Abstract
PURPOSE
Four programmed death ligand 1 (PD-L1) immunohistochemistry assays (28-8, 22C3, SP263, and SP142) have been approved for use by the US Food and Drug Administration (FDA). Analytical concordance between these assays has been evaluated in multiple studies. This systematic review included studies that investigated the analytical concordance of immunohistochemistry assays utilizing two or more PD-L1 antibodies from FDA-approved diagnostics for evaluation of PD-L1 expression on tumor or immune cells across a range of tumor types and algorithms.
METHODS
Literature searches were conducted in MEDLINE (via PubMed) and EMBASE to identify studies published between January 1, 2010, and March 31, 2019, that evaluated analytical concordance between two or more assays based on antibodies from FDA-approved assays. Proceedings of key oncology and pathology congresses that took place between January 2016 and March 2019 were searched for abstracts of studies evaluating PD-L1 assay concordance.
RESULTS
A total of 42 studies across a range of tumor types met the selection criteria. Concordance between 28-8-, 22C3-, and SP263-based assays in lung cancer, urothelial carcinoma, and squamous cell carcinoma of the head and neck was high when used to assess PD-L1 expression on tumor cells (TCs). SP142-based assays had overall low concordance with other approved assays when used to assess PD-L1 expression on TCs. Analytical concordance for assessment of PD-L1 expression on immune cells was variable and generally lower than for PD-L1 expression on TCs.
CONCLUSION
A large body of evidence supports the potential interchangeability of 28-8-, 22C3-, and SP263-based assays for the assessment of PD-L1 expression on TCs in lung cancer. Further studies are required in tumor types for which less evidence is available.
INTRODUCTION
Programmed death-1 (PD-1) and programmed death ligand 1 (PD-L1) inhibitors have been approved in the United States and globally for the treatment of a range of tumor types. PD-(L)1 inhibitors approved by the US Food and Drug Administration (FDA) include atezolizumab, avelumab, cemiplimab, durvalumab, nivolumab, and pembrolizumab.1-6 PD-L1 expression on tumor cells (TCs) and immune cells (ICs) is a mechanism of tumor immune escape through engagement and activation of the PD-1 receptor.7,8 The expression of PD-L1 on TCs or ICs is associated with enhanced response to PD-(L)1 inhibitor therapy in some tumor types.7 As of December 2020, four PD-L1 diagnostic immunohistochemistry (IHC) assays have been approved by the US FDA for assessment of PD-L1 expression on TCs or ICs in clinical practice (Table 1).
TABLE 1.
CONTEXT
Key Objective
Evaluate analytical concordance between programmed death ligand 1 (PD-L1) immunohistochemistry assays utilizing antibodies from US Food and Drug Administration–approved diagnostics across a range of tumor types, scoring algorithms, and PD-L1 expression cutoffs.
Knowledge Generated
Analytical concordance between 28-8-, 22C3-, and SP263-based assays was high when used to assess PD-L1 expression on tumor cells (TCs) in lung cancer, urothelial carcinoma, and squamous cell carcinoma of the head and neck. SP142-based assays had low concordance with other assays for assessment of PD-L1 expression on TCs. Analytical concordance for assessment of PD-L1 expression on immune cells was variable and generally lower than for PD-L1 expression on TCs.
Relevance
As the immune checkpoint inhibitor treatment landscape continues to become increasingly complex, PD-L1 assay analytical concordance, in context with data on the predictive performance, sensitivity, and specificity of assays, informs decisions around assay choice and interpretation.
PD-L1 assay approvals are specific to the tumor types and therapeutic regimens for which the FDA authorizes their use and are variable with regard to the scoring algorithms used and the cell types on which PD-L1 expression is evaluated (ie, TCs, ICs, or both). Currently, there is a lack of data supporting assay harmonization. Not all laboratories can provide multiple PD-L1 assays corresponding to the approved indication for several reasons, including high cost or limited access to IHC staining platforms. Consequently, not having the approved assay may hinder PD-L1 testing and/or result interpretation and potentially a physician's recommendation for treatment guidance. Defined assay performance criteria are critical to guide pathologists and oncologists in identifying the most appropriate assay for an intended use and for interpreting test results. A variety of factors should be incorporated into such decisions, including analytical concordance, predictive performance, and the sensitivity and specificity of available assays around the relevant clinical cutoffs.9
Three previous literature reviews have evaluated PD-L1 assay concordance in lung cancers. A review by Büttner et al10 found high concordance and reproducibility for assessment of PD-L1 expression on TCs in non–small-cell lung cancer (NSCLC) with the 28-8, 22C3, and SP263 assays, while the detection of PD-L1 expression on TCs with the SP142 assay was lower than with other assays.10 There was poor concordance between assays when measuring PD-L1 expression on ICs.10 Similar findings were reported in a review by Udall et al,11 which found that the 28-8, 22C3, and SP263 assays produced comparable results when used to evaluate PD-L1 expression on TCs. However, the authors concluded that there was a lack of standardization among PD-L1 assays in terms of expression cutoffs and scoring algorithms, and that information on the interchangeability of PD-L1 assays was limited.11 PD-L1 assay interchangeability was further evaluated in a meta-analysis of PD-L1 assay concordance by Torlakovic et al,9 which concluded that FDA-approved assay kits were generally more interchangeable with a well-developed, fit-for-purpose, laboratory-developed test (LDT) than with another FDA-approved kit developed for a different purpose.
This systematic review was undertaken to update previous literature reviews, with the goal of assessing analytical concordance between assays utilizing antibodies from FDA-approved diagnostics for assessment of PD-L1 expression on TCs and/or ICs across a range of tumor types, algorithms, and PD-L1 expression cutoffs.
METHODS
The methodology of this study adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines.12 Systematic searches were conducted in MEDLINE (via PubMed) and EMBASE (Elsevier) to identify studies published between January 1, 2010, and March 31, 2019, that evaluated concordance between two or more assays based on antibodies from FDA-approved diagnostics. The search string employed was (“PD-L1” OR “Programmed death ligand 1” OR “PDL1” OR “Programmed death ligand”) AND (“IHC” OR “immunohistochemistry”) AND (concordance OR validation OR sensitivity OR specificity OR correlat* OR reproducib* OR valid* OR agree*). Search results were limited to English-language publications only.
Proceedings of key oncology and pathology congresses that took place between January 1, 2016, and March 31, 2019, were searched for abstracts of studies evaluating concordance between assays utilizing antibodies from FDA-approved diagnostics. The congresses searched were the annual meetings of the American Association for Cancer Research (AACR), the Association for Molecular Pathology (AMP), the American Society of Clinical Oncology (ASCO), the College of American Pathologists (CAP), the European Society for Medical Oncology (ESMO), the Society for Immunotherapy of Cancer (SITC), and the United States and Canadian Academy of Pathology (USCAP). The ASCO Gastrointestinal Cancers Symposium (ASCO GI), the ASCO Genitourinary Cancers Symposium (ASCO GU), the ASCO-SITC Clinical Immuno-Oncology Symposium, the European Congress of Pathology (ECP), and the International Association for the Study of Lung Cancer World Conference on Lung Cancer (IASLC WCLC) were also searched.
The study inclusion and exclusion criteria are shown in Table 2. Publications and congress abstracts were compiled, and duplicates were removed manually. Congress abstracts were checked for subsequent publication, with those abstracts that had been subsequently published as full manuscripts excluded. Publications and abstracts were initially screened against two key inclusion criteria: evaluation of PD-L1 expression with at least two assays utilizing antibodies from FDA-approved diagnostics and evaluation of comparability or concordance between two or more assays. Publications meeting these criteria were read in full and scored against the remaining selection criteria. Studies that explicitly stated that assays were performed using materials and equipment other than those specified in the manufacturers' instructions (ie, an LDT) were excluded. Studies that did not explicitly state whether the FDA-approved assay or an LDT was used were assumed to have met the inclusion criteria. In addition, studies that evaluated analytical concordance in a tumor type for which the assay is not approved were included. Disagreements were resolved by majority opinion of the reviewing authors. A standardized template was used to extract key information, including the type of study, location, number of patients or samples evaluated, tumor types included, antibodies or tests used, PD-L1 scoring algorithms used, cell types assessed for PD-L1 expression, key concordance and agreement statistics, and training status of the scoring pathologist. Key data from the identified studies were analyzed descriptively with the aim of identifying trends in concordance statistics. Studies reporting concordance or agreement frequently qualified their results subjectively. Therefore, there is no convention for descriptive reporting of concordance between assays. To assist with evaluation, data were grouped into the following subjective categories: poor, fair, and strong. Concordance was described as poor for k values ≤ 0.4, fair for k values > 0.4 to < 0.7, and strong for k values ≥ 0.7; agreement was described as poor for overall percentage agreement (OPA) ≤ 60%, fair for OPA > 60% to < 75%, and strong for OPA ≥ 75%; correlation was described as poor for Pearson correlation coefficient (r2) and Spearman correlation coefficient (ρ) for r2 and ρ ≤ 0.6, fair for r2 and ρ > 0.60 to < 0.85, and strong for r2 and ρ ≥ 0.85. Meta-analyses or other statistical analyses of the results were not performed because of the heterogeneity of the studies identified in the search.
TABLE 2.
RESULTS
Part I: Screening of Reports and Studies' Details
Searches of MEDLINE (PubMed) and EMBASE identified 819 and 2,203 records, respectively, published between January 1, 2010, and March 31, 2019. Manual searches of the proceedings of key congresses identified an additional 521 abstracts presented between January 1, 2016, and March 31, 2019. A total of 3,477 unique records were screened against the two key inclusion criteria, of which 97 met both criteria and were reviewed in detail by the authors. A total of 42 publications and abstracts met all inclusion and exclusion criteria and were included in the review. Included and excluded manuscripts and abstracts, as well as the number of studies evaluating each assay by tumor type, are shown in Figure 1. Details of the studies included in the review are summarized in Appendix Table A1.
Part II: Analytical Concordance in Studies Assessing PD-L1 Expression on TCs Only
Analytical concordance for assessment of PD-L1 expression on TCs only using 28-8-, 22C3-, SP142-, and SP263-based assays is shown in Table 3. The training status of the pathologists was reported in six studies and was not specified in the remaining studies. The details of pathologist training were not reported in enough studies to enable an assessment of the impact of pathologist training on analytical concordance.
TABLE 3.
Data from studies in which PD-L1 expression agreement was assessed across multiple cutoffs suggested a trend for higher agreement with increasing cutoff in lung cancer and squamous cell carcinoma of the head and neck (SCCHN).13-16 However, none of the studies formally evaluated the changes in analytical concordance across PD-L1 expression cutoffs.
Overall, concordance between 28-8-, 22C3-, and SP263-based assays in lung cancer, urothelial carcinoma (UC), and SCCHN was high when used to assess PD-L1 expression on TCs. SP142-based assays had overall low concordance with other approved assays when used to assess PD-L1 expression on TCs.
Lung cancer
Most studies reported concordance results in NSCLC only, but a number of studies reported results across heterogeneous types of lung tumors, including small-cell lung cancer and NSCLC, or did not specify the types of lung tumors included. Across studies that evaluated 28-8-based assays, generally strong analytical concordance was seen with 22C3-based assays15,17-23 and fair-to-strong analytical concordance with SP263-based assays.15,18,24,25 Analytical concordance between 22C3- and SP263-based assays was variable across studies.15,16,18,20,26-31 In one study in which a six-category integrated proportion score was used to evaluate PD-L1 expression on TCs, a higher proportion of PD-L1–positive TCs was seen with both 22C3- and SP263-based assays versus 28-8-based assays in nonconcordant samples, and a higher proportion of TCs were stained with SP263-based assays versus 22C3-based assays.32
SP142-based assays showed generally poor-to-fair analytical concordance with 28-8-,15,25 22C3-,15,20,33-35 and SP263-based assays15,25,36-38 for assessment of PD-L1 expression on TCs, with nonconcordant cases showing stronger staining with comparator assays than with SP142-based assays.32
In the Blueprint studies, generally comparable distribution of TC staining with 28-8-, SP263-, and 22C3-based assays was seen across a series of samples.39,40 In Blueprint phase 1, SP142-based assays showed weaker staining of TCs and fewer positive TCs compared with other assays, while Blueprint phase 2 also found SP142 to have lower sensitivity for detection of PD-L1 expression on TCs than other assays. Although statistical analyses were performed in these studies, formal statistics for comparisons between assays have not been published.39,40
Nonlung tumor types
Analytical concordance data for TC scoring were limited in most nonlung tumor types. With the exception of SCCHN and melanoma, the majority of tumor types had a single publication. Generally strong agreement or concordance between 28-8-, 22C3-, and/or SP263-based assays was seen in breast cancer,41 melanoma,42 malignant pleural mesothelioma,43 SCCHN,13,14 thymic carcinoma,44 and UC.45 Analytical concordance for comparisons including SP142-based assays was variable, with fair-to-strong concordance and agreement with 22C3- or SP263-based assays in SCCHN and strong concordance in B- or T-cell lymphoma and thymic carcinoma.13,14,44,46
Multiple tumor types
Strong concordance between 28-8- and 22C3-based assays was observed in two real-world studies evaluating concordance for TC scoring across samples from multiple tumor types.17,47 In a third study, strong agreement was also seen between 22C3- and SP142-based assays in samples from multiple tumor types, with OPAs of 95%-100%; however, this study was published as a research letter, and information on the assay methodology used was limited.48
Part III: Analytical Concordance in Studies Assessing PD-L1 Expression on ICs or Combined ICs and TCs
Assay concordance in studies where evaluation of PD-L1 expression with 28-8-, 22C3-, SP263-, and SP142-based assays included ICs is shown, with algorithm definitions, in Table 4. Only one study reported on the training status of pathologists.
TABLE 4.
Analytical concordance for assessment of PD-L1 expression on ICs was variable and generally lower than for PD-L1 expression on TCs.
Lung cancer
Most studies reported concordance results in NSCLC only, but a number of studies reported results across heterogeneous types of lung tumors, including small-cell lung cancer and NSCLC, or did not specify the types of lung tumors included. Agreement and concordance for IC scoring was generally high between 28-8- and SP263-based assays25,37 and between 22C3- and SP263-based assays,37 although the number of studies where these assays and algorithms were compared was small. There were no studies directly comparing the 28-8- and 22C3-based assays using IC scoring. Generally poor concordance between 22C3- and SP142-based assays for scoring of ICs or combined ICs and TCs was seen in three studies in lung cancer.20,27,34 In separate studies, analytical concordance between SP142- and SP263-based assays for IC scoring was poor to fair,25,38 and no studies compared 28-8- and SP142-based assays using IC scoring, aside from the Blueprint studies.
In the Blueprint studies, IC staining was generally comparable between 28-8-, 22C3-, and SP263-based assays.39,40 Staining with an SP142-based assay was less sensitive than with 28-8-, 22C3-, or SP263-based assays.39,40 As with concordance analyses in TCs, formal statistics for comparisons between assays were not presented in the publications from the Blueprint studies.
Nonlung tumor types
As was the case for analytical concordance for TC scoring in nonlung tumor types, data were limited for IC scoring in nonlung tumor types. Single studies were identified in most tumor types, with the exception of three studies in UC. Among nonlung tumor studies, only one study in thymic carcinoma and one study in UC reported formal concordance statistics. In thymic carcinoma, strong concordance for IC scoring was seen between 28-8- and 22C3-based assays, whereas poor-to-fair analytical concordance for IC scoring was seen between all other possible combinations of 28-8-, 22C3-, SP263-, and SP142-based assays.44 In UC, concordance for IC scoring between SP142- and SP263-, 22C3- and SP263-, and 22C3- and SP142-based assays was generally poor to fair, and higher concordance between assays was reported with TC scoring than with IC scoring.49
Multiple tumor types
Concordance between SP263- and SP142-based assays for TC or IC scoring was poor in a cohort of patients with various tumor types.50
DISCUSSION
This systematic review identified 42 studies that assessed concordance between assays utilizing antibodies from FDA-approved diagnostics across a range of tumor types. Concordance between PD-L1 assays was most frequently evaluated in lung cancer, particularly NSCLC, reflecting the approval of multiple PD-(L)1 inhibitors and associated companion or complementary PD-L1 diagnostic assays across multiple treatment lines for the treatment of advanced lung cancers,51-53 the relatively early approval of PD-L1 assays in NSCLC compared with other tumor types,54 and the high incidence of lung cancers compared with other cancers in which PD-(L)1 inhibitors and PD-L1 assays have been approved.55 The combination of these factors would be expected to lead to greater interest in the analytical concordance between assays in lung cancer, as well as make it the tumor type of choice for concordance studies because of the higher absolute number of cases and widespread use of PD-L1 testing.
The current review was designed to focus on interassay concordance data, but a number of studies identified by the literature search also evaluated interobserver variability and concordance between sample types (eg, resections, core needle or bronchial biopsy samples, tumor-positive lymph node excision biopsy or resection samples, and cytology specimens).39,40 These studies used a variety of designs and assessment measures to investigate the contribution of these factors to PD-L1 test variability. Concordance between pathologists, centers, and sample types has been examined extensively in previous reviews of the literature.10,11,56 Interobserver reproducibility is generally good for assessment of PD-L1 expression on TCs but is variable for assessment of PD-L1 expression on ICs because of a range of factors, including assessment of both cytoplasmic and cell membrane staining of ICs and scoring of percentage area staining rather than the percentage of PD-L1–positive cells.10,11,32 Sample types may also play an important role in concordance between tests, with limited available data suggesting generally good concordance between cytology specimens and tumor tissue, as well as between core biopsy samples and surgical specimens.10 Studies evaluating concordance between original diagnostic material and newly acquired tissue and evaluating the effects of intertumoral and intratumoral heterogeneity also suggest that these factors can affect reproducibility of PD-L1 assessment.10,57 A possible limitation of this review is the absence of the assessment of the impact that these factors may have on concordance results. Variation in assay methodology across the included studies may represent another possible limitation, despite efforts to exclude studies that did not use assay manufacturers' specified materials and methods.
Concordance between 28-8-, 22C3-, and SP263-based assays was generally high for assessment of PD-L1 expression on TCs in tumor types for which one or more assays have been approved. Of note, there is a sizable body of evidence for generally high concordance between assays in lung cancers, reflecting pathologists' level of experience and supporting the potential interchangeability of approved assays in this tumor type. High concordance was also seen for TC and IC scoring in SCCHN and for TC scoring in UC, although more data are needed to allow comprehensive evaluation of analytical concordance in these tumor types. Data from studies in which PD-L1 expression agreement was assessed across multiple cutoffs suggested a trend for higher agreement with increasing cutoff, possibly because of variability in pathologist assessment at low expression levels.13,15,58 Adequately powered studies are required to confirm this observation. It is important to note that analytical concordance alone is insufficient to guide decisions around assay choice. Clinicians and pathologists should place these results in context with relevant data on assay predictive performance, sensitivity, and specificity, as well as performance around relevant clinical cutoffs, when making decisions for their laboratory and clinical practices and selecting treatment.
The causative factors for the low staining intensity obtained with SP142-based assays compared with other assays remain unclear but are suggested to be the result of differences in assay methodology rather than variation in the epitope binding site targeted by each antibody clone.59 Agreement between SP142-based assays and other assays was higher for TC scoring in lymphomas and thymic carcinoma and IC scoring in renal cell carcinoma than in other tumor types. However, the study comparing SP142- and SP263-based assays in B- and T-cell lymphomas included only 78 samples,46 whereas the study in thymic carcinoma included only 53 samples.44 Similarly, the study with renal cell carcinoma specimens had a small sample size (n = 32), and the reported results were limited to overall agreement at an unknown PD-L1 expression cutoff.60 The results of these studies should be confirmed in larger studies of concordance between the SP142 assay and other assays.
Assessment of PD-L1 expression on ICs generally showed lower concordance than TC scoring across all assays and tumor types evaluated. Reduced concordance for IC scoring may be related to greater subjectivity when interpreting IC staining compared with TC staining, due to the small size of ICs, ultimately reflected in the high interobserver variability reported.32,39,40 A relative lack of pathologist experience with IC scoring compared with TC scoring and less methodological standardization of IC scoring may also contribute to reduced concordance.32,39,40
Pembrolizumab has been approved for the treatment of PD-L1–expressing gastric cancer, cervical cancer, UC, esophageal squamous cell carcinoma, triple-negative breast cancer, and SCCHN based on assessment with the PD-L1 combined positive score (CPS) algorithm.6 One study in UC and one study in SCCHN identified in this systematic review assessed interassay concordance using the PD-L1 CPS algorithm, both of which found generally similar concordance to that seen with % TC scoring and/or % IC scoring.14,61 In those two studies, the training status of the pathologist and, hence, any potential impact on concordance were not reported. Reproducibility of scoring between pathologists appeared to be higher with the CPS algorithm than with the mononuclear IC density score using a 22C3-based assay in UC specimens.62 Future studies are needed to assess analytical concordance using the CPS algorithm across multiple tumor types and assays.
Although most of the studies included here did not report whether pathologists received specific training, it is reasonable to assume that pathologists participating in the cited studies completed training, since assay-specific training is frequently provided for pathologists.10 Effective training is an important part of efforts to improve scoring accuracy, which may be reflected in higher levels of concordance seen in lung tumors and for TC-based scoring methods. At present, it is unclear if pathologist training improves interassay concordance, but several studies have found strong concordance between observers in studies where practical training was required.10,32,63-67 Given the comparatively low concordance for IC scoring seen in this review and the likely introduction of scoring systems that incorporate ICs, such as CPS, in a wider range of tumor types in the future, it is important that effective training is put in place to aid in consistent and accurate interpretation. As well as supporting standardized assay interpretation, training should educate pathologists on how to approach tumor-specific challenges, such as scoring of PD-L1 staining in tumors with heterogeneous morphology.10 Provision of tumor type–specific training is another important consideration, as pathologists' familiarity with the tissue structures and cell types present in a sample is important for assessment of PD-L1 expression.10
Greater uptake of digital pathology might also promote a shift toward centralization of test interpretation by pathologists with subspecialty expertise.68 Adoption of artificial intelligence–based assessment may also improve the reproducibility of test results by supporting process harmonization, reducing interobserver and intraobserver variability, and assisting with interpretation and standardization of scoring.63,68-71 Of note, the uPath PD-L1 (SP263) image analysis algorithm suite received CE-IVD status in Europe in June 2020 for evaluation of PD-L1 expression in NSCLC samples,72 highlighting the importance of evaluating the potential benefits of these technologies for assisting in interpretation of PD-L1 immunostaining as they enter clinical practice. In line with these points, a possible limitation of this study is the exclusion of studies using digital images. Although a number of studies investigating these technologies were identified during the course of this systematic review, the inclusion criteria restricted the studies evaluated to those investigating “glass slide” pathology only, so as to reflect PD-L1 diagnostic assay approvals at the time the literature search was performed.
In summary, 28-8-, 22C3-, and SP263-based assays show strong analytical concordance for the assessment of PD-L1 expression on TCs in lung cancers and UC. The body of evidence in other tumor types was limited, preventing a conclusion on assay concordance. When placed in context with data for predictive performance, sensitivity, and specificity, the large body of evidence for analytical concordance in lung cancer supports the potential interchangeability of these assays in clinical practice. Care must be taken in tumor types where data for predictive value and/or analytical concordance are limited. As the body of evidence for PD-L1 as a predictor of response to PD-(L)1 inhibitor therapy expands, further studies assessing the comparability and interchangeability of PD-L1 assays with scoring algorithms such as CPS are necessary in additional tumor types.
ACKNOWLEDGMENT
Writing and editorial assistance were provided by Bernard Kerr, PGDipSci, and Jay Rathi, MA, of Spark Medica Inc, funded by Bristol Myers Squibb.
Appendix
TABLE A1.
Emily A. Prince
Employment: TG Therapeutics, Bristol Myers Squibb (Emily Prince was employed by Bristol Myers Squibb when this analysis was performed)
Stock and Other Ownership Interests: TG Therapeutics, Bristol Myers Squibb
Jenine K. Sanzari
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb
Travel, Accommodations, Expenses: Bristol Myers Squibb
Dimple Pandya
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb
Patents, Royalties, Other Intellectual Property: Bristol Myers Squibb
David Huron
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb
Robin Edwards
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb/Celgene
Patents, Royalties, Other Intellectual Property: I am an inventor on pending patent applications filed for work related to my employment at Bristol Myers Squibb. The patent applications are assigned to Bristol Myers Squibb.
Travel, Accommodations, Expenses: Bristol Myers Squibb/Celgene
No other potential conflicts of interest were reported.
SUPPORT
Supported by Bristol Myers Squibb.
AUTHOR CONTRIBUTIONS
Conception and design: All authors
Collection and assembly of data: Emily A. Prince, Dimple Pandya, David Huron, Robin Edwards
Data analysis and interpretation: All authors
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO’s conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Emily A. Prince
Employment: TG Therapeutics, Bristol Myers Squibb (Emily Prince was employed by Bristol Myers Squibb when this analysis was performed)
Stock and Other Ownership Interests: TG Therapeutics, Bristol Myers Squibb
Jenine K. Sanzari
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb
Travel, Accommodations, Expenses: Bristol Myers Squibb
Dimple Pandya
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb
Patents, Royalties, Other Intellectual Property: Bristol Myers Squibb
David Huron
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb
Robin Edwards
Employment: Bristol Myers Squibb
Stock and Other Ownership Interests: Bristol Myers Squibb/Celgene
Patents, Royalties, Other Intellectual Property: I am an inventor on pending patent applications filed for work related to my employment at Bristol Myers Squibb. The patent applications are assigned to Bristol Myers Squibb.
Travel, Accommodations, Expenses: Bristol Myers Squibb/Celgene
No other potential conflicts of interest were reported.
REFERENCES
- 1.Roche Products Limited . TECENTRIQ® (Atezolizumab) [package insert] https://www.gene.com/download/pdf/tecentriq_prescribing.pdf [Google Scholar]
- 2.Regeneron Pharmaceuticals Inc . LIBTAYO® (Cemiplimab) [package insert] https://www.regeneron.com/sites/default/files/Libtayo_FPI.pdf [Google Scholar]
- 3.AstraZeneca . IMFINZI® (Durvalumab) [package insert] https://www.azpicentral.com/imfinzi/imfinzi.pdf#page=1 [Google Scholar]
- 4.Bristol Myers Squibb . OPDIVO® (Nivolumab) [package insert] https://packageinserts.bms.com/pi/pi_opdivo.pdf [Google Scholar]
- 5.EMD Serono Inc, Pfizer Inc . BAVENCIO® (Avelumab) [package insert] https://www.emdserono.com/us-en/pi/bavencio-pi.pdf [Google Scholar]
- 6.Merck & Co Inc . KEYTRUDA® (Pembrolizumab) [package insert] https://www.merck.com/product/usa/pi_circulars/k/keytruda/keytruda_pi.pdf [Google Scholar]
- 7.Pardoll DM.The blockade of immune checkpoints in cancer immunotherapy Nat Rev Cancer 12252–2642012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Juneja VR, McGuire KA, Manguso RT, et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity J Exp Med 214895–9042017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Torlakovic E, Lim HJ, Adam J, et al. “Interchangeability” of PD-L1 immunohistochemistry assays: A meta-analysis of diagnostic accuracy Mod Pathol 334–172020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Büttner R, Gosney JR, Skov BG, et al. Programmed death-ligand 1 immunohistochemistry testing: A review of analytical assays and clinical implementation in non-small-cell lung cancer J Clin Oncol 353867–38762017 [DOI] [PubMed] [Google Scholar]
- 11.Udall M, Rizzo M, Kenny J, et al. PD-L1 diagnostic tests: A systematic literature review of scoring algorithms and test-validation metrics. Diagn Pathol. 2018;13:12. doi: 10.1186/s13000-018-0689-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Liberati A, Altman DG, Tetzlaff J, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. PLoS Med. 2009;6:e1000100. doi: 10.1371/journal.pmed.1000100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.De Meulenaere A, Vermassen T, Creytens D, et al. Importance of choice of materials and methods in PD-L1 and TIL assessment in oropharyngeal squamous cell carcinoma Histopathology 73500–5092018 [DOI] [PubMed] [Google Scholar]
- 14.Scott M, Wildsmith S, Ratcliffe M, et al. Comparison of patient populations identified by different PD-L1 assays in head and neck squamous cell carcinoma (HNSCC) Ann Oncol. 2018;29 abstr 1051PD. [Google Scholar]
- 15.Fujimoto D, Sato Y, Uehara K, et al. Predictive performance of four programmed cell death ligand 1 assay systems on nivolumab response in previously treated patients with non-small cell lung cancer J Thorac Oncol 13377–3862018 [DOI] [PubMed] [Google Scholar]
- 16.Fujimoto D, Yamashita D, Fukuoka J, et al. Comparison of PD-L1 assays in non-small cell lung cancer: 22C3 pharmDx and SP263 Anticancer Res 386891–68952018 [DOI] [PubMed] [Google Scholar]
- 17.Batenchuk C, Albitar M, Zerba K, et al. A real-world, comparative study of FDA-approved diagnostic assays PD-L1 IHC 28-8 and 22C3 in lung cancer and other malignancies J Clin Pathol 711078–10832018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ratcliffe MJ, Sharpe A, Midha A, et al. Agreement between programmed cell death ligand-1 diagnostic assays across multiple protein expression cutoffs in non-small cell lung cancer Clin Cancer Res 233585–35912017 [DOI] [PubMed] [Google Scholar]
- 19.Saito T, Tsuta K, Takeyasu Y, et al. Comparison study of PD-L1 immunohistochemistry assays with 22C3 and 28-8: Analysis on 147 surgically resected NSCLC. Ann Oncol. 2017;28 abstr O2-5-1. [Google Scholar]
- 20.Motoi N, Ishii G, Hayashi Y, et al. Nationwide comparative study of PD-L1 IHC assays on lung cancer: Initial report of LC-SCRUM-IBIS project J Thorac Oncol 13S324–S3252018 [Google Scholar]
- 21.Nakamura Y, Kobayashi T, Nishii Y, et al. Comparable immunoreactivity rates of PD-L1 in archival and recent specimens from non-small cell lung cancer Thorac Cancer 91476–14822018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Krigsfeld GS, Zerba K, Novotny J, Jr, et al. A comparative study of the PD-L1 IHC 22C3 and 28-8 assays on lung cancer samples. Int J Radiat Oncol Biol Phys. 2019;104 abstr 101. [Google Scholar]
- 23.Saito T, Tsuta K, Ishida M, et al. Comparative study of programmed cell death ligand-1 immunohistochemistry assays using 22C3 and 28-8 antibodies for non-small cell lung cancer: Analysis of 420 surgical specimens from Japanese patients Lung Cancer 125230–2372018 [DOI] [PubMed] [Google Scholar]
- 24.Zhang L, Begot C, McGee R, et al. Establishment of analytical performance features of two immunohistochemistry assays for PD-L1 expression in lung cancer. Cancer Res. 2018;78 abstr 1566. [Google Scholar]
- 25.Ilie M, Falk AT, Butori C, et al. PD-L1 expression in basaloid squamous cell lung carcinoma: Relationship to PD-1(+) and CD8(+) tumor-infiltrating T cells and outcome Mod Pathol 291552–15642016 [DOI] [PubMed] [Google Scholar]
- 26.Beck K, Lee KY, Hong SJ. CT-guided transthoracic needle biopsy for evaluation of PD-L1 expression: Comparison of 22C3 and SP263 assays. J Thorac Oncol. 2018;13 doi: 10.1111/1759-7714.13126. abstr MA13.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kim H, Kwon HJ, Park SY, et al. PD-L1 immunohistochemical assays for assessment of therapeutic strategies involving immune checkpoint inhibitors in non-small cell lung cancer: A comparative study Oncotarget 898524–985322017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Quinn AM, Gosney J, Bishop P, et al. Expression of PD-L1 on routine non-small cell lung carcinoma sections: Comparative assessment of SP263 (Ventana) and 22C3 (DAKO pharmDx) J Thorac Oncol. 2018;13 abstr P2.09-06. [Google Scholar]
- 29.Wilberger A, Aisner D, Merrick DT. Pitfalls of PD-L1 immunohistochemistry: A comparison of 22C3 and SP263 antibodies across laboratories and pathologists. Lab Invest. 2018;98 abstr 2118. [Google Scholar]
- 30.Cho JH, Hyeon J, Choi Y, et al. Comparison of PD-L1 immunohistochemical assays and clinical response to anti PD-1 checkpoint inhibitors in patients with lung cancer. J Thorac Oncol. 2018;13 abstr P3.01-18. [Google Scholar]
- 31.Villaruz LC, Ancevski Hunter K, Kurland BF, et al. Comparison of PD-L1 immunohistochemistry assays and response to PD-1/L1 inhibitors in advanced non-small-cell lung cancer in clinical practice Histopathology 74269–2752019 [DOI] [PubMed] [Google Scholar]
- 32.Scheel AH, Dietel M, Heukamp LC, et al. Harmonized PD-L1 immunohistochemistry for pulmonary squamous-cell and adenocarcinomas Mod Pathol 291165–11722016 [DOI] [PubMed] [Google Scholar]
- 33.Xu H, Li C, Gen L, et al. Heterogeneity assessment of two PD-L1 clones: 22C3 and SP142 in patients with non-small cell lung cancer (NSCLC) J Clin Oncol. 2017;35 suppl; abstr e14515. [Google Scholar]
- 34.Xu H, Lin G, Huang C, et al. Assessment of concordance between 22C3 and SP142 immunohistochemistry assays regarding PD-L1 expression in non-small cell lung cancer. Sci Rep. 2017;7:16956. doi: 10.1038/s41598-017-17034-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lisberg A, Hu-Lieskovan S, Grogan T, et al. Evaluation of PD-L1-stained tumor cells via the 22C3 and SP-142 antibodies in cohort of patients treated on KEYNOTE-001. J Thorac Oncol. 2018;13 abstr P2.09-05. [Google Scholar]
- 36.Pang C, Yin L, Zhou X, et al. Assessment of programmed cell death ligand-1 expression with multiple immunohistochemistry antibody clones in non-small cell lung cancer J Thorac Dis 10816–8242018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Scott M, Ratcliffe MJ, Sharpe A, et al. Concordance of tumor and immune cell staining with Ventana SP142, Ventana SP263, Dako 28-8 and Dako 22C3 PD-L1 tests in NSCLC patient samples. J Clin Oncol. 2017;35 suppl; abstr 7. [Google Scholar]
- 38.Conde E, Caminoa A, Dominguez C, et al. Aligning digital CD8(+) scoring and targeted next-generation sequencing with programmed death ligand 1 expression: A pragmatic approach in early-stage squamous cell lung carcinoma Histopathology 72270–2842018 [DOI] [PubMed] [Google Scholar]
- 39.Hirsch FR, McElhinny A, Stanforth D, et al. PD-L1 immunohistochemistry assays for lung cancer: Results from phase 1 of the Blueprint PD-L1 IHC assay comparison project J Thorac Oncol 12208–2222017 [DOI] [PubMed] [Google Scholar]
- 40.Tsao MS, Kerr KM, Kockx M, et al. PD-L1 immunohistochemistry comparability study in real-life clinical samples: Results of Blueprint phase 2 project J Thorac Oncol 131302–13112018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Karnik T, Kimler BF, Fan F, et al. PD-L1 in breast cancer: Comparative analysis of 3 different antibodies Hum Pathol 7228–342018 [DOI] [PubMed] [Google Scholar]
- 42.Krigsfeld GS, Zerba K, Novotny J, Jr, et al. A comparative study of the PD-L1 IHC 22C3 and 28-8 assays in melanoma samples. J Immunother Cancer. 2018;6 abstr P107. [Google Scholar]
- 43.Watanabe T, Okuda K, Murase T, et al. Four immunohistochemical assays to measure the PD-L1 expression in malignant pleural mesothelioma Oncotarget 920769–207802018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sakane T, Murase T, Okuda K, et al. A comparative study of PD-L1 immunohistochemical assays with four reliable antibodies in thymic carcinoma Oncotarget 96993–70092018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Krigsfeld GS, Prince E, Zerba K, et al. Analysis of real-world PD-L1 testing in patients with urothelial carcinoma. J Clin Oncol. 2019;37 suppl; abstr 447. [Google Scholar]
- 46.Vranic S, Ghosh N, Kimbrough J, et al. PD-L1 status in refractory lymphomas. PLoS One. 2016;11:e0166266. doi: 10.1371/journal.pone.0166266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Krigsfeld GS, Prince E, Zerba K, et al. Analysis of real-world PD-L1 IHC testing for clinical use in patients across multiple tumor types. J Clin Oncol. 2019;37 suppl; abstr 151. [Google Scholar]
- 48.Abdul Karim L, Kallakury BVS, Chahine JJ, et al. Collaborative multi-institutional experience in comparing PD-L1 immunohistochemistry assays: Concordance of SP142 and 22C3 immunoreactivity. Appl Immunohistochem Mol Morphol. 2018;26:e22. doi: 10.1097/PAI.0000000000000609. [DOI] [PubMed] [Google Scholar]
- 49.Zavalishina L, Tsimafeyeu I, Povilaitite P, et al. RUSSCO-RSP comparative study of immunohistochemistry diagnostic assays for PD-L1 expression in urothelial bladder cancer Virchows Arch 473719–7242018 [DOI] [PubMed] [Google Scholar]
- 50.Nakasaki M, Fadare O, Patel S, et al. Comparison of PD-L1 immunostain concordance of SP263 and SP142 across different tumor types. Lab Invest. 2017;97 abstr 2140. [Google Scholar]
- 51.Agilent Technologies . PD-L1 IHC 28-8 pharmDx [package insert] https://www.agilent.com/cs/library/packageinsert/public/PD04163_rev_04_SK00521-5_IFU%20Final.pdf [Google Scholar]
- 52.Ventana Medical Systems Inc . VENTANA PD-L1 (SP142) Assay [package insert] https://pim-eservices.roche.com/eLD/api/downloads/2456f3c8-2a9a-ea11-fc90-005056a71a5d?countryIsoCode=us [Google Scholar]
- 53.Agilent Technologies . PD-L1 IHC 22C3 pharmDx [package insert] https://www.agilent.com/cs/library/packageinsert/public/P03951E_18.pdf [Google Scholar]
- 54.Krigsfeld GS, Prince EA, Pratt J, et al. Analysis of real-world PD-L1 IHC 28-8 and 22C3 pharmDx assay utilisation, turnaround times and analytical concordance across multiple tumour types J Clin Pathol 73656–6642020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.GLOBOCAN: International Agency for Research on Cancer All cancers fact sheet. https://gco.iarc.fr/today/data/factsheets/cancers/39-All-cancers-fact-sheet.pdf
- 56.Koomen BM, Badrising SK, van den Heuvel MM, et al. Comparability of PD-L1 immunohistochemistry assays for non-small cell lung cancer: A systematic review Histopathology 76793–8022019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Boothman A-M, Scott M, Ratcliffe M, et al. Impact of patient characteristics, prior therapy, and sample type on tumor cell programmed cell death ligand 1 expression in patients with advanced NSCLC screened for the ATLANTIC study J Thorac Oncol 141390–13992019 [DOI] [PubMed] [Google Scholar]
- 58.Brunnström H, Johansson A, Westbom-Fremer S, et al. PD-L1 immunohistochemistry in clinical diagnostics of lung cancer: Inter-pathologist variability is higher than assay variability Mod Pathol 301411–14212017 [DOI] [PubMed] [Google Scholar]
- 59.Lawson NL, Dix CI, Scorer PW, et al. Mapping the binding sites of antibodies utilized in programmed cell death ligand-1 predictive immunohistochemical assays for use with immuno-oncology therapies Mod Pathol 33518–5302020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhu J, Labriola M, Cheris S, et al. Concordance between PD-L1 assays for metastatic renal cell carcinoma (mRCC) and metastatic urothelial carcinoma (mUC) J Clin Oncol. 2019;37:577. [Google Scholar]
- 61.Walker J, Zajac M, Ye J, et al. Impact of different programmed cell death ligand-1 (PD-L1) expression algorithms on patient selection and durvalumab efficacy in urothelial carcinoma (UC) Ann Oncol. 2018;29 abstr 904P. [Google Scholar]
- 62.Kulangara K, Zhang N, Corigliano E, et al. Clinical utility of the combined positive score for programmed death ligand-1 expression and the approval of pembrolizumab for treatment of gastric cancer Arch Pathol Lab Med 143330–3372019 [DOI] [PubMed] [Google Scholar]
- 63.Jasani B, Banfer G, Fish R, et al. Evaluation of an online training tool for scoring programmed cell death ligand-1 (PD-L1) diagnostic tests for lung cancer. Diagn Pathol. 2020;15:37. doi: 10.1186/s13000-020-00953-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Scheel AH, Baenfer G, Baretton G, et al. Interlaboratory concordance of PD-L1 immunohistochemistry for non-small-cell lung cancer Histopathology 72449–4592018 [DOI] [PubMed] [Google Scholar]
- 65.Vennapusa B, Baker B, Kowanetz M, et al. Development of a PD-L1 complementary diagnostic immunohistochemistry assay (SP142) for atezolizumab Appl Immunohistochem Mol Morphol 2792–1002019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Adam J, Hofman V, Mansuet-Lupo A, et al. Real-world concordance across pathologists for PD-L1 scoring in non-small cell lung cancer: Results from a large nationwide initiative. J Thorac Oncol. 2019;14 suppl; abstr P2.09-17. [Google Scholar]
- 67.Lantuejoul S, Sound-Tsao M, Cooper WA, et al. PD-L1 testing for lung cancer in 2019: Perspective from the IASLC pathology committee J Thorac Oncol 15499–5192020 [DOI] [PubMed] [Google Scholar]
- 68.Pell R, Oien K, Robinson M, et al. The use of digital pathology and image analysis in clinical trials J Pathol Clin Res 581–902019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bera K, Schalper KA, Rimm DL, et al. Artificial intelligence in digital pathology—new tools for diagnosis and precision oncology Nat Rev Clin Oncol 16703–7152019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Steele KE, Tan TH, Korn R, et al. Measuring multiple parameters of CD8+ tumor-infiltrating lymphocytes in human cancers by image analysis. J Immunother Cancer. 2018;6:20. doi: 10.1186/s40425-018-0326-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Aeffner F, Zarella MD, Buchbinder N, et al. Introduction to digital image analysis in whole-slide imaging: A white paper from the Digital Pathology Association. J Pathol Inform. 2019;10:9. doi: 10.4103/jpi.jpi_82_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Roche Diagnostics uPath PD-L1 (SP263) image analysis, NSCLC (CE-IVD) https://diagnostics.roche.com/global/en/products/instruments/upath-pd-l1-sp263-image-analysis.html
- 73.Ventana Medical Systems Inc . VENTANA PD-L1 (SP263) Assay [package insert] https://pim-eservices.roche.com/eLD/api/downloads/cb09dec4-3136-ea11-fc90-005056a71a5d?countryIsoCode=us [Google Scholar]
- 74.Skov BG, Skov T.Paired comparison of PD-L1 expression on cytologic and histologic specimens from malignancies in the lung assessed with PD-L1 IHC 28-8 pharmDx and PD-L1 IHC 22C3 pharmDx Appl Immunohistochem Mol Morphol 25453–4592017 [DOI] [PubMed] [Google Scholar]
- 75.Velcheti V, Patwardhan PD, Liu FX, et al. Real-world PD-L1 testing and distribution of PD-L1 tumor expression by immunohistochemistry assay type among patients with metastatic non-small cell lung cancer in the United States. PLoS One. 2018;13:e0206370. doi: 10.1371/journal.pone.0206370. [DOI] [PMC free article] [PubMed] [Google Scholar]