Abstract

The efficacy of programmed cell death-ligand 1 (PD-L1)/programmed cell death protein 1 (PD-1) blockade therapy has been demonstrated but is limited in patients with PD-L1low or immune desert tumors. This limitation can be overcome by combination therapies that include anti-vascular endothelial growth factor (VEGF) therapy. Such combinations have been investigated in clinical trials for a number of cancer types; however, evidence on the mechanisms underlying their effects in these types of patients is still not sufficient. Therefore, the present study investigated the efficacy and effects on CD8+ T cell and C-X-C motif chemokine receptor 3 (CXCR3) ligand expression in tumors by combining anti-PD-L1 and anti-VEGF antibodies using an OV2944-HM-1 mouse model with PD-L1low and immune desert-like phenotypes. Although the model exhibited anti-PD-L1 insensitivity, anti-PD-L1 antibody treatment combined with anti-VEGF antibody inhibited tumor growth compared with anti-VEGF monotherapy, which itself inhibited tumor growth compared with the control treatment on Day 25. In combination-treated mice, a higher percentage of CD8+ T cells and higher levels of CXCR3 ligands were observed in tumor tissues compared with those in the anti-VEGF antibody treatment group, which was not significantly different from control treatment on Day 8. The increase in the intratumoral percentage of CD8+ T cells following the combination treatment was reversed by CXCR3 blocking to the same level as the control. In an anti-PD-L1 insensitive model with PD-L1low and immune desert-like phenotypes, although anti-PD-L1 antibody alone was not effective, anti-PD-L1 antibody in combination with anti-VEGF antibody exhibited antitumor combination efficacy with an increase of CD8+ T cell infiltration, which was suggested to be dependent on the increase of intratumoral CXCR3 ligands. This mechanism could explain the efficacy of anti-PD-L1 antibody and anti-VEGF antibody combination therapy in the clinical setting.

Highlights

  • Programmed cell death‐ligand 1 (PD‐L1) is an immune check‐ point molecule expressed on tumor cells and tumor‐infiltrating immune cells, which is involved in the suppression of cancer immunity [1]

  • Anti‐PD‐L1 antibody combined with anti‐Vascular endothelial growth factor (VEGF) antibody improves tumor control compared with anti‐VEGF antibody alone in an anti‐PD‐L1 insensitive HM‐1 tumor model

  • In the HM‐1 model, the anti‐VEGF antibody alone significantly inhibited tumor growth compared with the control; the anti‐PD‐L1 antibody alone did not significantly inhibit the tumor growth compared with the control (Fig. 2B)

Read more

Summary

Introduction

Programmed cell death‐ligand 1 (PD‐L1) is an immune check‐ point molecule expressed on tumor cells and tumor‐infiltrating immune cells, which is involved in the suppression of cancer immunity [1]. Anti‐PD‐L1 antibody relieves T cell suppres‐ sion by inhibiting the binding of PD‐L1 to programmed cell death protein 1 (PD‐1) and B7.1 ( known as CD80), which are receptors on effector T cells, and exerts antitumor effects in various types of cancer [2]. Vascular endothelial growth factor (VEGF) has been reported to exert tumor angiogenesis‐inducing activity, and immunosuppressive activity which can attenuate the antitumor immunity elicited by anti‐PD‐L1/PD‐1 therapy through inhibition of dendritic cell (DC) maturation [9,10,11,12] and accumulation of myeloid‐derived suppressor cells (MDSCs) [13]. It has been reported that VEGF blockade may promote antitumor immunity by inhibiting the accumulation of regulatory T‐cells (Tregs) [14]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.