Abstract

Although the cytotoxic chemotherapeutic agent 5-fluorouracil (5-FU) is generally considered to directly kill cancer cells and exert immunostimulatory effects in advanced gastric cancer, accumulating evidence indicates that it upregulates the expression of PD-L1, a representative immune checkpoint blockade molecule involved in negative regulation of the immune response. It was reported that exosomes could transfer functional PD-L1 locally and distantly to suppress the antitumor immune response. However, whether 5-FU alters the expression of exosomal PD-L1 and induces immunosuppression in gastric cancer remains unclear. Herein, we found that 5-FU increased gastric cancer-derived exosomal PD-L1. Importantly, compared with baseline levels, circulating exosomal PD-L1 was significantly upregulated in 21 stage III–IV gastric cancer patients after two, four, and six repeated cycles of fluoropyrimidine treatment (P = 0.009, P = 0.047, and P = 0.023, respectively), accompanied by decreased amounts of IFN-γ, TNF-α, IL-2, IL-6, and GM-CSF (P = 0.014, P = 0.004, P = 0.009, P = 0.031, and P = 0.014, respectively). Additionally, circulating exosomal PD-L1 was increased more significantly in clinical non-responders compared with responders (P = 0.018). Furthermore, exosomal PD-L1 induced apoptosis in Jurkat T cells and inhibited T cell activation in PBMCs, which could be partly reversed by nivolumab. These results suggested that 5-FU-induced upregulation of exosomal PD-L1 causes systemic immunosuppression in advanced gastric cancer following multiple cycles of chemotherapy, especially after two cycles.

Highlights

  • Gastric cancer (GC) is the fifth most prevalent cancer and the third leading cause of cancer-related death worldwide

  • Aliquots (2 × 105) of peripheral blood mononuclear cells (PBMCs) were placed in wells of 96well-plates, and T cells in PBMCs were activated by human CD3/CD28 T cell activator (#10971, Stemcell Technologies) for 6 h at 37◦C and coincubated with MKN74-derived negative control (NC) exosomes or programmed death-ligand 1 (PD-L1)-KD exosomes for 48 h in the presence or absence of nivolumab (Bristol-Myers Squibb Company, Princeton, NJ, USA)

  • Immunotherapy, which is characterized by immune checkpoint blockade (ICB), has changed the standard of care for a subgroup of patients and improved survival compared with previous criteria in several cancer types, even though limitations remain [31, 32]

Read more

Summary

Introduction

Gastric cancer (GC) is the fifth most prevalent cancer and the third leading cause of cancer-related death worldwide. In 2018, GC caused 782,685 deaths [1]. Accumulating evidence indicates that, conventional chemotherapeutic drugs exert direct cytostatic/cytotoxic effects [3], they could alter tumor-reactive. Immunosuppressive Effects of 5-FU in Gastric Cancer immune responses and even display immunosuppressive features [4], which may affect the survival of patients. 5-FU was shown to selectively eliminate tumor-associated MDSCs resulting in enhanced T-celldependent antitumor immunity in several types of cancer [7]. A better understanding of the mechanism by which 5-FU may alter antitumor immune responses in gastric cancer patients, especially immune status changes after different cycles of 5-FU treatment, is important for the improvement of treatment efficacy

Objectives
Methods
Findings
Conclusion
Full Text
Published version (Free)

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