The tyrosine-kinase inhibitor sunitinib targets vascular endothelial (VE)-cadherin: a marker of response to antitumoural treatment in metastatic renal cell carcinoma

Helena Polena, Julie Creuzet, Maeva Dufies, Adama Sidibé, Abir Khalil-Mgharbel, Aude Salomon, Alban Deroux, Jean-Louis Quesada, Caroline Roelants, Odile Filhol, Claude Cochet, Ellen Blanc, Céline Ferlay-Segura, Delphine Borchiellini, Jean-Marc Ferrero, Bernard Escudier, Sylvie Négrier, Gilles Pages, Isabelle Vilgrain, Helena Polena, Julie Creuzet, Maeva Dufies, Adama Sidibé, Abir Khalil-Mgharbel, Aude Salomon, Alban Deroux, Jean-Louis Quesada, Caroline Roelants, Odile Filhol, Claude Cochet, Ellen Blanc, Céline Ferlay-Segura, Delphine Borchiellini, Jean-Marc Ferrero, Bernard Escudier, Sylvie Négrier, Gilles Pages, Isabelle Vilgrain

Abstract

Background: Vascular endothelial (VE)-cadherin is an endothelial cell-specific protein responsible for endothelium integrity. Its adhesive properties are regulated by post-translational processing, such as tyrosine phosphorylation at site Y685 in its cytoplasmic domain, and cleavage of its extracellular domain (sVE). In hormone-refractory metastatic breast cancer, we recently demonstrated that sVE levels correlate to poor survival. In the present study, we determine whether kidney cancer therapies had an effect on VE-cadherin structural modifications and their clinical interest to monitor patient outcome.

Methods: The effects of kidney cancer biotherapies were tested on an endothelial monolayer model mimicking the endothelium lining blood vessels and on a homotypic and heterotypic 3D cell model mimicking tumour growth. sVE was quantified by ELISA in renal cell carcinoma patients initiating sunitinib (48 patients) or bevacizumab (83 patients) in the first-line metastatic setting (SUVEGIL and TORAVA trials).

Results: Human VE-cadherin is a direct target for sunitinib which inhibits its VEGF-induced phosphorylation and cleavage on endothelial monolayer and endothelial cell migration in the 3D model. The tumour cell environment modulates VE-cadherin functions through MMPs and VEGF. We demonstrate the presence of soluble VE-cadherin in the sera of mRCC patients (n = 131) which level at baseline, is higher than in a healthy donor group (n = 96). Analysis of sVE level after 4 weeks of treatment showed that a decrease in sVE level discriminates the responders vs. non-responders to sunitinib, but not bevacizumab.

Conclusions: These data highlight the interest for the sVE bioassay in future follow-up of cancer patients treated with targeted therapies such as tyrosine-kinase inhibitors.

Trial registration: ClinicalTrials.gov NCT00619268 NCT00943839.

Conflict of interest statement

The authors declare no competing interest.

Figures

Fig. 1
Fig. 1
Sunitinib impairs VEGF-induced VE-cadherin tyrosine phosphorylation and cleavage: primary ECs were grown 3–4 days to reach confluency (3 × 106 cells). After 3 h of serum starvation, ECs were stimulated by VEGF alone (50 ng/mL) or in the presence of SUT (20 μM) or TEM (50 ng/mL) during indicated times. Lysates were analysed by SDS-PAGE on 10% polyacrylamide gels and western blotting (5 μg of total protein lysate/lane). a Analysis of phosphotyrosine-containing protein pattern using the anti-phosphotyrosine antibody 4G10. b, c Analysis of phospho-Y685VE-cadherin d. Conditioned media from untreated ECs (CTL), or VEGF-stimulated-ECs or VEGF-stimulated-ECs treated with SUT or TEM for 20 min (same amount of cells in each condition) were collected, centrifuged to discard floating cells, and concentrated on Centriprep Centrifugal Filter Units with an Ultracel YM-30 membrane. Volume of 5 μL of concentrated medium of each condition was analysed by SDS-PAGE and western blotting using the monoclonal anti-VE-cadherin antibody (BV9 clone). e Same experiment as in d with VEGF-treated ECs in the presence of either INFα2 A (7.5 ng/mL), Irinotecan (IRN, 10 μM) and SN-38 (10 μM). Results are representative of three independent experiments
Fig. 2
Fig. 2
SUT impairs cell invasion and migration in heterotypic spheroids. a Representative images of spheroids formed from 786-O (RCC) cells alone at the time of their incorporation into collagen gel (T0) and 24 h later (T24), or RCC + ECs at time 0 and 24 h later. b Area of the spheroids was quantified using Image J software (National Institutes of Health). c Quantification of ECs migration from spheroids co-cultured with RCC using the measurement of the Euclidean distance of each EC (n = 500) from the centre of the spheroid after 24 h of culture. Results are representative of three independent experiments. Values are the mean ± SD of the migration distance in μm. Statistical significance is indicated by the number of asterisks (*)
Fig. 3
Fig. 3
Effect of RCC tumour microenvironment on VE-cadherin cleavage: elevated levels of sVE in mRCC patients: a Phase contrast microscopy of RCC cells in culture. b Analysis of RCC culture media by zymography showed the presence of MMP2 and MMP9 activities. c The relative amounts of MMPs were measured by densitometry of autoradiographs using ImageJ software (National Institutes of Health). d Phase contrast microscopy of ECs in culture. e Release of sVE by ECs upon RCC conditioned media treatment (20 min) (left panel) alone or RCC conditioned media pre-incubated or not with bevacizumab (BEV) (5 μg/mL) (right panel). f, g The relative amounts of sVE were measured by densitometry of autoradiographs using ImageJ software (National Institutes of Health). Results are representative of three independent experiments. Values are the mean ± SD. h sVE levels in healthy donors (n = 96) and mRCC patients from the clinical trial TORAVA (n = 115) at diagnosis (Day 0, D0) was performed by ELISA. Statistical significance is indicated by the number of asterisks (*)
Fig. 4
Fig. 4
Analysis of sVE- levels in mRCC patients after the first cycle of treatment. a Serum from mRCC patient treated with SUT was analysed for sVE-cadherin level using the monoclonal anti-VE-cadherin antibody. b sVE levels in mRCC patients after the first cycle of treatment. c Variations of sVE expressed as % of the initial concentration at diagnosis (level of sVE after the first cycle/level of sVE at diagnosis × 100) in SUT group with stable disease and relapse. d Variations of sVE expressed as % of the initial concentration at diagnosis (level of sVE after the first cycle/level of sVE at diagnosis × 100) in BEV group with stable disease and relapse. Statistical significance is indicated by the number of asterisks (*)
Fig. 5
Fig. 5
Relationship between sVE-cadherin levels and PFS of mRCC patients: ac Kaplan–Meier analysis of PFS of patients with RCC. PFS was calculated from patient subgroups with variations of sVE expressed as % of the initial concentration at diagnosis (level of sVE after the first cycle/level of sVE at diagnosis × 100) that were less or greater than a cutoff ratio at 120%, for SUT group trial (a), (b) BEV group. Statistical significance (P-value) and the time of the median disease free are indicated
Fig. 6
Fig. 6
Proposed mechanisms of microenvironment-induced VE-cadherin structural modifications in kidney cancer. This working hypothesis was mainly built from our results obtained using in vitro EC cultures and VEGF as the major actor in angiogenesis., The involvement of other cytokines in these processes cannot be excluded since we previously showed that TNFα induced sVE release

References

    1. Ferlay J, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer. 2015;136:E359–E386. doi: 10.1002/ijc.29210.
    1. Afriansyah A, Hamid AR, Mochtar CA, Umbas R. Targeted therapy for metastatic renal cell carcinoma. Acta Med. Indones. 2016;48:335–347.
    1. Yang JC, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N. Engl. J. Med. 2003;349:427–434. doi: 10.1056/NEJMoa021491.
    1. Escudier B, et al. Axitinib versus sorafenib in advanced renal cell carcinoma: subanalyses by prior therapy from a randomised phase III trial. Br. J. Cancer. 2014;110:2821–2828. doi: 10.1038/bjc.2014.244.
    1. Motzer RJ, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N. Engl. J. Med. 2007;356:115–124. doi: 10.1056/NEJMoa065044.
    1. Motzer RJ, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J. Clin. Oncol. 2009;27:3584–3590. doi: 10.1200/JCO.2008.20.1293.
    1. Motzer RJ, Escudier B, Gannon A, Figlin RA. Sunitinib: ten years of successful clinical use and study in advanced renal cell carcinoma. Oncologist. 2017;22:41–52. doi: 10.1634/theoncologist.2016-0197.
    1. Huang D, et al. Sunitinib acts primarily on tumour endothelium rather than tumour cells to inhibit the growth of renal cell carcinoma. Cancer Res. 2010;70:1053–1062. doi: 10.1158/0008-5472.CAN-09-3722.
    1. Dufies M, et al. Sunitinib stimulates expression of VEGFC by tumour cells and promotes lymphangiogenesis in clear cell renal cell carcinomas. Cancer Res. 2017;77:1212–1226. doi: 10.1158/0008-5472.CAN-16-3088.
    1. Giuliano S, et al. Resistance to sunitinib in renal clear cell carcinoma results from sequestration in lysosomes and inhibition of the autophagic flux. Autophagy. 2015;11:1891–1904. doi: 10.1080/15548627.2015.1085742.
    1. Joosten SC, et al. Resistance to sunitinib in renal cell carcinoma: from molecular mechanisms to predictive markers and future perspectives. Biochim. Biophys. Acta. 2015;1855:1–16.
    1. Karashima T, et al. Expression of angiogenesis-related gene profiles and development of resistance to tyrosine-kinase inhibitor in advanced renal cell carcinoma: characterization of sorafenib-resistant cells derived from a cutaneous metastasis. Int. J. Urol. 2013;20:923–930. doi: 10.1111/iju.12084.
    1. Dejana E, Tournier-Lasserve E, Weinstein BM. The control of vascular integrity by endothelial cell junctions: molecular basis and pathological implications. Dev. Cell. 2009;16:209–221. doi: 10.1016/j.devcel.2009.01.004.
    1. Bouillet L, et al. Endothelial junctions: exploiting their instability in the development of biomarkers for vascular remodelling. Med. Sci. 2014;30:633–635.
    1. Wallez Y, et al. Src kinase phosphorylates vascular endothelial-cadherin in response to vascular endothelial growth factor: identification of tyrosine 685 as the unique target site. Oncogene. 2007;26:1067–1077. doi: 10.1038/sj.onc.1209855.
    1. Lambeng N, et al. Vascular endothelial-cadherin tyrosine phosphorylation in angiogenic and quiescent adult tissues. Circ. Res. 2005;96:384–391. doi: 10.1161/01.RES.0000156652.99586.9f.
    1. Sidibé A, et al. Dynamic phosphorylation of VE-cadherin Y685 throughout mouse estrous cycle in ovary and uterus. Am. J. Physiol. Heart Circ. Physiol. 2014;307:H448–H454. doi: 10.1152/ajpheart.00773.2013.
    1. Sidibé A, et al. Soluble vascular-endothelial (VE)-cadherin: toward a marker of endothelial dysfunction. In: Feige JJ, Pagès G, Soncin F, et al., editors. Molecular Mechanisms of Angiogenesis. France: Springer-Verlag; 2014. pp. 461–478.
    1. Sidibé A, et al. Soluble VE-cadherin in rheumatoid arthritis patients correlates with disease activity: evidence for tumour necrosis factor α-induced VE-cadherin cleavage. Arthritis Rheum. 2012;64:77–87. doi: 10.1002/art.33336.
    1. Vilgrain I, et al. Evidence for post-translational processing of vascular endothelial (VE)-cadherin in brain tumours: towards a candidate biomarker. PLoS ONE. 2013;8:e80056. doi: 10.1371/journal.pone.0080056.
    1. Rochefort P, et al. Soluble VE-cadherin in metastatic breast cancer: an independent prognostic factor for both progression-free survival and overall survival. Br. J. Cancer. 2017;116:356–361. doi: 10.1038/bjc.2016.427.
    1. Lê J, et al. Quantitative zymography of matrix metalloproteinases by measuring hydroxyproline: application to gelatinases A and B. Electrophoresis. 1999;20:2824–2829. doi: 10.1002/(SICI)1522-2683(19991001)20:14<2824::AID-ELPS2824>;2-H.
    1. Dufies M, et al. CXCL7 is a predictive marker of sunitinib efficacy in clear cell renal cell carcinomas. Br. J. Cancer. 2017;117:947–953. doi: 10.1038/bjc.2017.276.
    1. Kluger HM, et al. Classification of renal cell carcinoma based on expression of VEGF and VEGF receptors in both tumour cells and endothelial cells. Lab. Invest. 2008;88:962–972. doi: 10.1038/labinvest.2008.65.
    1. Cunningham D, et al. Randomised trial of irinotecan plus supportive care versus supportive care alone after fluorouracil failure for patients with metastatic colorectal cancer. Lancet. 1998;352:1413–1418. doi: 10.1016/S0140-6736(98)02309-5.
    1. Figlin RA. Mechanisms of disease: survival benefit of temsirolimus validates a role for mTOR in the management of advanced RCC. Nat. Clin. Pract. Oncol. 2008;5:601–609. doi: 10.1038/ncponc1173.
    1. Parker BS, Rautela J, Hertzog PJ. Antitumour actions of interferons: implications for cancer therapy. Nat. Rev. Cancer. 2016;16:131–144. doi: 10.1038/nrc.2016.14.
    1. Gialeli C, Theocharis AD, Karamanos NK. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J. 2011;278:16–27. doi: 10.1111/j.1742-4658.2010.07919.x.
    1. Senger DR, et al. Vascular permeability factor (VPF, VEGF) in tumour biology. Cancer Metastas. Rev. 1993;12:303–324. doi: 10.1007/BF00665960.
    1. Di Desidero T, et al. Antiproliferative and proapoptotic activity of sunitinib on endothelial and anaplastic thyroid cancer cells via inhibition of Akt and ERK1/2 phosphorylation and by down-regulation of cyclin-D1. J. Clin. Endocrinol. Metab. 2013;98:E1465–E1473. doi: 10.1210/jc.2013-1364.
    1. Motzer RJ, et al. Investigation of novel circulating proteins, germ line single-nucleotide polymorphisms, and molecular tumour markers as potential efficacy biomarkers of first-line sunitinib therapy for advanced renal cell carcinoma. Cancer Chemother. Pharmacol. 2014;74:739–750. doi: 10.1007/s00280-014-2539-0.
    1. Kamiyama H, Takano S, Tsuboi K, Matsumura A. Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. J. Cancer Res. Clin. Oncol. 2005;131:205–213. doi: 10.1007/s00432-004-0642-z.
    1. Tien WS, Chen JH, Wu KP. Sheddome DB: the ectodomain shedding database for membrane-bound shed markers. BMC Bioinform. 2017;18:1465–1467. doi: 10.1186/s12859-017-1465-7.
    1. De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nat. Rev. Cancer. 2017;17:457–474. doi: 10.1038/nrc.2017.51.
    1. Bartolomé RA, et al. VE-cadherin RGD motifs promote metastasis and constitute a potential therapeutic target in melanoma and breast cancers. Oncotarget. 2017;8:215–227. doi: 10.18632/oncotarget.13832.
    1. Stubbs C, et al. A study of angiogenesis markers in patients with renal cell carcinoma undergoing therapy with sunitinib. Anticancer Res. 2017;37:253–259. doi: 10.21873/anticanres.11315.
    1. Cohen RB, Oudard S. Antiangiogenic therapy for advanced renal cell carcinoma: management of treatment-related toxicities. Invest. New Drug. 2012;30:2066–2079. doi: 10.1007/s10637-012-9796-8.
    1. Pastore AL, et al. Serum and urine biomarkers for human renal cell carcinoma. Dis. Markers. 2015;2015:251403. doi: 10.1155/2015/251403.
    1. Bauman TM, Huang W, Lee MH, Abel EJ. Neovascularity as a prognostic marker in renal cell carcinoma. Hum. Pathol. 2016;57:98–105. doi: 10.1016/j.humpath.2016.07.005.
    1. Blanchet B, et al. Development and validation of an HPLC-UV-visible method for sunitinib quantification in human plasma. Clin. Chim. Acta. 2009;404:134–139. doi: 10.1016/j.cca.2009.03.042.
    1. Sabanathan D, et al. Dose individualization of sunitinib in metastatic renal cell cancer: toxicity-adjusted dose or therapeutic drug monitoring. Cancer Chemother. Pharmacol. 2017;80:385–393. doi: 10.1007/s00280-017-3362-1.
    1. Cabel L, et al. Drug monitoring of sunitinib in patients with advanced solid tumours: a monocentric observational French study. Fundam. Clin. Pharmacol. 2018;32:98–107. doi: 10.1111/fcp.12327.
    1. Fernando NT, et al. Tumor escape from endogenous, extracellular matrix-associated angiogenesis inhibitors by up-regulation of multiple proangiogenic factors. Clin. Cancer Res. 2008;14:1529–1539. doi: 10.1158/1078-0432.CCR-07-4126.
    1. Négrier S, et al. Temsirolimus and bevacizumab, or sunitinib, or interferon alfa and bevacizumab for patients with advanced renal cell carcinoma (TORAVA): a randomised phase 2 trial. Lancet Oncol. 2011;12:673–680. doi: 10.1016/S1470-2045(11)70124-3.
    1. Bhojani N, et al. Toxicities associated with the administration of sorafenib, sunitinib, and temsirolimus and their management in patients with metastatic renal cell carcinoma. Eur. Urol. 2008;53:917–930. doi: 10.1016/j.eururo.2007.11.037.
    1. Noé G, et al. Clinical and kinomic analysis identifies peripheral blood mononuclear cells as a potential pharmacodynamic biomarker in metastatic renal cell carcinoma patients treated with sunitinib. Oncotarget. 2016;7:67507–67520. doi: 10.18632/oncotarget.11686.

Source: PubMed

3
Suscribir