receives salaries from the company ABA GmbH & Co. KG that may be affected by the research reported in the enclosed paper. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Basic principle of CTC isolation.
Fig 1. Basic principle of CTC isolation.
1. Leucosep tube: 2. Separation media; 3. Whole blood/PBMNC mixture; 4. Plasma; 5. Separation media after centrifugation; 6. Erythocytes; 7. Buffy coat incl. CTCs; 8. Anti-CD45 beads; 9. Anti-EpCAM beads (positive isolation) or anti-CD15 beads (negative isolation); 10. Washing buffer; 11. EpCAM-bead bound CTC suspension for qRT-PCR; 12i. Depleted bead free cell suspension containing CTCs for Cellspin and immunofluorescence staining. 12 ii. Depleted bead free cell suspension containing CD45-/EpCAM- CTC for qRT-PCR.
Fig 2. CTC detection.
Fig 2. CTC detection.
A) Positive control consisting of PBMNC mixed with CD133-expressing K562 cells which were stably transduced with lentiviral vectors endocoding an internal ribosomal entry site (IRES)-mediated co-expression cassette of CD133 and enhanced green fluorescent protein (eGFP) (K562-CD133:IEG). Cells were stained with DAPI (nucleus; blue) and for CD133 (pseudo-color red). The antibodies used for anti-CD133 immunostaining specifically bound to K562-CD133:IEG cells, visualized by Cy3 (red) counterstaining (40x magnification). B) CTC isolated from mRCC patients stained with DAPI (blue), for pan-CK (epithelial; red) and for CD45 (hematopoietic; green; 20x magnification). The cell marked with a white arrow shows a DAPI-positive (blue)/CD45-negative staining and was positive for pan-CK (red) and subsequently considered as CTC.
Fig 3. Detection of CTC subtypes.
Fig 3. Detection of CTC subtypes.
CTC isolated from mRCC patients were stained with DAPI (nucleus; blue) and for CD45 (hematopoietic; green), pan-CK (epithelial; red) and N-cadherin (mesenchymal; yellow) on one slide and with DAPI and for CD45, pan-CK and CD133 (stem cell; yellow) on a second slide. Cells marked with a white arrow were considered as CTC. The image displays various CTC subtypes with epithelial, mesenchymal and/or stem cell-like features such as N-cadherin+/CK-/CD45-; N-cadherin-/CK+/CD45-; CD133+/CK+/CD45+ and CD133-/CK+/CD45 (low) cells.
Fig 4. CTC subtypes were associated to…
Fig 4. CTC subtypes were associated to clinical outcome.
A) Kaplan-Meier test showed that the number of N-cadherin+/CK- cells was significantly associated to progression free survival (PFS) of mRCC patients during first-line treatment with anti-angiogenesis therapy (PFS; 7 vs. 15 months; p = 0.03; [HR] = 0.31; CI: 0.06–1.59). B) Mann-Whitney test revealed a significantly increased number of CD133+ cells in the presence of N-cadherin+ cells (p = 0.05).
Fig 5. Gene expression analysis.
Fig 5. Gene expression analysis.
A) Visualization of significant interrelationships between gene expressions. B and C) Mann-Whitney test showed that mRNA expression levels of HIF1A (B) and KDR1 (VEGFR) (C) were significantly decreased (p = 0.05) in the EpCAM-/CD45- fraction when CD133+ cells were present in the blood sample.

References

    1. Lianidou ES, Markou A, (2011) Circulating tumor cells as emerging tumor biomarkers in breast cancer. Clin Chem Lab Med 49: 1579–1590. 10.1515/CCLM.2011.628
    1. O'Flaherty JD, Gray S, Richard D, Fennell D, O'Leary JJ, Blackhall FH, et al. (2012) Circulating tumour cells, their role in metastasis and their clinical utility in lung cancer. Lung Cancer 76: 19–25. 10.1016/j.lungcan.2011.10.018
    1. Nel I, David P, Gerken GH, Schlaak J, Hoffmann A-C (2014) Role of circulating tumor cells and cancer stem cells in hepatocellular carcinoma. Hepatology International 8: 321–329. 10.1007/s12072-014-9539-3
    1. Alix-Panabieres C, Pantel K (2013) Circulating tumor cells: liquid biopsy of cancer. Clin Chem 59: 110–118. 10.1373/clinchem.2012.194258
    1. Yu M, Bardia A, Wittner BS, Stott SL, Smas ME,Ting DT, et al. (2013) Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 339: 580–584. 10.1126/science.1228522
    1. Hernandez-Yanez M, Heymach JV, Zurita AJ (2012) Circulating biomarkers in advanced renal cell carcinoma: clinical applications. Curr Oncol Rep 14: 221–229. 10.1007/s11912-012-0231-2
    1. Bluemke K, Bilkenroth U, Meye A, Fuessel S, Lautenschlaeger C, Goebel S,et al. (2009) Detection of circulating tumor cells in peripheral blood of patients with renal cell carcinoma correlates with prognosis. Cancer Epidemiol Biomarkers Prev 18: 2190–2194. 10.1158/1055-9965.EPI-08-1178
    1. Gradilone A, Iacovelli R, Cortesi E, Raimondi C, Gianni W, Nicolazzo C, et al. (2011) Circulating tumor cells and "suspicious objects" evaluated through CellSearch(R) in metastatic renal cell carcinoma. Anticancer Res 31: 4219–4221.
    1. Nel I, Jehn U, Gauler T, Hoffmann AC (2014) Individual profiling of circulating tumor cell composition in patients with non-small cell lung cancer receiving platinum based treatment. Transl Lung Cancer Res 3: 100–106. 10.3978/j.issn.2218-6751.2014.03.05
    1. Nel I, Baba HA, Ertle J, Weber F, Sitek B, Eisenacher M, et al. (2013) Individual profiling of circulating tumor cell composition and therapeutic outcome in patients with hepatocellular carcinoma. Transl Oncol 6: 420–428.
    1. Weller P, Nel I, Hassenkamp P, Gauler T, Schlueter A, Lang S, et al. (2014) Detection of Circulating Tumor Cell Subpopulations in Patients with Head and Neck Squamous Cell Carcinoma (HNSCC). PLoS One 9.
    1. Nel I, Baba HA, Weber F, Sitek B, Eisenacher M, Meyer HE, et al. (2014) IGFBP1 in epithelial circulating tumor cells as a potential response marker to selective internal radiation therapy in hepatocellular carcinoma. Biomarkers in Medicine 8: 687–698. 10.2217/bmm.14.23
    1. Nel I, Gauler T, Hoffmann AC (2014) Circulating tumor cell composition and outcome in patients with solid tumors. International Journal of Clinical Pharmacology and Therapeutics 52: 74–75. 10.5414/CPXCES13EA01
    1. Ahn HS, Lee HJ, Hahn S, Kim WH, Lee KU, Sano T, et al. (2010) Evaluation of the seventh American Joint Committee on Cancer/International Union Against Cancer Classification of gastric adenocarcinoma in comparison with the sixth classification. Cancer 116: 5592–5598. 10.1002/cncr.25550
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45: 228–247. 10.1016/j.ejca.2008.10.026
    1. Tuveson DA, Willis NA, Jacks T, Griffin JD, Singer S, Fletcher CDM, et al. (2001) STI571 inactivation of the gastrointestinal stromal tumor c-KIT oncoprotein: biological and clinical implications. Oncogene 20: 5054–5058.
    1. Goergens A, Ludwig AK, Moellmann M, Krawczyk A, Duerig J, Hanenberg H, et al. (2014) Multipotent Hematopoietic Progenitors Divide Asymmetrically to Create Progenitors of the Lymphomyeloid and Erythromyeloid Lineages. Stem Cell Reports 3: 1058–1072. 10.1016/j.stemcr.2014.09.016
    1. Leurs C, Jansen M, Pollok KE, Heinkelein M, Schmidt M, Wissler M, et al. (2003) Comparison of three retroviral vector systems for transduction of nonobese diabetic/severe combined immunodeficiency mice repopulating human CD34+ cord blood cells. Hum Gene Ther 14: 509–519.
    1. Mochizuki H, Schwartz JP, Tanaka K, Brady RO, Reiser J (1998) High-titer human immunodeficiency virus type 1-based vector systems for gene delivery into nondividing cells. J Virol 72: 8873–8883.
    1. Müllers E, Uhlig T, Stirnnagel K, Fiebig U, Zentgraf H, Lindemann D. (2011) Novel functions of prototype foamy virus Gag glycine- arginine-rich boxes in reverse transcription and particle morphogenesis. J Virol 85: 1452–1463. 10.1128/JVI.01731-10
    1. Hoffmann AC, Wild P, Leicht C, Bertz S, Danenberg KD, Danenberg PV, et al. (2010) MDR1 and ERCC1 expression predict outcome of patients with locally advanced bladder cancer receiving adjuvant chemotherapy. Neoplasia 12: 628–636.
    1. Hoffmann AC, Danenberg KD, Taubert H, Danenberg PV, Wuerl P (2009) A three-gene signature for outcome in soft tissue sarcoma. Clin Cancer Res 15: 5191–5198. 10.1158/1078-0432.CCR-08-2534
    1. Hoffmann AC, Mori R, Vallbohmer D, Brabender J, Klein E, Drebber U, et al. (2008) High expression of HIF1a is a predictor of clinical outcome in patients with pancreatic ductal adenocarcinomas and correlated to PDGFA, VEGF, and bFGF. Neoplasia 10: 674–679.
    1. Sarkar FH, Li Y, Wang Z, Kong D (2009) Pancreatic cancer stem cells and EMT in drug resistance and metastasis. Minerva Chir 64: 489–500.
    1. Pirozzi G, Tirino V, Camerlingo R, La Rocca A, Martucci N, Scognamiglio R, et al. (2013) Prognostic value of cancer stem cells, epithelial-mesenchymal transition and circulating tumor cells in lung cancer. Oncol Rep 29: 1763–1768. 10.3892/or.2013.2294
    1. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al. (2007) Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1: 313–323. 10.1016/j.stem.2007.06.002
    1. Jiang YS, Jiang T, Huang B, Chen PS, Ouyang J (2013) Epithelial-mesenchymal transition of renal tubules: Divergent processes of repairing in acute or chronic injury? Med Hypotheses 81: 73–75. 10.1016/j.mehy.2013.03.020
    1. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. (2008) The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133: 704–715. 10.1016/j.cell.2008.03.027
    1. Armstrong AJ, Marengo MS, Oltean S, Kemeny G, Bitting RL, Turnbull JD, et al. (2011) Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol Cancer Res 9: 997–1007. 10.1158/1541-7786.MCR-10-0490
    1. Nakajima S, Doi R, Toyoda E, Tsuji S, Wada M, Koizumi M, et al. (2004) N-cadherin expression and epithelial-mesenchymal transition in pancreatic carcinoma. Clin Cancer Res 10: 4125–4133.
    1. Thiery JP (2003) Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 15: 740–746.
    1. Gradilone A, Raimondi C, Nicolazzo C, Petracca A, Gandini O, Vincenzi B, et al. (2011) Circulating tumour cells lacking cytokeratin in breast cancer: the importance of being mesenchymal. J Cell Mol Med 15: 1066–1070. 10.1111/j.1582-4934.2011.01285.x
    1. Raimondi C, Gradilone A, Naso G, Vincenzi B, Petracca A, Nicolazzo C, et al. (2011) Epithelial-mesenchymal transition and stemness features in circulating tumor cells from breast cancer patients. Breast Cancer Res Treat 130: 449–455. 10.1007/s10549-011-1373-x
    1. Chong PP, Selvaratnam L, Abbas AA, Kamarul T (2012) Human peripheral blood derived mesenchymal stem cells demonstrate similar characteristics and chondrogenic differentiation potential to bone marrow derived mesenchymal stem cells. J Orthop Res 30: 634–642. 10.1002/jor.21556
    1. Kassis I, Zangi L, Rivkin R, Levdansky L, Samuel S, Marx G, et al. (2006) Isolation of mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads. Bone Marrow Transplant 37: 967–976.
    1. Wexler SA, Donaldson C, Denning-Kendall P, Rice C, Bradley B, Hows JM (2003) Adult bone marrow is a rich source of human mesenchymal 'stem' cells but umbilical cord and mobilized adult blood are not. Br J Haematol 121: 368–374.
    1. Roufosse CA, Direkze NC, Otto WR, Wright NA (2004) Circulating mesenchymal stem cells. Int J Biochem Cell Biol 36: 585–597.
    1. Yang FC, Watanabe S, Tsuji K, Xu MJ, Kaneko A, Ebihara Y, et al. (1998) Human granulocyte colony-stimulating factor (G-CSF) stimulates the in vitro and in vivo development but not commitment of primitive multipotential progenitors from transgenic mice expressing the human G-CSF receptor. Blood 92: 4632–4640.
    1. Cesselli D, Beltrami AP, Rigo S, Bergamin N, D'Aurizio F, Verardo R, et al. (2009) Multipotent progenitor cells are present in human peripheral blood. Circ Res 104: 1225–1234. 10.1161/CIRCRESAHA.109.195859
    1. Klopp AH, Spaeth EL, Dembinski JL, Woodward WA, Munshi A, Meyn RE, et al. (2007) Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res 67: 11687–11695.
    1. Yu M, Stott S, Toner M, Maheswaran S, Haber DA (2011) Circulating tumor cells: approaches to isolation and characterization. J Cell Biol 192: 373–382. 10.1083/jcb.201010021
    1. Joly E, Hudrisier D (2003) What is trogocytosis and what is its purpose? Nat Immunol 4: 815
    1. Chen S, Hou JH, Feng XY, Zhang XS, Zhou ZW, Yun JP, et al. (2013) Clinicopathologic Significance of Putative Stem Cell Marker, CD44 and CD133, in Human Gastric Carcinoma. J Surg Oncol 107: 799–806. 10.1002/jso.23337
    1. Gruenwald V, Beutel G, Schuch-Jantsch S, Reuter C, Ivanyi P, Ganser A, et al. (2010) Circulating endothelial cells are an early predictor in renal cell carcinoma for tumor response to sunitinib. BMC Cancer 10: 695 10.1186/1471-2407-10-695
    1. Bitting RL, Boominathan R, Rao C, Kemeny G, Foulk B, Garcia-Blanco MA, et al. (2013) Development of a method to isolate circulating tumor cells using mesenchymal-based capture. Methods 64: 129–136. 10.1016/j.ymeth.2013.06.034

Source: PubMed

3
Suscribir