Context.—

The rapid advances in targeted therapies in non–small cell lung cancer (NSCLC) make the optimization and implementation of cytology specimens for molecular testing a priority. Up to 70% of patients with NSCLC are diagnosed at advanced stages and tissue biopsies often cannot be taken. Although cytology samples provide high-quality material for molecular testing, molecular cytopathology is not yet well known or widely used.

Objective.—

To report the many advances in molecular cytopathology and the suitability and utility of cytology samples in molecular and genetic testing of NSCLC.

Data Sources.—

Data sources comprised published peer-reviewed literature and personal experience of the authors.

Conclusions.—

Molecular testing can be performed on cytologic specimens, especially on direct smears. Rapid on-site evaluation by cytopathologists has improved the adequacy and the management of cytology samples for molecular testing. Mutational profiling of NSCLC using next-generation sequencing can be performed on cytology samples from very small amounts of DNA. Fluorescence in situ hybridization assays on cytology specimens, including stained direct smear, offer some distinct advantages over their histologic counterpart, and are used to detect ALK and ROS1 rearrangements in NSCLC. Cytology specimens allow assessment of the entire tumor cell nucleus, avoiding signal loss from truncation artifacts. The use of cytology samples for assessing programmed death ligand-1 protein expression is currently being developed. Protocols for bisulfite conversion and DNA droplet digital polymerase chain reaction assays have been optimized for cytology smear to investigate aberrant DNA methylation of several NSCLC-related genes.

Lung cancer is one of the most prevalent cancers and results in the largest number of cancer-related deaths in the world.1  More than 85% of those cases are currently classified as non–small cell lung cancer (NSCLC), for which the predicted 5-year survival rate is 15.9%—a figure that has only marginally improved during the past few decades.2  Non–small cell lung cancer is currently defined by histopathologic features (Figure 1, A through E). The 2 most common NSCLC histopathologic phenotypes are adenocarcinoma (ADC; ∼50%; Figure 1, B) and squamous cell carcinoma (SCC; ∼40%; Figure 1, C). Adenocarcinomas often have glandular histology and express markers that are consistent with an origin in the distal airways, including thyroid transcription factor 1 (also known as NKX2-1; Figure 1, D) and cytokeratin 7.3  Squamous cell carcinomas are characterized by squamous differentiation, which is more reminiscent of the pseudostratified columnar epithelium that lines the trachea and upper airways.3,4  Squamous cell carcinomas are distinguished from ADCs by immunostaining for p63 or p40 (Figure 1, E) and high-molecular-weight keratins (cytokeratin 5/6).3,5 

Figure 1

Next-generation sequencing (NGS) analysis on direct smears. A, Stained cytologic direct smear with selected areas for NGS analysis. Macro-dissection can be easily performed on smears. B and D, Area with lung adenocarcinoma diagnosed at rapid on-site examination with nuclear expression of thyroid transcription factor 1 (TTF-1). C and E, Area with squamous cell carcinoma with nuclear expression of p40. F and G, Representations of the reads aligned to the reference genome (Integrative Genomics Viewer software v 2.1, Broad Institute, Cambridge, Massachusetts) for an epidermal growth factor receptor exon 19 deletion (c.2235_2249del p.E746_A750del) in both components, adenocarcinoma and squamous cell carcinoma (Papanicolaou, original magnifications ×20 [B and C] and ×40 [D and E]).

Figure 1

Next-generation sequencing (NGS) analysis on direct smears. A, Stained cytologic direct smear with selected areas for NGS analysis. Macro-dissection can be easily performed on smears. B and D, Area with lung adenocarcinoma diagnosed at rapid on-site examination with nuclear expression of thyroid transcription factor 1 (TTF-1). C and E, Area with squamous cell carcinoma with nuclear expression of p40. F and G, Representations of the reads aligned to the reference genome (Integrative Genomics Viewer software v 2.1, Broad Institute, Cambridge, Massachusetts) for an epidermal growth factor receptor exon 19 deletion (c.2235_2249del p.E746_A750del) in both components, adenocarcinoma and squamous cell carcinoma (Papanicolaou, original magnifications ×20 [B and C] and ×40 [D and E]).

Close modal

Recent advances in next-generation sequencing (NGS) and other high-throughput genomic profiling platforms have transformed the understanding of NSCLC from histopathologic descriptions to precise molecular and genetic identities, thereby forming the rationales for novel therapeutic strategies (Figure 1, A through G).3  Additionally, the very encouraging progress of immunotherapy in patients with NSCLC has advanced the codevelopment of diagnostics assays required for the use of associated drugs.6  There are several drugs approved by the US Food and Drug Administration and the European Medicines Agency with companion diagnostics that assist with therapeutic selection for treatment of patients with NSCLC.7,8  For instance, programmed death ligand-1 (PD-L1) protein expression, as detected by immunohistochemistry (IHC) testing, has been used as a predictive biomarker of response to programmed death receptor-1 (PD-1) and PD-L1 immune checkpoint inhibitors in NSCLC.9,10 

The 2013 College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology clinical practice guidelines for molecular testing of lung cancer include classifying NSCLC in ADC and SCC by morphology, by limited immunoperoxidase staining, or by both methods, as well as saving adequate material for molecular testing.11  Consequently, pathologists are increasingly facing the challenges of performing more tests on smaller specimens.

Patients with NSCLC are often diagnosed with advanced-stage disease, and are not, therefore, candidates for surgical resection of the primary tumor.12  For these patients, the only pathologic material guiding systemic therapy should be small biopsies or cytologic specimens.13  Several studies have demonstrated and validated the utility of cytologic specimens in molecular testing.1417  The rapid advances in targeted therapies in NSCLC make the optimization and implementation of molecular studies in cytology samples a priority. Although cytology samples provide high-quality material for molecular analysis, molecular cytopathology is not yet well known or widely used.

Molecular cytopathology is a promising field able to determine the molecular profile of a tumor and may eliminate the need for invasive tissue biopsies, allowing the detection of genetic alterations throughout the course of therapy.15  This is particularly important in lung cancer, where up to 70% of patients are diagnosed at advanced stages and tissue biopsies often cannot be taken.11  Additionally, a third of patients may have biopsies with insufficient tumor tissue for molecular studies.18  In such a scenario, cytology, especially fine-needle aspiration (FNA), is placed at the front line in the management of patients with NSCLC. In this review, we describe the use of cytology samples, especially direct smears, in molecular studies, including epidermal growth factor receptor (EGFR), KRAS, and BRAF mutational testing; ALK and ROS1 fluorescence in situ hybridization (FISH) testing; NGS; PD-L1 IHC; and DNA methylation detection.

Lung ADC is characterized by recurrent mutations in EGFR kinase and rearrangements involving anaplastic lymphoma kinase (ALK).3  These genetic alterations are successfully being targeted with specific drugs. Molecular testing for EGFR and ALK is required to select patients with lung ADC for EGFR- and ALK-targeted tyrosine kinase inhibitor therapy, respectively.12  KRAS and BRAF function downstream of EGFR in the signaling pathway, and activating mutations have been described to be usually mutually exclusive in EGFR-mutated tumors.19 

Activating EGFR mutations are located in the tyrosine kinase domain (exon 18–21). Deletions in exon 19 and the point mutation L858R in exon 21 are the most common activating mutations usually found in lung ADCs, never smokers, females, and Asian patients.20  Secondary EGFR mutations have also been shown to be involved in resistance to EGFR-directed tyrosine kinase inhibitor therapy.21  About 50% of cases of acquired resistance are related to a secondary T790M mutation in exon 20 of the EGFR gene.22  ALK gene rearrangements are often found in never smokers or light ex-smokers and in younger patients.23 

KRAS, a Ras GTPase acting as a key molecular switch in the EGFR signaling pathway, is mutated at codons 12, 13, and 61 in 16% to 40% of patients with lung ADC.19,24  KRAS mutations have been shown to be associated with poor response to EGFR-directed tyrosine kinase inhibitor therapy.12 

BRAF, a serine/threonine kinase, is activated by somatic point mutations in exon 15, including V600E, in 1% to 2% of patients with lung cancer.25 

Further recurrent mutations and amplifications in many potentially targetable oncogenes have been found in lung ADC, including human epidermal growth factor receptor 2 (HER2; also known as ERBB2), mesenchymal-epithelial transition factor (MET), and fibroblast growth factor receptors 1 and 2 (FGFR1 and FGFR2), as well as fusion oncogenes involving the c-ros oncogene 1 (ROS1) receptor tyrosine kinase, neuregulin 1 (NRG1), neurotrophic tyrosine kinase receptor type 1 (NTRK1), and rearranged during transfection (RET) gene.3  These oncogenic changes may predict sensitivity to clinical inhibitors and jointly account for most cases of lung ADC.3  Because of the similarity between the tyrosine kinase domains of ALK and ROS1, many ALK inhibitors also inhibit ROS1. Lung cancers with ROS1 translocations respond to crizotinib and alectinib, which are also used in ALK-positive lung cancers.8 

Activating mutations in EGFR and ALK gene rearrangement are typically not present in squamous NSCLC.3  Lung SCCs are characterized by complex genomic alterations with recurrent mutations in several genes, including TP53, NFE2L2, KEAP1, BAI3, FBXW7, GRM8, MUC16, RUNX1T1, STK11, ERBB4, DDR2, and FGFR1 amplification.26 

It has been described that DNA hypomethylation leads to elevated mutation rates and is suggested to be an early step in carcinogenesis.3  Epigenetic silencing of genes through DNA promoter methylation of cytosines (C) affects the expression of several genes during initiation and development of lung cancer. Aberrant DNA methylation consists of the addition of a methyl group to the 5′ carbon of C within C-guanine dinucleotide sequences (CpGs), which are mainly concentrated in regions known as CpG islands. In general, methylation studies are based on DNA bisulfite conversion.27,28  The C in CpG is resistant to bisulfite conversion and remains a C upon bisulfite treatment. The C outside CpG will be changed to thymidine by bisulfite treatment.27,28 

The terms DNA hypermethylation and hypomethylation are applied to describe the aberrant methylated state of most CpG sites in a specific sequence. DNA methylation level is often described to be inversely correlated to gene expression level.29 

Silencing of genes by aberrant promoter DNA hypermethylation is likely to influence the transformation of lung epithelial cells.28  In lung cancer, CDKN2A, which encodes INK4A (also known as p16) and ARF (also known as p14), is often silenced through aberrant promoter hypermethylation. The CDKN2A gene is inactivated in ∼67% of ADCs and ∼70% of SCCs.30  Aberrant hypermethylation of p16/CDKN2A is an early event in lung cancer and a potential biomarker for early diagnosis.27,28 

Aberrant patterns of DNA methylation of certain genes are associated with specific histologic phenotypes and are currently used as diagnostic markers. Aberrant short stature homeobox 2 (SHOX2) DNA methylation is being used in lung cancer as a prognostic biomarker as well as a diagnostic marker.3133  Patients with lung cancer and aberrant SHOX2 methylation have an adverse overall survival and the hypermethylation levels significantly correlate with a worse outcome.33  Recently, our research group has described that the transmembrane protease serine 4 (TMPRSS4) promoter region is often hypomethylated in ADC and SCC lung cancer subtypes compared with normal lung tissue, suggesting that the methylation levels of the TMPRSS4 promoter might be useful as a diagnostic tool in lung cancer.34 

Tumor heterogeneity—genetic, cellular, and epigenetic—characterizes lung cancer.3  For patients with multiple, apparently separate, primary lung ADCs, each tumor may be tested but testing of multiple different areas within a single tumor is not necessary.35 

Paraffin-embedded tumor biopsy, generally taken from the primary tumor or metastatic lesion, is a diagnostic technique commonly used to determine the molecular profile of a tumor, for instance at a single time point before treatment commences. Most patients with lung cancer are diagnosed with advanced-stage disease or with metastatic lesions, which precludes them from surgical tumor resection. For these patients, the diagnosis is based on the analysis of cytology samples, especially FNA, obtained through minimally invasive procedures.

Cytology samples offer advantages over tissue biopsies for routine molecular testing: (1) cytology direct smears can be either air dried or fixed in alcohol; (2) they allow for rapid on-site adequacy assessment; and (3) DNA is readily extractable, providing high-quality DNA even if archived for a prolonged period.17,36  Alcohol-fixed and air-dried smears are used for DNA extraction. In both cytologic preparations, the number of intact nuclei that are suitable for FISH analysis is higher and DNA appears to be better preserved.16,37  Direct smear allows assessment of tumor content, ensuring adequacy for DNA extraction for molecular studies.17 

There are a variety of minimally invasive procedures depending on the location of the tumor and/or metastasis. The most-used sampling procedures are computed tomography–guided FNA of the lung (frequently coupled with core needle biopsies), bronchoscopy-guided FNA with or without electromagnetic navigation, endobronchial ultrasound–guided FNAs of lymph nodes, endoscopic ultrasound–guided FNA, and direct aspiration of superficial metastatic lesions.38,39 

Cytology samples also offer some advantages over their histologic counterpart. Rapid on-site evaluation can redirect the procedure in order to obtain samples adequate for ancillary studies. Rapid on-site evaluation done by a cytopathologist has improved the adequacy of FNA, allowing the assessment of cellular content, tumor cell yields, and adequate material for molecular testing (Figure 2, A through D). Rapid on-site evaluation maximizes cytology samples for molecular testing, including important steps for samples acquisition, processing, and selection.40 

Figure 2

Anaplastic lymphoma kinase (ALK) gene rearrangement by fluorescence in situ hybridization (FISH) in non–small cell lung cancer on direct smears. A, Stained cytologic direct smear with a selected area for hybridization. B, Lung adenocarcinoma diagnosed at rapid on-site examination. C, ALK FISH testing is interpreted as positive when 15% or more tumor cell nuclei demonstrate isolated green and red signals or isolated red signals. D, ALK-positive signal patterns (arrows) (Papanicolaou, original magnification ×40 [B]; original magnifications ×63 [C] and ×100 [D]).

Figure 2

Anaplastic lymphoma kinase (ALK) gene rearrangement by fluorescence in situ hybridization (FISH) in non–small cell lung cancer on direct smears. A, Stained cytologic direct smear with a selected area for hybridization. B, Lung adenocarcinoma diagnosed at rapid on-site examination. C, ALK FISH testing is interpreted as positive when 15% or more tumor cell nuclei demonstrate isolated green and red signals or isolated red signals. D, ALK-positive signal patterns (arrows) (Papanicolaou, original magnification ×40 [B]; original magnifications ×63 [C] and ×100 [D]).

Close modal

Although there are many studies validating any form of cytology samples as a reliable source for high-quality DNA,17,39  in reality, we commonly face the challenge of cytology samples with limited amounts of tumor cells. Therefore, validation studies are essential steps for any molecular testing on cytologic samples.38,41  Cytologic samples pose unique challenges in validation, partly because of the lack of standardization among laboratories for specimen collection, processing, and staining methodologies.11,38,42 

We have previously demonstrated that on-site evaluation by a cytopathologist is crucial for obtaining adequate cytology samples for molecular studies.37  When preanalytic factors are correctly implemented, cytology samples are an excellent source of DNA for molecular testing. When needed, microdissection or macro-dissection can be easily performed (Figure 1, A). However, some types of specimens, such as brushing and exfoliative samples, have less neoplastic content, and therefore microdissection is not an option. Good-quality DNA can be extracted from stained smears, fresh fluids, cell block, and ThinPrep (Hologic Inc, Marlborough, Massachusetts). Previously, we have demonstrated that the amount of DNA extracted from cytology samples is similar to that extracted from core needle biopsies.37  Core needle biopsy samples yield a higher concentration of DNA than FNA samples, but the DNA quality is similar.37  Although most commercially available DNA extraction kits have been validated for unstained slides from formalin-fixed, paraffin-embedded samples, they can also be used in cytology samples with excellent results.16 

Tissue biopsy is often not an option for patients with advanced stage cancer or metastatic NSCLC.43  Then, FNA cytology offers a minimally invasive, accurate, simple, and cost-effective method for diagnosing NSCLC.39  Moreover, rapid on-site evaluation helps to overcome the limitations of sampling in FNA cytology. The relatively lower amount of DNA yielded by a cytologic smear is compensated for by more sensitive genomic platforms, including NGS, that require less sample material and time.15,36 

We have previously demonstrated that the sensitivity and specificity of EGFR and KRAS mutation analyses by Sanger sequencing, real-time polymerase chain reaction (PCR), and Cobas (Roche Molecular Systems Inc, Pleasanton, California) EGFR mutation test from cytologic smears and core needle biopsies are similar.16,37 

It has been shown that PCR analysis using cytologic slides has better results when compared with biopsy specimens, regardless of the amount of tumor cells analyzed.44,45  This could be explained by differences in fixation method used in the 2 types of specimens, favoring alcohol fixatives over formalin for mutational studies.44  Therefore, as cytology samples are often collected in non–formalin-based fixatives, they offer the possibility of testing of higher-quality nucleic acids.42 

In general, the amount of extracted DNA depends on the technique used.16  For DNA extraction, cells from cytology smears can be removed from glass slides by scraping. It has been shown that scraping yields significantly more DNA than cell lifting.46  We have previously shown that cytology-based EGFR and KRAS mutational tests are as effective as tumor tissue–based tests. In addition, various types of cytology specimens, including FNA, bronchoscopy-guided FNA, direct aspirations, and pleural effusion, are shown to be suitable for molecular testing.37,45  Direct smear preparations provide a high overall tumor fraction and higher average DNA yields than cell block preparation.37  Routinely in our laboratory, after classic xylene coverslip removal, Papanicolaou-stained smears are first rinsed in alcohol and then scraped into Eppendorf tubes. Slides are not destained prior to DNA extraction.16 

If possible, it is recommended that cytology smears be digitally scanned before performing a molecular analysis to record the most representative and diagnostic slide.14,36,40 

Next-generation sequencing technology is a high-throughput method that allows massive parallel sequencing of multiple targeted genomic regions in multiple samples.47  It requires a very low input of DNA, in contrast to traditional sequencing methods.47  A variety of genomic alterations can be detected with high accuracy and sensitivity. Moreover, it allows quantitative evaluation of the mutated allele.15,47 

Next-generation sequencing results from stained cytologic samples are similar to the ones from their snap-frozen, dry pellet counterparts, indicating that preanalytical factors, such as fixation and staining of cytologic specimens, do not induce significant detectable modifications in nucleic acid.48 

In a proof-of-concept study, an NGS gene panel that targets 568 clinically relevant mutations in 6 genes (EGFR, KRAS, NRAS, BRAF, cKIT, and PDGFRα) was designed and tested using samples with a very low DNA content.41  The NGS assay showed excellent sensitivity and specificity, being able to detect relevant mutations in cell-free DNA purified from samples with a very low DNA, including serum and plasma.14 

In the last 2 years, our institution has adopted the Oncomine Focus Assay (Thermo Fisher Scientific, San Francisco, California), a targeted, multi-biomarker NGS assay that analyzes clinically relevant gene alterations including single-nucleotide variants, short insertions and deletions, somatic copy number variations, and gene fusions from DNA and RNA in a single workflow (Figure 1, F and G). This commercially available assay with low DNA requirements (∼10 ng, approximately 1500 intact cells) is being increasingly adopted in many laboratories for testing samples with limited material. This assay is able to detect amplifications and fusions in addition to mutations, including ALK and ROS1 gene fusions.

The FISH assay is used for detection of ALK and ROS1 rearrangements, using dual-labeled, break-apart probes.11,49  Direct smears are currently being used for FISH assays. Nuclei on direct smears are not truncated, which allows for the detection of the true number of FISH signals (Figure 2, A and B).50 

Immunohistochemistry detects the expression of ALK and ROS1 protein and is a screening tool for testing NSCLC samples for ALK and ROS1 rearrangement.11,18,49  ROS1 IHC should be followed by confirmatory FISH testing.18  ALK or ROS1 FISH testing is positive if rearrangement is seen in at least 15% of cancer cells.18  In specimens positive for ALK (Figure 2, C and D), (1) if ALK rearrangement has occurred, nuclei will contain “broken-apart” red and green signals, which appear separated by at least 2 signal diameters; and (2) if ALK deletion has occurred, nuclei will contain single red signals.23  In specimens positive for ROS1, (1) in the classical pattern, 1 fusion signal (native ROS1) and broken-apart green and red signals are seen; (2) in the atypical pattern, 1 fusion signal (native ROS1) and 1 green signal without the corresponding red signal are seen.49 

Cell block preparation, Diff-Quik– and Papanicolaou-stained smears, and ThinPrep slides are used for ALK rearrangement analysis by FISH.38  Cell blocks have been traditionally used for ALK rearrangement analysis, because the same protocols as for histology can be used. Ethanol-based fixation of cytologic smears is suitable for FISH assay, although laboratory protocols may well differ from those for formalin-fixed tissue.35 

In our laboratory, we favor direct smears over cell blocks for FISH analysis (Figure 2, A through D). On direct smears, the entire cell nucleus is analyzed, eliminating signal loss from truncation artifacts.45  We select an area of the smear where cells are not overlapped in order to analyze entire and individual nuclei. When performing a FISH assay, we mark the area previously selected and add the hybridization probe only in this area. It is neither necessary nor cost-effective to use the whole slide. In our experience, overlapped cells and areas of nuclear debris should be avoided. Monolayered areas with at least 100 tumor cells with entire nuclei should be used to assess the FISH signals.

The pathologic diagnosis of lung cancer has become a multistep process.43  Although assays have not been validated on cytology specimens, cytopathologists are increasingly being asked to assess the expression of PD-L1 in NSCLC to select patients for treatment with PD-1/PD-L1 inhibitors.

PD-L1 protein expression, as detected by IHC testing, has been used as a predictive biomarker assay for anti–PD-1/PD-L1 therapies.51  An assay for assessing PD-L1 expression is approved by the US Food and Drug Administration and the European Medicines Agency for both first-line and second-line therapy with pembrolizumab.7 

The tissue-handling procedure for PD-L1 IHC does not differ from that for other diagnostic or predictive IHC markers.9  There are several recommendations regarding preanalytic conditions for preparation and storage of tissues for future PD-L1 staining. Critical steps are (1) minimum fixation time (6–48 hours for biopsies and 24–48 hours for resections), (2) storage time for cut sections (fewer than 2 months), and (3) storage time for blocks (less than 3 years).52  PD-L1 IHC on decalcified tissue should be avoided or interpreted with great caution until further validation studies on PD-L1 IHC have become available.

PD-L1 protein expression is seen on dendritic cells, macrophages, mast cells, T- and B-lymphocytes, endothelial cells, and tumor cells. The different commercial assays available define PD-L1 staining as complete circumferential or partial linear cell membrane staining of tumor cells at any intensity.9,43  Only cytoplasmic staining in tumor cells is not considered positive for scoring purposes. PD-L1 expression on tumor cells and immune cells is scored as a percentage of total cells.

The use of cytology samples for assessing PD-L1 protein expression is not advocated because none of the assays are validated for this purpose. PD-L1 staining and quantitation on cytology samples could be feasible, provided that appropriate protocols and validation studies are in place. In the case of alcohol-fixed samples, the PD-L1 IHC protocol may require some adjustments.

Emerging data from cell blocks and matched histologic specimens suggest that cytologic material is as good as histologic material for PD-L1 IHC.53  Actually, several preliminary studies have shown that the development of cytologic criteria and interpretation of the PD-L1 IHC on cell blocks is feasible, but may require adjustments of standard PD-L1 interpretation parameters to account for differences in processing method.6,53  In cytology samples, quantification of PD-L1–positive immune cells will likely be challenging.9  In our institution, cytology samples are an important method for diagnosing lung cancer. We are currently optimizing PD-L1 staining on direct smear (Figure 3). The PD-L1 IHC is being performed and evaluated in the same way as the corresponding clinically validated commercial assay. We are evaluating 100 or more viable tumor cells, and positive PD-L1 staining is defined as complete circumferential or partial linear cell membrane staining of tumor cells at any intensity (Figure 3).

Figure 3

Programmed death ligand-1 (PD-L1) immunohistochemistry analysis of cytology direct smears. PD-L1–positive tumor cells with complete circumferential and/or partial linear membrane staining (original magnification ×40).

Figure 4. Methylation detection by droplet digital polymerase chain reaction (ddPCR). A FAM-labeled probe was designed to bind methylated and a HEX-labeled probe to bind nonmethylated cytosine-guanine dinucleotide sequence islands in the promoter region of the transmembrane protease, serine 4 (TMPRSS4) gene. Very low input of bisulfite-converted genomic DNA (as low as 0.5 ng) can be used to detect TMPRSS4 promoter methylation/nonmethylation. The blue dots represent individual droplets that contain at least 1 copy of the methylated DNA; the green dots represent droplets that contain at least 1 copy of the nonmethylated DNA.

Figure 3

Programmed death ligand-1 (PD-L1) immunohistochemistry analysis of cytology direct smears. PD-L1–positive tumor cells with complete circumferential and/or partial linear membrane staining (original magnification ×40).

Figure 4. Methylation detection by droplet digital polymerase chain reaction (ddPCR). A FAM-labeled probe was designed to bind methylated and a HEX-labeled probe to bind nonmethylated cytosine-guanine dinucleotide sequence islands in the promoter region of the transmembrane protease, serine 4 (TMPRSS4) gene. Very low input of bisulfite-converted genomic DNA (as low as 0.5 ng) can be used to detect TMPRSS4 promoter methylation/nonmethylation. The blue dots represent individual droplets that contain at least 1 copy of the methylated DNA; the green dots represent droplets that contain at least 1 copy of the nonmethylated DNA.

Close modal

Investigating promoter methylation in patients with lung cancer is, in general, challenging. In the majority of cases, lung cancer is linked to smoking, and smoking itself leads to gene methylation changes.54  Therefore, finding good and reliable methylation-specific biomarkers may rely on the choice of an appropriate control group or on having a combination of 2 or more genes that could reduce the probability of having a false-positive or false-negative result.

The detection of the methylation level of certain genes is currently being used as a diagnostic tool and a prognostic biomarker in lung cancer, but only a few have been validated in cytology samples. In general, methylation studies are based on DNA bisulfite conversion, which leads to DNA fragmentation. After bisulfite conversion, the fraction of circulating tumor cell DNA obtained from a cytology sample is often very low and can be difficult to detect with current approaches. Protocols for bisulfite conversion and DNA methylation–specific PCR have been optimized for cytology smear.55 

Quantitative methylation-specific real-time PCR is currently being used to detect aberrant patterns of DNA methylation of several genes, including p16-INK4a and SHOX2.33,56,57  This technique needs a relatively high amount of bisulfite-converted DNA (∼50 ng), which is often difficult to obtain from small specimens. Additionally, bisulfite conversion will reduce even more the final fraction of good DNA needed for methylation studies. Therefore, the development of new assays using more sensitive and quantitative techniques with a very low input of DNA are needed.

Recently, droplet digital PCR (ddPCR) has been shown to reliably detect genomic alteration at low allele frequencies and with high sensitivity (Figure 4).58  Droplet digital PCR is achieved by partitioning a standard PCR into many replicate reactions prior to thermal cycling.59  This results in the physical separation of DNA molecules, thus allowing binary classification of compartments that either contain or do not contain the amplified target sequence.59  Copy numbers can then be calculated by applying the Poisson distribution.60  Therefore, ddPCR allows absolute measure of target DNA.60 

Using ddPCR for the detection of DNA methylation in the TMPRSS4 promoter gene in bisulfite-converted samples, we were able to detect TMPRSS4 promoter hypomethylation with very low input of DNA, as low as 0.5 ng (3.3 copies/μL; Figure 4). It has also been shown that ddPCR can detect amounts as low as 0.5% methylated DNA in the background of a nonmethylated control DNA. Therefore, ddPCR is a very promising tool, suitable for measuring methylation in samples with low DNA concentrations, including cytology samples.

The promoter DNA methylation ddPCR assay for cytology sample analysis could improve lung cancer diagnosis and staging.

Adequate management of cytology samples, including direct smears, is beneficial for patients with NSCLC, especially when tissue biopsy is not an option. Cytology samples are suitable for determining the genetic profile of a tumor, eliminating the need for invasive tissue biopsies. This is particularly important in lung cancer, where a very high percentage of patients may have no biopsies taken or have biopsies with insufficient tumor tissue for molecular studies. Rapid on-site evaluation from FNA procedures has improved the adequacy and management of cytology samples.

More sensitive approaches, such as NGS and ddPCR, combined with minimally invasive procedures, such as FNA, may be used to better understand the disease, treat patients, and monitor treatment response in NSCLC patients.

We thank Nerea Gomez, BSc; Mariam Maset, BSc; María Mercedes Aguirre, BSc; and Maria Eugenia Echarri, BSc, for the expert technical help. We also thank Maria Villalba, MSc; Miriam Redrado, MSc, PhD; and Francisco Expósito, MSc, for the methylation studies. We thank CIMA Labs Diagnostics for assistance in the molecular work. This work was supported by PIUNA (University of Navarra; ref. 12028701) and ERA-NET on Translational Cancer Research (TRANSCAN; ref. AC14/00034).

1
Siegel
R.
Naishadham
D.
Jemal
A.
Cancer statistics, 2013
.
CA Cancer J Clin
.
2013
;
63
(
1
):
11
30
.
2
Li
B.
Lu
Q.
Song
ZG.
et al.
Functional analysis of DNA methylation in lung cancer
.
Eur Rev Med Pharmacol Sci
.
2013
;
17
(
9
):
1191
1197
.
3
Chen
Z.
Fillmore
CM.
Hammerman
PS.
Kim
CF.
Wong
KK.
Non-small-cell lung cancers: a heterogeneous set of diseases
.
Nat Rev Cancer
.
2014
;
14
(
8
):
535
546
.
4
Davidson
MR.
Gazdar
AF.
Clarke
BE.
The pivotal role of pathology in the management of lung cancer
.
J Thorac Dis
.
2013
;
5
(
suppl 5
):
S463
S478
.
5
Rekhtman
N.
Ang
DC.
Sima
CS.
Travis
WD.
Moreira
AL.
Immunohistochemical algorithm for differentiation of lung adenocarcinoma and squamous cell carcinoma based on large series of whole-tissue sections with validation in small specimens
.
Mod Pathol
.
2011
;
24
(
10
):
1348
1359
.
6
Kerr
KM.
Tsao
MS.
Nicholson
AG.
et al.
Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art?
J Thorac Oncol
.
2015
;
10
(
7
):
985
989
.
7
Reck
M.
Rodriguez-Abreu
D.
Robinson
AG.
et al.
Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer
.
N Engl J Med
.
2016
;
375
(
19
):
1823
1833
.
8
Mazieres
J.
Zalcman
G.
Crino
L.
et al.
Crizotinib therapy for advanced lung adenocarcinoma and a ROS1 rearrangement: results from the EUROS1 cohort
.
J Clin Oncol
.
2015
;
33
(
9
):
992
999
.
9
Cree
IA.
Booton
R.
Cane
P.
et al.
PD-L1 testing for lung cancer in the UK: recognizing the challenges for implementation
.
Histopathology
.
2016
;
69
(
2
):
177
186
.
10
Shien
K.
Papadimitrakopoulou
VA.
Wistuba II. Predictive biomarkers of response to PD-1/PD-L1 immune checkpoint inhibitors in non-small cell lung cancer
.
Lung Cancer
.
2016
;
99
:
79
87
.
11
Lindeman
NI.
Cagle
PT.
Beasley
MB.
et al.
Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology
.
Arch Pathol Lab Med
.
2013
;
137
(
6
):
828
860
.
12
Azzoli
CG.
Temin
S.
Aliff
T.
et al.
2011 focused update of 2009 American Society of Clinical Oncology clinical practice guideline update on chemotherapy for stage IV non-small-cell lung cancer
.
J Clin Oncol
.
2011
;
29
(
28
):
3825
3831
.
13
Rekhtman
N.
Brandt
SM.
Sigel
CS.
et al.
Suitability of thoracic cytology for new therapeutic paradigms in non-small cell lung carcinoma: high accuracy of tumor subtyping and feasibility of EGFR and KRAS molecular testing
.
J Thorac Oncol
.
2011
;
6
(
3
):
451
458
.
14
Malapelle
U.
Mayo de-Las-Casas
C.
Rocco
D.
et al.
Development of a gene panel for next-generation sequencing of clinically relevant mutations in cell-free DNA from cancer patients
.
Br J Cancer
.
2017
;
116
(
6
):
802
810
.
15
Kanagal-Shamanna
R.
Portier
BP.
Singh
RR.
et al.
Next-generation sequencing-based multi-gene mutation profiling of solid tumors using fine needle aspiration samples: promises and challenges for routine clinical diagnostics
.
Mod Pathol
.
2014
;
27
(
2
):
314
327
.
16
Lozano
MD.
Zulueta
JJ.
Echeveste
JI.
et al.
Assessment of epidermal growth factor receptor and K-ras mutation status in cytological stained smears of non-small cell lung cancer patients: correlation with clinical outcomes
.
Oncologist
.
2011
;
16
(
6
):
877
885
.
17
Knoepp
SM.
Roh
MH.
Ancillary techniques on direct-smear aspirate slides: a significant evolution for cytopathology techniques
.
Cancer Cytopathol
.
2013
;
121
(
3
):
120
128
.
18
DiStasio
M.
Chen
Y.
Rangachari
D.
Costa
DB.
Heher
YK.
VanderLaan
PA.
Molecular testing turnaround time for non-small cell lung cancer in routine clinical practice confirms feasibility of CAP/IASLC/AMP guideline recommendations: a single-center analysis
.
Clin Lung Cancer
.
2017
.
18
(
5
):
e349
e356
.
19
Ding
L.
Getz
G.
Wheeler
DA.
et al.
Somatic mutations affect key pathways in lung adenocarcinoma
.
Nature
.
2008
;
455
(
7216
):
1069
1075
.
20
Sharma
SV.
Bell
DW.
Settleman
J.
Haber
DA.
Epidermal growth factor receptor mutations in lung cancer
.
Nat Rev Cancer
.
2007
;
7
(
3
):
169
181
.
21
Kobayashi
S.
Boggon
TJ.
Dayaram
T.
et al.
EGFR mutation and resistance of non-small-cell lung cancer to gefitinib
.
N Engl J Med
.
2005
;
352
(
8
):
786
792
.
22
Gazdar
AF.
Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors
.
Oncogene
.
2009
;
28
(
suppl 1
):
S24
S31
.
23
Kim
H.
Chung
JH.
Overview of clinicopathologic features of ALK-rearranged lung adenocarcinoma and current diagnostic testing for ALK rearrangement
.
Transl Lung Cancer Res
.
2015
;
4
(
2
):
149
155
.
24
Suzuki
Y.
Orita
M.
Shiraishi
M.
Hayashi
K.
Sekiya
T.
Detection of ras gene mutations in human lung cancers by single-strand conformation polymorphism analysis of polymerase chain reaction products
.
Oncogene
.
1990
;
5
(
7
):
1037
1043
.
25
Schmid
K.
Oehl
N.
Wrba
F.
Pirker
R.
Pirker
C.
Filipits
M.
EGFR/KRAS/BRAF mutations in primary lung adenocarcinomas and corresponding locoregional lymph node metastases
.
Clin Cancer Res
.
2009
;
15
(
14
):
4554
4560
.
26
Cancer Genome Atlas Research Network
.
Comprehensive genomic characterization of squamous cell lung cancers
.
Nature
.
2012
;
489
(
7417
):
519
525
.
27
Tsou
JA.
Hagen
JA.
Carpenter
CL.
Laird-Offringa
IA.
DNA methylation analysis: a powerful new tool for lung cancer diagnosis
.
Oncogene
.
2002
;
21
(
35
):
5450
5461
.
28
Belinsky
SA.
Gene-promoter hypermethylation as a biomarker in lung cancer
.
Nat Rev Cancer
.
2004
;
4
(
9
):
707
717
.
29
Lokk
K.
Vooder
T.
Kolde
R.
et al.
Methylation markers of early-stage non-small cell lung cancer
.
PLoS One
.
2012
;
7
(
6
):
e39813
.
30
Merlo
A.
Herman
JG.
Mao
L.
et al.
5′ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers
.
Nat Med
.
1995
;
1
(
7
):
686
692
.
31
Zhao
QT.
Guo
T.
Wang
HE.
et al.
Diagnostic value of SHOX2 DNA methylation in lung cancer: a meta-analysis
.
Onco Targets Ther
.
2015
;
8
:
3433
3439
.
32
Schmidt
B.
Beyer
J.
Dietrich
D.
Bork
I.
Liebenberg
V.
Fleischhacker
M.
Quantification of cell-free mSHOX2 plasma DNA for therapy monitoring in advanced stage non-small cell (NSCLC) and small-cell lung cancer (SCLC) patients
.
PLoS One
.
2015
;
10
(
2
):
e0118195
.
33
Dietrich
D.
Jung
M.
Puetzer
S.
et al.
Diagnostic and prognostic value of SHOX2 and SEPT9 DNA methylation and cytology in benign, paramalignant and malignant pleural effusions
.
PLoS One
.
2013
;
8
(
12
):
e84225
.
34
Villalba
M.
Diaz-Lagares
A.
Redrado
M.
et al.
Epigenetic alterations leading to TMPRSS4 promoter hypomethylation and protein overexpression predict poor prognosis in squamous lung cancer patients
.
Oncotarget
.
2016
;
7
(
16
):
22752
22769
.
35
Kerr
KM.
Bubendorf
L.
Edelman
MJ.
et al.
Second ESMO consensus conference on lung cancer: pathology and molecular biomarkers for non-small-cell lung cancer
.
Ann Oncol
.
2014
;
25
(
9
):
1681
1690
.
36
Killian
JK.
Walker
RL.
Suuriniemi
M.
et al.
Archival fine-needle aspiration cytopathology (FNAC) samples: untapped resource for clinical molecular profiling
.
J Mol Diagn
.
2010
;
12
(
6
):
739
745
.
37
Lozano
MD.
Labiano
T.
Echeveste
J.
et al.
Assessment of EGFR and KRAS mutation status from FNAs and core-needle biopsies of non-small cell lung cancer
.
Cancer Cytopathol
.
2015
;
123
(
4
):
230
236
.
38
Roy-Chowdhuri
S.
Aisner
DL.
Allen
TC.
et al.
Biomarker testing in lung carcinoma cytology specimens: a perspective from members of the Pulmonary Pathology Society
.
Arch Pathol Lab Med
.
2016
;
140
(
11
):
1267
1272
39
Rafael
OC.
Aziz
M.
Raftopoulos
H.
Vele
OE.
Xu
W.
Sugrue
C.
Molecular testing in lung cancer: fine-needle aspiration specimen adequacy and test prioritization prior to the CAP/IASLC/AMP Molecular Testing Guideline publication
.
Cancer Cytopathol
.
2014
;
122
(
6
):
454
458
.
40
Roy-Chowdhuri
S.
Stewart
J.
Preanalytic variables in cytology: lessons learned from next-generation sequencing—the MD Anderson experience
.
Arch Pathol Lab Med
.
2016
;
140
(
11
):
1191
1199
.
41
Malapelle
U.
Mayo-de-Las-Casas
C.
Molina-Vila
MA.
et al.
Consistency and reproducibility of next-generation sequencing and other multigene mutational assays: a worldwide ring trial study on quantitative cytological molecular reference specimens
.
Cancer
.
2017
.
125
(
8
):
615
626
.
42
Tian
SK.
Killian
JK.
Rekhtman
N.
et al.
Optimizing workflows and processing of cytologic samples for comprehensive analysis by next-generation sequencing: Memorial Sloan Kettering Cancer Center experience
.
Arch Pathol Lab Med
.
2016
;
140
(
11
):
1200
1205
.
43
Bubendorf
L.
Lantuejoul
S.
de Langen
AJ.
Thunnissen
E.
Nonsmall cell lung carcinoma: diagnostic difficulties in small biopsies and cytological specimens
.
Eur Respir Rev
.
2017
;
26
(
144
):
170007
.
44
Fukui
T.
Ohe
Y.
Tsuta
K.
et al.
Prospective study of the accuracy of EGFR mutational analysis by high-resolution melting analysis in small samples obtained from patients with non-small cell lung cancer
.
Clin Cancer Res
.
2008
;
14
(
15
):
4751
4757
.
45
Lozano
MD.
Labiano
T.
Zudaire
I.
et al.
Variations in molecular profile in NSCLC can be analyzed using cytological samples: development of EGFR resistance mutations and coexistence of ALK-EML4 translocation in an EGFR-sensitive patient
.
Int J Surg Pathol
.
2015
;
23
(
2
):
111
115
.
46
Roy-Chowdhuri
S.
Chow
CW.
Kane
MK.
et al.
Optimizing the DNA yield for molecular analysis from cytologic preparations
.
Cancer Cytopathol
.
2016
;
124
(
4
):
254
260
.
47
Frampton
GM.
Fichtenholtz
A.
Otto
GA.
et al.
Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing
.
Nat Biotechnol
.
2013
;
31
(
11
):
1023
1031
.
48
Piqueret-Stephan
L.
Marcaillou
C.
Reyes
C.
et al.
Massively parallel DNA sequencing from routinely processed cytological smears
.
Cancer Cytopathol
.
2016
;
124
(
4
):
241
253
.
49
Bubendorf
L.
Buttner
R.
Al-Dayel
F.
et al.
Testing for ROS1 in non-small cell lung cancer: a review with recommendations
.
Virchows Arch
.
2016
;
469
(
5
):
489
503
.
50
Savic
S.
Bubendorf
L.
Common fluorescence in situ hybridization applications in cytology
.
Arch Pathol Lab Med
.
2016
;
140
(
12
):
1323
1330
.
51
Masucci
GV.
Cesano
A.
Hawtin
R.
et al.
Validation of biomarkers to predict response to immunotherapy in cancer, volume I: pre-analytical and analytical validation
.
J Immunother Cancer
.
2016
;
476
.
52
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
.
2017
;
23
(
14
):
3585
3591
.
53
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 28-8pharmDx and PD-L1 IHC 22C3pharmDx
.
Appl Immunohistochem Mol Morphol
.
2017
;
25
(
7
):
453
459
.
54
Ostrow
KL.
Michailidi
C.
Guerrero-Preston
R.
et al.
Cigarette smoke induces methylation of the tumor suppressor gene NISCH
.
Epigenetics
.
2013
;
8
(
4
):
383
388
.
55
Kurdyukov
S.
Bullock
M.
DNA methylation analysis: choosing the right method
.
Biology (Basel)
.
2016
;
5
(
1
):
3
.
56
Branchi
V.
Schaefer
P.
Semaan
A.
et al.
Promoter hypermethylation of SHOX2 and SEPT9 is a potential biomarker for minimally invasive diagnosis in adenocarcinomas of the biliary tract
.
Clin Epigenetics
.
2016
;
8
:
133
.
57
Ilse
P.
Biesterfeld
S.
Pomjanski
N.
Wrobel
C.
Schramm
M.
Analysis of SHOX2 methylation as an aid to cytology in lung cancer diagnosis
.
Cancer Genomics Proteomics
.
2014
;
11
(
5
):
251
258
.
58
Newman
AM.
Bratman
SV.
To
J.
et al.
An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage
.
Nat Med
.
2014
;
20
(
5
):
548
554
.
59
Siravegna
G.
Marsoni
S.
Siena
S.
Bardelli
A.
Integrating liquid biopsies into the management of cancer
.
Nat Rev Clin Oncol
.
2017
;
14
(
9
):
531
548
.
60
Hindson
CM.
Chevillet
JR.
Briggs
HA.
et al.
Absolute quantification by droplet digital PCR versus analog real-time PCR
.
Nat Methods
.
2013
;
10
(
10
):
1003
1005
.

Author notes

The authors have no relevant financial interest in the products or companies described in this article.

Competing Interests

Presented in part at the V Molecular Cytopathology: Focus on Next Generation Sequencing in Cytopathology meeting; October 18, 2016; Napoli, Italy.