Abstract

BackgroundTumor cells express programmed death ligand 1 (PD-L1) and is a key immune evasion mechanism. PD-L1 expression in multiple breast cancer cell lines was evaluated to identify intrinsic differences that affect their potential for immune evasion.MethodsPD-L1 expression was analyzed in six breast cancer cell lines: AU565&MCF7 (luminal), BT20&HCC1143 (basal A), MDA231&HCC38 (basal B). Surface and intracellular PD-L1 expression +/− interferon γ for 48 hours was measured by flow cytometry. PD-L1 gene expression data for all breast cancer cell lines in the Comprehensive Cell Line Encyclopedia (CCLE) was analyzed. Correlation between PD-L1 levels and clinicopathologic parameters was analyzed within Oncomine datasets. A tissue microarray containing 61 invasive breast cancer primary tumor cores was stained for PD-L1 expression and analyzed.ResultsBasal breast cancer cells constitutively express the highest levels of PD-L1. All cell lines increased PD-L1 expression with interferon γ, but basal B cells (MDA-231 and HCC38) demonstrated the largest increases. There were no differences in protein localization between cell lines. In the CCLE data, basal cell lines demonstrated higher mean PD-L1 expression compared to luminal cell lines. High PD-L1 expressing basal cell lines over-express genes involved in invasion, proliferation, and chemoresistance compared to low PD-L1 basal cell lines. High PD-L1 basal cell lines had lower expression of IRF2BP2 and higher STAT1 levels compared to low PD-L1 expressing cell lines. Within Oncomine datasets PDL1 mRNA levels were higher in basal type tumors. The TMA analysis demonstrated that lymph node positive cases had higher levels of PD-L1 protein expression compared to lymph node negative cases.ConclusionsBasal type breast cancer (especially basal B) express greater levels of PD-L1 constitutively and with IFN γ. High PD-L1 basal cells over-express genes involved in invasion, motility, and chemoresistance. Targeting PD-L1 may enhance eradication of aggressive breast cancer cells by the immune system.

Highlights

  • Programmed cell death 1 ligand 1 (PD-L1, CD274, B7-H1) is encoded by the CD274 gene on chromosome nine under the control of an interferon regulatory factor 1 (IRF1) and Signal Transducer Activation of Transcription 1 (STAT1) response elements within its promoter. [1] programmed death ligand 1 (PD-L1) is a 40kDa transmembrane protein that is expressed on a wide variety of normal tissues including natural killer cells, macrophages, myeloid dendritic cells, B cells, epithelial cells, and vascular endothelial cells. [2] Its normal physiologic role is to bind programmed death 1 receptors (PD-1) expressed on the surface of activated cytotoxic T cells

  • This study identified a subset of basal breast cancer cells lines with much higher PD-L1 expression compared to other basal and luminal cell lines

  • Both basal B cell lines had the greatest levels of IFNc inducible PD-L1 expression, especially with HCC38 cells

Read more

Summary

Introduction

Programmed cell death 1 ligand 1 (PD-L1, CD274, B7-H1) is encoded by the CD274 gene on chromosome nine under the control of an interferon regulatory factor 1 (IRF1) and Signal Transducer Activation of Transcription 1 (STAT1) response elements within its promoter. [1] PD-L1 is a 40kDa transmembrane protein that is expressed on a wide variety of normal tissues including natural killer cells, macrophages, myeloid dendritic cells, B cells, epithelial cells, and vascular endothelial cells. [2] Its normal physiologic role is to bind programmed death 1 receptors (PD-1) expressed on the surface of activated cytotoxic T cells. [2] Its normal physiologic role is to bind programmed death 1 receptors (PD-1) expressed on the surface of activated cytotoxic T cells. The expression of PD-L1 has been evaluated in a number of different tumor types including breast cancer.[5,6,7] Ghebeh et al reported that PD-L1 expression was associated with a variety of adverse features such as higher grade, negative estrogen receptor status, and increased infiltration with T regulatory cells. Since our understanding of breast cancer biology has evolved with the advent of genomic classification schemes, it is imperative to understand how PD-L1 behaves within different genomic subtypes as well. This is especially important given the fact that while there is overlap between classical and genomic subtypes PD-L1 expression in multiple breast cancer cell lines was evaluated to identify intrinsic differences that affect their potential for immune evasion

Methods
Results
Discussion
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