Choice of PD-L1 immunohistochemistry assay influences clinical eligibility for gastric cancer immunotherapy

J Yeong, HYJ Lum, CB Teo, BKJ Tan, YH Chan… - Gastric Cancer, 2022 - Springer
J Yeong, HYJ Lum, CB Teo, BKJ Tan, YH Chan, RYK Tay, JRE Choo, AD Jeyasekharan
Gastric Cancer, 2022Springer
Background Immune checkpoint inhibitors (ICI) are now standard-of-care treatment for
patients with metastatic gastric cancer (GC). To guide patient selection for ICI therapy,
programmed death ligand-1 (PD-L1) biomarker expression is routinely assessed via
immunohistochemistry (IHC). However, with an increasing number of approved ICIs, each
paired with a different PD-L1 antibody IHC assay used in their respective landmark trials,
there is an unmet clinical and logistical need for harmonization. We investigated the …
Background
Immune checkpoint inhibitors (ICI) are now standard-of-care treatment for patients with metastatic gastric cancer (GC). To guide patient selection for ICI therapy, programmed death ligand-1 (PD-L1) biomarker expression is routinely assessed via immunohistochemistry (IHC). However, with an increasing number of approved ICIs, each paired with a different PD-L1 antibody IHC assay used in their respective landmark trials, there is an unmet clinical and logistical need for harmonization. We investigated the interchangeability between the Dako 22C3, Dako 28–8 and Ventana SP-142 assays in GC PD-L1 IHC.
Methods
In this cross-sectional study, we scored 362 GC samples for PD-L1 combined positive score (CPS), tumor proportion score (TPS) and immune cells (IC) using a multiplex immunohistochemistry/immunofluorescence technique. Samples were obtained via biopsy or resection of gastric cancer.
Results
The percentage of PD-L1-positive samples at clinically relevant CPS ≥ 1, ≥ 5 and ≥ 10 cut-offs for the 28–8 assay were approximately two-fold higher than that of the 22C3 (CPS ≥ 1: 70.3 vs 49.4%, p < 0.001; CPS ≥ 5: 29.1 vs 13.4%, p < 0.001; CPS ≥ 10: 13.7 vs 7.0%, p = 0.004). The mean CPS score on 28–8 assay was nearly double that of the 22C3 (6.39 ± 14.5 vs 3.46 ± 8.98, p < 0.001). At the clinically important CPS ≥ 5 cut-off, there was only moderate concordance between the 22C3 and 28–8 assays.
Conclusion
Our findings suggest that scoring PD-L1 CPS with the 28–8 assay may result in higher PD-L1 scores and higher proportion of PD-L1 positivity compared to 22C3 and other assays. Until stronger evidence of inter-assay concordance is found, we urge caution in treating the assays as equivalent.
Springer