The clinicopathological significance and predictive value for immunotherapy of programmed death ligand-1 expression in Epstein-Barr virus-associated gastric cancer

Oncoimmunology. 2021 Jun 17;10(1):1938381. doi: 10.1080/2162402X.2021.1938381. eCollection 2021.

Abstract

The effect of anti-programmed cell death 1 (PD-1) antibody in Epstein-Barr virus-associated gastric cancer (EBVaGC) was debatable, and no predictive biomarkers for efficacy have been reported. Public reports on anti-PD-1 antibody monotherapy-treated EBVaGC with available programmed death ligand-1 (PD-L1) expression status were summarized and analyzed. Relevance with clinicopathologic characteristics of PD-L1 expression by immunohistochemistry was analyzed in 159 patients diagnosed with EBVaGC. Relevance with genomic transcriptome and mutation profile of PD-L1 status in EBVaGC was assessed with three datasets, the cancer genome atlas (TCGA), Gene Expression Omnibus (GEO) GSE51575, and GSE62254. Based on the data from 8 reports, patients with positive PD-L1 expression (n = 30) had significantly superior objective response rate (ORR) than patients with negative PD-L1 expression (n = 9) (63.3% vs. 0%, P = .001) in EBVaGC receiving anti-PD-1 antibody monotherapy. PD-L1 positivity was associated with less aggressive clinicopathological characteristics and was an independent predictor for a longer disease-free survival (hazard ratio [HR] and 95% CI: 0.45 [0.22-0.92], P = .03) and overall survival (HR and 95% CI: 0.17 [0.06-0.43], P < .001). Analysis of public EBVaGC transcriptome and mutation datasets revealed enhanced immune-related signal pathways in PD-L1high EBVaGC and distinct mutation patterns in PD-L1low EBVaGC. PD-L1 positivity indicates a subtype of EBVaGC with 'hot' immune microenvironment, lower aggressiveness, better prognosis, and higher sensitivity to anti-PD-1 immunotherapy.

Keywords: Epstein-Barr virus-associated gastric cancer; Programmed death ligand-1; biomarker; immunotherapy.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • B7-H1 Antigen / genetics
  • Epstein-Barr Virus Infections* / complications
  • Herpesvirus 4, Human / genetics
  • Humans
  • Immunotherapy
  • Stomach Neoplasms* / genetics
  • Tumor Microenvironment

Substances

  • B7-H1 Antigen

Grants and funding

This work was supported by National Key R&D Program of China [2018YFC1313300]; the National Natural Science Foundation of China [82002476]; National Natural Science Foundation of China [81801895]; Guangdong Basic and Applied Basic Research Foundation [2020A1515110034]; Guangdong Basic and Applied Basic Research Foundation [2019A1515110171].