PD-L1 expression in Epstein-Barr virus-infected gastric cancers.

Authors

null

Sarah Derks

dana-farber cancer institute, Boston, MA

Sarah Derks , Xiaoyun Liao , Xinsen Xu , M. Constanza Camargo , Anna M Chiaravalli , Enrico Solcia , Tania Fleitas , Gordon J. Freeman , Scott J. Rodig , Charles S. Rabkin , Adam Joel Bass

Organizations

dana-farber cancer institute, Boston, MA, Dana-Farber Cancer Institute, Center for Immuno-Oncology, Boston, MA, Dana-Farber Cancer Institute, Boston, MA, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, Dept. of Pathology, Ospedale di Circolo, Varese, Italy, Dept. of Molecular Medicine, University of Pavia and Policlinico S. Matteo, Pavia, Italy, Department of Pathology, Division of Hematopathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA

Research Funding

Other Foundation

Background: Gastric cancer (GC) is a deadly disease with limited treatment options. Recent studies with PD-1 inhibitor Pembrolizumab have shown promising results in GC. Key questions remain which GC subclass may respond best. In a comprehensive molecular analysis of gastric adenocarcinomas as part of The Cancer Genome Atlas (TCGA) we identified that 15% of Epstein Barr Virus (EBV)+ GCs have amplification of chromosomal region 9p24.1, the locus of PD-1 ligands PD-L1 and PD-L2. As EBV+ cancers have multiple mechanisms to induce PD-L1 expression, we hypothesize that PD-L1 status may differ specifically in EBV+ GCs, regardless of 9p24.1 status, indicating a broader potential role for PD-1 blockade in this GC subtype. Methods: PD-L1 expression was analyzed with immunohistochemistry (IHC) using whole tissues slides of EBV+ (n = 32) and EBV negative (n = 49) GCs. The transcriptional landscape of EBV+ and EBV negative GCs was determined using mRNA sequencing results from the TCGA study. Results: With IHC we showed that EBV+ GCs have PD-L1 expression in tumor and immune cells in 50% (16/32) and 94% (30/32) of GCs respectively. This in contrast to EBV negative GCs that have PD-L1+ tumor and immune cells in only 10% (5/49) and 39% (19/49) of cases respectively (P< 0.001). Furthermore, PD-L1+ immune cells infiltrated the center of the majority of EBV+ GCs, while in EBV negative GCs PD-L1+ immune cells remained at the invasive margin (P = 0.007). mRNA sequencing analyses of 12 EBV+ GCs and 10 EBV- GCs showed that PD-L1+/EBV+ GCs have enrichment of an interferon-γ driven gene signature. Single-sample gene set enrichment analyses on a bigger series of 214 individual GCs demonstrated that besides EBV+ GCs (n = 19), microsatellite instable (MSI) GCs (n = 47) have strong enrichment of IFN-γ response gene expression, in contrast to genomic stable (GS) (n = 42) and chromosomal instable (CIN) (n = 106) gastric cancer subtypes (P< 0.001). Conclusions: As the presence of PD-L1+ tumor and/or tumor infiltrating immune cells as well as an interferon-mediated adaptive immune response are associated with PD-1 blockade vulnerability in other cancers, we here provide a strong rationale for testing of PD-1 blockade in EBV+ GC and for evaluating EBV and MSI status as variables for response.

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2016 ASCO Annual Meeting

Session Type

Poster Session

Session Title

Gastrointestinal (Noncolorectal) Cancer

Track

Gastrointestinal Cancer—Gastroesophageal, Pancreatic, and Hepatobiliary

Sub Track

Esophageal or Gastric Cancer

Citation

J Clin Oncol 34, 2016 (suppl; abstr 4052)

DOI

10.1200/JCO.2016.34.15_suppl.4052

Abstract #

4052

Poster Bd #

44

Abstract Disclosures

Similar Abstracts

First Author: Hidekazu Hirano

First Author: Sun Mi Lee

Abstract

2022 ASCO Annual Meeting

Pan-cancer landscape of CD274 (PD-L1) and PDCD1LG2 (PD-L2) structural variations.

First Author: Emily Louise Hoskins