Dynamic changes of live/apoptotic circulating tumour cells as predictive marker of response to sunitinib in metastatic renal cancer

E Rossi, M Fassan, M Aieta, F Zilio, R Celadin, M Borin, A Grassi, L Troiani, U Basso, C Barile, T Sava, C Lanza, L Miatello, A Jirillo, M Rugge, S Indraccolo, M Cristofanilli, A Amadori, R Zamarchi, E Rossi, M Fassan, M Aieta, F Zilio, R Celadin, M Borin, A Grassi, L Troiani, U Basso, C Barile, T Sava, C Lanza, L Miatello, A Jirillo, M Rugge, S Indraccolo, M Cristofanilli, A Amadori, R Zamarchi

Abstract

Background: Recently, we developed an apoptotic assay for expanding the monitoring capabilities of the circulating tumour cells (CTC) test during therapy. An automated platform for computing CTCs was integrated with a mAb (M30) targeting a neoepitope disclosed by caspase cleavage at cytokeratin 18 in early apoptosis; we showed that live CTCs were associated with progression, consistent with enhanced cell migration and invasion. The test was first applied here to mRCC.

Methods: Live/apoptotic CTCs changes were measured in mRCC patients receiving first-line Sunitinib and compared with circulating endothelial cell (CEC) levels.

Results: The presence of EpCAM-positive, live CTCs predicts progression in individual mRCC patient, being associated with distant metastasis under first-line Sunitinib. Synchronous detection of CTCs and CEC levels discloses for the first time an association between their dynamic changes and outcome: a rapid increase of the CEC number as early as the first cycle of therapy is associated with CTC decrease in non-progressed patients, whereas a delayed response of CECs is related to higher CTC values in the progressed group indicating treatment failure.

Conclusion: We demonstrated that a delayed response to antiangiogenic treatment indicated by persistent detection of CECs correlates with persistent live CTCs and more aggressive disease.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
EpCAM expression in mRCC primary tumours, metastasis and CTC. (A) EpCAM expression was analysed by IHC in primary tumours (T, n=28) and metastasis (M, n=17) obtained from 34 patients, for comparing IHC data with CTC results. The white bars represent the EpCAM-negative IHC in T (19 out of 28) and in M (10 out of 17) and patients that were CTC-negative by CellSearch assay (5 out of 34). The light brown bars represent weak staining in T (6 out of 9) and M (4 out of 7); the dark brown bars represent Moderate staining in T (3 out of 9) and M (3 out of 7). The light blue bar symbolises patients that had 1–4 CTC (20 out of 29), the dark blue bar symbolises patients that had 5–10 CTC (5 out of 29), the deep blue bar symbolises patients that had more than 10 CTC (4 out of 29). (B) The EpCAM staining profile is consistent in secondary lesions and synchronous CTC detection; one representative case of 4 is shown. The primary renal lesion (patient number 58) does not feature any EpCAM immunostaining (Original magnification 20X). (C) Characteristic EpCAM membranous immunoreaction appears in the tumour biopsy obtained from the surrenalic metastasis (patient number 58). (Original magnification × 20). (D) Analysis of three rare cells in baseline blood sample of patient number 58 using an Analyzer II device (Veridex, Raritan, NJ, USA). Horizontally, the photos show the same cell stained for the combination (Comp) of CK (green) and DAPI (violet); CK PE only; DAPI only; CD45 APC only; and M30 FITC only. The red squares indicate positively stained cells: events #1 and #7 are live CTC, exhibiting strong and irregular CK staining, respectively; in the photo #19 based on M30 staining profile (sufficient signal relative to background) the event on the right is classified as apoptotic CTC; the event on the left is a leukocyte (CD45-positive).
Figure 2
Figure 2
Progression-free survival (PFS) for patients with negative or equal 0 vs positive ΔAUC. Progression-free survival was measured as the time between the baseline CTC assessment and documentation of first radiographic evidence of disease progression or death. The ΔAUC criterion represents the difference between live and apoptotic CTC concentration-time area and was calculated in all patients according to the following formula:
Figure 3
Figure 3
CEC and CTC changes in mRCC patients under Sunitinib. (A) CEC and CTC values (mean +1 s.d., white and black symbols, respectively) synchronously determined in 30 mRCC patients receiving Sunitinib treatment are plotted in relation to time. (B) The bars represent the number of CEC and CTC samples (white and black symbols, respectively) collected at each time point. (C) CEC and CTC longitudinal graphs (white and black symbols, respectively) are separately plotted for non-progressed and progressed patients (○ and ∇ symbols, respectively). Time points with n ⩾14 were showed.

References

    1. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, Tibbe AG, Uhr JW, Terstappen LW (2004) Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res 10: 6897–6904
    1. Amadori A, Rossi E, Zamarchi R, Carli P, Pastorelli D, Jirillo A (2009) Circulating and disseminated tumor cells in the clinical management of breast cancer patients: unanswered questions. Oncology 76: 375–386
    1. Amadori A, Zamarchi R, De Silvestro G, Forza G, Cavatton G, Danieli GA, Clementi M, Chieco-Bianchi L (1995) Genetic control of the CD4/CD8 T-cell ratio in humans. Nat Med 1: 1279–1283
    1. Bidard FC, Mathiot C, Degeorges A, Etienne-Grimaldi MC, Delva R, Pivot X, Veyret C, Bergougnoux L, de Cremoux P, Milano G, Pierga JY (2010) Clinical value of circulating endothelial cells and circulating tumor cells in metastatic breast cancer patients treated first line with bevacizumab and chemotherapy. Ann Oncol 21: 1765–1771
    1. Blumke K, Bilkenroth U, Schmidt U, Melchior A, Fussel S, Bartel F, Heynemann H, Fornara P, Taubert H, Wirth MP, Meye A (2005) Detection of circulating tumor cells from renal carcinoma patients: experiences of a two-center study. Oncol Rep 14: 895–899
    1. Budd GT, Cristofanilli M, Ellis MJ, Stopeck A, Borden E, Miller MC, Matera J, Repollet M, Doyle GV, Terstappen LW, Hayes DF (2006) Circulating tumor cells versus imaging--predicting overall survival in metastatic breast cancer. Clin Cancer Res 12: 6403–6409
    1. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW, Hayes DF (2004) Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 351: 781–791
    1. Danila DC, Heller G, Gignac GA, Gonzalez-Espinoza R, Anand A, Tanaka E, Lilja H, Schwartz L, Larson S, Fleisher M, Scher HI (2007) Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer. Clin Cancer Res 13: 7053–7058
    1. de Haas EC, di Pietro A, Simpson KL, Meijer C, Suurmeijer AJ, Lancashire LJ, Cummings J, de Jong S, de Vries EG, Dive C, Gietema JA (2008) Clinical evaluation of M30 and M65 ELISA cell death assays as circulating biomarkers in a drug-sensitive tumor, testicular cancer. Neoplasia 10: 1041–1048
    1. Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS (2009) Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15: 232–239
    1. Gosens MJ, van Kempen LC, van de Velde CJ, van Krieken JH, Nagtegaal ID (2007) Loss of membranous Ep-CAM in budding colorectal carcinoma cells. Mod Pathol 20: 221–232
    1. Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, Matera J, Allard WJ, Doyle GV, Terstappen LW (2006) 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 12: 4218–4224
    1. Huang D, Ding Y, Li Y, Luo WM, Zhang ZF, Snider J, Vandenbeldt K, Qian CN, Teh BT (2010) Sunitinib acts primarily on tumor endothelium rather than tumor cells to inhibit the growth of renal cell carcinoma. Cancer Res 70: 1053–1062
    1. Hutson TE, Davis ID, Machiels JP, De Souza PL, Rottey S, Hong BF, Epstein RJ, Baker KL, McCann L, Crofts T, Pandite L, Figlin RA (2010) Efficacy and safety of pazopanib in patients with metastatic renal cell carcinoma. J Clin Oncol 28: 475–480
    1. Johannsen M, Florcken A, Bex A, Roigas J, Cosentino M, Ficarra V, Kloeters C, Rief M, Rogalla P, Miller K, Grunwald V (2009) Can tyrosine kinase inhibitors be discontinued in patients with metastatic renal cell carcinoma and a complete response to treatment? A multicentre, retrospective analysis. Eur Urol 55: 1430–1438
    1. Krishnamurthy S, Cristofanilli M, Singh B, Reuben J, Gao H, Cohen EN, Andreopoulou E, Hall CS, Lodhi A, Jackson S, Lucci A (2010) Detection of minimal residual disease in blood and bone marrow in early stage breast cancer. Cancer 116: 3330–3337
    1. Liu L, Qian J, Singh H, Meiers I, Zhou X, Bostwick DG (2007) Immunohistochemical analysis of chromophobe renal cell carcinoma, renal oncocytoma, and clear cell carcinoma: an optimal and practical panel for differential diagnosis. Arch Pathol Lab Med 131: 1290–1297
    1. Maghzal N, Vogt E, Reintsch W, Fraser JS, Fagotto F (2010) The tumor-associated EpCAM regulates morphogenetic movements through intracellular signaling. J Cell Biol 191: 645–659
    1. Marshall FF, Dietrick DD, Baumgartner WA, Reitz BA (1988) Surgical management of renal cell carcinoma with intracaval neoplastic extension above the hepatic veins. J Urol 139: 1166–1172
    1. McDermott DF, Regan MM, Clark JI, Flaherty LE, Weiss GR, Logan TF, Kirkwood JM, Gordon MS, Sosman JA, Ernstoff MS, Tretter CP, Urba WJ, Smith JW, Margolin KA, Mier JW, Gollob JA, Dutcher JP, Atkins MB (2005) Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J Clin Oncol 23: 133–141
    1. McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM (2005) Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst 97: 1180–1184
    1. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA (2007) Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 356: 115–124
    1. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, Inoue M, Bergers G, Hanahan D, Casanovas O (2009) Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15: 220–231
    1. Patil S, Figlin RA, Hutson TE, Michaelson MD, Negrier S, Kim ST, Huang X, Motzer RJ (2011) Prognostic factors for progression-free and overall survival with sunitinib targeted therapy and with cytokine as first-line therapy in patients with metastatic renal cell carcinoma. Ann Oncol 22: 295–300
    1. Pierga JY, Hajage D, Bachelot T, Delaloge S, Brain E, Campone M, Dieras V, Rolland E, Mignot L, Mathiot C, Bidard FC (2012) High independent prognostic and predictive value of circulating tumor cells compared with serum tumor markers in a large prospective trial in first-line chemotherapy for metastatic breast cancer patients. Ann Oncol 23: 618–624
    1. Ratain MJ, Eisen T, Stadler WM, Flaherty KT, Kaye SB, Rosner GL, Gore M, Desai AA, Patnaik A, Xiong HQ, Rowinsky E, Abbruzzese JL, Xia C, Simantov R, Schwartz B, O'Dwyer PJ (2006) Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma. J Clin Oncol 24: 2505–2512
    1. Riethdorf S, Fritsche H, Muller V, Rau T, Schindlbeck C, Rack B, Janni W, Coith C, Beck K, Janicke F, Jackson S, Gornet T, Cristofanilli M, Pantel K (2007) Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res 13: 920–928
    1. Rini BI, Cohen DP, Lu DR, Chen I, Hariharan S, Gore ME, Figlin RA, Baum MS, Motzer RJ (2011) Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst 103: 763–773
    1. Rossi E, Basso U, Celadin R, Zilio F, Pucciarelli S, Aieta M, Barile C, Sava T, Bonciarelli G, Tumolo S, Ghiotto C, Magro C, Jirillo A, Indraccolo S, Amadori A, Zamarchi R (2010) M30 neoepitope expression in epithelial cancer: quantification of apoptosis in circulating tumor cells by CellSearch analysis. Clin Cancer Res 16: 5233–5243
    1. Rowand JL, Martin G, Doyle GV, Miller MC, Pierce MS, Connelly MC, Rao C, Terstappen LW (2007) Endothelial cells in peripheral blood of healthy subjects and patients with metastatic carcinomas. Cytometry A 71: 105–113
    1. Sandri MT, Zorzino L, Cassatella MC, Bassi F, Luini A, Casadio C, Botteri E, Rotmensz N, Adamoli L, Nole F (2010) Changes in circulating tumor cell detection in patients with localized breast cancer before and after surgery. Ann Surg Oncol 17: 1539–1545
    1. Sastre J, Maestro ML, Puente J, Veganzones S, Alfonso R, Rafael S, Garcia-Saenz JA, Vidaurreta M, Martin M, Arroyo M, Sanz-Casla MT, Diaz-Rubio E (2008) Circulating tumor cells in colorectal cancer: correlation with clinical and pathological variables. Ann Oncol 19: 935–938
    1. Seligson DB, Pantuck AJ, Liu X, Huang Y, Horvath S, Bui MH, Han KR, Correa AJ, Eeva M, Tze S, Belldegrun AS, Figlin RA (2004) Epithelial cell adhesion molecule (KSA) expression: pathobiology and its role as an independent predictor of survival in renal cell carcinoma. Clin Cancer Res 10: 2659–2669
    1. van der Gun BT, Melchers LJ, Ruiters MH, de Leij LF, McLaughlin PM, Rots MG (2010) EpCAM in carcinogenesis: the good, the bad or the ugly. Carcinogenesis 31: 1913–1921
    1. Visvader JE, Lindeman GJ (2008) Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8: 755–768
    1. Went P, Dirnhofer S, Salvisberg T, Amin MB, Lim SD, Diener PA, Moch H (2005) Expression of epithelial cell adhesion molecule (EpCam) in renal epithelial tumors. Am J Surg Pathol 29: 83–88
    1. Winter MJ, Nagelkerken B, Mertens AE, Rees-Bakker HA, Briaire-de Bruijn IH, Litvinov SV (2003) Expression of Ep-CAM shifts the state of cadherin-mediated adhesions from strong to weak. Exp Cell Res 285: 50–58
    1. Xin H, Zhang C, Herrmann A, Du Y, Figlin R, Yu H (2009) Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res 69: 2506–2513

Source: PubMed

3
S'abonner