Volume 12, Issue 4 pp. 4042-4063
RESEARCH ARTICLE
Open Access

Utilization of cytologic cell blocks for targeted sequencing of solid tumors

Erica Vormittag-Nocito

Erica Vormittag-Nocito

Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA

Contribution: Conceptualization (supporting), Methodology (supporting), Writing - original draft (supporting), Writing - review & editing (supporting)

Search for more papers by this author
Ravindra Kumar

Ravindra Kumar

Department of Pathology, Laboratory of Genomic Medicine, University of Illinois at Chicago, Chicago, Illinois, USA

Contribution: Data curation (supporting)

Search for more papers by this author
Kunwar Digvijay Narayan

Kunwar Digvijay Narayan

Department of Pathology, Laboratory of Genomic Medicine, University of Illinois at Chicago, Chicago, Illinois, USA

Contribution: ​Investigation (supporting), Project administration (supporting)

Search for more papers by this author
Zhengjia Chen

Zhengjia Chen

Department of Biostatistics, University of Illinois at Chicago, Chicago, Illinois, USA

Contribution: Software (lead)

Search for more papers by this author
Odile David

Odile David

Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA

Contribution: Project administration (supporting), Resources (supporting), Supervision (supporting)

Search for more papers by this author
Frederick Behm

Frederick Behm

Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA

Contribution: Resources (lead), Supervision (supporting)

Search for more papers by this author
Gayatry Mohapatra

Corresponding Author

Gayatry Mohapatra

Department of Pathology, Laboratory of Genomic Medicine, University of Illinois at Chicago, Chicago, Illinois, USA

Correspondence

Gayatry Mohapatra, Department of Pathology, Laboratory of Genomic Medicine, University of Illinois at Chicago, 840 S Wood Street, CSN Suite 130, Chicago, IL 60612, USA.

Email: [email protected]

Contribution: Conceptualization (equal), Data curation (equal), Formal analysis (equal), Project administration (equal), Writing - original draft (lead), Writing - review & editing (lead)

Search for more papers by this author
First published: 20 September 2022
Citations: 2

Abstract

Background

Targeted sequencing of cytologic samples has significantly increased in recent years. With increasing numbers of clinical trials for variant specific therapeutics, validating a comprehensive assay for cytologic samples has become clinically important.

Aim

For this study, a retrospective review of cytologic cell blocks from fine needle aspirations and fluid specimens was performed.

Methods

Two hundred twenty six total cases of solid tumor malignancies were identified, of which 120 cases and 20 lymph node negative controls were sequenced for the Oncomine Comprehensive Assay. Cytology and surgical specimen correlation was performed in a subset of cases. Statistical analysis to determine variant concordance was performed.

Results

Within the 117 cases sequenced, a total of 347 pathogenic variants were detected. Of the 117 cases, 32 cases (27.4%) would qualify for FDA approved targeted therapy according to the current guidelines, and an additional 23 cases (19.7%) would qualify for clinical trial based on pathogenic variants detected.

Discussion

With over 27% of cases in our cohort qualifying for some form of targeted therapy, our study shows the importance of providing comprehensive molecular diagnostic options. Despite only half of the cytology cases in the review period having enough material to be sequenced, overall approximately 27% of patients in this cohort would have benefitted from this service.

1 INTRODUCTION

Utilization of cytologic samples for clinical testing in the molecular pathology laboratory has greatly increased over recent years. As next-generation sequencing (NGS) technologies have become dominant in the diagnostic and therapeutic arenas, it is essential to optimize assays for which low input of DNA can be used to sequence large numbers of clinically actionable targets in a highly accurate and reproducible manner. This will permit comprehensive overviews of the driver mutations for targeted therapy even in very small samples of tumor. Several publications have addressed the use of cytologic smear preparations and the minimum amount of tissue needed for adequate DNA/RNA extraction from a cytology sample.1-3 More recent studies have shown the potential of formalin-fixed and paraffin-embedded (FFPE) cytology cell blocks as an additional source of molecular testing material without the destruction of diagnostic cytologic smears.4, 5 In addition, there are preanalytic variables that are specific to cytology samples.1 With a small amount of tissue as seen in cytologic samples, variables such as tumor heterogeneity and low percentage of tumor cells may lead to false-negative results. However, in cases of metastasis at the time of presentation or in cases where a fine needle aspiration (FNA) is the only procedure the patient can tolerate for sample collection, utilization of these small samples is invaluable for patient care.

For utilizing small samples, targeted panels are an ideal way to assess a tumor for clinically actionable mutations compared to PCR and Sanger sequencing methods. With many FDA-approved drugs available for various types of solid tumors harboring mutations in EGFR, BRAF, BRCA, NTRK, PIK3CA, etc., sequencing these targets has never been more important for patient care. At our institution, a medium-sized university hospital, we have elected to use a comprehensive solid tumor panel that targets important driver genes for therapy, tumor progression, and prognosis across all types of solid tumors. To our knowledge, this is the first study utilizing cytology cell blocks across multiple cancer types for comprehensive targeted sequencing for solid tumors to simulate routine diagnostic molecular oncology workflow. In the present study, we reviewed cytology cell blocks from 2013–2017 to identify cases with remaining malignant cells to be used for targeted panel sequencing. In order to test the viability of cytologic cell block samples as an alternative to surgical pathology material, archival cytology cell blocks from cytology fluid specimens and FNA material was utilized to simulate samples in a standard practice workflow.

2 MATERIALS AND METHODS

2.1 Case selection

Institutional Review Board approval for this study was obtained from the University of Illinois at Chicago. A case retrieval search was performed at the University of Illinois at Chicago pathology archives for all FNAs and cytology fluid samples that had a corresponding cytology cell block from August 2013 through May 2017. FNA specimens and fluid samples were chosen as all of these cases reflexively have a cell block performed. All samples were initially fixed in CytoLyt and transferred in pellet form into formalin for a minimum of 6 h prior to being placed on the processor in the histology laboratory. A total of 2702 cases were identified during this time period. The diagnoses of these cases were reviewed to identify all cases that had a malignant solid tumor neoplasm (this included carcinomas and soft tissue tumors). A total of 451 cases were identified as being positive for a solid tumor malignancy, and the cell block H&E slides for these cases were reviewed to confirm presence of malignant cells. Approximately 30% of these cases were found to have no malignant cells within the cell block sections and were confirmed to have had the diagnosis of malignancy rendered on the cytospin or FNA smear preparations; these cases were excluded from the study. Another 20% of cases identified were excluded from the study due to cell blocks or slides missing from the archives.

Of the remaining 226 cases, 106 (47%) cases were found to have inadequate numbers of tumor cells on cell block H&E sections (cases with less than 200 tumor cells per H&E section were considered inadequate and not included in the study) or inadequate tissue remaining on the paraffin block for 16 unstained slides to be cut for the molecular analysis performed in this study. A total of 120 cases were identified with enough archival tissue remaining to proceed with molecular analysis (Figure 1).

Details are in the caption following the image
Sample selection schema

In addition to the cytology cell blocks for 120 cases, we reviewed the pathology archival material for any surgical pathology case for the patient cohort that had the same diagnosis as the cytology sample. Many of our cases within the cohort were cytologic fluid samples that represented metastatic disease where surgical resection was not an option (Table 1). In addition, our center is a tertiary care referral center and many smaller community practices send patients to our clinics for endoscopic diagnostic procedures, and therefore, resection specimens are not present in our archives for comparison in many lung and gastrointestinal malignancies. Due to these conditions, we were able to find surgical samples for only 18 cases that corresponded to the cytology sample and were included in the study for comparison of NGS results.

TABLE 1. Clinical characteristics of the samples included in the study
Case ID Diagnosis Age @ diagnosis Gender Tissue Type Clinical/Pathologic Stage at Diagnosis Clinical Hx (Hx) Primary/ Metastasis
Case 1 Lung Adenocarcinoma 69 F Pleural fluid, left Stage IVB; pT4pM1c Contralateral lung, bone, and brain metastasis at the time of diagnosis Met
Case 2 Lung Adenocarcinoma 58 F FNA, right hilar mass pT2apN0 Recurrence
Case 3 Lung Adenocarcinoma 73 F Pericardial fluid Stage IVA; pT4pN3pM1a Pleural fluid and pericardial fluid positive for malignancy at the time of diagnosis Met
Case 4 Lung Adenocarcinoma 64 F Pleural fluid Stage IVB Supraclavicular lymph node and brain metastasis at the time of diagnosis Met
Case 5 Lung Adenocarcinoma 54 F Pleural fluid Stage IV Spine metastasis at the time of diagnosis Met
Case 6 Lung Adenocarcinoma 66 M Pericardial fluid Stage IIIA Pericardial malignant effusion and positive lymph nodes at the time of diagnosis Met
Case 7 Lung Adenocarcinoma 58 F Pleural fluid Stage IV Bone metastasis at the time of diagnosis Met
Case 8 Lung Adenocarcinoma 52 F FNA, lymph node at least Stage IIIB Met
Case 9 Lung Adenocarcinoma 37 M Pleural fluid Malignant pleural effusion at the time of diagnosis Met
Case 10 Lung Adenocarcinoma 80 F Pleural fluid Met
Case 11 Lung adenocarcinoma 37 F Pericardial fluid Stage IVB Met
Case 12 Lung Adenocarcinoma 71 F TBNA-EBUS guided Stage IV Primary
Case 13 Squamous cell carcinoma 62 F TBNA-EBUS guided Stage IIIB Primary
Case 14 Lung Adenocarcinoma 60 F TBNA-EBUS guided Stage IVA Brain metastasis at the time of diagnosis Primary
Case 15 Lung Adenocarcinoma 72 F Pleural fluid Stage IV Met
Case 16 Lung Adenocarcinoma 73 M TBNA-EBUS guided, lung mass Primary
Case 17 Lung Adenocarcinoma 55 F TBNA-EBUS guided, right bronchus intermedius mass Stage IVB; pT2b pN3 pM1c Primary
Case 18 Lung Adenocarcinoma 69 F TBNA-EBUS guided, right lower lobe lung mass Stage IIIB, pT2b pN3 Original tumor diagnosed 7 years before presentation at an outside institution Recurrence
Case 19 Lung Adenocarcinoma 61 F TBNA-EBUS guided Stage IV Metastasis known at the time of presentation Primary
Case 20 Lung Adenocarcinoma 63 F TBNA-EBUS guided, mediastinal mass Stage I, pT1 pN0 Original tumor 15 years prior to presentation Recurrence
Case 21 Lung Adenocarcinoma 71 M Pleural fluid Stage IVB Malignant pleural effusion at diagnosis Met
Case 22 Lung Adenocarcinoma 65 M Pleural fluid at least Stage III Met
Case 23 Lung adenocarcinoma 66 M TBNA-EBUS guided, left lower lobe lung mass Stage IIIA Primary
Case 24 Lung adenocarcinoma 68 M TBNA-EBUS guided, right hilar mass Stage IV Brain metastasis at the time of diagnosis Primary
Case 25 Lung adenocarcinoma 50 M Pleural fluid Stage IVB Malignant pleural effusion at the time of diagnosis Recurrence
Case 26 Lung adenocarcinoma 75 M TBNA-EBUS guided, lymph node Stage IV Met
Case 27 Lung SqCC 61 M TBNA-EBUS guided, hilar mass Stage IIIB Hx of TB Recurrence
Case 28 Lung SqCC 65 F TBNA-EBUS guided, lymph node Stage IVA Hx of LUL lung mass Met
Case 29 Lung SqCC 64 M TBNA-EBUS guided, lymph node At least Stage IIIB Hx of right lower lobe lung mass Met
Case 30 Lung SqCC 72 F TBNA-EBUS guided, mediastinal lymph node Stage IIIB Hx of smoking with lung mass Met
Case 31 Lung SqCC 69 M TBNA-EBUS guided, right lung mass Stage IV Primary
Case 32 Lung SqCC 55 M TBNA-EBUS guided, lung mass Stage IIIA Primary
Case 33 Lung SqCC 62 M TBNA-EBUS guided, RUL mass At least Stage IIIA Primary
Case 34 Lung SqCC 82 F TBNA-EBUS guided, left lung mass At least Stage III Primary
Case 35 Small cell carcinoma, Lung 59 M Pleural fluid Stage IV Met
Case 36 Small cell carcinoma, Lung 62 M TBNA-EBUS guided, lymph node Stage IV Mediastinal lymphadenopathy & spinal lesions at the time of presentation Met
Case 37 Small cell carcinoma, Lung 57 F Pericardial fluid Stage IV Brain metastasis at the time of diagnosis Met
Case 38 Small cell carcinoma, Lung 63 M TBNA-EBUS guided, lymph node Stage IV Brain metastasis at the time of diagnosis Met
Case 39 Small cell carcinoma, Lung 71 M TBNA-EBUS guided, lymph node Confined to chest cavity at the time of diagnosis Met
Case 40 Small cell carcinoma, Lung 76 F TBNA-EBUS guided, right hilar mass T2N1 Confined to chest cavity at the time of diagnosis Primary
Case 41 Poorly differentiated lung carcinoma 50 F TBNA-EBUS guided, lymph node Stage IV Adrenal, hilar lymph nodes, and brain metastasis at the time of diagnosis Met
Case 42 Poorly differentiated lung carcinoma 54 F TBNA-EBUS guided, lymph node Stage IV Mediastinal lymph nodes, brain, and spinal cord metastasis at the time of diagnosis Met
Case 43 Poorly differentiated lung cancer 64 M TBNA-EBUS guided, RUL lung mass Stage IIIC Primary
Case 44 Adenocarcinoma unknown origin 56 F Pleural fluid Stage IV Brain and spinal cord metastasis at the time of diagnosis. Met
Case 45 Adenocarcinoma unknown origin 59 F Pleural fluid Stage IV Positive lymph nodes and pancreatic lesion at diagnosis; presumed primary was lung mass Met
Case 46 Adenocarcinoma unknown origin 60 M Ascitic fluid Malignant ascites on presentation Met
Case 47 Adenocarcinoma unknown origin 49 F Ascitic fluid Stage IV Omentum lesions and malignant ascites on presentation Met
Case 48 Adenocarcinoma unknown origin 49 M Pleural fluid Stage IV Liver and bilateral lung lesions on presentation Met
Case 49 Mucinous adenocarcinoma unknown primary 70 F Pleural fluid Stage IV Spinal metastasis at presentation Met
Case 50 High-grade serous ovarian adenocarcinoma 46 F Ascitic fluid pT3cpN1b Met
Case 51 High-grade serous ovarian adenocarcinoma 86 F Pleural fluid Stage IVA Pelvic lymph nodes positive with abdominal and peritoneal implants on presentation Met
Case 52 High-grade serous ovarian adenocarcinoma 65 F EUS FNA, gastric mass Stage IV Vaginal wall invasion and stomach and spleen lesions at the time of diagnosis Met
Case 53 High-grade serous ovarian adenocarcinoma 56 F Pleural fluid Stage IV Lung and Liver metastasis at the time of diagnosis Met
Case 54 High-grade serous ovarian adenocarcinoma 77 F Ascitic fluid Stage III Omentum and abdominal wall lesions at the time of diagnosis Met
Case 55 High-grade serous ovarian adenocarcinoma 59 F Pleural fluid Stage IV Omentum lesions at the time of diagnosis Met
Case 56 High-grade serous ovarian adenocarcinoma 68 F Pleural fluid Stage IIIC Carcinomatosis at the time of diagnosis Met
Case 57 High-grade serous ovarian adenocarcinoma 60 F Ascitic fluid Stage IIIC Carcinomatosis at the time of diagnosis Met
Case 58 High-grade serous ovarian adenocarcinoma 49 F Pleural fluid Stage IV Abdominal wall implants at the time of diagnosis Met
Case 59 Low-grade serous ovarian carcinoma 41 F Pelvic washing Stage IIIB, pT3bpN1a Met
Case 60 Endometrial adenocarcinoma 60 F Ascitic fluid Stage IV, pT3apN0pM1 Serous endometrial carcinoma Met
Case 61 Endometrial adenocarcinoma 49 F Ascitic fluid Stage IV Presented with carcinomatosis Met
Case 62 Endometrial adenocarcinoma 78 F Ascitic fluid Stage IV Clear cell endometrial carcinoma, presented with carcinomatosis Met
Case 63 Endometrial adenocarcinoma 59 F Ascitic fluid Stage IV Serous endometrial carcinoma Met
Case 64 Endometrial adenocarcinoma 58 F FNA, left lower back soft tissue mass Stage IV Serous endometrial carcinoma; back mass and small intestine mass at the time of diagnosis Met
Case 65 Endometrial adenocarcinoma 53 F Pelvic washing Stage IV Endometrioid adenocarcinoma with lung and diaphragm metastasis at the time of diagnosis Met
Case 66 Endometrial adenocarcinoma 62 F Ascitic fluid Stage IVB, pT3pM1 Serous endometrial carcinoma with lung metastasis at presentation Met
Case 67 Endometrial adenocarcinoma 64 F Pleural fluid Stage IV Pleural effusion and lung metastasis at the time of presentation Met
Case 68 Cervical SqCC 62 F FNA, left supraclavicular mass Unknown Original tumor treated at outside hospital. Presented 10 years after original diagnosis with positive lymph node Met
Case 69 Cervical SqCC 59 F Ascitic fluid Stage IV Hx of recurrent low saag ascites, Hx of Cervical cancer Met
Case 70 Cervical adenocarcinoma 37 F Ascitic fluid Stage IIB History of treatment at outside hospital. Presented with omentum lesions and supraclavicular lymph node positive. Met
Case 71 Breast adenocarcinoma 55 F Pleural fluid Stage IV Bone, lung, brain, and gastric metastasis at the time of diagnosis Met
Case 72 Breast adenocarcinoma 65 F Pleural fluid Unknown Diagnosis at another institution more than 15 years before presentation. Presented to our institution with lung and liver metastasis Met
Case 73 Bilateral breast adenocarcinoma 57 F Pleural fluid pT1c Presented more than 10 years after initial diagnosis with spine and brain metastasis and pleural effusion Met
Case 74 Breast adenocarcinoma 48 F Pleural fluid Stage IIB Cytologic sample 4 years after original diagnosis of lung metastasis Met
Case 75 Breast adenocarcinoma 56 F FNA, left breast mass at Least Stage III Positive lymph nodes at the time of presentation Primary
Case 76 Breast adenocarcinoma 62 F Ultrasound-guided FNA, liver Unknown Presented to our institution 12 years after initial diagnosis with spine metastasis Met
Case 77 Breast adenocarcinoma 73 F TBNA-EBUS guided, RLL mass Unknown Hx of breast cancer with unknown time of diagnosis, presented with lung nodule Met
Case 78 Pancreatic adenocarcinoma 91 M EUS-guided FNA, pancreatic body Stage IV Liver and lung metastasis at the time of diagnosis Primary
Case 79 Pancreatic adenocarcinoma 68 F EUS-guided FNA, pancreas Stage IA Primary
Case 80 Pancreatic adenocarcinoma 63 M EUS-guided FNA, liver Stage IV Liver and lymph node metastasis at the time of diagnosis Met
Case 81 Pancreatic adenocarcinoma 79 F EUS-guided FNA, liver Stage IV Liver lesions at the time of diagnosis Met
Case 82 Pancreatic carcinoma with squamoid features 64 F EUS-guided FNA, pancreas Stage IV Liver lesions at the time of diagnosis Primary
Case 83 Pancreatic adenocarcinoma 75 F EUS-guided FNA, pancreatic head Stage IV Liver and lung metastasis at the time of diagnosis Primary
Case 84 Pancreatic adenocarcinoma 71 F EUS-guided FNA, pancreas Unknown Only seen for diagnostic procedure Primary
Case 85 Pancreatic adenocarcinoma 59 M EUS-guided FNA, pancreatic head T3 Primary
Case 86 Pancreatic adenocarcinoma 58 M EUS-guided FNA, pancreas Unknown Primary
Case 87 Gastrointestinal stromal tumor (GIST) 45 F EUS-guided FNA, stomach pT2pN0 Primary
Case 88 Gastrointestinal stromal tumor (GIST) 75 M EUS-guided FNA, splenic hilum Unknown Primary
Case 89 Gastrointestinal stromal tumor (GIST) 61 F EUS-guided FNA, mediastinal mass Stage IV Liver metastasis at the time of diagnosis Primary
Case 90 Gastrointestinal stromal tumor (GIST) 59 M EUS-guided FNA, gastric mass Unknown Primary
Case 91 Gastrointestinal stromal tumor (GIST) 67 M EUS-guided FNA, stomach pT2 Primary
Case 92 Gastrointestinal stromal tumor (GIST) 48 M EUS-guided FNA, abdominal mass Unknown Patient diagnosed ten years earlier than the presentation. FNA performed on recurrence Primary
Case 93 Extrahepatic cholangiocarcinoma 52 M EUS-guided FNA, pancreas Stage IIIA Primary
Case 94 Cholangiocarcinoma 82 F Ascites fluid Stage IV Malignant ascites at the time of diagnosis Met
Case 95 Gastric adenocarcinoma 67 F Ascites fluid Stage IV Lymph nodes positive and malignant ascites at the time of diagnosis Met
Case 96 Gastric adenocarcinoma 73 M Pleural fluid Stage IIIA, pT2pN3a Met
Case 97 Low-grade appendiceal mucinous tumor (LAMN) 52 M Peritoneal fluid pT4b pN2 Liver lesions and peritoneal implants at the time of presentation Met
Case 98 Head & Neck SqCC 58 M Pleural fluid Unknown Original tumor treated at outside institution. Presented with pleural effusion and lung nodule. Met
Case 99 EBV-associated nasopharyngeal carcinoma 56 M FNA, left neck mass pT2pN2b Lymph Node positivity at time of diagnosis Met
Case 100 Laryngeal SqCC 54 M FNA, RUL mass pT4apN0 Hx of laryngectomy for SqCC; Biopsy of lung mass found to be metastasis Met
Case 101 Left frontal sinus SqCC 49 M TBNA-EBUS guided, lymph node pT4apN0pM0 Hx of sinus carcinoma. FNA of lung metastasis Met
Case 102 Tongue/tonsil/FOM SqCC 63 M FNA, right neck mass Stage IVB Met
Case 103 SqCC of face (skin) 87 M FNA, submandibular region, left pT2pN0 Hx of Mycosis Fungoides Met
Case 104 SqCC of larynx 64 F FNA, right neck mass Stage IVA; pT2pN2b Met
Case 105 Conjunctival SqCC 78 F FNA, right preauricular region pT4pN1 Neck lymph node FNA Met
Case 106 Metastatic papillary thyroid carcinoma 66 F TBNA-EBUS guided, posterior tracheal mass Stage III, pT4 pN1a Current presentation after treatment with radioactive iodine and resection of thyroid Met
Case 107 Papillary thyroid Carcinoma 27 F US-guided FNA, left cervical lymph node Met
Case 108 High-grade salivary carcinoma 74 M TBNA-EBUS guided, lymph node At least Stage III Hx of salivary duct carcinoma; mediastinal lymph node positive Met
Case 109 Melanoma 38 M Ascites fluid Unknown Presented 5 year after original diagnosis with malignant ascites and spine lesions Met
Case 110 Melanoma 43 M Peritoneal fluid Unknown Presented 12 years after original diagnosis with metastatic bone lesions Met
Case 111 Metastatic renal cell carcinoma 57 M Right rib fluid Unknown Prior outside treatment of original tumor. Presented with bone metastasis Met
Case 112 Oncocytic renal cell carcinoma 65 M CT-guided FNA, right kidney Stage I, kidney confined lesion. Hx of right lung cancer, radioablated renal mass; no resection performed Primary
Case 113 Urothelial adenocarcinoma 64 M CT-guided FNA, Left acetabulum pT3apN0 (Bladder tumor); Stage IV (DLBCL) Hx of Diffuse large B cell lymphoma with prior treatment and BCG treatment of urothelial carcinoma before resection. This sample was at the time of recurrence in bone Met
Case 114 Malignant mesothelioma 65 M Pleural fluid Met
Case 115 Pancreatic neuroendocrine tumor 71 M EUS-guided FNA, pancreas Primary
Case 116 Neuroendocrine tumor, duodenum 66 F EUS-guided FNA, duodenal bulb Stage IV; pT3pN1pM1 Liver metastasis at the time of diagnosis Primary
Case 117 Neuroendocrine tumor 77 F EUS-guided FNA, ampulla Stage IV; pM1 Spine and liver metastatic lesions at the time of diagnosis Met

A total of 20 negative controls were identified in FNA lymph node samples; 10 negative lymph nodes from patients with no history of malignancy, and 10 negative lymph nodes from patients with a history of malignancy were included.

2.2 DNA and RNA extraction

FFPE cytology cell blocks were sectioned at 7 μm. A total of 15 unstained and unbaked serial sections were used for both DNA and RNA extraction. A 5 μm section was stained with hematoxylin and eosin. Tumor area was marked, and the percentage of tumor cells was estimated by a pathologist. Tumor cells were manually macrodissected to enrich for tumor fraction to achieve ≥20% tumor cells in the DNA/RNA sample. The Promega semi-automated FFPE DNA and RNA extraction kits were used with the Promega Maxwell RSC system (Promega Corporation). Quantitation of DNA and RNA by Qubit (Thermo Fisher Scientific) was performed prior to library preparation.

2.3 Targeted panel sequencing

Library preparation and sequencing using the Oncomine Comprehensive Assay version 3 was performed on all cases, using 20 ng of DNA and 20 ng of RNA for each sample, as has been previously described.6 The assay targets 161 unique genes including 84 genes for hotspot mutations, 43 genes for focal copy number gains, 48 full coding sequences for deletion mutations, and 51 fusion drivers. Data analysis was performed using the Torrent Suite software version v5.10. Ion Reporter version v5.10 was used for variant calling. A minimum average depth of coverage of 600X was considered adequate for each sample. Fusion analysis was performed with Ion Reporter version v5.10 fusion analysis workflow. Variants were classified as benign, likely benign, variant of undetermined significance, likely pathogenic or pathogenic based on the clinical criteria set by the College of American Pathologists and the Association of Molecular Pathology.7

2.4 Statistical analysis of outcome data

For cytologic and surgical comparison, results from a total of 17 paired samples were included. For each subject, the locus, genes, AA change, and values for variants (surgical and cytology) were recorded. The goal of this analysis was to determine the correlation between variants detected in surgical and cytology samples. Pearson's and Spearman's correlations were measured between variants. Parametric paired t-test was performed to compare the means of variants. Nonparametric paired test (Wilcoxon Signed Rank Test) was used to compare the medians of variants. The significance levels were set at 0.05 for all tests. The SAS 9.4 Version (SAS Institute, Inc.) was used for data management and analyses.

A clinical chart review was performed for all cases that passed quality metrics. A total of 117 subjects were included in this analysis. The charts were reviewed for the following information: date of diagnosis, final pathology diagnosis, treatment regimen, date of recurrence, date of metastasis diagnosis, date of death, and date of last known contact. Time of overall survival (OS) was calculated as the time from study enrollment to death or last contact. Time of progression-free survival (PFS) was calculated as the time from study enrollment to disease progression date, death date, or last contact whichever comes first. The survivor functions for PFS or OS were estimated by Kaplan-Meier survival analysis. Cox proportional hazards model was employed to estimate the adjusted effect of variants, diagnosis, and metastasis status on PFS or OS after adjustment for all other factors.

3 RESULTS

3.1 Case cohort

A total of 120 cases met the criteria for inclusion in the study. Two samples did not yield enough DNA for library preparation, and one case had too much formalin-induced artifacts for reliable analysis, leaving 117 samples in our study. There were 31 primaries, 6 recurrences, and 80 metastatic cases included in the study (Table 1), and the percentage of tumor cells ranged from 20–100%. The largest portion of the cohort consisted of lung carcinomas (35.8%) including 26 adenocarcinomas, 8 squamous cell carcinomas (SCCs), 6 small cell carcinomas, and 3 poorly differentiated carcinomas, which adequately reflects the frequency of thoracic specimens sent to our university's cytology service. The remaining cases consisted of 6 carcinomas of unknown origin, 9 high-grade serous carcinoma (HGSC) of the ovary and fallopian tube, 1 low-grade serous carcinoma (LGSC), 8 endometrial carcinomas (ECs) (including endometrioid, serous, and clear cell subtypes), 3 cervical carcinoma (1 squamous and 2 adenocarcinoma), 7 breast adenocarcinoma, 9 pancreatic adenocarcinoma, 6 gastrointestinal stromal tumors (GIST), 2 cholangiocarcinoma, 2 gastric adenocarcinoma, 1 low-grade appendiceal mucinous tumor, 7 head and neck SCCs (including larynx, pharynx, ocular, and oral cavity), 2 thyroid carcinoma, 1 salivary gland neoplasms, 2 melanoma, 2 renal cell carcinoma, 1 mesothelioma, 1 urothelial carcinoma, and 3 well-differentiated neuroendocrine tumors. A breakdown of each tumor type is represented in the pie chart (Figure 2).

Details are in the caption following the image
Cohort by diagnosis

3.2 Surgical-Cytologic correlation

There were 18 cases within the cohort that had paired surgical and cytology samples. These samples were from different time points over the course of patient treatment (ie: surgical case at primary diagnosis and cytology case at the time of metastasis), and this difference in time was noted in our correlation results table as 1st diagnosis and 2nd diagnosis (Table 2). The Spearman correlation between 1st and 2nd diagnosis was 0.65056 with a p-value of <0.0001, and the Pearson correlation between 1st and 2nd diagnosis was 0.54763 with a p-value of <0.0001. Both methods show a significant correlation between the surgical-cytologic paired samples. A detailed comparison of variants detected in each of the surgical and cytology cases is shown in Figure 3. Ten out of 18 cases had discrepancy in variants between the surgical and cytology samples. In 8 cases, the cytology samples had extra variants compared to surgical samples. All 8 samples were from metastatic disease with additional copy number, missense, or nonsense variants that are likely associated with tumor progression and/or metastasis. In case 15, the surgical sample had a nonsense mutation (R58*) in CDKN2A (VAF 74.9%) and a promoter mutation (c.-146C > T) in TERT (VAF 50.7%) that were not present in the cytology sample. The cytology sample had an indel (W557_K558del) in KIT (VAF 11.3%) and a BRAF V600E (VAF 5.3%) mutation that were not detected in the surgical sample (Figure 3). It is possible that the cytology sample represents a different clone that expanded in the tumor metastasis. Finally, for case 11, there were two surgical samples and one cytology sample. The first surgical sample and cytology samples were both from the primary breast cancer, whereas the second surgical sample was from the metastasis. In this case, the second surgical sample had a pathogenic ESR1 D538G mutation known to be associated with endocrine therapy resistance in breast cancer.8 The rest of the variants were identical in both surgical and cytology samples (Figure 3).

TABLE 2. List of variants detected in surgical and cytology paired samples
Case # Variants at 1st diagnosis Variants at 2nd diagnosis
1 TP53 p.Y220C; ATRX p.N1860S TP53 p.Y220C; ATRX p.N1860S; AXL amplification
2 PIK3CA p.E542K; ERBB3 p.V104L; ERBB2 p.R678Q; SETD2 p. P1962L; SETD2 p.M1080I; FGFR4 p.G388R PIK3CA p.E542K; ERBB3 p.V104L; ERBB2 p.R678Q; SETD2 p. P1962L; SETD2 p.M1080I; FGFR4 p.G388R; KRAS p.G12V
3 KRAS p.G12V; FGFR4 p. G388R; FNDC3B-PIK3CA fusion KRAS p.G12V; FGFR4 p. G388R; FNDC3B-PIK3CA fusion
4 MET p. T1010I; BRCA2 p. L3326* MET p. T1010I; BRCA2 p. L3326*; PMS2 p.H479Q; SMO p.V270I; ATM p.L1420F
5 KRAS p.G12V; TP53 p.L132R; ARID1A p.P120S; SETD2 p.P1962L; TERT p.A279T; FGFR4 p.G388R; CDK12 p.L1189Q; CCND1 Amplification; FGF19 amplification; FGF3 amplification; CDKN2A hemizygous loss; CDKN2B hemizygous loss KRAS p.G12V; TP53 p.L132R; ARID1A p.P120S; SETD2 p.P1962L; TERT p.A279T; FGFR4 p.G388R; CDK12 p.L1189Q; CCND1 Amplification; FGF19 amplification; FGF3 amplification; CDKN2A hemizygous loss; CDKN2B hemizygous loss
6 FBXW7 p.R465Ll TP53 p.G244S; RET p.E867D; FLT3 p.I417L; PALB2 p.P210L; NOTCH3 p.A1020P FBXW7 p.R465Ll TP53 p.G244S; RET p.E867D; FLT3 p.I417L; PALB2 p.P210L; NOTCH3 p.A1020P
7 BRCA2 p.R2318*; NOTCH3 p.Y1106fs; FGFR4 p.G388R; CDK12 p.I1131V BRCA2 p.R2318*; NOTCH3 p.Y1106fs; FGFR4 p.G388R; CDK12 p.I1131V; MDM4 amplification, MYC amplification, AKT1 amplification
8 TP53 p.G244V; MET p.I316M; FLT3 p.A988P; PALB2 p.P210L; NOTCH3 p.A1020P; ERBB2 amplification TP53 p.G244V; MET p.I316M; FLT3 p.A988P; PALB2 p.P210L; NOTCH3 p.A1020P; ERBB2 amplification; PDGFRA amplification; KIT amplification; CNNE1 amplification
9 NRAS p.Q61R; MSH2 p.L449N; SETD2 p.P1962L; POLE p.R1556W; CDKN2A homozygous loss; CDKN2B homozygous loss; TP53 hemizygous loss NRAS p.Q61R; MSH2 p.L449N; SETD2 p.P1962L; POLE p.R1556W; CDKN2A homozygous loss; CDKN2B homozygous loss; TP53 hemizygous loss
10 BRCA2 p.L3326*; TP53 p.R282W; PTCH1 p.T728M; SLX4 p.R1761C BRCA2 p.L3326*; TP53 p.R282W; PTCH1 p.T728M; SLX4 p.R1761C; ESR1 p.D538G; IGF1R amplification
11

1st Surgery: FGFR1 amplification, MYC amplification, MYC p.V185I, POLE P697S, TP53 splice site;

2nd Surgery: ESR1 p.D538G, FGFR1 amplification, MYC amplification, MYC p.V185I, POLE P697S, TP53 splice site

FGFR1 amplification, MYC amplification, MYC p.V185I, POLE P697S, TP53 splice site
12 FBXW7 p.R479P; FGFR4 p.G388R; SLX4 p.T919I; SLX4 p.W546C; TSC2 splice site ARID1A p.E1718*; FBXW7 p.R479P; FGFR4 p.G388R; SLX4 p.T919I; SLX4 p.W546C; TSC2 splice site
13 TP53 p.R248fs; BRCA1 p.Q1806*; FGFR4 p.G388R; ATM p.B122T TP53 p.R248fs; BRCA1 p.Q1806*; FGFR4 p.G388R; ATM p.B122T
14 MSH2 p.I770V; FAND2 p.Q65H; SETD2 p.P1962L; BRCA2 p.H1561N; BRCA2 p.V2138F; CDK12 p.T1195M; STK11 p.F354L; NOTCH3 p.A1020P; TSC2 splice site; KIT deletion MSH2 p.I770V; FAND2 p.Q65H; SETD2 p.P1962L; BRCA2 p.H1561N; BRCA2 p.V2138F; CDK12 p.T1195M; STK11 p.F354L; NOTCH3 p.A1020P; TSC2 splice site; KIT deletion
15 RAC1 p.P29F; RAC1 p.P29L; CDKN2A p.R58*; TP53 p.E286K; TP53 p.A159V; SETD2 p.P1962L; RAD50 p.D767N; TSC1 p.K587R; POLE p.G6R; STK11 p.D350N; TERT promoter RAC1 p.P29F; RAC1 p.P29L; TP53 p.E286K; TP53 p.A159V; SETD2 p.P1962L; RAD50 p.D767N; TSC1 p.K587R; POLE p.G6R; STK11 p.D350N; BRAF p. V600E; KIT p.W557_K558del
16 ARID1A p.V1817fs; PIK3CA p.E81K; PIK3CA p.R88Q; PTEN p.Y68H; TP53 p.R273H; CDKN2A p.H123Q; PALB2 p.T386A; NOTCH3 p.A1020P; TSC2 splice site ARID1A p.V1817fs; PIK3CA p.E81K; PIK3CA p.R88Q; PTEN p.Y68H; TP53 p.R273H; CDKN2A p.H123Q; PALB2 p.T386A; NOTCH3 p.A1020P; TSC2 splice site
17 KRAS p.G12F; KRAS p.G12C; TP53 p.R273L; SETD2 p.P1962L; PTCH1 p.T728M; ATM p.M1040V; SLX4 p.P975L; NF2 p.E463L; TSC2 In/Del KRAS p.G12F; KRAS p.G12C; TP53 p.R273L; SETD2 p.P1962L; PTCH1 p.T728M; ATM p.M1040V; SLX4 p.P975L; NF2 p.E463L; TSC2 In/Del
18 CDKN2A p.D108H; MAP2K1 p.P124L; TP53 p.L194R; NF1 p.Y80fs; MSH6 p.S63P; SETD2 p.P1962L; ATR p.S1607N; NOTCH3 p.A1020P; TSC2 splice site; CCND2 amplification CDKN2A p.D108H; MAP2K1 p.P124L; TP53 p.L194R; NF1 p.Y80fs; MSH6 p.S63P; SETD2 p.P1962L; ATR p.S1607N; NOTCH3 p.A1020P; TSC2 splice site; CCND2 amplification; KRAS amplification
  • Note: Bolded text identifies variants not identified in both specimens.
Details are in the caption following the image
Surgical cytologic correlation. Summary of variants identified in 18 pairs of surgical and cytology samples. Each pair is separated by a double line. Disease status, primary or metastatic is indicated in the second column. Case 11 had two surgical (one primary and one metastasis) and one cytology sample sequenced. Case 5 and 14 had both samples from primary tumors, show 100% concordance. Discrepant variants are shown in black arrows (surgical) and red arrows (cytology). Each row represents a sample, columns represent genes.

3.3 Cytology cases

Within the 117 cases sequenced, the depth of coverage ranged from 648X to 2694X (Figure 4A). A total of 711 variants were detected, including 505 single nucleotide variants (SNVs), 2 multinucleotide variants (MNVs), 73 insertion/deletions (indels), 126 copy number variants (CNVs), and 5 fusions (Figure 4B, Figure 5, Table S1). There was a total of 347 pathogenic variants and 345 variants of undetermined significance (VUS) detected in all cases (Figure 4C). The 20 negative control samples were sequenced with adequate depth of coverage and no pathogenic variants were detected.

Details are in the caption following the image
Performance metrics and variants detected in the case cohort. (A) Histogram showing average depth of coverage in all samples (range 648x–2694x). Cases are grouped by diagnosis and are in the same order as shown in Figure 1; X-axis: cases 1–117; Y-axis: Depth of coverage; (B) Pie chart showing the types of variants detected in the cohort; (C) Histogram showing the number of pathogenic variants and VUS for each case as well as the total number of variants identified for each case. Each case has a single bar representing the total number of variants and color difference within the bar indicates the types of variants present; (D) Clinically actionable variants detected in the lung adenocarcinoma.
Details are in the caption following the image
Heatmap of all variants detected in the cohort of 117 cases. Cases are grouped by diagnosis and are shown in the same order as in Figure 1. Each row represents a case, columns represent genes. Second column represents diagnosis and third column shows clinical relevance based on the presence of variants. Multiple variants of the same gene are indicated by stripes if the two variants are of the same type or by triangles if they are of different types.

In the entire cohort of 117 cases, TP53 was the most frequently mutated (58%) gene (Figure 5). Among the 26 lung adenocarcinomas, 18 cases had clinically actionable variants for which FDA-approved drugs are available. Seven cases had EGFR mutations (4 cases with exon 19 deletion, 1 case with R776H and L858R, 1 case with R831C, and 1 case with P794T). Four cases with KRAS G12C/S/V and TP53 variants, 2 cases with EML4-ALK fusion, 3 cases with BRAF variants (1 V600E, 1 G469V, and 1 exon 15 indel), 1 case with PIK3CA E542K, and 1 case with ERBB2 exon 20 indel. In the remaining 8 cases variants in NF1, STK11, ERBB4, SETD2, BAP1, and CDKN2A genes were detected (Figure 4D, Table S1). One case with KRAS G12S also had a WHSC1L1-FGFR1 fusion. The squamous cell lung carcinomas showed two cases with ARID1A loss of function variants (E1718* and G1848fs), one FGFR3 S249C variant, one MAP2K1 P124L, and one case with PIK3CA amplification and E545A variant. One case of poorly differentiated carcinoma of the lung had an ARID1A G836fs variant. The small cell carcinoma cases and remaining lung carcinoma cases did not have clinically relevant pathogenic variants.9

Of the 6 carcinomas of unknown origin, 2 cases showed a KRAS G12D/V variant and one case showed an amplification of the same gene. One case had an ERBB2 amplification with a SMARCA4 loss of function G1232C variant. One case had many amplifications present.

Our cohort of gynecologic tumors included 9 high-grade serous ovarian/fallopian tube cancers (HGSC), one LGSC, 8 ECs, one SCC of the cervix, and two adenocarcinomas of the cervix. Of the HGSC, two cases had a BRCA1 pathogenic variant (W1733* and Q1806*), and one case had a BRCA2 pathogenic variant (L1227fs). Additionally, one case showed a FGFR2 amplification and another case showed a FGFR1 pathogenic variant (D166del). The LGSC showed a PIK3CA-FNDC3B fusion and a KRAS G12V variant. Of the 8 ECs, two had the most common hotspot PIK3CA variants (Q546K and E542A), one case with two different hotspot PIK3CA variants (E81K and R88Q) and an additional case with a PIK3CA amplification. One case of EC had a BRCA1 frameshift variant (E1210fs), and one case showed ERBB2 amplification. FGFR3 amplification and gain-of-function variant in one case appeared to be the driving alterations. Within the 7 breast cancer cases, two cases showed a BRCA2 nonsense variant (p.R2318* and p.K3326*), one case showed ERBB3 G234R, one case showed KRAS G12V, and one case showed ARID1A P453fs. No PIK3CA, BRCA1, or ERBB2 alterations were identified in the breast carcinoma cases.10, 11 Amplifications of AKT1, MYCN, MYC, FGFR1, FGFR4, FGF3, FGF19, IGF1R, CCND1, CCNE1, TERT, and NTRK3 were detected in 4/7 cases (Figure 5).

All nine pancreatic adenocarcinomas harbored exon 2 pathogenic gain-of-function variants in KRAS and 6/9 cases had a deleterious loss-of-function variant in TP53. In addition to these variants, one case showed a frameshift mutation in BRCA1 (Q867fs), and another case showed a less common G1049R gain-of-function variant in PIK3CA. All six cases of GIST harbored a pathogenic variant in KIT—three indels including a splice site variant and 3 gain of function missense variants. Of the two cholangiocarcinoma cases, one case had a SMAD4 R361H pathogenic variant and the other case had a pathogenic KRAS, TP53, and additional CTNNB1 S45F variant. The two gastric carcinoma cases showed an ERBB2 R678Q pathogenic variant with a PIK3CA E542K and KRAS G12V variants.

Within the SCCs of the head and neck region, there was variability in locations that was reflected in the variants detected. One case from the face showed two loss of function variants in BRCA1 (E17758* and E1491*). A frontal sinus-based neoplasm had a loss of function BRCA2 K3326* variant. A nasopharyngeal-based lesion harbored a pathogenic HRAS Q61R variant and a FGFR1 amplification. A laryngeal SCC was found to have a PIK3CA H1047R pathogenic variant. The additional larynx-based tumor and the floor of mouth tumor included in our cohort showed significant number of copy number variations (CNVs).12

In the smaller diagnosis cohort, one melanoma case had a NRAS Q61R variant and the other case of melanoma had a BRAF V600E variant. Both papillary thyroid carcinoma cases harbored BRAF V600E variants. One case of renal cell carcinoma harbored a KRAS G12D variant and showed an oncocytic papillary phenotype. The salivary gland malignancy showed PIK3CA H1047R and HRAS Q61R pathogenic variants.

Of note, there were no significant pathogenic variants detected in the urothelial carcinoma, the mesothelioma, the low-grade appendiceal mucinous tumor, and neuroendocrine tumors. A summary of variants along with diagnosis and clinical relevance is shown in Figure 5, and for a full list of variants detected in our cohort, please refer to Table S1.

4 DISCUSSION

In recent years, cytology samples are a growing proportion of samples that are sent for routine molecular diagnostics. Our study demonstrates that NGS-based targeted sequencing can be performed in a highly accurate and reproducible manner with small quantity cytology samples. Within our university cytology service, we found that approximately 27% (120/451) of our malignant cytologic samples had cell block material adequate for molecular testing via a NGS comprehensive assay. Of the 117 cases included in this study, 18 cases had a surgical pathology sample available and showed good concordance of results between cytologic and surgical pathology specimens validating the utility of cytologic specimens in the molecular diagnostics laboratory.

One of the challenges of using comprehensive NGS-based assays for routine diagnostic testing is the quality and quantity of tissue required for detecting clinically relevant mutations. Our study demonstrates that we had a very small failure rate (2.5%; 3/120) due to either sample size or formalin-induced artifacts. Our assay detected variants (SNVs) with 5% or higher allele frequency, CNVs (amplification, single copy loss and homozygous loss) and gene fusions efficiently. For many tumors, the percent tumor cells present in the section amounted to 20% but we were able to enrich the tumor fraction by macrodissecting to yield sufficient DNA and RNA for running the assay successfully.

Genomic heterogeneity plays a significant role in eventual drug resistance and treatment failure resulting from the generation of subpopulations within a tumor. The Cancer Genome Atlas (TCGA) studies performed by inter- and intra-tumor comparisons have shown tumor heterogeneity in many types of tumors including lung, breast, prostate, glioblastoma and colon cancers.10, 13-16 Furthermore, TCGA pan-cancer analysis of genomic landscapes of 12 tumor types from more than 3000 tumors identified 127 significantly mutated genes with both established and emerging links to cancer, indicating that the number of driver mutations required for oncogenesis is relatively small.17 Although a common set of driver mutations exists in a given cancer type, the combination of mutations within a patient tumor and their distribution within the founding clone and subclones will be critical for optimizing their treatment. We sequenced the samples at a higher depth of coverage to account for the sub-populations of cells that might be contributing toward the makeup of the tumor.

One of the limitations with cytology samples is the small quantity of cellular material, which raises concerns about capturing tumor heterogeneity. In FNA biopsy samples, multiple planes of the tumor and the resulting specimen comprises multiple passes through the tumor. In body fluid specimens, concern for heterogeneity should be low because malignant cells in cavity fluids typically represent the most aggressive and metastatic subclone of the tumor and would be the ideal subpopulation to study. Methodologically, increasing the percent of tumor cells in a sample by selective microdissection proportionally increases the yield of tumor cells coming from the major subpopulations, thereby increasing the yield of both driver and passenger mutations.18

In our cohort, we identified 32 cases (27.4%) that would qualify for FDA-approved targeted therapy according to the current guidelines. An additional 23 cases (19.7%) would qualify for a clinical trial based on pathogenic variants detected by NGS (Table 3). With approximately 45% of cases in our cohort qualifying for some form of targeted therapy, the critical importance of providing high complexity NGS testing of cytologic samples is undeniable. Currently available treatments include imatinib for KIT alterations in GIST, gefitinib, osimertinib, sotorasib, crizotinib, for alterations in EGFR, KRAS, and ALK fusion in NSCLC, PARP inhibitors, and alpelisib for BRCA1/2 and PIK3CA variants in gynecologic malignancies, and dabrafenib and trametinib for BRAF V600E in melanoma and metastatic papillary thyroid carcinoma.19-24Our cohort does not include enough samples for each diagnosis to reach statistical significance with regard to clinical outcome. But comparing cases with and without actionable variants in our overall cohort, there was a significant correlation between cases with targetable variants with both PFS and OS (Figure S1).

TABLE 3. Cases with variants for which targeted therapy or clinical trial available
Case ID Diagnosis Clinical/Pathologic Stage at Diagnosis Primary/Metastasis Gene Exon Variant type AA Change FDA-Approved Therapy Options Clinical Trial Options
1 Lung Adeno Stage IVB; pT4pM1c Met EGFR 19 INDEL p.E746_A750del Yes
2 Lung Adeno pT2apN0 Recurrence KRAS 2 SNV p.G12C Yes
4 Lung Adeno Stage IVB Met EGFR 19 INDEL p.E746_A750del Yes
5 Lung Adeno Stage IV Met ERBB2 20 INDEL p.A771_Y772insYVMA Yes
6 Lung Adeno Stage IIIA Met BRAF 15 SNV p.V600E Yes
11 Lung Adeno Stage IVB Met EML4-ALK e13|e20 FUSION Read count: 27218 Yes
12 Lung Adeno Stage IV Primary EGFR 21 SNV p.L858R Yes
14 Lung Adeno Stage IVA Primary PIK3CA 10 SNV p.E542K Yes
15 Lung Adeno Stage IV Met BRAF 11 SNV p.G469V Yes
16 Lung Adeno Unknown Primary EGFR 19 INDEL p.E746_A750del Yes
17 Lung Adeno Stage IVB; pT2b pN3 pM1c Primary EML4-ALK e13|e20 FUSION Read count: 131144 Yes
20 Lung Adeno Stage I, pT1 pN0 Recurrence NF1 37 INDEL p.C1682Ter Yes
21 Lung Adeno Stage IVB Met EGFR 19 INDEL p.E746_A750del Yes
22 Lung Adeno At least Stage III Met EGFR 21 SNV p.R831C Yes
23 Lung Adeno Stage IIIA Primary NF1 42 INDEL p.L2103fs Yes
26 Lung Adeno Stage IV Met KRAS 2 SNV p.G12C Yes
27 Lung SqCC Stage IIIB Recurrence ARID1A 20 SNV p.G1848fs Yes
30 Lung SqCC Stage IIIB Met ARID1A 20 SNV p.E1718* Yes
32 Lung SqCC Stage IIIA Primary FGFR3 7 SNV p.S249C Yes
33 Lung SqCC At least Stage IIIA Primary PIK3CA 10 SNV p.E545K Yes
41 Poorly differentiated lung ca Stage IV Met ARID1A 8 SNV p.G836fs Yes
51 High-grade serous ovarian ca Stage IVA Met BRCA2 11 SNV p.L1227fs Yes
52 High-grade serous ovarian ca Stage IV Met BRCA1 18 SNV p.W1733* Yes
54 High-grade serous ovarian ca Stage IV Met BRCA1 22 SNV p.Q1806Ter Yes
57 High grade serous ovarian ca Stage IIIC Met ARID1A 1 SNV p.A246fs Yes
61 Endometrial adenocarcinoma Stage IV Met PIK3CA 10 SNV p.E542A Yes
62 Endometrial adenocarcinoma Stage IV Met ERBB2 Amp CNV Copy number: 16 Yes
63 Endometrial adenocarcinoma Stage IV Met BRCA1 10 SNV p.E1210fs Yes
64 Endometrial adenocarcinoma Stage IV Met ARID1A 20 SNV p.V1817fs Yes
65 Endometrial adenocarcinoma Stage IV Met PIK3CA 10 SNV p.Q546K Yes
66 Endometrial adenocarcinoma Stage IVB, pT3pM1 Met ERBB2 Amp CNV Copy number: 8.4 Yes
68 Cervical SqCC Unknown Met PIK3CA 10 SNV p.E545K Yes
69 Cervical SqCC Stage IV Met PIK3CA 10 SNV p.E542V Yes
71 Breast adenocarcinoma Stage IV Met ERBB3 7 SNV p.G284R Yes
72 Breast adenocarcinoma Unknown Met ARID1A 3 SNV p.P453fs Yes
73 Bilateral breast adenocarcinoma pT1c Met BRCA2 13 SNV p.R2318* Yes
74 Breast adenocarcinoma Stage IIB Met BRCA2 27 SNV p.K3326* Yes
87 Gastro intestinal stromal tumor pT2pN0 Primary KIT 11 INDEL p.W557_K558del Yes
88 Gastro intestinal stromal tumor Unknown Primary KIT 11 SNV p.V559G Yes
89 Gastro intestinal stromal tumor Stage IV Primary KIT 11 INDEL p.W557_K558del Yes
90 Gastro intestinal stromal tumor Unknown Primary KIT 11 SNV p.W557G Yes
91 Gastro intestinal stromal tumor pT2 Primary KIT 11 SNV p.V559G Yes
92 Gastro intestinal stromal tumor Unknown Recurrence KIT 11 INDEL Splice site Yes
94 Cholangiocarcinoma Stage IV Met RET 13 SNV p.Y791F Yes
100 Laryngeal SCC pT4apN0 Met PIK3CA 21 SNV p.H1047R Yes
101 Left frontal sinus SCC pT4apN0pM0 Met MET 14 SNV p.T1010I Yes
102 Tongue/tonsil/FOM SCC Stage IVB Met ARID1A 9 SNV p.Q944* Yes
103 SCC of face (skin) pT2pN0 Met BRCA1 20 SNV p.E1775* Yes
104 SCC of larynx Stage IVA; pT2pN2b Met MSH2 15 SNV p.C873* Yes
105 Conjunctival SCC pT4pN1 Met KIT 11 INDEL p.W557_K558del Yes
106 Metastatic papillary thyroid ca Stage III, pT4 pN1a Recurrence BRAF 15 SNV p.V600E Yes
107 Papillary thyroid ca Unknown Met BRAF 15 SNV p.V600E Yes
108 High-grade salivary ca At least Stage III Met PIK3CA 21 SNV p.H1047R Yes
109 Melanoma Unknown Met BRAF 15 SNV p.V600E Yes
110 Melanoma Unknown Met NRAS 3 SNV p.Q61R Yes

It is important to note that 50% of the cases that came through the cytology service during the period of archival review were not included in this study due to lack of material to review or due to insufficient tissue for further analysis. Because of the retrospective nature of this study, some samples had cell blocks that were exhausted or had very little tissue left that was inadequate for molecular testing. Additional prospective studies would need to be done to get a more accurate success rate of cytologic cell blocks in light of tissue preservation methods used in laboratories today to improve ancillary testing results. Alternative forms of cell preservation and additional FNA passes dedicated for molecular laboratories could also increase the number of cytologic cases for successful molecular analysis. At our institution, a high number of patients diagnosed by the cytology service as “positive for malignancy” are high stage at presentation or have recurrent disease, and these patients would benefit most from molecular analysis. Although intra-tumor heterogeneity is a concern, molecular analysis of cytologic specimens can adequately identify driver mutations for which targeted therapy options are available. As shown in our comparison of surgical samples and cytologic cell blocks, the major drivers of oncogenesis in each tumor pair were identified in both samples. As most of the patients in our cohort either were not eligible for surgical resection or had not responded to first-line chemotherapy at the time of the cytologic sample collection, this is exactly the cohort that would most benefit from approved targeted therapies or clinical trial options. With this in mind, and extrapolating from our study, approximately 30% more of the patients in our population with similar clinical histories would qualify for currently approved targeted therapy regimens or would qualify for a clinical trial. We continue to optimize our service for such patients and have developed confidence in the methods and yield of targeted sequencing of solid tumor in FNA and body fluid cytology cell block specimens.

AUTHOR CONTRIBUTIONS

Erica Vormittag-Nocito: Conceptualization (supporting); methodology (supporting); writing – original draft (supporting); writing – review and editing (supporting). Ravindra Kumar: Data curation (supporting). Kunwar Narayan: Investigation (supporting); project administration (supporting). Zhengjia Chen: Software (lead). Odile David: Project administration (supporting); resources (supporting); supervision (supporting). Frederick Behm: Resources (lead); supervision (supporting). Gayatry Mohapatra: Conceptualization (equal); data curation (equal); formal analysis (equal); project administration (equal); writing – original draft (lead); writing – review and editing (lead).

ACKNOWLEDGMENTS

None.

    CONFLICT OF INTEREST

    None.

    ETHICAL APPROVAL STATEMENT

    This study was approved by the University of Illinois Institutional Review Board with an exemption granted for written informed consent.

    CLINICAL TRIAL REGISTRATION

    N/A

    DATA AVAILABILITY STATEMENT

    Data will be shared once the manuscript is accepted.

      The full text of this article hosted at iucr.org is unavailable due to technical difficulties.