Dual Vascular Endothelial Growth Factor Receptor and Fibroblast Growth Factor Receptor Inhibition Elicits Antitumor Immunity and Enhances Programmed Cell Death-1 Checkpoint Blockade in Hepatocellular Carcinoma

Liver Cancer - Tập 9 Số 3 - Trang 338-357 - 2020
Haijing Deng1,2, Anna Kan3,2, Ning Lv1,2, Luwen Mu4, Yi Han2, Longzhong Liu5,2, Yanyu Zhang2, Youfa Duan2, Shuangye Liao2, Shaolong Li2, Qingling Xie2,6, Tianxiao Gao7,2, Yanrong Li1,2, Zhenfeng Zhang8, Ming Zhao1,2
1Division of Minimally Invasive Interventional, Sun Yat-sen University Cancer Center, Guangzhou, China
2State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
3Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
4Department of Vascular Interventional Radiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
5Department of Ultrasonics, Sun Yat-sen University Cancer Center, Guangzhou, China
6VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
7Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
8Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China

Tóm tắt

<b><i>Background and Aims:</i></b> Combining anti-angiogenic therapy with immune checkpoint blockade with anti-programmed cell death-1 (PD-1) antibodies is a promising treatment for hepatocellular carcinoma (HCC). Tyrosine kinase inhibitors are well-known anti-angiogenic agents and offer potential for combination with anti-PD-1 antibodies. This study investigated the possible underlying immunomodulatory mechanisms of combined therapy. <b><i>Methods:</i></b> HCC tissue samples for RNA-sequencing (RNA-seq) were obtained from patients with differential prognoses following anti-PD-1 treatment. Recombinant basic fibroblast growth factor (bFGF) and vascular endothelial growth factor A (VEGFA) were used to stimulate T cells following lenvatinib or sorafenib treatment, respectively. T cell function was analyzed by flow cytometry and lactate dehydrogenase assay. In vivo experiments were conducted in murine H22 and Hepa 1–6 competent models of HCC. Local immune infiltration in the tumor microenvironment (TME) was assessed using multicolor flow cytometry. Gene regulation was evaluated by RNA-seq. Microvascular density was measured by immunohistochemistry, and PD-1 ligand (PD-L1) induction was quantified by western blot. <b><i>Results:</i></b> The baseline expression of VEGF and fibroblast growth factor (FGF) in patients with progressive disease was significantly higher than in patients achieving stable disease following anti-PD-1 treatment. VEGFA and bFGF significantly upregulated the expression of PD-1, cytotoxic T-lymphocyte-associated protein-4, and Tim-3 on T cells, while inhibiting the secretion of interferon gamma (IFNG) and granzyme B and suppressing T cell cytotoxicity. This immunosuppressive effect was reverted by lenvatinib but not sorafenib. Furthermore, dual lenvatinib/anti-PD-1 antibody therapy led to better antitumor effects than either sorafenib or fibroblast growth factor receptor (FGFR) inhibitor (BGJ398) in H22 murine models of HCC. Combined lenvatinib/anti-PD-1 treatment also led to long-term immune memory formation, while synergistically modulating the TME and enhancing the cytotoxic effect of T cells. Finally, lenvatinib inhibited PD-L1 expression on human umbilical vein endothelial cells, which improved the function of T cells. <b><i>Conclusions:</i></b> Inhibition of vascular endothelial growth factor receptor and FGFR augmented the efficacy of anti-PD-1 antibodies. Combined lenvatinib/anti-PD-1 treatment appears to exert antitumor activity by synergistically modulating effector T cell function in the TME and by mutually regulating tumor vessel normalization.

Từ khóa


Tài liệu tham khảo

10.1056/NEJMoa1200690

10.1056/NEJMoa1305133

10.1016/S0140-6736(17)31046-2

10.1080/14737140.2018.1535315

10.1016/j.intimp.2017.03.015

10.1136/esmoopen-2017-000213

10.1038/bjc.2017.434

10.3322/caac.20075

10.1073/pnas.1215397109

10.1111/cas.13889

10.1002/hep.27236

10.1159/000487148

10.1080/14712598.2018.1499722

10.2217/imt.15.126

10.1126/scitranslmed.aak9670

10.1038/nature21724

10.1016/S0140-6736(18)30207-1

10.2217/fon-2018-0745

10.1016/S1470-2045(19)30020-8

10.1056/NEJMoa1717002

10.3390/cancers10110412

10.1084/jem.20140559

10.1136/gutjnl-2015-309501

10.1186/2045-824X-6-18

10.4049/jimmunol.1801199

10.1371/journal.pone.0212513

10.1016/j.cell.2017.07.024

10.1016/j.cell.2018.09.035

10.1126/scitranslmed.aak9679

10.1159/000492533

10.1016/j.bbrc.2019.02.015