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

Haijing Deng, Anna Kan, Ning Lyu, Luwen Mu, Yi Han, Longzhong Liu, Yanyu Zhang, Youfa Duan, Shuangye Liao, Shaolong Li, Qiankun Xie, Tianxiao Gao, Yanrong Li, Zhenfeng Zhang, Ming Zhao, Haijing Deng, Anna Kan, Ning Lyu, Luwen Mu, Yi Han, Longzhong Liu, Yanyu Zhang, Youfa Duan, Shuangye Liao, Shaolong Li, Qiankun Xie, Tianxiao Gao, Yanrong Li, Zhenfeng Zhang, Ming Zhao

Abstract

Background and aims: 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.

Methods: 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.

Results: 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.

Conclusions: 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.

Keywords: Immune checkpoint; Immunomodulatory; Lenvatinib; Liver cancer; Programmed cell death-1 antibodies; Sorafenib; Vascular endothelial growth factor A; bFGF.

Conflict of interest statement

The authors have no conflict of interest.

Copyright © 2020 by S. Karger AG, Basel.

Figures

Fig. 1
Fig. 1
The expression of angiogenesis-related factors in patients with HCC before initiating anti-PD-1 antibody treatment. Baseline tumor tissue samples from HCC patients treated with PD-1 antibodies were analyzed using RNA-seq. According to treatment response assessed using imaging evaluation (mRECIST), the RNA expression was analyzed (3 patients with partial response [PD] vs. 2 with SD). a Treatment pattern diagram and disease progression for HCC patients. b Relative mRNA expression of angiogenesis-related factors. c FGF family. Tyrosine kinase receptors (d) and chemokines (e) in patients achieving a PD and SD as tested by RNA-seq. fThe secretion level of VEGFA in HCC cell lines tested by enzyme-linked immunosorbent assay and read by microplate reader at 450 nm. * p < 0.05, ** p < 0.01, **** p < 0.0001 (two-way ANOVA). PD-1, programmed cell death-1; VEGFA, vascular endothelial growth factor A; VEGFB, vascular endothelial growth factor B; VEGFC, vascular endothelial growth factor C; FGF, fibroblast growth factor; FGFR1, FGF receptor 1; IL, interleukin; PDGFD, platelet-derived growth factor D; PDGFC, platelet-derived growth factor C; MMP9, matrix metalloprotein; SD, stable disease; PD, progressive disease; TKR, tyrosine kinase receptors; KDR, kinase insert domain receptor; PDGFRA, platelet-derived growth factor receptor A; PDGFRB, platelet-derived growth factor receptor B; CXCL, chemokine (C-X-C motif) ligand.
Fig. 2
Fig. 2
The effect of stimulation of VEGFA and bFGF on T cells derived from healthy donors and patients with HCC with or without PD-1 antibody treatment were analyzed for PD-1, CTLA-4, and Tim-3 expression and IFNG or GZMB secretion. a Representative images and histogram showing the expression of PD-1 on T cells stimulated by VEGFA (50 ng/mL), bFGF (50 ng/mL), or the combination of both. b Healthy donor-derived T cells were separated and RNA expression of bFGF (FGF2) was analyzed in remaining T cells and activated T cells by CD3 antibody (5 ng/mL) and CD28 antibody (2.5 ng/mL) and activated T cells treated with lenvatinib (100 nM), sorafenib (100 nM), and BGJ398 (100 nM). c Healthy donor-derived T cells were cultured with HepG2 supernatant and then stimulated by bFGF with gradient concentrations (0, 20, 50, 100 ng/mL). d Representative images showing CTLA-4 and Tim-3 expression on PD-1+/− T cells. * p < 0.05, ** p < 0.01, *** p < 0.001 (two-way ANOVA). VEGF, vascular endothelial growth factor; FGF2, fibroblast growth factor 2; PD-1, programmed cell death-1; HCC, hepatocellular carcinoma; LEN, lenvatinib; SOR, Sorafenib; CD8, cluster of differentiation 8; CTLA4, cytotoxic lymphocyte antigen 4; FSC-H, forward scatter height.
Fig. 3
Fig. 3
The effect of dual targeting of VEGFR and FGFR by lenvatinib on T cell function. HCC-patient-derived T cells were used to assess the effect of targeting VEGFR and FGFR on T cell function and cytotoxicity. a Representative images showing the expression of PD-1 on T cells stimulated by VEGFA (50 ng/mL) combined with bFGF (50 ng/mL), and then treated with lenvatinib (100 nM) and sorafenib (100 nM), respectively. b The effect of lenvatinib with different concentration gradients (0, 30, 60, 120, 240, 480, 960, 2,000, 3,000 nM) on T cell proliferation, tested by CCK8 and read by microplate reader at 450 nm. c HCC-patient-derived T cells were co-cultured with or without HepG2 cells and treated with or without lenvatinib (100 nM); T cells were cultured with HepG2 cells and treated with Blank, VEGFA (20 ng/mL), VEGFA (20 ng/mL) + lenvatinib (100 nM), and VEGFA (20 ng/mL) + sorafenib (100 nM). The LDH level was tested and read by microplate reader at 450 nm. Representative images showing the expression of IFNG (d) and GZMB (e) on T cells stimulated by VEGFA or bFGF, respectively, and the combination, and then treated with lenvatinib (100 nM), sorafenib (100 nM), and BGJ398 (FGFRi; 100 nM), respectively. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 (two-way ANOVA). VEGF, vascular endothelial growth factor; LEN, lenvatinib; PD-1, programmed cell death-1; LDH, lactate dehydrogenase; IFNG, interferon gamma; GZMB, granzyme B; SSC-A, side scatter; PE-A, KO525, VF, and ECD-A are fluorescent dyes; HepG2 is a hepatoma cell line; BLANK is referred to the control group used in this experiment.
Fig. 4
Fig. 4
The effect of lenvatinib, sorafenib, or BGJ398 combined with anti-PD-1 antibodies in vivo. In vivo antitumor experiments in murine models of HCC were performed to compare the anti-tumor effects of lenvatinib, sorafenib, and BGJ398 combined with PD-1 antibodies. a Tumor growth curves over 10 days for the control group (n = 5), lenvatinib combined with PD-1 antibodies group (10 mg/kg, 200 µg/3 days; n = 5), sorafenib combined with PD-1 antibodies group (50 mg/kg, 200 µg/3 days; n = 5) and BGJ398 combined with PD-1 antibodies group (10 mg/kg, 200 µg/3 days; n = 5) in Balb/C mice models subcutaneously implanted with H22 cells. b Mean weights for different treatment groups versus control in Balb/C mice models. c Tumor volume changes during treatment in the different groups and the final mean tumor volume at day 10 in the different treatment groups; individual tumor growth curves over the 10 days of treatment in the control group (d); lenvatinib combined with anti-PD-1 antibodies group (e); sorafenib combined with anti-PD-1 antibodies group (f); and BGJ398 combined with anti-PD-1 antibodies group (g). * p < 0.05, ** p < 0.01, **** p < 0.0001 (one-way/two-way ANOVA). LEN, lenvatinib; PD-1, programmed cell death-1.
Fig. 5
Fig. 5
Antitumor and immunoregulatory effect of combination treatment with lenvatinib and anti-PD-1 antibodies on murine models of HCC. The effect of combination treatment with lenvatinib and anti-PD-1 antibodies was investigated in 2 HCC murine models and the immune TME was tested by flow cytometry. a Mean tumor weights for the lenvatinib group (10 mg/kg), PD-1 antibodies group (100 µg/3 days), and lenvatinib combined with PD-1 antibodies group (10 mg/kg, 100 µg/3 days) versus control group treated for 21 days in Balb/C mice models subcutaneously implanted with H22 cells (n = 7 per group). b Mean tumor volume for the different treatment groups versus control over 21 days of treatment in C57BL/6 mice models subcutaneously implanted with Hepa 1–6 cells (n = 5 per group); survival curve of mice over 24 days of treatment for Balb/C (c) and C57BL/6 (d) mice in the different treatment groups; tumor re-challenge for CR mice in each group after a 50-day washout without treatment in Balb/C mice and C57BL/6 mice versus normal control (e). Histogram showing infiltration of CD4, CD8, NK, and the proportion of PD-1+ NK cells, and infiltration of dendritic cells and the portion of MHC+/PD-L1+ dendritic cells of Balb/C (f) and C57BL/6 mice (g) measured by flow cytometry. h Representative flow cytometry image showing the change of CD8 T cells in lenvatinib group versus control group; infiltration of PD-1+ CD8, PD-1+ CD4, and T regulatory cell of Balb/C and C57BL/6 mice measured by flow cytometry. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 (two-way ANOVA). LEN, lenvatinib; PD-1, programmed cell death-1; NK, natural killer; MHC+, major histocompatibility complex-positive; PD-L1, PD-1 ligand; DC, dendritic cell; PE-Cy7, a fluorescent dye; CON, control group.
Fig. 6
Fig. 6
The effect of combination lenvatinib/anti-PD-1 therapy on the TME. Profiler Mice XL cytokine analysis, RNA-seq, IHC, and TSA-IHC were used to evaluate the effect of lenvatinib combined with PD-1 antibodies therapy on the TME. a Profiler Mice XL cytokine analysis of peripheral blood plasma from tumor-bearing Balb/C mice in the different treatment groups. b Venn diagram analysis of gene expression profiles in tumors in the different treatment groups tested by RNA-seq. c Markers of activated T cells in the different treatment groups tested by RNA-seq. d Gene expression of Cluster 4 in the different treatment groups tested by RNA-seq. e Representative images and histogram of CD31 immunohistochemical staining in the different treatment groups in Balb/C mice and C57BL/6 mice. f Representative images of CD8 and PD-1 tyramide signal amplification immunohistochemical staining in tumor tissues sampled from tumor-bearing Balb/C mice in the different treatment groups and representative images for PD-1+ CD8 distribution in the combination group versus the control group. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 (two-way ANOVA). PD-1, programmed cell death-1; LEN, lenvatinib; ANGPT, angiopoietin 1; TNF-a, tumor necrosis factor-a; ACTB, beta actin, GAPDH, glyceraldehyde-3-phosphate dehydrogenase; Si-NC, the negative control of small interference RNA.
Fig. 7
Fig. 7
The effect of lenvatinib on PD-L1 expression on HUVEC. HUVEC were used to test the effect of lenvatinib on PD-L1 expression in vitro. a The effect of VEGFA (20 ng/mL) and lenvatinib with gradient concentration (4, 30, 300, 3,000 nM) on PD-L1 expression following stimulation by IFNG (10 ng/mL); the effect of HepG2 supernatant on PD-L1 expression in HUVEC. b The effect of downregulating VEGFR2 (sh21, sh22, sh23; si-01, si-02) in HUVEC on p-STAT1 and PD-L1 following stimulation by IFNG (10 ng/mL). c The effect of downregulating PD-L1 (si-01, si-02, si-03) in HUVEC on p-VEGFR2 following stimulation by VEGFA (50 ng/mL). d Diagram showing that dual targeting VEGFR and FGFR elicits antitumor immunity and enhances PD-1 checkpoint blockade in HCC. VEGF, vascular endothelial growth factor; LEN, lenvatinib; PD-1, programmed cell death-1; IFNG, interferon gamma; PD-L1, PD-1 ligand; HN and NH are both amines

Source: PubMed

3
Se inscrever