Abstract
Over the past decade, immunotherapy has emerged as one of the most promising cancer treatments. Several monoclonal antibodies targeting the programmed death 1 (PD‐1)/ programmed death ligand‐1 (PD‐L1) pathway have been integrated into standard‐of‐care treatments for a wide range of cancer types. Although all the available PD‐L1 immunohistochemistry (IHC) assays have been developed on formalin‐fixed histological specimens, a growing body of research has recently suggested the feasibility of PD‐L1 testing on cytological samples. Although promising results have been reported, several important issues still need to be addressed. Among these are pre‐analytical issues, cyto‐hystological correlation, and inter‐observer agreement. This review will briefly summarise the knowledge gaps and future directions of cytopathology in the immuno‐oncology scenario.
Keywords: cytopathology, immune oncology, immunotherapy, PD‐L1
Short abstract
Programmed death ligand‐1 (PD‐L1) assays have been developed on formalin‐fixed histological specimens, and a growing body of research is being dedicated to confirming the feasibility of applying these assays to cytological samples. Promising results have been reported but several important issues still need to be addressed. This review summarizes the current status, knowledge gaps and future directions of cytopathology in the context of immuno‐oncology.
1. INTRODUCTION
Over the past decade, immunotherapy, particularly the clinical development of immune‐checkpoint inhibitors (ICIs), has emerged as one of the most promising cancer treatments. To date, monoclonal antibodies targeting the Programmed Death 1 (PD‐1)/ Programmed Death Ligand‐1 (PD‐L1) axis have been integrated into standard treatments for a wide range of cancer types.1, 2 Despite having proven effective, ICI treatments seem to work only for a subset of patients. Not surprisingly, the identification of new predictive biomarkers for targeted treatments has become a major goal of immuno‐oncology.
In June 2020, the FDA approved pembrolizumab for the treatment of adult and pediatric patients with unresectable or metastatic solid tumour mutational burden‐high (TMB‐H) (greater than or equal to 10 mutations/megabase [mut/Mb]).3 At the time PD‐L1, evaluated by immunohistochemistry (IHC), was the only predictive biomarker available for PD‐L1/PD1 immunotherapy.4
Recently, numerous articles have been published on the topic of PD‐L1 assays, addressing factors such as clone harmonization, analytical validation, and scoring reproducibility issues.5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 However, most of these studies involve only patients with available satisfactory formalin‐fixed paraffin‐embedded (FFPE) tissue specimens. Unfortunately, clinical research does not always reflect routine clinical practice. Indeed, PDL‐1 testing of collected tissue specimens may often be unworkable, primarily because tissue biopsies from advanced cancer patients, including those from non‐small cell lung cancer (NSCLC), are highly challenging if not impossible to obtain. Consequently, cytopathologists have no choice but to resort to cytological samples for both morphological characterisation and predictive testing. It is in this context that molecular cytopathology has emerged as a major player in diagnostic and predictive pathology. Indeed, the growing popularity of molecular cytopathology stems from the fact that most molecular tests are highly versatile and can, therefore, be applied to a wide range of cytological preparations. However, the feasibility of PD‐L1 IHC evaluation on cytological specimens still warrants thorough investigation. In fact, as of today, the commercially available PD‐L1 assays have never been validated on cytological samples.18 Nonetheless, since both immunostaining and predictive testing are routinely performed in cytopathology practice, pathologists have been exploring the feasibility and reliability of assessing PD‐L1 expression in cytological samples.
In this review, we will briefly summarise the knowledge gaps and future directions of cytopathology in the immuno‐oncology scenario.
2. PRE‐ANALYTIC ISSUES: DOES THE SAMPLE TYPE MATTER?
Several types of cytological samples are used in routine practice. However, being characterized by distinct pre‐analytical issues, each specimen should be considered as a separate entity. In particular, the common reluctance to use cytological samples for PD‐L1 evaluation primarily stems from the notion that alcohol‐based fixatives might compromise IHC staining.6, 19, 20 Consequently, since PD‐L1 IHC procedures have been validated only on FFPE samples, formalin‐fixed cell block (CB) preparations are generally recommended. However, not all CBs are processed in the same way. Indeed, CB preparatory techniques may vary significantly depending on several factors, that is, the choice of the fluid medium used for the FNA needle rinse (formalin, saline or alcohol‐based fixatives followed by formalin post‐fixation), the fixation time, and the method of concentration.19, 21, 22, 23, 24 Despite the lack of standardized preparation protocols, several lines of evidence have demonstrated that the type of fixative does not affect PD‐L1 staining. In fact, Wang et al21 observed that fixation with formalin only, methanol/alcohol only, or both did not affect PD‐L1 expression. Moreover, Gosney et al25 indicated that paired CBs fixed in either alcohol‐based solutions (CytoRich Red or CytoLyt) or neutral buffered formalin (NBF) yielded concordant PD‐L1 expression. Likewise, Lou et al26 observed that specimen prefixation with CytoLyt had only a negligible impact on PD‐L1 IHC staining. Table 1 presents a summary of the literature on the effects of different types of fixatives, except formalin, on PD‐L1 evaluation.
TABLE 1.
Summary of available literature assessing the effect of different fixation type, other than formalin, on PD‐L1 evaluation
| Authors (ref.) | Sample type | Preparation type | No. | Fixatives/preservatives | Antibody clone |
|---|---|---|---|---|---|
| Lloyd et al19 | Cell lines | CB | nr |
PreservCyt CytoLyt Roswell Park Memorial Institute (RPMI) cell culture media Saline |
28‐8 |
| Wang et al21 | FNA, fluids, BAL | CB | 261 |
Methanol/alcohol only Formalin and methanol/alcohol |
22C3 |
| Gosney et al25 | EBUS | CB | 50 |
CytoRich Red CytoLyt |
22C3 |
| Lou et al26 | Fluids, EBUS‐TBNA | CB | 52 | CytoLyt | 22C3 |
| Jain et al27 | Bronchial brushing/washing | LBC | 26 | CytoRich Red | SP263 |
| Capizzi et al28 | FNA | Smears | 49 | MicroFix spray | SP263 |
| Lozano et al29 | FNA | Smears | 62 | Alcohol | 22C3, SP263 |
| Noll et al30 | FNA | Smears | 41 | Alcohol | 22C3 |
Abbreviations: BAL, broncho‐alveolar lavage; CB, cell‐block; EBUS‐TBNA, endobronchial ultrasound‐guided transbronchial needle aspiration; FNA, fine needle aspiration; LBC, liquid‐based cytology; No., number of samples; nr, not reported; ref, reference number.
Evidence that the type of fixative does not compromise PD‐L1 staining is also confirmed by studies assessing the feasibility of using “traditional” non‐formalin fixed cytological preparations, including direct smears or liquid‐based cytology specimens (LBC) for PD‐L1 IHC testing.27, 28, 29, 30 Indeed, although some studies have indicated that FFPE samples and corresponding non‐formalin fixed cytological smears show a good concordance rate of PD‐L1 expression, these preparations may lead to cytopathological misinterpretation. For instance, the presence of a non‐specific staining of neoplastic cell cytoplasms, extracellular mucus, background cellular debris,31 and inflammatory cells may result in an overestimation of PD‐L1 expression on direct smears. Moreover, appreciation of true membranous staining, which is perceived as distinct from cytoplasmic staining, and the presence of false‐positive staining in large three‐dimensional cell groups entrapping reagents, may also lead to a misinterpretation of PD‐L1 expression on direct cytological smears.32 (Figure 1).
FIGURE 1.

(A) Hematoxylin and eosin‐stained cell block section (original magnification 20×) and corresponding PD‐L1–stained cell block section (B): a circumferential pattern of membrane staining in neoplastic cells was observed. PD‐L1 positive lymphocytes showed indistinguishable membrane and cytoplasmic staining, due to a high nuclear to cytoplasmic ratio (arrow). (C) PD‐L1–stained ethanol‐fixed direct smear: a partial circumferential pattern of membrane staining in neoplastic cells was observed (arrows). The presence of false‐positive staining in three‐dimensional cell groups entrapping reagents (arrowhead) and staining in inflammatory cells (asterisk, histiocyte) can lead to an overestimation of the PD‐L1 expression (original magnification 20×). (D) PD‐L1–stained ethanol‐fixed direct smear: high amount of non‐specific staining of extracellular mucus (original magnification 2.5×)
3. CYTO‐HISTOLOGICAL CORRELATION
To assess whether cytological samples are as reliable as histological samples for PD‐L1 testing, several authors have intensively investigated the concordance rates between matched cytological and histological samples. Indeed, several research groups have evaluated both cytological and histological samples in terms of adequacy rates and PD‐L1 expression levels. More specifically, the adequacy criteria state that a sample must contain a minimum of 100 viable tumour cells to be eligible for quantification of PD‐ L1 expression. It is worth noting that the literature has shown that the adequacy rate of cytological samples is generally higher than 80%21, 42 and only occasionally lower than 70%44 (Table 2). These data are remarkable if one considers the great difficulty of obtaining a sufficient number of tumour cells in small biopsies.38 It is also of note that rapid on‐site evaluation (ROSE) can be used to assess specimen adequacy and possibly improve CB quality in terms of tumour cellularity.45 However, conclusive evidence regarding the use of ROSE on downstream ancillary testing outcomes is still lacking.46, 47 Under this scenario, cytopathologists play a key role, not only in carrying out on‐site evaluation of cytological material, but also in ensuring the proper triage of available material.
TABLE 2.
Summary of available literature assessing adequacy rate of PD‐L1 evaluation on cytological samples
| Authors (ref.) | Sample type | Preparation type | No. | Adequacy rate % |
|---|---|---|---|---|
| Wang et al21 | FNA, fluids, BAL | CB | 371 | 92 |
| Noll et al30 | FNA | CB/smears | 41 | 92.6/90.2 |
| Zou et al33 | Fluids | CB | 124 | 91.9 |
| Torous et al34 | TBNA, pleural effusion, bronchial washing | CB | 94 | 93.6 |
| Evans et al35 | nr | CB | 2276 | 84 |
| Bubendorf et al36 | Fluids, washing, brushing, FNA, ex vivo FNA | CB | 165 | 86.6 |
| Vigliar et al37 | nr | CB/smears/LBC | 48 | 85.4 |
| Heymann et al38 | FNA, fluids | CB | 40 | 90 |
| Mei et al39 | Fluids, FNA | CB | 100 | 96 |
| Skov et al41 | nr | CB | 86 | 80.3 |
| Stoy et al41 | TBNA | CB | 22 | 90.9 |
| Dong et al42 | FNA, brushing | CB | 112 | 70.5 |
| Kravstov et al43 | nr | CB | 75 | 84 |
| Hendry et al44 | Bronchial brushing, FNA | CB | 60 | 50 |
Abbreviations: BAL, broncho‐alveolar lavage; CB, cell‐block; FNA, fine needle aspiration; LBC, liquid‐based cytology; No., number of samples; nr, not reported; ref, reference number; TBNA, transbronchial needle aspiration.
As for PD‐L1 evaluation, since 2017 several single institutional studies have reported comparable PD‐L1 expression on matched cytological and histological (small biopsy/surgical resection) specimens.21, 47, 48, 49, 50, 51 In light of these findings, in a systemic review, Gosney et al52 painstakingly evaluated the concordance rate of PD‐L1 staining in matched histological and cytological samples from patients with advanced NSCLC. Based on a total of 428 paired specimens collected across nine studies, the authors reported an overall concordance rate of 88.3% at a clinically relevant tumour proportion score (TPS) cut‐off greater than 1% and of 89.7% for specimens with TPS greater than or equal to 50%. Interestingly, these values closely reflect sample heterogeneity in real‐life cytology practice. In fact, the review evaluated data from both CBs and direct smears obtained from different sampling types (endobronchial ultrasound, computed tomography and ultrasound guided FNA, washing, brushing and fluid collection). Moreover, it also examined different PD‐L1 antibody clones (22C3 [Dako] and SP263 [Ventana]) using both pharmDx assays and laboratory developed tests (LDTs). The clear concordant results confirm once again the reliability of using cytological material for PD‐L1 evaluation. Interestingly, Dong et al's study42 pointed out that CBs with higher cellularity show better agreement scores between cytology and histology. Indeed, PD‐L1 expression levels in resected specimens were nearly equivalent to those in CBs with abundant cellularity (greater than 400 cells). Altogether, these studies clearly indicate that cytological materials constitute a reliable source for PD‐L1 evaluation in NSCLC patients.
4. INTEROBSERVER AGREEMENT
Cytopathologists should take into account interobserver variability rates before deeming cytological specimens suitable for PD‐L1 assessment. However, as of today, data on interobserver agreement are still limited to a few studies involving varying numbers of pathologists and analyzed samples. Overall, though, reproducibility has been remarkable. For example, Russell‐Goldman et al53 reported a high interobserver agreement (intraclass correlation coefficient, ICC equal to 0.96) between two pathologists who evaluated 56 cytological specimens. Similarly, Gagnè et al54 reported substantial or almost perfect interobserver agreement rates (Fleiss' kappa, k equal to 0.74‐0.82) among four pathologists who evaluated 46 CBs. Consistently, the Blueprint (BP) PD‐L1 Immunohistochemistry Comparability Project phase 26 reported a good ICC at all cutoff levels (k equal to 0.60‐0.80), for both glass (0.78) and digital (0.85) slides among 24 pathologists who analyzed 22 CBs. More recently, quite similar interobserver agreement rates were reported by Sinclair et al55 (k equal to 0.74) and Kravsotv et al43 (k equal to 0.66) (Table 3). Despite such encouraging agreement rates, some studies have highlighted the fact that that variability among observers is generally more pronounced in cytological samples than in biopsies and surgical specimens,6, 56 suggesting that the interpretation of PD‐L1 in cytological samples is more challenging. The main difficulties arise primarily from the presence of background aspecific staining and the difficulty of differentiating tumour cells from benign ones, including macrophages, especially in cases presenting discohesive cells. Moreover, these pitfalls are more pronounced in traditional, non‐formalin fixed cytological preparations, for which data on interobserver agreement are still lacking. For this reason, deciding whether a cytological sample is appropriate for PD‐L 1 IHC assessment requires considerable expertise and specialized training.43, 56
TABLE 3.
Summary of literature studies assessing interobserver agreement for PD‐L1 scoring on cytological specimens
| Authors (ref.) | Preparation type | No. | Number of pathologists | Antibody clone | Statistical test | Interobserver agreement |
|---|---|---|---|---|---|---|
| Tsao et al6 | CB | 22 | 24 | 22C3, 28‐8, SP142, SP263, 73‐10 |
ICC Fleiss's kappa |
ICC =0.78‐0.85 k = 0.6‐0.85 |
| Kravstov et al43 | CB | 50 | 3 | 22C3 | Fleiss's kappa | k = 0.66 |
| Russel‐Goldman et al53 | CB | 56 | 2 | E1L3N | ICC | 0.96 |
| Gagnè et al54 | CB | 46 | 4 | SP263, 28‐8 | Fleiss's kappa | k = 0.74 to 0.82 |
| Sinclair et al55 | CB | 86 | 5 | 22C3 |
Fleiss's kappa Cohen's kappa |
0.74‐0.79 0.49‐0.83 to 0.63‐0.90 |
Abbreviations: CB, cell block; ICC, intraclass correlation coefficient; No., number of samples; ref, reference number.
5. GUIDELINES
The literature has clearly established that cytology specimens (smears, CBs, LBC) are valuable sources for ancillary techniques, provided that careful validation of the samples is carried out.57 Consequently, recommendations for proper management of cytological material have been included in biomarker testing guidelines for patient selection in immuno‐oncology. For example, the Canadian Association of Pathologists‐Association Canadienne Des Pathologistes (CAP‐ACP) recommends that FDA‐approved or CE‐marked PD‐L1 IHC kits, validated for FFPE samples, be used for cytology samples only if they are processed according to the pre‐analytical conditions provided by the kit and the readout is compatible with the type of cytology samples.58 For NSCLC cases, the International Association for the Study of Lung Cancer Pathology Committee (IASCL) requires that protocols for cytological materials be fully validated and submitted to quality‐control measures. Thus, it stands to reason that validation processes ought to be carried out separately for any type of cytological preparation.18
6. FUTURE PERSPECTIVES
Current advances in both digital image analysis (DIA) technologies and multiplex immunofluorescence (IF)/IHC could be a powerful strategy for PD‐L1 assessment. In fact, the application of a high throughput image analysis pipeline to multiplex IF or IHC to assess PD‐L1, the epithelial cell marker cytokeratin, the macrophage marker CD68, and the T‐cell marker CD8 has been shown to yield a high diagnostic level of confidence in the identification of specific cell types co‐expressing PD‐L1. Therefore, a multiplex approach may enable cytopathologists to refine PD‐L1 scores in neoplastic cells, especially in cases close to clinical thresholds. Nonetheless, cytological samples pose practical issues due to a lack of tissue architecture. Therefore, further investigations are warranted to investigate the diagnostic accuracy of the PD‐L1 multiplex image analysis on cytological specimens.59, 60, 61, 62
It is widely known that predicting ICI therapy outcome on the basis of a single biomarker, such as PD‐L1, is far from perfect. Therefore, promising predictive biomarkers are currently under investigation, including TMB, defined as the total number of somatic mutations per tumour genome. Although most of the data on TMB are derived from the evaluation of FFPE histological samples,63, 64 some authors have provided preliminary results on the feasibility of assessing TMB on cytological material. For example, Pepe et al65 recently demonstrated the technical feasibility of assessing TMB on FFPE CBs in a pilot study evaluating 16 paired histological and CB samples from eight NSCLC patients. Interestingly, Alborelli et al,66 who compared TMB values in matched FFPE and cytological specimens, demonstrated that cytological smears provide even more consistent TMB values than their histological counterparts. Therefore, considering the high quality of DNA and lack of formalin‐fixation induced artifacts, the authors concluded that ethanol‐fixed cytological specimens allow a more robust TMB estimation than histological samples.
However, immunotherapy outcomes may significantly vary among patients, regardless of PD‐L1 expression and TMB values. Thus, major efforts are being made to identify co‐occurring mutations. For example, Marinelli et al67 identified four genes (KEAP1, PBRM1, SMARCA4 and STK11) that potentially reduce the efficacy of immunotherapy in patients with lung adenocarcinoma. Thus, the dynamic nature of immuno‐oncology highlights the relevance of managing cytological materials appropriately to maximise their use for comprehensive predictive testing.
Finally, in addition to tumour cells, the tumour microenvironment (TME) and its dynamic reshaping have emerged as major players in cancer progression and treatment outcomes. The importance of this line of research is reflected in the recent development of ultra‐fast cycling for multiplexed cellular fluorescence imaging for the analysis of single cell populations, such as those analyzable in cytological samples.68 This new approach could break new ground in the evaluation of immunological dynamics by exploiting the ability of cytopathologists to perform serial cytological tumour sampling.69
In conclusion, this review clearly indicates that cytological samples constitute a reliable source for PDL‐1 IHC analysis (Figure 2), as evidenced by the remarkable specimen adequacy and concordance rate seen between cytological and histological specimens. Moreover, the fact that that cytological fixatives do not compromise PD‐L1 staining further attests to the utility of cytological specimens for PD‐L1 testing in routine clinical practice. However, there are few challenges which still need to be addressed. In particular, training programs should be provided to ensure adequacy assessment and proper sample management, and preparation protocols must be validated and standardized across individual laboratories. Moreover, the value of dedicated expertise in PD‐L1 interpretation in cytological samples cannot be underestimated.
FIGURE 2.

Schematic representation of challenges and solutions in PD‐L1 evaluation on cytological material. IHC, immunohistochemistry; ROSE, rapid on‐site evaluation
Finally, although much of the new evidence regarding TMB and TME is still preliminary, we are confident that cytological samples will have great utility in precision immuno‐oncology.
CONFLICT OF INTEREST
Umberto Malapelle received personal fees (as speaker's bureau or advisor) from Boehringer Ingelheim, AstraZeneca, Roche, MSD, Amgen and Merck, for work unrelated to the current paper. Giancarlo Troncone received personal fees (as speaker's bureau or advisor) from Roche, MSD, Pfizer and Bayer, for work unrelated to the current paper. Elena Vigliar received personal fees as advisor from Diaceutics, for work unrelated to the current study. The other authors declare no potential conflicts of interest.
AUTHOR CONTRIBUTIONS
Antonino Iaccarino, Maria Salatiello, Giancarlo Troncone and Elena Vigliar conceived the review. All authors collected the literature data, wrote the original draft, and approved the final version of the manuscript.
ACKNOWLEDGMENTS
We thank Paola Merolla for English language editing.
Iaccarino A, Salatiello M, Migliatico I, et al. PD‐L1 and beyond: Immuno‐oncology in cytopathology. Cytopathology. 2021;32:596–603. 10.1111/cyt.12982
Antonino Iaccarino and Maria Salatiello contributed equally.
DATA AVAILABILITY STATEMENT
Data sharing not applicable to this article as no datasets were generated or analysed during the current study.
REFERENCES
- 1.Cancer Research Institute (CRI) . PD‐1/PD‐L1 landscape analysis. [Internet]. https://www.cancerresearch.org/scientists/immuno‐oncology‐landscape/pd‐1‐pd‐l1‐landscape. Accessed December 14, 2020.
- 2.Cancer Research Institute (CRI) . FDA approval timeline of active immunotherapies. [Internet]. https://www.cancerresearch.org/scientists/immuno‐oncology‐landscape/fda‐approval‐timeline‐of‐active‐immunotherapies. Accessed December 14, 2020.
- 3.FDA . FDA approves pembrolizumab for adults and children with TMB‐H solid tumors. [Internet]. https://www.fda.gov/drugs/drug‐approvals‐and‐databases/fda‐approves‐pembrolizumab‐adults‐and‐children‐tmb‐h‐solid‐tumors. Accessed October 23, 2020.
- 4.FDA . List of cleared or approved companion diagnostic devices (In vitro and imaging tools). [Internet]. https://www.fda.gov/medical‐devices/vitro‐diagnostics/list‐cleared‐or‐approved‐companion‐diagnostic‐devices‐vitro‐and‐imaging‐tools. Accessed 2020 December 14, 2020.
- 5.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. 2017;12:208‐222. [DOI] [PubMed] [Google Scholar]
- 6.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. 2018;13:1302‐1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hendry S, Byrne DJ, Wright GM, et al. Comparison of four PD‐L1 immunohistochemical assays in lung cancer. J Thorac Oncol. 2018;13:367‐376. [DOI] [PubMed] [Google Scholar]
- 8.Marchetti A, Barberis M, Franco R, et al. Multicenter comparison of 22C3 PharmDx (Agilent) and SP263 (Ventana) assays to test PD‐L1 expression for NSCLC patients to be treated with immune checkpoint inhibitors. J Thorac Oncol. 2017;12:1654‐1663. [DOI] [PubMed] [Google Scholar]
- 9.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Scheel AH, Baenfer G, Baretton G, et al. Interlaboratory concordance of PD‐L1 immunohistochemistry for non‐small‐cell lung cancer. Histopathology. 2017;72(3):449‐459. [DOI] [PubMed] [Google Scholar]
- 11.Scheel AH, Dietel M, Heukamp LC, et al. Harmonized PD‐L1 immunohistochemistry for pulmonary squamous‐cell and adenocarcinomas. Mod Pathol. 2016;29(10):1165‐1172. [DOI] [PubMed] [Google Scholar]
- 12.Li C, Huang C, Mok TS, et al. Comparison of 22C3 PD‐L1 expression between surgically resected specimens and paired tissue microarrays in non‐small cell lung cancer. J Thorac Oncol. 2017;12:1536‐1543. [DOI] [PubMed] [Google Scholar]
- 13.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. 2017;30:1411‐1421. [DOI] [PubMed] [Google Scholar]
- 14.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. 2018;71:1078‐1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Adam J, Le Stang N, Rouquette I, et al. Multicenter harmonization study for PD‐L1 IHC testing in non‐small‐cell lung cancer. Ann Oncol. 2018;29:953‐958. [DOI] [PubMed] [Google Scholar]
- 16.Cooper WA, Russell PA, Cherian M, et al. Intra‐ and interobserver reproducibility assessment of PD‐L1 biomarker in non‐small cell lung cancer. Clin Cancer Res. 2017;23:4569‐4577. [DOI] [PubMed] [Google Scholar]
- 17.Rehman JA, Han G, Carvajal‐Hausdorf DE, et al. Quantitative and pathologist‐read comparison of the heterogeneity of programmed death‐ligand 1 (PD‐L1) expression in non‐small cell lung cancer. Mod Pathol. 2017;30:340‐349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lantuejoul S, Damotte D, Hofman V, Adam J. Programmed death ligand 1 immunohistochemistry in non‐small cell lung carcinoma. J Thorac Dis. 2019;11:S89‐S101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lloyd IE, Zhou W, Witt BL, Chadwick BE. Characterization of PD‐L1 immunohistochemical expression in cell blocks with different specimen fixation and processing methods. Appl Immunohistochem Mol Morphol. 2019;27:107‐113. [DOI] [PubMed] [Google Scholar]
- 20.Gruchy JR, Barnes PJ, Dakin Haché KA. CytoLyt ® fixation and decalcification pretreatments alter antigenicity in normal tissues compared with standard formalin fixation. Appl Immunohistochem Mol Morphol. 2015;23:297‐302. [DOI] [PubMed] [Google Scholar]
- 21.Wang H, Agulnik J, Kasymjanova G, et al. Cytology cell blocks are suitable for immunohistochemical testing for PD‐L1 in lung cancer. Ann Oncol. 2018;29:1417‐1422. [DOI] [PubMed] [Google Scholar]
- 22.Nambirajan A, Jain D. Cell blocks in cytopathology: an update. Cytopathology. 2018;29:505‐524. [DOI] [PubMed] [Google Scholar]
- 23.Crapanzano J, Heymann J, Monaco S, Nassar A, Saqi A. The state of cell block variation and satisfaction in the era of molecular diagnostics and personalized medicine. Cytojournal. 2014;11:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Vigliar E, Iaccarino A, Campione S, et al. PD‐L1 expression in cell‐blocks of non‐small cell lung cancer: the impact of prolonged fixation. Diagn Cytopathol. 2020;48:595‐603. [DOI] [PubMed] [Google Scholar]
- 25.Gosney JR, Haragan A, Chadwick C, et al. Programmed death ligand 1 expression in EBUS aspirates of non–small cell lung cancer: is interpretation affected by type of fixation? Cancer Cytopathol. 2020;128:100‐106. [DOI] [PubMed] [Google Scholar]
- 26.Lou SK, Ko HM, Kinoshita T, et al. Implementation of PD‐L1 22C3 IHC pharmDx in cell block preparations of lung cancer: concordance with surgical resections and technical validation of CytoLyt® prefixation. Acta Cytol. 2020;64:1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jain D, Sukumar S, Mohan A, Iyer VK. Programmed death‐ligand 1 immunoexpression in matched biopsy and liquid‐based cytology samples of advanced stage non‐small cell lung carcinomas. Cytopathology. 2018;29:550‐557. [DOI] [PubMed] [Google Scholar]
- 28.Capizzi E, Ricci C, Giunchi F, et al. Validation of the immunohistochemical expression of programmed death ligand 1 (PD‐L1) on cytological smears in advanced non small cell lung cancer. Lung Cancer. 2018;126:9‐14. [DOI] [PubMed] [Google Scholar]
- 29.Lozano MD, Abengozar‐Muela M, Echeveste JI, et al. Programmed death–ligand 1 expression on direct Pap‐stained cytology smears from non–small cell lung cancer: Comparison with cell blocks and surgical resection specimens. Cancer Cytopathol. 2019;127:470‐480. [DOI] [PubMed] [Google Scholar]
- 30.Noll B, Wang W‐L, Gong Y, et al. Programmed death ligand 1 testing in non‐small cell lung carcinoma cytology cell block and aspirate smear preparations. Cancer Cytopathol. 2018;126(5):342‐352. [DOI] [PubMed] [Google Scholar]
- 31.Roy‐Chowdhuri S. Immunocytochemistry of cytology specimens for predictive biomarkers in lung cancer. Transl Lung Cancer Res. 2020;9:898‐905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Jain D, Nambirajan A, Borczuk A, et al. Immunocytochemistry for predictive biomarker testing in lung cancer cytology. Cancer Cytopathol. 2019;127(5):325‐339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zou Y, Xu L, Tang Q, et al. Cytology cell blocks from malignant pleural effusion are good candidates for PD‐L1 detection in advanced NSCLC compared with matched histology samples. BMC Cancer. 2020;20:344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Torous VF, Rangachari D, Gallant BP, Shea M, Costa DB, VanderLaan PA. PD‐L1 testing using the clone 22C3 pharmDx kit for selection of patients with non–small cell lung cancer to receive immune checkpoint inhibitor therapy: are cytology cell blocks a viable option? J Am Soc Cytopathol. 2018;7:133‐141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Evans M, O'Sullivan B, Hughes F, et al. The clinicopathological and molecular associations of PD‐L1 expression in non‐small cell lung cancer: analysis of a series of 10,005 cases tested with the 22C3 assay. Pathol Oncol Res. 2018;26(1):79‐89. [DOI] [PubMed] [Google Scholar]
- 36.Bubendorf L, Conde E, Cappuzzo F, et al. A noninterventional, multinational study to assess PD‐L1 expression in cytological and histological lung cancer specimens. Cancer Cytopathol. 2020;128:928‐938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Vigliar E, Malapelle U, Iaccarino A, et al. PD‐L1 expression on routine samples of non‐small cell lung cancer: results and critical issues from a 1‐year experience of a centralised laboratory. J Clin Pathol. 2019;72:412‐417. [DOI] [PubMed] [Google Scholar]
- 38.Heymann JJ, Bulman WA, Swinarski D, et al. PD‐L1 expression in non‐small cell lung carcinoma: comparison among cytology, small biopsy, and surgical resection specimens. Cancer Cytopathol. 2017;125:896‐907. [DOI] [PubMed] [Google Scholar]
- 39.Mei P, Shilo K, Wei L, Shen R, Tonkovich D, Li Z. Programmed cell death ligand 1 expression in cytologic and surgical non–small cell lung carcinoma specimens from a single institution: association with clinicopathologic features and molecular alterations. Cancer Cytopathol. 2019;127:447‐457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Skov B, Skov T. P2.01‐048 paired comparison of PDL1 assessment on cytology and histology from malignancies in the lung. J Thorac Oncol. 2017;12(1):1‐48. [Google Scholar]
- 41.Stoy SP, Rosen L, Mueller J, Murgu S. Programmed death‐ligand 1 testing of lung cancer cytology specimens obtained with bronchoscopy. Cancer Cytopathol. 2018;126:122‐128. [DOI] [PubMed] [Google Scholar]
- 42.Dong Z, Liu Y, Jiang T, et al. Cell block as a surrogate for programmed death‐ligand 1 staining testing in patients of non‐small cell lung cancer. J Cancer. 2020;11:551‐558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kravtsov O, Hartley CP, Sheinin Y, Hunt BC, Felix JC, Giorgadze T. Utility of PD‐L1 testing on non‐small cell lung cancer cytology specimens: an institutional experience with interobserver variability analysis. Ann Diagn Pathol. 2020;48:e151602. [DOI] [PubMed] [Google Scholar]
- 44.Hendry S, Byrne DJ, Christie M, et al. Adequate tumour cellularity is essential for accurate PD‐L1 immunohistochemistry assessment on cytology cell‐block specimens. Cytopathology. 2020;31:90‐95. [DOI] [PubMed] [Google Scholar]
- 45.Collins BT, Garcia TC, Hudson JB. Rapid on‐site evaluation improves fine‐needle aspiration biopsy cell block quality. J Am Soc Cytopathol. 2016;5:37‐42. [DOI] [PubMed] [Google Scholar]
- 46.Trisolini R, Cancellieri A, Tinelli C, et al. Randomized trial of endobronchial ultrasound‐guided transbronchial needle aspiration with and without rapid on‐site evaluation for lung cancer genotyping. Chest. 2015;148:1430‐1437. [DOI] [PubMed] [Google Scholar]
- 47.Roy‐Chowdhuri S, Aisner DL, Allen TC, et al. Special articles biomarker testing in lung carcinoma cytology specimens a perspective from members of the pulmonary pathology society. Arch Pathol Lab Med. 2016;140:1267‐1272. [DOI] [PubMed] [Google Scholar]
- 48.Pak MG, Roh MS. Cell‐blocks are suitable material for programmed cell death ligand‐1 immunohistochemistry: comparison of cell‐blocks and matched surgical resection specimens in lung cancer. Cytopathology. 2019;30:578‐585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ilie M, Juco J, Huang L, Hofman V, Khambata‐Ford S, Hofman P. Use of the 22C3 anti‐programmed death‐ligand 1 antibody to determine programmed death‐ligand 1 expression in cytology samples obtained from non‐small cell lung cancer patients. Cancer Cytopathol. 2018;126:264‐274. [DOI] [PubMed] [Google Scholar]
- 50.Humphries MP, Mcquaid S, Craig S, et al. Critical appraisal of PD‐L1 reflex diagnostic testing: current standards and future opportunities. J Thorac Oncol. 2019;14:45‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Xu H, Bratton L, Nead M, Russell D, Zhou Z. Comparison of programmed death‐ligand 1 (PD‐L1) immunostain for nonsmall cell lung carcinoma between paired cytological and surgical specimens. CytoJournal. 2018;15:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gosney JR, Boothman AM, Ratcliffe M, Kerr KM. Cytology for PD‐L1 testing: a systematic review. Lung Cancer. Elsevier Ireland Ltd. 2020;141:101‐106. [DOI] [PubMed] [Google Scholar]
- 53.Russell‐Goldman E, Kravets S, Dahlberg SE, Sholl LM, Vivero M. Cytologic‐histologic correlation of programmed death‐ligand 1 immunohistochemistry in lung carcinomas. Cancer Cytopathol. 2018;126:253‐263. [DOI] [PubMed] [Google Scholar]
- 54.Gagné A, Orain M, Ionescu D, Tsao MS, Joubert D, Joubert P. Comprehensive assessment of PD‐L1 immunohistochemistry on paired tissue and cytology specimens from non‐small cell lung cancer. Lung Cancer. 2020;146:276‐284. [DOI] [PubMed] [Google Scholar]
- 55.Sinclair W, Kobalka P, Ren R, et al. Interobserver agreement in programmed cell death‐ligand 1 immunohistochemistry scoring in nonsmall cell lung carcinoma cytologic specimens. Diagn Cytopathol. 2021;49(2):219‐225. [DOI] [PubMed] [Google Scholar]
- 56.Kuempers C, van der Linde LIS, Reischl M, et al. Comparison of PD‐L1 expression between paired cytologic and histologic specimens from non‐small cell lung cancer patients. Virchows Arch. 2020;476:261‐271. [DOI] [PubMed] [Google Scholar]
- 57.Roy‐Chowdhuri S, Dacic S, Ghofrani M, et al. Collection and handling of thoracic small biopsy and cytology specimens for ancillary studies. Arch Pathol Lab Med. 2020;144:933‐958. [DOI] [PubMed] [Google Scholar]
- 58.Cheung CC, Barnes P, Bigras G, et al. Fit‐for‐purpose PD‐L1 biomarker testing for patient selection in immuno‐oncology: guidelines for clinical laboratories from the Canadian Association of Pathologists‐Association Canadienne des Pathologistes (CAP‐ACP). Appl Immunohistochem Mol Morphol. 2019;27:699‐714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Koelzer VH, Sirinukunwattana K, Rittscher J, Mertz KD. Precision immunoprofiling by image analysis and artificial intelligence. Virchows Archiv. 2019;474(4):511‐522.Springer Verlag. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lu S, Stein JE, Rimm DL, et al. Comparison of biomarker modalities for predicting response to PD‐1/PD‐L1 checkpoint blockade. JAMA Oncol. 2019;5(8):1195. American Medical Association. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Humphries MP, Bingham V, Abdullahi Sidi F, et al. Improving the diagnostic accuracy of the PD‐L1 test with image analysis and multiplex hybridization. Cancers (Basel). 2020;12(5):1114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sidi FA, Bingham V, Craig SG, et al. PD‐L1 multiplex and quantitative image analysis for molecular diagnostics. Cancers (Basel). 2021;13:1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Allgäuer M, Budczies J, Christopoulos P, et al. Implementing tumor mutational burden (TMB) analysis in routine diagnostics—a primer for molecular pathologists and clinicians. Transl Lung Cancer Res. 2018;7(5):703‐715.AME Publishing Company. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Büttner R, Longshore JW, López‐Ríos F, et al. Implementing TMB measurement in clinical practice: considerations on assay requirements. ESMO Open. 2019;4(1):e000442. BMJ Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Pepe F, Pisapia P, Gristina V, et al. Tumor mutational burden cytological samples: a pilot study. Cancer Cytopathol. 2020. 10.1002/cncy.22400. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 66.Alborelli I, Bratic Hench I, Chijioke O, et al. Robust assessment of tumor mutational burden in cytological specimens from lung cancer patients. Lung Cancer. 2020;149:84‐89. [DOI] [PubMed] [Google Scholar]
- 67.Marinelli D, Mazzotta M, Scalera S, et al. KEAP1‐driven co‐mutations in lung adenocarcinoma unresponsive to immunotherapy despite high tumor mutational burden. Ann Oncol. 2020;31:1746‐1754. [DOI] [PubMed] [Google Scholar]
- 68.Ko J, Oh J, Ahmed MS, Carlson JCT, Weissleder R. Ultra‐fast cycling for multiplexed cellular fluorescence imaging. Angew Chemie Int Ed. 2020;59:6839‐6846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pai SI, Faquin WC, Sadow PM, Pittet MJ, Weissleder R. New technology on the horizon: fast analytical screening technique FNA (FAST‐FNA) enables rapid, multiplex biomarker analysis in head and neck cancers. Cancer Cytopathol. 2020;128:782‐791. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing not applicable to this article as no datasets were generated or analysed during the current study.
