Abstract

There is currently no reliable biomarker to predict who would benefit from anti-PD-1/PD-L1 inhibitors. We comprehensively analyzed the immunogenomic properties in The Cancer Genome Atlas (TCGA) according to the classification of tumor into four groups based on PD-L1 status and tumor-infiltrating lymphocyte recruitment (TIL), a combination that has been suggested to be a theoretically reliable biomarker of anti-PD-1/PD-L1 inhibitors. The RNA expression levels of PD-L1 and CD8A in the samples in the pan-cancer database of TCGA (N = 9,677) were analyzed. Based on their median values, the samples were classified into four tumor microenvironment immune types (TMIT). The mutational profiles, PD-L1 amplification, and viral association of the samples were compared according to the four TMITs. The proportions of TMIT I, defined by high PD-L1 and CD8A expression, were high in lung adenocarcinoma (67.1%) and kidney clear cell carcinoma (64.8%) among solid cancers. The number of somatic mutations and the proportion of microsatellite instable-high tumor in TMIT I were significantly higher than those in other TMITs, respectively (P < 0.001). PD-L1 amplification and oncogenic virus infection were significantly associated with TMIT I, respectively (P < 0.001). A multivariate analysis confirmed that the number of somatic mutations, PD-L1 amplification, and Epstein-Barr virus/human papillomavirus infection were independently associated with TMIT I. TMIT I is associated with a high mutational burden, PD-L1 amplification, and oncogenic viral infection. This integrative analysis highlights the importance of the assessment of both PD-L1 expression and TIL recruitment to predict responders to immune checkpoint inhibitors. Clin Cancer Res; 22(9); 2261-70. ©2016 AACRSee related commentary by Schalper et al., p. 2102.

Highlights

  • A recent strategy targeting immune checkpoints such as CTLA-4 and PD-1/PD-L1 shows promising clinical benefits and introduces a paradigm shift in cancer treatment

  • A multivariate analysis confirmed that the number of somatic mutations, PD-L1 amplification, and Epstein–Barr virus/human papillomavirus infection were independently associated with tumor microenvironment immune types (TMIT) I

  • TMIT I is associated with a high mutational burden, PD-L1 amplification, and oncogenic viral infection

Read more

Summary

Introduction

A recent strategy targeting immune checkpoints such as CTLA-4 and PD-1/PD-L1 shows promising clinical benefits and introduces a paradigm shift in cancer treatment. Recent advances in immuno-genomics have shown that tumors with a high mutational burden, abundant neoantigen, and microsatellite (MSI)-high status are associated with a good response to anti-PD-1/PD-L1 therapy [8,9,10,11,12,13,14]. Oncogenic viruses such as Epstein–Barr virus (EBV) or human papillomavirus (HPV) are associated with an inflamed TME, and a favorable clinical outcome in response to anti-PD-1/PD-L1 therapy would be expected [10, 15]. Because of the complex nature of tumor immunity, a comprehensive immuno-genomic analysis that include analyses of the interaction between the tumor and TIL in the TME and investigations of the underlying reasons for the promotion of tumor immunogenicity is needed

Objectives
Methods
Results
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