VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors

Thibault Voron, Orianne Colussi, Elie Marcheteau, Simon Pernot, Mevyn Nizard, Anne-Laure Pointet, Sabrina Latreche, Sonia Bergaya, Nadine Benhamouda, Corinne Tanchot, Christian Stockmann, Pierre Combe, Anne Berger, Franck Zinzindohoue, Hideo Yagita, Eric Tartour, Julien Taieb, Magali Terme, Thibault Voron, Orianne Colussi, Elie Marcheteau, Simon Pernot, Mevyn Nizard, Anne-Laure Pointet, Sabrina Latreche, Sonia Bergaya, Nadine Benhamouda, Corinne Tanchot, Christian Stockmann, Pierre Combe, Anne Berger, Franck Zinzindohoue, Hideo Yagita, Eric Tartour, Julien Taieb, Magali Terme

Abstract

Immune escape is a prerequisite for tumor development. To avoid the immune system, tumors develop different mechanisms, including T cell exhaustion, which is characterized by expression of immune inhibitory receptors, such as PD-1, CTLA-4, Tim-3, and a progressive loss of function. The recent development of therapies targeting PD-1 and CTLA-4 have raised great interest since they induced long-lasting objective responses in patients suffering from advanced metastatic tumors. However, the regulation of PD-1 expression, and thereby of exhaustion, is unclear. VEGF-A, a proangiogenic molecule produced by the tumors, plays a key role in the development of an immunosuppressive microenvironment. We report in the present work that VEGF-A produced in the tumor microenvironment enhances expression of PD-1 and other inhibitory checkpoints involved in CD8(+) T cell exhaustion, which could be reverted by anti-angiogenic agents targeting VEGF-A-VEGFR. In view of these results, association of anti-angiogenic molecules with immunomodulators of inhibitory checkpoints may be of particular interest in VEGF-A-producing tumors.

© 2015 Voron et al.

Figures

Figure 1.
Figure 1.
VEGF-A–VEGFR inhibition decreases PD-1 expression on intratumoral CD8+ T cells. CT26 tumor-bearing mice were treated with sunitinib or DMSO (as a control; a), or anti–VEGF-A antibody or mouse serum (as a control; b), or masitinib or DMSO (as a control; c). Treatments were started when tumors reached 9–10 mm2. Tumor growth (left) was shown. Mice were sacrificed at day 22 after 2 wk of treatment, and PD-1 expression on tumor-infiltrating CD8+ T cells was analyzed (right). Each dot represents an individual mouse, and histograms represent mean ± SEM of 2–4 pooled experiments with at least 4 mice/group. *, P = 0.0439; ***, P < 0.001. (d) Depletion of CD8+ T cells decreases anti-VEGFA–induced antitumor effects. (e) Correlation of VEGF-A concentration/gram of tumor tissue and number of PD-1-expressing CD8+ T cells/gram of tumor tissue (determined by flow cytometry). Each dot represents an individual tumor sample. (f) PD-1 expression on tumor-infiltrating CD8+ T cells was analyzed on wild-type MEF-tumor bearing mice (MEF WT) and VEGF-A–deleted MEF cells (MEF KO) after 14 d of tumor growth. Each dot represents an individual mouse. Histograms represent mean ± SEM of 2 pooled experiments with 4–5 mice/group. For tumor growths, one representative experiment out of 3 is shown with 5 mice/group. **, P < 0.01; ***, P < 0.001.
Figure 2.
Figure 2.
VEGF-A enhances PD-1 expression on CD8+ T cells in vitro. (a) VEGF-R1 (left) and -R2 (right) expression is shown on CD8+ T cells from tumors and spleen of tumor-free (naive) and CT26 tumor-bearing mice. Each dot represents an individual mouse, histograms represent mean ± SEM of 2 pooled experiments with 3–5 mice/group. (b) VEGF-R1 (left) and -R2 (right) expression on purified CD8+ T cells after 24 and 48 h of culture with various doses of plate-bound anti-CD3 antibody. (c and d) Same experimental setting as in b, but showing a representative staining of VEGF-R2 by flow cytometry (c) and confocal microscopy (d). Isotype control of anti-VEGFR2 antibody is shown (blue line for c and top for d). (e) PD-1 expression on purified CD8+ T cells after 48 h of culture with plate-bound anti-CD3 with or without VEGF-A (50 ng/ml). (f) Same experiment as in e but with 10 µg/ml of plate-bound anti-CD3 and various doses of VEGF-A. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Figure 3.
Figure 3.
VEGF-A enhances co-expression of inhibitory receptors involved in CD8+ T cell exhaustion in a VEGF-R2 and NFAT-dependent manner. Percentages of Tim-3 (a), CTLA-4 (b), and Lag-3 (c) expression on purified CD8+ T cells after 48 h of culture with plate-bound anti-CD3 (10 µg/ml) and various doses of VEGF-A. Histograms represent means ± SEM of 3 pooled experiments. (d) Same experimental settings as in (a) but mean fluorescence intensity (MFI) is shown. (e) The simultaneous expression of inhibitory receptors (PD-1, Tim-3, CTLA-4, and Lag-3) was examined on stimulated CD8+ T cells. (f) Same experimental setting as in (e) but in the presence of anti-VEGF-R1 or -R2 antibodies. (g) Transcriptional analyses of gene products linked to T cell exhaustion and VEGF-R2 signaling in CD8+ T cells stimulated or not with VEGF-A using a microfluidic card designed for qRT-PCR (TaqMan Low Density Mouse Immune Array from Applied Biosystems). Graph represents log fold changes (relative to nontreated controls, calculated with the ΔΔCT method (normalization with RNA18s as endogenous control) of transcripts. (h) Same experimental setting as in (e) in the presence of 11R-VIVIT. For simultaneous expression of inhibitory receptors, one representative experiment out of three is shown. *, P < 0.05.
Figure 4.
Figure 4.
VEGF-A neutralization decreases expression of inhibitory receptors involved in exhaustion on CD8+ T cells in heterotopic and orthotopic mouse tumor models. Mice bearing subcutaneous CT26 tumors were treated with anti–VEGF-A antibody or mouse serum (as a control). Co-expressions of PD-1/Tim-3 (a), PD-1/CTLA-4 (b), or PD-1/Lag-3 (c) on intratumoral CD8+ T cells have been analyzed. 3 pooled experiments are shown with 5 mice/group. *, P < 0.05; **, P < 0.01. (d) Co-expression of PD-1, Tim-3, CTLA-4, and Lag-3 has been determined on intratumoral CD8+ T cells after 14 d of anti-VEGFA treatment (day 22). (e) Anti–VEGF-A was given to mice bearing CT26 hepatic metastases. Representative pictures of hepatic metastases (left panel) and the percentages of intratumoral CD8+ T cells expressing PD-1/Tim-3 (right panel) were shown. Two pooled experiments are shown with 3–4 mice/group. *, P < 0.05 (f). Same experimental setting as in d, but on CT26 hepatic metastases. (g) Co-expression of inhibitory receptors on CD8+ T from MC38 and MEF KO tumors. (h–i) VEGF-KO MEF (h) and wild-type MEF tumor-bearing mice (i) were treated with anti–PD-1 alone twice a week starting at day 7. (j) CT26 tumor-bearing mice were treated with anti-VEGFA or anti–PD-1 alone or both. Tumor growth was monitored twice a week. One representative experiment out of three is shown, with five mice per group. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

References

    1. Adotevi O., Pere H., Ravel P., Haicheur N., Badoual C., Merillon N., Medioni J., Peyrard S., Roncelin S., Verkarre V., et al. . 2010. A decrease of regulatory T cells correlates with overall survival after sunitinib-based antiangiogenic therapy in metastatic renal cancer patients. J. Immunother. 33:991–998 10.1097/CJI.0b013e3181f4c208
    1. Appay V., Jandus C., Voelter V., Reynard S., Coupland S.E., Rimoldi D., Lienard D., Guillaume P., Krieg A.M., Cerottini J.-C., et al. . 2006. New generation vaccine induces effective melanoma-specific CD8+ T cells in the circulation but not in the tumor site. J. Immunol. 177:1670–1678 10.4049/jimmunol.177.3.1670
    1. Baitsch L., Baumgaertner P., Devêvre E., Raghav S.K., Legat A., Barba L., Wieckowski S., Bouzourene H., Deplancke B., Romero P., et al. . 2011. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Invest. 121:2350–2360 10.1172/JCI46102
    1. Blackburn S.D., Shin H., Haining W.N., Zou T., Workman C.J., Polley A., Betts M.R., Freeman G.J., Vignali D.A.A., and Wherry E.J.. 2009. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 10:29–37 10.1038/ni.1679
    1. Cao M., Xu Y., Youn J.I., Cabrera R., Zhang X., Gabrilovich D., Nelson D.R., and Liu C.. 2011. Kinase inhibitor Sorafenib modulates immunosuppressive cell populations in a murine liver cancer model. Lab. Invest. 91:598–608 10.1038/labinvest.2010.205
    1. Dubreuil P., Letard S., Ciufolini M., Gros L., Humbert M., Castéran N., Borge L., Hajem B., Lermet A., Sippl W., et al. . 2009. Masitinib (AB1010), a potent and selective tyrosine kinase inhibitor targeting KIT. PLoS ONE. 4:e7258 10.1371/journal.pone.0007258
    1. Duraiswamy J., Kaluza K.M., Freeman G.J., and Coukos G.. 2013. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 73:3591–3603 10.1158/0008-5472.CAN-12-4100
    1. Finke J.H., Rini B., Ireland J., Rayman P., Richmond A., Golshayan A., Wood L., Elson P., Garcia J., Dreicer R., and Bukowski R.. 2008. Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clin. Cancer Res. 14:6674–6682 10.1158/1078-0432.CCR-07-5212
    1. Gabrilovich D.I., Chen H.L., Girgis K.R., Cunningham H.T., Meny G.M., Nadaf S., Kavanaugh D., and Carbone D.P.. 1996. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med. 2:1096–1103 10.1038/nm1096-1096
    1. Gavalas N.G., Tsiatas M., Tsitsilonis O., Politi E., Ioannou K., Ziogas A.C., Rodolakis A., Vlahos G., Thomakos N., Haidopoulos D., et al. . 2012. VEGF directly suppresses activation of T cells from ascites secondary to ovarian cancer via VEGF receptor type 2. Br. J. Cancer. 107:1869–1875 10.1038/bjc.2012.468
    1. Gibson H.M., Hedgcock C.J., Aufiero B.M., Wilson A.J., Hafner M.S., Tsokos G.C., and Wong H.K.. 2007. Induction of the CTLA-4 gene in human lymphocytes is dependent on NFAT binding the proximal promoter. J. Immunol. 179:3831–3840 10.4049/jimmunol.179.6.3831
    1. Hamid O., Robert C., Daud A., Hodi F.S., Hwu W.-J., Kefford R., Wolchok J.D., Hersey P., Joseph R.W., Weber J.S., et al. . 2013. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 369:134–144 10.1056/NEJMoa1305133
    1. Huang Y., Chen X., Dikov M.M., Novitskiy S.V., Mosse C.A., Yang L., and Carbone D.P.. 2007. Distinct roles of VEGFR-1 and VEGFR-2 in the aberrant hematopoiesis associated with elevated levels of VEGF. Blood. 110:624–631 10.1182/blood-2007-01-065714
    1. Ko J.S., Zea A.H., Rini B.I., Ireland J.L., Elson P., Cohen P., Golshayan A., Rayman P.A., Wood L., Garcia J., et al. . 2009. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin. Cancer Res. 15:2148–2157 10.1158/1078-0432.CCR-08-1332
    1. Le Roy C., Deglesne P.-A., Chevallier N., Beitar T., Eclache V., Quettier M., Boubaya M., Letestu R., Lévy V., Ajchenbaum-Cymbalista F., and Varin-Blank N.. 2012. The degree of BCR and NFAT activation predicts clinical outcomes in chronic lymphocytic leukemia. Blood. 120:356–365 10.1182/blood-2011-12-397158
    1. Liu Z.-J., Shirakawa T., Li Y., Soma A., Oka M., Dotto G.P., Fairman R.M., Velazquez O.C., and Herlyn M.. 2003. Regulation of Notch1 and Dll4 by vascular endothelial growth factor in arterial endothelial cells: implications for modulating arteriogenesis and angiogenesis. Mol. Cell. Biol. 23:14–25 10.1128/MCB.23.1.14-25.2003
    1. Oestreich K.J., Yoon H., Ahmed R., and Boss J.M.. 2008. NFATc1 regulates PD-1 expression upon T cell activation. J. Immunol. 181:4832–4839 10.4049/jimmunol.181.7.4832
    1. Ozao-Choy J., Ma G., Kao J., Wang G.X., Meseck M., Sung M., Schwartz M., Divino C.M., Pan P.-Y., and Chen S.-H.. 2009. The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res. 69:2514–2522 10.1158/0008-5472.CAN-08-4709
    1. Sakuishi K., Apetoh L., Sullivan J.M., Blazar B.R., Kuchroo V.K., and Anderson A.C.. 2010. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 207:2187–2194 10.1084/jem.20100643
    1. Schreiber R.D., Old L.J., and Smyth M.J.. 2011. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 331:1565–1570 10.1126/science.1203486
    1. Schweighofer B., Testori J., Sturtzel C., Sattler S., Mayer H., Wagner O., Bilban M., and Hofer E.. 2009. The VEGF-induced transcriptional response comprises gene clusters at the crossroad of angiogenesis and inflammation. Thromb. Haemost. 102:544–554.
    1. Stockmann C., Doedens A., Weidemann A., Zhang N., Takeda N., Greenberg J.I., Cheresh D.A., and Johnson R.S.. 2008. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature. 456:814–818 10.1038/nature07445
    1. Terme M., Pernot S., Marcheteau E., Sandoval F., Benhamouda N., Colussi O., Dubreuil O., Carpentier A.F., Tartour E., and Taieb J.. 2013. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 73:539–549 10.1158/0008-5472.CAN-12-2325
    1. Topalian S.L., Hodi F.S., Brahmer J.R., Gettinger S.N., Smith D.C., McDermott D.F., Powderly J.D., Carvajal R.D., Sosman J.A., Atkins M.B., et al. . 2012. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366:2443–2454 10.1056/NEJMoa1200690
    1. Wherry E.J.2011. T cell exhaustion. Nat. Immunol. 12:492–499 10.1038/ni.2035
    1. Wherry E.J., Ha S.-J., Kaech S.M., Haining W.N., Sarkar S., Kalia V., Subramaniam S., Blattman J.N., Barber D.L., and Ahmed R.. 2007. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity. 27:670–684 10.1016/j.immuni.2007.09.006
    1. Whiteside T.L., and Parmiani G.. 1994. Tumor-infiltrating lymphocytes: their phenotype, functions and clinical use. Cancer Immunol. Immunother. 39:15–21 10.1007/BF01517175
    1. Woo S.-R., Turnis M.E., Goldberg M.V., Bankoti J., Selby M., Nirschl C.J., Bettini M.L., Gravano D.M., Vogel P., Liu C.L., et al. . 2012. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 72:917–927 10.1158/0008-5472.CAN-11-1620
    1. Yamazaki T., Akiba H., Koyanagi A., Azuma M., Yagita H., and Okumura K.. 2005. Blockade of B7-H1 on macrophages suppresses CD4+ T cell proliferation by augmenting IFN-gamma-induced nitric oxide production. J. Immunol. 175:1586–1592 10.4049/jimmunol.175.3.1586
    1. Ziogas A.C., Gavalas N.G., Tsiatas M., Tsitsilonis O., Politi E., Terpos E., Rodolakis A., Vlahos G., Thomakos N., Haidopoulos D., et al. . 2012. VEGF directly suppresses activation of T cells from ovarian cancer patients and healthy individuals via VEGF receptor Type 2. Int. J. Cancer. 130:857–864 10.1002/ijc.26094

Source: PubMed

3
Abonnere