scholarly journals 38 Clinical validation of an image analysis assay for determining programmed death-ligand 1 (22C3) in non-small cell lung cancer

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A39-A39
Author(s):  
Roberto Gianani ◽  
Will Paces ◽  
Elliott Ergon ◽  
Kristin Shotts ◽  
Vitria Adisetiyo ◽  
...  

BackgroundDetermination of programmed death-ligand 1 (PD-L1) level in tumor by immunohistochemistry (IHC) is widely used to predict response to check point inhibitor therapy. In particular, the Dako PD-L1 (22C3) antibody is a common companion diagnostic to the monoclonal antibody drug Keytruda® (pembrolizumab) in non-small cell lung cancer (NSCLC).1 However, for the practicing pathologist, interpretation of the PD-L1 (22C3) assay is cumbersome and time consuming. Manual pathologist scoring also suffers from poor intra- and inter-pathologist precision, particularly around the cut-off point.2 In this clinical validation study, we developed an image analysis (IA) based solution to accurately and precisely score digital images obtained from PD-L1 stained NSCLC tissues for making clinical enrollment decisions.Methods10 NSCLC tissue samples were purchased from a qualified vendor and IHC stained for PD-L1; 4 of these samples had serial sections stained on two separate days. Stained slides were scanned at 20X magnification and analyzed using Flagship Biosciences’ IA solutions that quantify PD-L1 expression and separate tumor and stromal compartments. Resulting image markups of cell detection and PD-L1 expression were reviewed by an MD pathologist for acceptance. PD-L1 staining was evaluated by digital IA in the sample’s tumor compartment for Total Proportion Score (TPS,%). Assay specificity was defined by ≥ 90% of the tissue cohort exhibiting appropriate cell recognition (≥ 90% cells correctly recognized as determined by the pathologist), with ≤ 10% false positive rate for staining classification. Sensitivity was defined by ≥ 90% of the cohort exhibiting appropriate cell identification (≥ 90% cells correctly identified), with ≤ 10% false negative rate for staining classification. Accuracy was defined by the combination of sensitivity and specificity and precision was defined by concordance of the binned TPS (<1%, ≥ 1%, ≥ 50%) in ≥ 80% of the samples stained on multiple days.ResultsThe preliminary results show that IA can yield high analytical sensitivity, specificity, accuracy, and precision in the determination of the PD-L1 score. 100% of the tissue cohort met criteria for analytical specificity, sensitivity, and accuracy and 100% of the samples stained on multiple days met the precision criteria.ConclusionsThis data demonstrates the feasibility of an IA approach as applied to PD-L1 (22C3) scoring. Ongoing experiments include application of the developed 22C3 algorithm on a separate cohort of 20 NSCLC samples to determine the correlation of digital scoring and scoring obtained by three pathologists. Additionally, we will evaluate the precision obtained by digital scoring in relation to the intra- and inter-pathologist concordance.ReferencesIncorvaia L, Fanale D, Badalamenti G, et al. Programmed death ligand 1 (PD-L1) as a predictive biomarker for pembrolizumab therapy in patients with advanced non-small-cell lung cancer (NSCLC). Adv Ther 2019;36:2600–2617.Rimm DL, Han G, Taube JM, et al. A prospective, multi-institutional, pathologist-based assessment of 4 immunohistochemistry assays for PD-L1 expression in non–small cell lung cancer. JAMA Oncol 2017;3:1051–1058.

2021 ◽  
pp. jclinpath-2021-207448
Author(s):  
Jianghua Wu ◽  
Luning Mao ◽  
Wei Sun ◽  
Xin Yang ◽  
Haiyue Wang ◽  
...  

AimsThis study aimed to validate the application of combined multiplex immunofluorescence (mIF) and digital image analysis (DIA) in formalin-fixed and paraffin-embedded tissues for the quantitative assessment of programmed death-ligand 1(PD-L1) and immune cells (ICs) in non-small cell lung cancer (NSCLC).MethodsFifty resected samples of NSCLC were sequentially stained with a DNA-tagged mIF (panel including PD-L1, CKpan, CD8, CD68 and 4′,6-diamidino-2-phenylindole (DAPI)) and conventional immunohistochemistry (cIHC). The assessment of cell density and consistency of tumour proportion score (TPS) via DIA were compared with those by pathologists.ResultsA strong correlation in the cell population of immune markers was obtained between mIF and cIHC (for PD-L1: R=0.9304, CKpan: R=0.8231, CD8: R=0.9314 and CD68: R=0.8366) within 95% limits of agreement. The continuous TPS calculated using mIF was highly consistent with the IHC staining results which were evaluated by pathologists (R=0.9362). However, in the comparison of TPS using interval variables, a poor agreement was obtained at a cut-off of 1% (κ=0.197), whereas excellent agreement was achieved at cut-offs of 50% (κ=0.908) and 5% (κ=0.823). DIA on mIF showed that PD-L1 commonly colocalised with CD68+ macrophages and CD8+ cytotoxic cells were closer to PD-L1-/CK+ tumour cells (TCs) than to PD-L1+/CK+ TCs in spatial distribution.ConclusionsA combination of mIF and DIA is useful for the quantification of PD-L1 expression and IC populations in NSCLC. Further validation of TPS at a cut-off of 1% and assay harmonisation is essential for translating this method in a diagnostic setting.


2019 ◽  
Vol 36 (3) ◽  
pp. 184-189
Author(s):  
Songül Şahin ◽  
Şebnem Batur ◽  
Övgü Aydın ◽  
Tülin Öztürk ◽  
Akif Turna ◽  
...  

2016 ◽  
Vol 140 (4) ◽  
pp. 341-344 ◽  
Author(s):  
Lynette M. Sholl ◽  
Dara L. Aisner ◽  
Timothy Craig Allen ◽  
Mary Beth Beasley ◽  
Alain C. Borczuk ◽  
...  

The binding of programmed death ligand-1 and ligand-2 (PD-L1 and PD-L2) to PD-1 blocks T-cell–mediated immune response to tumor. Antibodies that target programmed death receptor-1 (PD-1) will block the ligand-receptor interface, thereby allowing T cells to attack the tumor and increase antitumor immune response. In clinical trials, PD-1 inhibitors have been associated with an approximately 20% overall response rate in unselected patients with non–small cell lung cancer, with sustained tumor response in a subset of patients treated by these immune checkpoint inhibitors. Facing a proliferation of PD-L1 immunohistochemistry clones, staining platforms, and scoring criteria, the pathologist must decide on the feasibility of introducing a newly approved companion diagnostic assay that may require purchase not only of a specific antibody kit but of a particular staining platform. Given the likely reality that clinical practice may, in the near future, demand access to 4 different PD-L1 antibodies coupled with different immunohistochemistry platforms, laboratories will be challenged with deciding among this variety of testing methods, each with its own potential benefits. Another immediate challenge to PD-L1 testing in lung cancer patients is that of access to adequate tumor tissue, given that non–small cell lung cancer samples are often extremely limited in size. With PD-L1 testing it has become clear that the historically used US regulatory approach of one assay–one drug will not be sustainable. One evolving concept is that of complementary diagnostics, a novel regulatory pathway initiated by the US Food and Drug Administration, which is distinct from companion diagnostics in that it may present additional flexibility. Although pathologists need to face the practical reality that oncologists will be asking regularly for the PD-L1 immunohistochemistry status of their patients' tumors, we should also keep in mind that there may be room for improvement of biomarkers for immunotherapy response. The field is rich with opportunities for investigation into biomarkers of immunotherapy response, particularly in the form of collaborative, multidisciplinary studies that incorporate oncologists, pathologists, and basic scientists. Pathologists must take the lead in the rational incorporation of these biomarkers into clinical practice.


Sign in / Sign up

Export Citation Format

Share Document