anlotinib alters tumor immune microenvironment by downregulating PD-L1 expression on vascular endothelial cells

Shaochuan Liu, Tingting Qin, Zhujun Liu, Jing Wang, Yanan Jia, Yingfang Feng, Yuan Gao, Kai Li, Shaochuan Liu, Tingting Qin, Zhujun Liu, Jing Wang, Yanan Jia, Yingfang Feng, Yuan Gao, Kai Li

Abstract

Aberrant vascular network is a hallmark of cancer. However, the role of vascular endothelial cells (VECs)-expressing PD-L1 in tumor immune microenvironment and antiangiogenic therapy remains unclear. In this study, we used the specimens of cancer patients for immunohistochemical staining to observe the number of PD-L1+ CD34+ VECs and infiltrated immune cells inside tumor specimens. Immunofluorescence staining and flow cytometry were performed to observe the infiltration of CD8+ T cells and FoxP3+ T cells in tumor tissues. Here, we found that PD-L1 expression on VECs determined CD8+ T cells', FoxP3+ T cells' infiltration, and the prognosis of patients with lung adenocarcinoma. Anlotinib downregulated PD-L1 expression on VECs through the inactivation of AKT pathway, thereby improving the ratio of CD8/FoxP3 inside tumor and remolding the immune microenvironment. In conclusion, our results demonstrate that PD-L1 high expression on VECs inhibits the infiltration of CD8+ T cells, whereas promotes the aggregation of FoxP3+ T cells into tumor tissues, thus becoming an "immunosuppressive barrier". Anlotinib can ameliorate the immuno-microenvironment by downregulating PD-L1 expression on VECs to inhibit tumor growth.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1. PD-L1 high expression on tumor…
Fig. 1. PD-L1 high expression on tumor VECs is closely related to the prognosis of patients.
a, b Typical images of CD34/PD-L1 (endothelial cells highly expressing PD-L1) immunostaining in lung adenocarcinoma (n = 35), kidney (n = 30), and colon cancer (n = 30), scale bars: 50 μm. Kaplan–Meier survival curves of OS c and PFS c of patients with lung adenocarcinoma exhibiting high (red) or low (green) VEC-PD-L1. d The correlation of PD-L1+ vessels and PD-L1+ tumor tissues (n = 106). Data are mean ± SD, *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, Student’s t test.
Fig. 2. PD-L1 high expression on VECs…
Fig. 2. PD-L1 high expression on VECs is closely related to the infiltration of immune cells.
a, c Representative images of CD8+T cells and FoxP3+ T cells in sections taken from subjects with either PD-L1+ or PD-L1− vessels in lung, kidney, and colon cancer. Original magnification, ×200. The number of intratumoral CD8+ T cells and FoxP3+ T cells from patients’ sections (kidney or colon cancer) associated with the percentage of PD-L1+ vessels. The determination as high and low were grouped by the median values. Lung adenocarcinoma (n = 41) b, colon cancer (n = 50) e and kidney cancer (n = 32) d; scale bars: 100 µm. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, Student’s t test.
Fig. 3. PD-L1 expression on endothelial cells…
Fig. 3. PD-L1 expression on endothelial cells affects the proliferation and activation of immune cells.
a PBMC was extracted from normal blood and treated with 2.5 µg/ml anti-CD3 antibody and IL-2 (100 U/ml) for 72 h, and co-cultured with tumor-derived endothelial cells (Td-EC) for 48 h. b HUVECs were treated with the supernatant of tumor cells (CM) for 48 h. c PBMC and Td-EC were treated with control, anlotinib (0.1 μm) or anti-PD-L1 antibody (4 μg/ml). d Detection of the CD8+ IFN-γ+, CD8+Ki67+, CD8+GranzymeB+, and FoxP3+ T cells by flow cytometry. e Quantification of the above positive cells in the immune cells. f Quantification of cytokine levels in co-culture supernatants (2 days) by liquid microarray. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, Student’s t test.
Fig. 4. anlotinib can inhibit endothelial PD-L1…
Fig. 4. anlotinib can inhibit endothelial PD-L1 expression via the AKT pathway.
a Western blot analysis showed p-AKT and p-ERK levels in with- or without-anlotinib (0.1 µm) treated HUVECs at the indicated time points. b Western blot analysis showed PD-L1 expression levels in HUVECs treated by U0126 (ERK inhibitor,10 µm), Ly294002 (AKT inhibitor, 10 µm), or anlotinib (0.1 µm) at 24 h. c, e Western blot analysis showed p-AKT and PD-L1 expression in MRMEC and bEnd.3 treated as shown. d, f Western blot analysis showed p-AKT and PD-L1 expression in HUVECs treated as shown. (SC-79, one of activator of AKT). Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, Student’s t test.
Fig. 5. anlotinib inhibits endothelial PD-L1 expression…
Fig. 5. anlotinib inhibits endothelial PD-L1 expression and inhibits tumor growth.
a C57BL/6 mice were injected with 1 × 106 B16 cells and tumors grew. On day 11, mice were divided into four groups (n = 10 per group), and treated with bevacizumab, anlotinib, or both as shown. b Kaplan–Meier survival curves of B16 mice treated as indicated. c Representative images of B16 tumors in different groups. d Left: tumor growth curve of the various treatment groups; right: statistics of the weight of B16 tumors treated as indicated. e Representative flow images of CD31+ PD-L1+ cells taken from mice treated with anlotinib (n = 5), bevacizumab (n = 4), or both (n = 5). f Statistics of the percentage of CD31+ PD-L1+ cells in the B16 or MC38 tumors and the percentage of PD-L1+ non -VECs in B16 tumors. g Statistics of the percentage of CD45−CD31+ cells in the B16 or MC38 tumors. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, Student’s t test.
Fig. 6. anlotinib improves the immune microenvironment…
Fig. 6. anlotinib improves the immune microenvironment via increasing the ratio of CD8/FoxP3.
a Representative flow images of CD4+ CD25+ FoxP3+ T cells taken from B16 tumors treated as above indicated. b, c Statistics of the percentage of CD4+T cells, CD8+ T cells, FoxP3+ T cells and ratio of CD8/FoxP3 in the B16 or MC38 tumors. d Representative images of CD31 immunostaining (red), 4′,6-diamidino-2-phenylindole (DAPI) nuclear staining (blue) and markers of immune cells (CD4, CD8, and FoxP3) (green) of B16 tumors (n = 5 per group, except for Bev group n = 4) treated as above indicated. Scale bars,100 mm. e Quantification of CD4+ T cells, CD8+ T cells, FoxP3+ T cells in B16 tumors. f Statistics of perivascular and without perivascular CD8+ T cells in the B16 tumors. g Statistics of ratio of CD8/FoxP3 in the B16 or MC38 tumors. All data are exhibited as mean ± SD. Statistical differences were assessed using the unpaired Student’s test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Fig. 7. Endothelial PD-L1 expression affects the…
Fig. 7. Endothelial PD-L1 expression affects the efficacy of anlotinib.
a C57BL/6 mice were injected with 1 × 106 B16 cells and tumors grew. On day 12, mice were divided into four groups (n = 8 per group), and treated with bEnd.3-vector, bEnd.3-CD274, PBS or anlotinib as shown. b Representative images of B16 tumor tissues treated as above indicated. c Left: tumor growth curve of the various treatment groups; right: Statistics of the weight of B16 tumors treated as indicated. d Representative flow images of CD4+ CD25+ FoxP3+ T cells taken from B16 tumors treated as indicated. e Statistics of the percentage of CD8+ T cells, CD8+ IFN-γ+ T cells, FoxP3+ T cells and ratio of CD8/FoxP3 in the tumors. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Fig. 8. A schematic representation of effects…
Fig. 8. A schematic representation of effects of PD-L1 expression on VECs in tumor microenvironment.
a Pattern diagram of tumor immune microenvironment. b PD-L1 highly expressed on VECs in tumor tissues inhibits activation of CD8+ T cells and promotes immune escape of tumors. c anlotinib inhibits the expression of PD-L1 on VECs through the AKT pathway. d anlotinib improved the ratio of CD8/FoxP3 and broke the immune barrier by inhibiting the VEC-PD-L1.

References

    1. Johansson-Percival A, He B, Ganss R. Immunomodulation of tumor vessels: it takes two to tango. Trends Immunol. 2018;39:801–814. doi: 10.1016/j.it.2018.08.001.
    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. Jourde-Chiche N, et al. Endothelium structure and function in kidney health and disease. Nat. Rev. Nephrol. 2019;15:87–108. doi: 10.1038/s41581-018-0098-z.
    1. Motz GT, et al. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 2014;20:607–615. doi: 10.1038/nm.3541.
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331.
    1. Chen WJ, et al. Human umbilical vein endothelial cells promote the inhibitory activation of CD4(+)CD25(+)Foxp3(+) regulatory T cells via PD-L1. Atherosclerosis. 2016;244:108–112. doi: 10.1016/j.atherosclerosis.2015.11.002.
    1. Pittet CL, Newcombe J, Prat A, Arbour N. Human brain endothelial cells endeavor to immunoregulate CD8 T cells via PD-1 ligand expression in multiple sclerosis. J. Neuroinflamm. 2011;8:155. doi: 10.1186/1742-2094-8-155.
    1. Grabie N, et al. Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart. Circulation. 2007;116:2062–2071. doi: 10.1161/CIRCULATIONAHA.107.709360.
    1. Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity. 2018;48:434–452. doi: 10.1016/j.immuni.2018.03.014.
    1. Schmittnaegel, M. et al. Dual angiopoietin-2 and VEGFA inhibition elicits antitumor immunity that is enhanced by PD-1 checkpoint blockade. Sci. Transl. Med.9, pii: eaak9670 (2017).
    1. Allen, E. et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci. Transl. Med.9, eaak9679 (2017).
    1. Shen G, et al. Anlotinib: a novel multi-targeting tyrosine kinase inhibitor in clinical development. J. Hematol. Oncol. 2018;11:120. doi: 10.1186/s13045-018-0664-7.
    1. Zhou, A. P. et al. Anlotinib versus sunitinib as first-line treatment for metastatic renal cell carcinoma: a randomized phase ii clinical trial. Oncologist24, e702-e708 (2019).
    1. Han B, et al. anlotinib as a third-line therapy in patients with refractory advanced non-small-cell lung cancer: a multicentre, randomised phase II trial (ALTER0302) Br. J. Cancer. 2018;118:654–661. doi: 10.1038/bjc.2017.478.
    1. Liu S, Qin T, Jia Y, Li K. PD-L1 expression is associated With VEGFA and LADC patients’ s.urvival. Front Oncol. 2019;9:189. doi: 10.3389/fonc.2019.00189.
    1. Fusi A, et al. PD-L1 expression as a potential predictive biomarker. Lancet Oncol. 2015;16:1285–1287. doi: 10.1016/S1470-2045(15)00307-1.
    1. Chen Y, et al. PD-L1 expression and tumor mutational burden status for prediction of response to chemotherapy and targeted therapy in non-small cell lung cancer. J. Exp. Clin. Cancer Res. 2019;38:193. doi: 10.1186/s13046-019-1192-1.
    1. Sato E, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl Acad. Sci. USA. 2005;102:18538–18543. doi: 10.1073/pnas.0509182102.
    1. Cavalleri T, et al. Combined low densities of FoxP3+ and CD3+ tumor-infiltrating lymphocytes identify stage II colorectal cancer at high risk of progression. Cancer Immunol. Res. 2019;7:751–758. doi: 10.1158/2326-6066.CIR-18-0661.
    1. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 2018;15:325–340. doi: 10.1038/nrclinonc.2018.29.
    1. Juneja VR, et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 2017;214:895–904. doi: 10.1084/jem.20160801.
    1. Tang H, et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J. Clin. Invest. 2018;128:580–588. doi: 10.1172/JCI96061.
    1. Tian L, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature. 2017;544:250–254. doi: 10.1038/nature21724.
    1. Huang Y, et al. Improving immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 2018;18:195–203. doi: 10.1038/nri.2017.145.
    1. Lane RS, et al. IFNgamma-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J. Exp. Med. 2018;215:3057–3074. doi: 10.1084/jem.20180654.
    1. Lomas-Neira J, et al. Novel role for PD-1:PD-L1 as mediator of pulmonary vascular endothelial cell functions in pathogenesis of indirect ARDS in mice. Front. Immunol. 2018;9:3030. doi: 10.3389/fimmu.2018.03030.
    1. Wyss J, et al. Stromal PD-1/PD-L1 expression predicts outcome in colon cancer patients. Clin. Colorectal Cancer. 2019;18:e20–20e38. doi: 10.1016/j.clcc.2018.09.007.
    1. Vila N, et al. Pretreatment of RPE cells with lutein can mitigate bevacizumab-induced increases in angiogenin and bFGF. Ophthalmic Res. 2017;57:48–53. doi: 10.1159/000449252.
    1. Tomita, Y. et al. Oncolytic herpes virus armed with vasculostatin in combination with bevacizumab abrogate glioma invasion via the CCN1 and AKT signaling pathways. Mol. Cancer Ther. 18, 1418–1429 (2019).
    1. Jia Y, et al. Effect of bevacizumab on the tight junction proteins of vascular endothelial cells. Am. J. Transl. Res. 2019;11:5546–5559.

Source: PubMed

Подписаться