Abstract

Four immunohistochemistry (IHC) diagnostic assays have been approved for tumour PD-L1 protein assessment in the clinic. However, mRNA detection by in situ hybridisation (ISH) could be utilised as an alternative to protein detection. Detecting spatial changes in gene expression provides vital prognostic and diagnostic information, particularly in immune oncology where the phenotype, cellular infiltration and immune activity status may be associated with patient survival. Translation of mRNA expression to a clinically relevant cut off or threshold is challenging due to variability between assays and the detection of different analytes. These studies aim to confirm the suitability of formalin fixed paraffin embedded (FFPE) tissue sections for use with RNA ISH. A comparison of mRNA expression and protein expression may inform the suitability of mRNA as a patient selection biomarker in a similar manner to IHC and provide evidence of a suitable scoring algorithm. Ninety patient samples, thirty for each indication of non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC) and urothelial carcinoma (UC), previously assessed using the VENTANA PD-L1 (SP263) Assay were chosen to represent a wide dynamic range of percentage tumour cell staining (TCIHC). Expression of mRNA was assessed by ISH using the RNAScope 2.5 assay and probe CD274/PD-L1 (Advanced Cell Diagnostics) including kit provided positive and negative control probes. Brightfield whole slide images of tissues were captured. The percentage of tumour cells with PD-L1 mRNA expression (%TCmRNA) and mean punctate dots/tumour cell were determined using image analysis. Differences in RNA expression between the IHC derived TCIHC≥25% and <25% groups were assessed using t-tests. For each indication, a receiver-operating characteristic (ROC) analysis identified thresholds for patient classification using %TCmRNA and dots/tumour cell, with reference to TCIHC≥25%. Eighty-six samples were successfully tested; 3 failed due to insufficient control probe staining, 1 due to lack of tumour. Percent TCmRNA staining using RNAScope demonstrated statistical significance (at α = 0.05) in the PD-L1 high (TCIHC ≥25%) vs the PD-L1 low (TCIHC <25%) groups for NSCLC, HNSCC, and UC. The number of punctate dots/tumour cell was significantly higher in the PD-L1 high vs the PD-L1 low groups for NSCLC and HNSCC but not UC. For %TCmRNA; ROC analysis identified thresholds of: NSCLC 18.0%, HNSCC 31.8%, UC 25.8%. For dots/tumour cell, thresholds were: NSCLC 0.26, HNSCC 0.53, UC 0.45. Routine tissue fixation and processing is suitable for RNA detection using RNAScope. PD-L1 mRNA extent and level is associated with PD-L1 status determined by IHC. Threshold optimisation for %TCmRNA and mean dots/tumour cell results in high specificity to IHC PD-L1 classification, but only moderate sensitivity.

Highlights

  • Programmed cell death ligand 1 (PD-L1) is part of a complex system of receptors and ligands that are involved in controlling T cell activation

  • Ninety (90) formalin fixed paraffin embedded (FFPE) cases from non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC) and urothelial carcinoma (UC) were assessed for analysis with RNAScope

  • Tissue for this study represented a variety of indications and sample acquisition methods which routine laboratories may encounter. mRNA was quantifiable in 96% of cases, demonstrating, as has been shown previously, that routine tissue processing is suitable for the preservation of RNA [18]. mRNA was not quantifiable in 4% of cases due to the absence of tumour in one case and the absence of positive control probe staining in 3 cases

Read more

Summary

Introduction

Programmed cell death ligand 1 (PD-L1) is part of a complex system of receptors and ligands that are involved in controlling T cell activation. PD-L1 acts at multiple sites in the body to help regulate immune responses by delivering inhibitory signals to T cells through the programmed cell death 1 (PD-1) and cluster of differentiation (CD) 80 receptors. The binding of the ligand PD-L1 to its receptor PD-1 on activated T cells delivers an inhibitory signal to the T cells, preventing them from killing target tumour cells, and protecting the tumour from immune elimination [3]. The binding of an anti-PD-L1 therapeutic agent to the PD-L1 receptor inhibits the interaction of PD-L1 with the PD-1 and CD80 receptors expressed on immune cells; restoring the host immune response, a mechanism which has been extensively reviewed [5, 6]. The activity of PD-1/PD-L1 therapy overcomes PD-L1-mediated inhibition of antitumor immunity

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