Next Article in Journal / Special Issue
BRAF and MLH1 Analysis Algorithm for the Evaluation of Lynch Syndrome Risk in Colorectal Carcinoma Patients: Evidence-Based Data from the Analysis of 100 Consecutive Cases
Previous Article in Journal
Micro-RNA in Cholangiocarcinoma: Implications for Diagnosis, Prognosis, and Therapy
Previous Article in Special Issue
Evaluation of the TruSight Tumor 170 Assay and Its Value in Clinical Diagnostics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Use of the Biocartis Idylla™ Platform for the Detection of Epidermal Growth Factor Receptor, BRAF and KRAS Proto-Oncogene Mutations in Liquid-Based Cytology Specimens from Patients with Non-Small Cell Lung Carcinoma and Pancreatic Adenocarcinoma

Diagnostic Cytopathology, Royal Cornwall Hospital NHS Trust, Truro TR1 3LJ, UK
*
Author to whom correspondence should be addressed.
J. Mol. Pathol. 2022, 3(2), 104-114; https://doi.org/10.3390/jmp3020010
Submission received: 19 April 2022 / Revised: 9 May 2022 / Accepted: 10 May 2022 / Published: 14 May 2022
(This article belongs to the Collection Feature Papers in Journal of Molecular Pathology)

Abstract

:
The study aimed to demonstrate rapid and effective molecular testing on liquid-based cytology (LBC) samples for EGFR, KRAS and BRAF mutations using the Biocartis Idylla™. Rapid on-site evaluation (ROSE) LBC samples for patients with non-small cell lung carcinoma (NSCLC) or pancreatic ductal adenocarcinoma (PDAC) were tested for EGFR, KRAS and BRAF mutations based on the relevance to tumour subtype. The quantification values (Cq values) and mutation detection status were compared between LBC samples and routine formalin-fixed paraffin-embedded (FFPE) clot samples. ROSE LBC samples (n = 54) showed a higher yield of well-preserved tumour and wild type (WT) DNA, demonstrated by lower quantification cycles, no false positives or false negatives, and a higher sensitivity for low allele frequency mutations when compared with FFPE clot samples. The Biocartis Idylla™ provides highly sensitive, reliable and rapid testing for LBC samples for the detection of EFGR and KRAS mutations. BRAF mutations were not detected in the participant cohort; however, all LBC WT BRAF results correlated with the results from the FFPE clot samples. Access to rapid molecular testing using LBC samples can detect the most frequent driver mutations closer to the time of diagnosis, enabling the selection of the most effective first-line targeted therapy sooner, reducing delays or side effects from suboptimal treatments, patient anxiety and costs to healthcare systems, whilst improving patient outcomes.

1. Introduction

Worldwide, lung cancer is the leading cause of cancer-related death [1,2]. Prognosis remains poor, with average five-year survival rates of 16.2% [3]. Half of all lung cancers present stage 4 disease [1]. Non-small cell lung cancer (NSCLC) accounts for 85% of diagnosed lung cancer cases [2], and approximately 60% of advanced NSCLC subtypes have an actionable molecular target [4]. Current guidelines emphasise testing for epidermal growth factor receptor (EGFR), ALK receptor tyrosine kinase (ALK), Programmed death ligand 1 (PD-L1), ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) and B-Raf proto-oncogene, serine/threonine kinase (BRAF) driver mutations prior to systemic anti-cancer treatment [5,6]. However, with the emergence of effective targeted therapies, this panel is expanding. KRAS proto-oncogene, GTPase (KRAS) mutations G12C, G12V and G12D are arguably the most prevalent gain-of-function alterations found in 20–40% of lung adenocarcinomas [7]. G12C mutations alone are found in 13% of NSCLC cases [8], and rates of concurrent mutations with other actionable molecular targets such as EGFR, BRAF and ALK are incredibly uncommon [9,10]. Agents to target KRAS mutations are desired and are currently in development [11]. Sotorasib is a G12C GTPase inhibitor with a median overall survival of 12.5 months in patients with previously treated NSCLC harbouring the G12C KRAS mutation [12]. Sotorasib is approved by the National Institute for Health and Care Excellence (NICE) for early access to medicine on the National Health Service (NHS) [13].
Pancreatic adenocarcinoma (PDAC) carries a poor prognosis. Five-year survival for pancreatic cancer increased from 0.9% in 1975 to only 4.2% in 2011 in patients of all stages, while in surgically resected patients, it rose from 1.5% to 17.4% [14]. The disease often presents at an advanced and inoperable stage. Current treatment involves multi-agent combination chemotherapy regimens with limited targeted therapies to a small subset of patients; 1–2% of patients that have microsatellite instability (MSI)/mismatch repair deficiency (MMR) are eligible for immunotherapy [15]. Anti-EGFR therapies are used for patients presenting with BRAF mutations (11% of KRAS wildtype), and treatments for germline BRCA mutations (5–7% of patients) are also available; however, progression-free and overall survival rates remain poor [16].
KRAS driver mutations are found in 90–93% of PDAC [16,17]. The most common mutations are G12D, G12V, G12R, Q61H, Q61L and G12C, respectively [17]. Unlike NSCLC, G12C mutations are rare in PDAC, presenting in roughly 1% of cases [18]. PDACs associated with KRAS mutations are aggressive [19], and improved survival rates have been seen in Q61 mutations compared with other KRAS mutations [20]. Therapeutic targets for KRAS isoforms are being researched with the aim to provide more effective personalised treatments. Positive outcomes have been observed with GTPases inhibitors (KRAS G12C) and lipid nanoparticle (LNP)-formulated mRNA-based cancer vaccines that elicit an adaptive immune response against G12V and G12D mutated PDAC. Oligonucleotide therapies targeted against G12D mutations are also in phase II trials [14].
As the need for diagnostic panels expands, contradicting the shift to minimally invasive procedures to obtain samples, maximising the efficiency of the diagnostic yield is paramount to preventing repetitive invasive procedures for patients [5]. Rapid on-site evaluation (ROSE)-assessed cytology samples from endobronchial ultrasound (EBUS), transbronchial needle aspiration (TBNA) and endoscopic ultrasound (EUS) are the first-line investigation for the sampling of mediastinal, perihilar lymph nodes, peribronchial or peritracheal masses, and pancreatic lesions [21,22]. Performing highly specialised molecular testing on residual ROSE-assessed liquid-based cytology (LBC) specimens could provide an efficient approach for meeting the competing aims of expanding testing requirements from smaller samples. Recent studies have shown that cytology samples in CytoLyt® solution can be used effectively for NGS [23]; however, NGS involves significant infrastructures, skills and resources. The Biocartis Idylla™ provides a highly sensitive and specific platform for local rapid molecular testing, which is vital for improved clinical outcomes in patients with advanced disease [24]. The IdyllaTM is a fully automated, real-time polymerase chain reaction (PCR)-based molecular diagnostic system. Formalin-fixed paraffin-embedded (FFPE) biopsies and clots made from cytology samples are routinely used to detect EGFR, KRAS and BRAF mutations on the Idylla™ platform. Sample to result is obtained in 1.5 to 3 h. Initial multi-test studies have demonstrated the use of residual cytology specimens on the IdyllaTM for EGFR [25,26]; however, with the recent developments in NSCLC and potential future requirements for KRAS in PDACs, a broader panel is needed for adoption into routine practice.

Objectives

The objectives are to determine whether LBC samples provide a feasible and reliable alternative to FFPE samples for detecting EGFR, KRAS and BRAF mutations using the Idylla™ platform.

2. Methods

Patients attending for EUS- or EBUS-guided fine-needle aspirations with a clinical suspicion of non-small cell lung carcinoma or pancreatic adenocarcinoma were included for participation in the study (n = 55). Currently, EGFR, KRAS and BRAF testing can be conducted on the IdyllaTM platform. Due to the high incidence of KRAS mutations in PDACs, and with EGFR, KRAS and BRAF being the most common driver mutations in NSCLC, both patient cohorts were recruited to increase the sample size and statistical validity of the study. Cytology samples were obtained with ROSE [27] for routine diagnostics. Diagnosis was carried out microscopically by a consultant pathologist as per RcPath guidelines for reporting NSCLC and PDAC by means of an interpretation of morphology (PAP/HE) and was supplemented with immunohistochemistry where applicable. The inclusion criteria was based on the appropriate diagnosis and available residual samples for testing. Cases were excluded from the study if material was exhausted following routine diagnostics. Molecular analysis (EGFR, KRAS and BRAF for the lung and KRAS for the pancreas) was conducted on both FFPE clot samples and LBC samples.

Preparation of Samples

Cytology samples were transported to the laboratory in CytoLyt® Solution for the preparation of a Thinprep PAP slide and a thrombin and plasma clot for diagnosis, routine molecular testing and additional molecular testing. From the FFPE clot samples, two sections were cut at a 10 µm thickness using the Leica RM2235, and the samples were tested using IdyllaTM cartridges as per individual manufacturers’ instructions for EGFR, KRAS and BRAF.
Both the PAP slide from LBC and the HE slide from the FFPE clot sample were assessed microscopically to obtain a count of neoplastic cells and the neoplastic cell content (%).
The residual LBC samples were tested for an additional molecular analysis (EGFR/KRAS/BRAF). Pellets were formed by centrifuging the remaining sample and processed by placing them straight onto the lysis pad of the FFPE IdyllaTM cartridges (20 µL).
The IdyllaTM EGFR mutation test (Biocartis, Mechelen, Belgium) is an in-vitro diagnostic (IVD) test that uses real-time PCR to qualitatively detect exon 18 (G719A/C/S), exon 21 (L858R, L861Q), exon 20 (T790M, S768I) mutations, exon 19 deletions and exon 20 insertions in the EGFR oncogene. The testing platform has been IVD-validated by the manufacturer for FFPE human tissue samples of NSCLC. The test is also validated at the Royal Cornwall Hospital for use on FFPE sections from human NSCLC against Sanger Sequencing [26]. The limit of detection of this assay is between 1–5% (mutation-dependent) with a background of genomic wild type DNA, the local sensitivity being 90% and specificity 100%.
The KRAS assay is used to detect mutations in codons 12, 13, 59, 61, 117 or 146 of the KRAS oncogene, verified for use at RCHT by comparison Pyrosequencing (100% specificity and sensitivity) technology.
The BRAF assay identifies V600E/E2/D and V600K/R/M mutations in codon 600 of the BRAF gene, verified locally against the Cobas Roche platform 4800 (100% specificity and sensitivity).
Each assay (EGFR/KRAS/BRAF) independently performs liquefaction, cell lysis, DNA extraction and amplification of the target regions. The testing package includes a software analysis of the DNA primer amplification, which will differentiate between wild-type and mutated amplicons. This is achieved by the dissociation of the annealed fluorescent beacons using heat. The fluorescent profile is then interpreted by the in-built algorithm so as to display any identified mutation(s) on the console.
The results of the mutations (IdyllaTM output) and integrity of the DNA were collected to study the limit of detection between LBC and FFPE clot samples per assay type. For each valid PCR curve, a cycle of quantification value (Cq) is calculated. Cq values are inverse to the amount of target nucleic acid that is in the sample and correlate to the number of target copies. It is a pseudo value that can be used to estimate the integrity of the DNA that is being sequenced. Samples for which the wild-type Cq was within a predefined range were given a genetic call [no mutation or a group of mutations]. A call of an INVALID result is where the output was not able to be determined. The delta Cq value relates to the relative gene expression value. The lower the value, the higher the amount of targeted nucleic acid in the sample. The Cq values for both methodologies were put through a t-test to establish whether there was a significant difference in the cycles of PCR (Cq) on the Idylla platform.
This study received medical ethical approval on 18 June 2020. IRAS Number: 275530.

3. Results

55 patients were recruited, 45 had both LBC and Clot FFPE samples for analysis. From these, a total of 59 assays were carried out, as shown in Table 1.
The 29 LBC samples tested for EGFR mutations demonstrated 26 WT DNA and three EGFR exon 19 mutations. The 22 LBC samples tested for KRAS showed 13 WT and nine KRAS mutations. 7/9 KRAS mutations were detected in the PDAC cohort (87.5%), three G12D, two Q61H and two G12R mutations. 2/9 KRAS mutations were detected in the NSCLC (Adenocarcinoma) cohort (25%), one G12C and one G12V (Table 2). 0/6 KRAS mutations were detected in the lung Squamous cell carcinomas. The eight LBC samples tested for BRAF showed eight WT DNA results.
The study showed a 50% invalid rate in the BRAF FFPE clot samples (Table 3). Four cases did not have sufficient samples remaining in the block, a reflection of having exhausted the samples due to the clot samples routinely having immunocytochemistry (TTF1, P40, ROS1, PDL and ALK) prior to the molecular testing for the study. BRAF was run last after EGFR and KRAS. There were no invalids in any of the LBC samples.
There was a 97% (28/29) correlation of LBC with Clot FFPE results for EGFR testing. An S768I mutation (L015) (false positive) was erroneously detected in the FFPE sample as a result of over-amplification (Table 2). This has been previously observed in other studies for this mutation on the IdyllaTM platform [28] and is being addressed in an updated version of the IdyllaTM EGFR test. The sample was referred for next-generation sequencing (NGS), and by this method no mutation was identified. Retrospectively, comparing the sample size and DNA integrity can help with identifying the rationale behind the discordant result. The clot WT EGFR Cq (26.1) required more PCR cycles to amplify the DNA that was present in the sample, and though the mutation call was made, it was very near the cut-off of a true mutation call (26), highlighting the importance of scrutinising the Cq values. The LBC Cq value of 18.8 indicated a high level of well-preserved DNA and a high tumour yield in the LBC sample, confirming a reliable WT result.
The analysis of samples for KRAS demonstrated a 90% (20/22) correlation of LBC with the Clot FFPE results. One case that was discordant (L038) demonstrated a KRAS G12V mutation with a WT DNA Cq value of 18 and a high delta Cq of 8.4 detected in the LBC sample, which was not detected in the FFPE sample (Table 2). The green circle in the graph on the left in Figure 1 shows the LBC sample G12V mutation as being well within the threshold of detection. The initial FFPE clot sample (red circle) and the repeat FFPE clot run (red square) are both outside the Biocartis algorithm cut-off for detection. Despite there being a sufficient neoplastic cell content (HE confirmed 80% and >200 cells), the call on the Idylla was ‘no mutation’. The lack of available DNA in the sample meant that the limit of detection of the test was reduced and that the mutation detected call was just missed in both cases. The study demonstrated the detection for low allele frequency mutations in the LBC sample.
The second case that was non-correlated was the case of Panc 5B. Figure 2 demonstrates high levels of WT KRAS with amplification at a Cq of 20 with a G12R mutation detected at a Cq of 24 in the LBC sample. The tumour cell count was 40% > 100 cells. The Clot FFPE sample that was reported as no mutation detected (WT only) had only 5% tumour and <0 cells. The KRAS mutation was detected after a Cq of >33, past the Idylla internal cut-off threshold, highlighting that 5% and <50 cells is too low for a reliable detection in FFPE clot samples.
The lowest tumour yield in LBC samples for the detection of a mutation was 5% neoplastic cell content and >100 tumour cells for EGFR WT with a Cq of <26, and 5% neoplastic cell content and >50 tumour cells for KRAS WT with a Cq of <33 for WT (Table 2). Necrosis or excessive amounts of inflammatory cell populations did not impact the results, provided that sufficient tumour cells were present.
The 8/8 WT BRAF results for LBC and FFPE showed a concordance.
Using the criteria in Table 4 provides an effective way of assessing sample suitability and an analysis for an assurance of the reliability and integrity of the results. The IdyllaTM test cartridges have thresholds that are applied to determine if a PCR curve is a valid (true) amplification curve and whether a mutation is detected, and these are based on a number of parameters within the software decision tree and may vary depending on factors such as the total DNA input.
The results (Table 2) demonstrate the limit of detection for the techniques used with LBC samples and are summarised in Table 4. BRAF mutations were not detected in this study, and thus the thresholds provided by the manufacturer for FFPE were used for the assessment of WT BRAF.
The LBC samples demonstrated higher levels of well-preserved DNA. Lower Cq values were consistently seen in the LBC samples when compared with the Clot FFPE samples (Table 5).
The difference between the LBC KRAS WT and Clot KRAS WT Cq values was significant at p < 0.01 with a t-value of −6.42925 (n = 22). The difference between the LBC EGFR WT and Clot EGFR WT Cq values was significant at p < 0.01 with a t-value of −10.92458 (n = 29). The difference between the LBC KRAS WT and Clot KRAS WT Cq values was significant at p < 0.01 with a t-value of −4.00241 (n = 7). The difference between the LBC BRAF WT and Clot FFPE BRAF WT Cq values was significant at p < 0.01 with a t-value of −4.00241 (n = 7).
The difference between the Cq values for WT DNA in all the LBC samples and Clot FFPE samples was significant at p < 0.01. (n = 54) The t-value was −5.62499. The p-value was <0.00001. There were significantly higher levels of well-preserved DNA in the LBC samples for each test type.

4. Discussion

The sample recruitment was lower than anticipated due to a reduced patient presentation during the coronavirus pandemic, leading to the BRAF cartridges expiring, which resulted in fewer tests being performed (Table 1). The initial study focused on EGFR in LBC samples only. The expansion of the project resulted in KRAS and BRAF for LBC samples, resulting in seven more EFGR cases than KRAS cases. In cases where ROSE deemed the sample insufficient, the entire sample was made into a clot to preserve material for the diagnosis, resulting in a PAP not being made for a comparison.
Using the LBC PAP slides provides an indication of the neoplastic cell count; however, this must be considered against the remaining samples in the vial after processing, as paucicellular samples will appear more cellular on the slide.
Cytology samples are often the only modality to obtain diagnostic material. Using residual LBC samples provides an advantageous medium for molecular testing (Figure 3), demonstrating increased sensitivity compared to FFPE clot samples, with no invalids. LBC Cq values were lower, illustrating high levels of well-preserved DNA. The use of whole cells fixed in alcohol, rather than sections of cells fixed in formalin, results in a higher nucleic acid yield, superior DNA preservation and the removal of the formalin fixation artefact [24]. This in turn reduces the likelihood of false positive or false negative results, increasing the sensitivity for detecting low allele frequency mutations. Using LBC samples eliminates concerns about erroneous DNA contamination associated with thrombin and plasma clots and removes multiple processing steps, reducing the likelihood of cross contamination. ROSE is recommended to ensure that sufficient material is obtained and that the DNA is preserved optimally to effectively cover the full scope of testing requirements.
KRAS testing in NSCLC has recently been FDA approved and is available to a subset of patients on an early access scheme. Due to the mutual exclusivity of EGFR, KRAS and BRAF, laboratories can streamline workflows by testing the most common mutation first (KRAS), and if only WT KRAS is detected they can then proceed to EGFR and BRAF testing, optimally preserving samples and resources.

5. Conclusions

The Biocartis Idylla™ provides a highly sensitive, reliable and rapid testing platform for LBC samples for the detection of EGFR and KRAS mutations. BRAF mutations were not detected in the participant cohort; however, all LBC WT BRAF results correlated with the results from the FFPE clot samples.
Using the criteria demonstrated in Table 4 provides an effective way of assessing sample suitability and an analysis for the assurance of the reliability and integrity of the results. The Idylla™ cartridges are currently not IVD-labelled for the use of liquid-based cytology, but our research showed that they had an enhanced performance when compared with FFPE clot samples.
The benefits of utilising residual LBC samples are demonstrated in Figure 3.
Using the IdyllaTM to conduct rapid molecular testing on LBC cytology samples enables localised rapid testing without the need for significant infrastructures, skills and resources. The study recognises the need for NGS at larger centres, but turnaround times are often prolonged (several weeks), which has significant consequences for patients [29]. Testing locally on residual LBC samples for the most common mutation facilitates timely first-line treatments for the majority of patients, reserving the FFPE samples for NGS testing at centralised sites. Detecting the most common mutations sooner helps to reduce patient anxiety, and, in cases where a mutation is detected, the best treatment plan can commence, thereby removing unnecessary delays as well as suboptimal or unnecessary treatments [30].

Author Contributions

Conceptualization, L.W.; methodology, L.W. and M.J.; validation, L.W., M.J. and J.G.; formal analysis, L.W. and M.J.; investigation, L.W.; data curation, L.W.; writing—original draft preparation, L.W., M.J., B.P. and D.S.; writing—review and editing, L.W., M.J., B.P., D.S. and J.G.; visualization, L.W.; supervision, L.W. and J.G.; funding acquisition, L.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Biocartis for the supply of the IdyllaTM cartridges, grant number PN-1992.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board (or Ethics Committee) of Royal Cornwall Hospital NHS Trust and HRA and Health and Care Research Wales (HCRW) protocol code 275530 and date of approval 18/06/2020 for studies involving humans.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data that supported the findings in this study is available on reasonable request from the corresponding author. The data is not publically available due to ethical restrictions.

Acknowledgments

Thank you to Gerard Little and Chantelle Adams for processing the samples. Thank you to Geert Maertens for advice and updates on molecular advances, and to Lize Bollen and Sarah Saunders for supporting the study and providing scientific advice. Thank you to the respiratory team for providing consenting participants and obtaining the samples. Thank you to the patients that participated in the study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cancer Research UK. 2021 Lung Cancer Statistics Based on: Cancer Incidence from Cancer Intelligence Statistical Information Team at Cancer Research UK (201520142017 UK Average). Available online: https://www.cancerresearchuk.org/health-professional/cancer-statistics-for-the-uk (accessed on 17 December 2021).
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  3. Office for National Statistics. Cancer Survival by Stage at Diagnosis for England. Available online: https://www.gov.uk/government/statistics/cancer-survival-in-england-for-patients-diagnosed-between-2014-and-2018-and-followed-up-until-2019/cancer-survival-in-england-for-patients-diagnosed-between-2014-and-2018-and-followed-up-to-2019 (accessed on 12 August 2021).
  4. National Comprehensive Cancer Network. NCCN Guidelines: Non-Small Cell Lung Cancer. Version 1.2020. 2019. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 8 January 2020).
  5. Liam, C.K.; Mallawathantri, S.; Fong, K.M. Is tissue still the issue in detecting molecular alterations in lung cancer? Respirology 2020, 25, 933–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. NICE guideline [NG122] Lung Cancer: Diagnosis and Management. Available online: https://www.nice.org.uk/guidance/ng122 (accessed on 9 November 2021).
  7. Wood, K.; Hensing, T.; Malik, R.; Salgia, R. Prognostic and Predictive Value inKRASin Non–Small-Cell Lung Cancer. JAMA Oncol. 2016, 2, 805–812. [Google Scholar] [CrossRef]
  8. Arbour, K.C.; Jordan, E.; Kim, H.R.; Dienstag, J.; Yu, H.A.; Sanchez-Vega, F.; Lito, P.; Berger, M.; Solit, D.B.; Hellmann, M.; et al. Effects of Co-occurring Genomic Alterations on Outcomes in Patients with KRAS-Mutant Non–Small Cell Lung Cancer. Clin. Cancer Res. 2017, 24, 334–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Scheffler, M.; Ihle, M.A.; Hein, R.; Merkelbach-Bruse, S.; Scheel, A.H.; Siemanowski, J.; Brägelmann, J.; Kron, A.; Abedpour, N.; Ueckeroth, F.; et al. K-ras Mutation Subtypes in NSCLC and Associated Co-occuring Mutations in Other Oncogenic Pathways. J. Thorac. Oncol. 2019, 14, 606–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Shaw, A.T.; Solomon, B. Targeting Anaplastic Lymphoma Kinase in Lung Cancer. Clin. Cancer Res. 2011, 17, 2081–2086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Araujo, L.H.; Souza, B.M.; Leite, L.R.; Parma, S.A.F.; Lopes, N.P.; Malta, F.S.V.; Freire, M.C.M. Molecular profile of KRAS G12C-mutant colorectal and non-small-cell lung cancer. BMC Cancer 2021, 21, 193. [Google Scholar] [CrossRef]
  12. Amgen. Amgen’s Investigational KRAS G12C Inhibitor Sotorasib Demonstrated Rapid, Deep and Durable Responses in Previously Treated Patients with Advanced Non-Small Cell Lung Cancer. Available online: https://prn.to/3px7knl (accessed on 28 January 2021).
  13. Sotorasib–Medicines–SPS–Specialist Pharmacy Service–The First Stop for Professional Medicines Advice. Available online: https://www.sps.nhs.uk/medicines/sotorasib/ (accessed on 28 January 2021).
  14. Lee, M.S.; Pant, S. Personalizing Medicine with Germline and Somatic Sequencing in Advanced Pancreatic Cancer: Current Treatments and Novel Opportunities. Am. Soc. Clin. Oncol. Educ. Book 2021, 41, e153–e165. [Google Scholar] [CrossRef]
  15. Kenney, C.; Kunst, T.; Webb, S.; Christina, D.; Arrowood, C.; Steinberg, S.M.; Mettu, N.B.; Kim, E.J.; Rudloff, U. Phase II study of selumetinib, an orally active inhibitor of MEK1 and MEK2 kinases, in KRASG12R-mutant pancreatic ductal adenocarcinoma. Investig. New Drugs 2021, 39, 821–828. [Google Scholar] [CrossRef]
  16. Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.-M.; Gingras, M.-C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.C.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [Google Scholar] [CrossRef]
  17. Waters, A.M.; Der, C.J. KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb. Perspect. Med. 2018, 8, a031435. [Google Scholar] [CrossRef]
  18. Bengtsson, A.; Andersson, R.; Ansari, D. The actual 5-year survivors of pancreatic ductal adenocarcinoma based on real-world data. Sci. Rep. 2020, 10, 16425. [Google Scholar] [CrossRef]
  19. Buscail, L.; Bournet, B.; Cordelier, P. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 153–168. [Google Scholar] [CrossRef]
  20. Witkiewicz, A.K.; McMillan, E.A.; Balaji, U.; Baek, G.; Lin, W.-C.; Mansour, J.; Mollaee, M.; Wagner, K.-U.; Koduru, P.; Yopp, A.; et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat. Commun. 2015, 6, 6744. [Google Scholar] [CrossRef]
  21. Du Rand, I.A.; Barber, P.V.; Goldring, J.; Lewis, R.A.; Mandal, S.; Munavvar, M.; Rintoul, R.; Shah, P.; Singh, S.; Slade, M.G.; et al. Summary of the British Thoracic Society Guidelines for advanced diagnostic and therapeutic flexible bronchoscopy in adults. Thorax 2011, 66, 1014–1015. [Google Scholar] [CrossRef] [Green Version]
  22. Shetty, D.; Bhatnagar, G.; Sidhu, H.S.; Fox, B.M.; Dodds, N.I. The increasing role of endoscopic ultrasound (EUS) in the man-agement of pancreatic and biliary disease. Clin. Radiol. 2013, 68, 323–335. [Google Scholar] [CrossRef]
  23. Tian, S.K.; Killian, J.K.; Rekhtman, N.; Benayed, R.; Middha, S.; Ladanyi, M.; Lin, O.; Arcila, M.E. 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, 1200–1205. [Google Scholar] [CrossRef] [Green Version]
  24. Arcila, M.E.; Yang, S.-R.; Momeni, A.; Mata, D.A.; Salazar, P.; Chan, R.; Elezovic, D.; Benayed, R.; Zehir, A.; Buonocore, D.J.; et al. Ultrarapid EGFR Mutation Screening Followed by Comprehensive Next-Generation Sequencing: A Feasible, Informative Approach for Lung Carcinoma Cytology Specimens with a High Success Rate. JTO Clin. Res. Rep. 2020, 1, 100077. [Google Scholar] [CrossRef]
  25. Petiteau, C.; Robinet-Zimmermann, G.; Riot, A.; Dorbeau, M.; Richard, N.; Blanc-Fournier, C.; Bibeau, F.; Deshayes, S.; Bergot, E.; Gervais, R.; et al. Contribution of the IdyllaTM System to Improving the Therapeutic Care of Patients with NSCLC through Early Screening of EGFR Mutations. Curr. Oncol. 2021, 28, 4432–4445. [Google Scholar] [CrossRef]
  26. Evrard, S.M.; Taranchon-Clermont, E.; Rouquette, I.; Murray, S.; Dintner, S.; Nam-Apostolopoulos, Y.-C.; Bellosillo, B.; Varela, M.; Nadal, E.; Wiedorn, K.H.; et al. Multicenter Evaluation of the Fully Automated PCR-Based Idylla EGFR Mutation Assay on Formalin-Fixed, Paraffin-Embedded Tissue of Human Lung Cancer. J. Mol. Diagn. 2019, 21, 1010–1024. [Google Scholar] [CrossRef]
  27. Glinski, L.; Shetty, D.; Iles, S.; Diggins, B.; Garvican, J. Single slide assessment: A highly effective cytological rapid on-site evaluation technique for endobronchial and endoscopic ultrasound-guided fine needle aspiration. Cytopathology 2018, 30, 164–172. [Google Scholar] [CrossRef]
  28. Pisapia, P.; Pepe, F.; Sgariglia, R.; Nacchio, M.; Russo, G.; Conticelli, F.; Girolami, I.; Eccher, A.; Bellevicine, C.; Vigliar, E.; et al. Next generation sequencing in cytology. Cytopathology 2021, 32, 588–595. [Google Scholar] [CrossRef]
  29. Finall, A.; Davies, G.; Jones, T.; Emlyn, G.; Huey, P.; Mullard, A. Integration of rapid PCR testing as an adjunct to NGS in diagnostic pathology services within the UK: Evidence from a case series of non-squamous, non-small cell lung cancer (NSCLC) patients with follow-up. J. Clin. Pathol. 2022, 18, 2021–207987. [Google Scholar] [CrossRef]
  30. Gregg, J.P.; Li, T.; Yoneda, K.Y. Molecular testing strategies in non-small cell lung cancer: Optimizing the diagnostic journey. Transl. Lung Cancer Res. 2019, 8, 286–301. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Output supplied by Biocartis following the review of the discordant FFPE result (L038). The graph on the left shows the threshold for positive and negative detection. The green circle is LBC sample. Red circle and red square are FFPE clot samples (one repeat). The graph on the right demonstrates the validity of the runs. The initial FFPE run was just inside the threshold for a valid result. The repeat run (red square) was considered invalid. The green circle (LBC) is well within range.
Figure 1. Output supplied by Biocartis following the review of the discordant FFPE result (L038). The graph on the left shows the threshold for positive and negative detection. The green circle is LBC sample. Red circle and red square are FFPE clot samples (one repeat). The graph on the right demonstrates the validity of the runs. The initial FFPE run was just inside the threshold for a valid result. The repeat run (red square) was considered invalid. The green circle (LBC) is well within range.
Jmp 03 00010 g001
Figure 2. Comparison of the Idylla output PCR graphs LBC vs. Clot FFPE for the case of Panc 5B. KRAS wild-type amplification is indicated by the black curves. KRAS G12R mutation is indicated by the magenta curves.
Figure 2. Comparison of the Idylla output PCR graphs LBC vs. Clot FFPE for the case of Panc 5B. KRAS wild-type amplification is indicated by the black curves. KRAS G12R mutation is indicated by the magenta curves.
Jmp 03 00010 g002
Figure 3. Benefits of utilising LBC samples compared with FFPE clot samples.
Figure 3. Benefits of utilising LBC samples compared with FFPE clot samples.
Jmp 03 00010 g003
Table 1. Number of LBC samples tested for each cancer type.
Table 1. Number of LBC samples tested for each cancer type.
KRASEGFRBRAFTOTAL
PANCREATIC ADENOCARCINOMA8--8
LUNG ADENOCARCINOMA829845
LUNG SQUAMOUS CELL CARCINOMA6--6
TOTAL22298 *59
* There were fewer numbers of BRAF tests due to expiration of the cartridges as a result of fewer cases during the pandemic.
Table 2. Comparison of LBC and Clot FFPE mutations detected.
Table 2. Comparison of LBC and Clot FFPE mutations detected.
Study NumberLBC Slide Tumour %LBC ResultLBC WT Cq ValueLBC Mutation Cq ValueLBC Delta Cq ValueClot Slide Tumour %Clot ResultClot WT Cq ValueClot Mutation Cq ValueClot Delta Cq Value
Panc 15% >50 cellsKRAS Q61H mutation detected21.7431.689.73<100 cells <5% tumourKRAS Q61H mutation detected25.3929.934.35
Panc 25% >100 cellsKRAS Q61H mutation detected22.3530.127.65100 cells 10% tumourKRAS Q61H mutation detected25.5329.233.8
Panc 350% >100 cellsKRAS G12D mutation detected25.1528.033.04<100 cells 5% tumourKRAS G12D mutation detected25.4530.855.67
Panc 5 ASlide not foundKRAS G12R mutation detected19.823.413.59>100 cells 60% tumourKRAS G12R mutation detected24.3031.126.86
Panc 5 B40% >100 cellsKRAS G12R mutation detected20.2424.894.77<50 cells 5% tumourNo mutation detected28.36n/an/a
Panc 6 A10% >100 cellsKRAS G12D mutation detected18.922.94.05>100cells 80% tumourKRAS G12D mutation detected21.6425.393.75
Panc 6 B10% >100 cellsKRAS G12D mutation detected18.3721.343.03<100 cells <5% tumourKRAS G12D mutation detected25.3929.934.35
Lung adeno L02140%>20 cellsKRAS G12Cmutation detected22.4127.616.24<200 cells >50% tumourKRAS G12Cmutation detected23.828.085.00
Lung SCC L03890% >100 cellsKRAS G12V mutation detected18.2126.728.44>200 cells >80%No mutation detected24.55n/an/a
Lung adeno L00250% >200 cellsEGFR
Mutation detected Exon 19
15.4204.8780% 500 cellsEGFR
Mutation detected Exon 19
21265.66
Lung adeno L01230% >200 cellsEGFR
Mutation detected Exon 19 deletion
20.725.24.1880% 100 cells.EGFR
Mutation detected Exon 19
21.526.324.62
Lung adeno L01590% >200 cellsNo mutation detected18.8n/an/a90%. >200EGFR Mutation detected S768I * false positive26.128.242.94
Lung adeno L0445%>100 cellsEGFR Mutation detected Exon 1919.1527.387.8575% >200 cellsEGFR Mutation detected Exon 1925.229.013.80
Green represents correlation of results between LBC and FFPE. Orange highlights where non correlations occurred.
Table 3. The number of mutations detected and the invalid rate for each test type.
Table 3. The number of mutations detected and the invalid rate for each test type.
Total Tests Number of Mutations DetectedInvalids LBCInvalids Clot FFPE
KRAS LBC2290/22
(0%)
0/22
(0%)
KRAS Clot227
EFGR LBC2930/29
(0%)
0/29
(0%)
EGFR Clot293
BRAF LBC800/8
(0%)
4/8
(50%)
BRAF Clot80
Table 4. LBC sample prerequisites and data scrutiny criteria.
Table 4. LBC sample prerequisites and data scrutiny criteria.
TestNeoplastic Cell Content
(LBC PAP)
Pellet VolumeWT Cq ValueDelta CqValid Amplification Curve
KRAS5%>50 cells20 μL<33Low Jmp 03 00010 i001
EGFR5%>100 cells20 μL<26Low Jmp 03 00010 i001
BRAF>50% 20 μL<33 *IFULow Jmp 03 00010 i001
* Instructions for use (IFU) for BRAF recommend 50% tumour for BRAF as macro section cannot be per-formed.
Table 5. Comparison of average Cq values by test type.
Table 5. Comparison of average Cq values by test type.
LBC Average
Cq Value
Clot FFPE Average Cq ValueLBC Average Mutation Cq ValueClot FFPE Average Mutation Cq Value
EGFR17.622.424.127.3
KRAS21.424.525.828.7
BRAF30.934.29--
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wheeldon, L.; Jones, M.; Probyn, B.; Shetty, D.; Garvican, J. Use of the Biocartis Idylla™ Platform for the Detection of Epidermal Growth Factor Receptor, BRAF and KRAS Proto-Oncogene Mutations in Liquid-Based Cytology Specimens from Patients with Non-Small Cell Lung Carcinoma and Pancreatic Adenocarcinoma. J. Mol. Pathol. 2022, 3, 104-114. https://doi.org/10.3390/jmp3020010

AMA Style

Wheeldon L, Jones M, Probyn B, Shetty D, Garvican J. Use of the Biocartis Idylla™ Platform for the Detection of Epidermal Growth Factor Receptor, BRAF and KRAS Proto-Oncogene Mutations in Liquid-Based Cytology Specimens from Patients with Non-Small Cell Lung Carcinoma and Pancreatic Adenocarcinoma. Journal of Molecular Pathology. 2022; 3(2):104-114. https://doi.org/10.3390/jmp3020010

Chicago/Turabian Style

Wheeldon, Leonie, Mary Jones, Ben Probyn, Dushyant Shetty, and James Garvican. 2022. "Use of the Biocartis Idylla™ Platform for the Detection of Epidermal Growth Factor Receptor, BRAF and KRAS Proto-Oncogene Mutations in Liquid-Based Cytology Specimens from Patients with Non-Small Cell Lung Carcinoma and Pancreatic Adenocarcinoma" Journal of Molecular Pathology 3, no. 2: 104-114. https://doi.org/10.3390/jmp3020010

Article Metrics

Back to TopTop