Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Biology

Ultra-Fast Amplicon-Based Next-Generation Sequencing in Non-Squamous Non-Small Cell Lung Cancer

Published: September 8, 2023 doi: 10.3791/65190

Summary

The increase of molecular biomarkers to be tested for non-squamous non-small cell lung cancer (NS-NSCLC) care management has prompted the development of fast and reliable molecular detection methods. We describe a workflow for genomic alteration assessment for NS-NSCLC patients using an ultra-fast-next generation sequencing (NGS) approach.

Abstract

The number of molecular alterations to be tested for targeted therapy of non-squamous non-small cell lung cancer (NS-NSCLC) patients has significantly increased these last few years. The detection of molecular abnormalities is mandatory for the optimal care of advanced or metastatic NS-NSCLC patients, allowing targeted therapies to be administrated with an improvement in overall survival. Nevertheless, these tumors develop mechanisms of resistance that are potentially targetable using novel therapies. Some molecular alterations can also modulate the treatment response. The molecular characterization of NS-NSCLC has to be performed in a short turnaround time (TAT), in less than 10 working days, as recommended by the international guidelines. In addition, the origin of the tissue biopsies for genomic analysis is diverse, and their size is continuously decreasing with the development of less invasive methods and protocols. Consequently, pathologists are being challenged to perform effective molecular technics while maintaining an efficient and rapid diagnosis strategy. Here, we describe the ultra-fast amplicon-based next-generation sequencing (NGS) workflow used in daily routine practice at diagnosis for NS-NSCLC patients. We showed that this system is able to identify the current molecular targets used in precision medicine in thoracic oncology in an appropriate TAT.

Introduction

Over the last decade, the development of targeted and immuno-therapies has significantly increased the overall survival (OS) of non-squamous non-small cell lung cancer (NS-NSCLC)1,2. In this regard, the ,number of mandatory genes and molecular targets to analyze when treating NS-NSCLC has increased over the last few years3,4.

Current international guidelines recommend testing EGFR, ALK, ROS1, BRAF, NTRK, RET, and MET at diagnosis of advanced NS-NSCLC5. Moreover, as new drugs have recently given very promising results in clinical trials, additional genomic alterations will shortly be screened in a number of additional genes, notably KRAS and HER2, along with BRAC1/BRAC2, PI3KA, NRG1, and NUT6,7,8,9. In addition, the status of different associated genes, such as STK11, KEAP1, and TP53 may be of strong interest for a better prediction of the response or resistance to some targeted therapies and/or immune checkpoint inhibitors (ICIs)10,11,12.

Importantly, the molecular alterations must be reported without significant delay to ensure careful clinical decision-making. The absence of molecular characterization of a tumor may lead to the initiation of non-targeted therapies such as chemotherapy with/without immunotherapy, leading to a suboptimal treatment strategy, as chemotherapy response is limited in patients with actionable alterations, such as EGFR mutations or gene fusions13.

Moreover, the current development of targeted therapies/immunotherapies in neoadjuvant and/or adjuvant settings could lead to systematically looking for, at least, EGFR and ALK alterations in early-stage NS-NSCLC as ICIs should be administered only in tumors that are wild-type for EGFR and ALK14. It is now also mandatory to test for the presence of EGFR mutations in early-stage NS-NSCLC, since osimertinib (a third-generation EGFR tyrosine kinase inhibitor) can be used as adjuvant therapy in EGFR-mutant NS-NSCLC15.

The strategy for the assessment of the different biomarkers in predicting the response to different targeted therapies and/or immunotherapies in NS-NSCLC patients is moving fast, which makes the identification of these biomarkers sequentially difficult3,16. In this regard, Next-Generation Sequencing (NGS) is now the optimal approach for high throughput parallel assessment of gene alterations in NS-NSCLC5,17.

However, NGS workflow can be difficult to master and may conduct to longer TAT18,19. Thus, many centers still perform sequential approaches (immunohistochemistry (IHC), fluorescence in situ hybridization (FISH) and/or targeted sequencing). However, this strategy is limited in case of small sample size and, above all, because of the increased number of actionable mutations that are required to be tested in NS-NSCLC20. Thus, ultra-fast and straightforward testing methods allowing the rapid assessment of gene alterations have become increasingly important for optimal clinical decision-making. Moreover, approved and accreditated systems for molecular testing are becoming mandatory for the prescription of specific targeted therapies.

Here, we describe an ultra-fast and automated amplicon-based DNA/RNA NGS assay for molecular testing of NS-NSCLC that is used in the Laboratory of Clinical and Experimental Pathology Laboratory (LPCE), Nice University Hospital, France and is accredited according to the ISO 15189 norm by the French Accreditation Committee (COFRAC) (https://www.cofrac.fr/). The COFRAC certifies that the laboratory fulfills the requirements of the standard ISO 15189 and COFRAC rules of application for the activities of testing/calibration in molecular analysis in automated NGS on a sequencer with the panel performed by the laboratory. Accreditation per the recognized international standard ISO 15189 demonstrates the laboratory’s technical competence for a defined scope and the proper operation of an appropriate management system in this laboratory. The benefits and limitations of this workflow, starting from the preparation of tissue biopsy samples to obtaining the report, are discussed.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

All procedures have been approved by the local ethics committee (Human Research Ethics Committee, Centre Hospitalier Universitaire de Nice, Tumorothèque BB-0033-00025). Informed consent was obtained from all patients to use samples and generated data. All samples were obtained from patients diagnosed with NS-NSCLC in LPCE (Nice, France) between 20 September and 31 January 2022 as part of the medical care.

1. Preparation of FFPE DNA and RNA samples using automated purification instrument ( API ) (Processing time: 5 h 15 min)

  1. Run planification on the instrument.
    1. Power on the instrument and sign in with the user name and password (Table of Materials). Create a purification run plan by clicking Run, then Add Plan, and by giving a name to the run plan.
    2. Then, select the appropriate purification kit and protocol for sequentially purifying DNA and RNA from formalin-fixed paraffin-embedded (FFPE) samples. Enable quantitation after purification.
    3. Select the desired elution volume (50 µL) from the dropdown list, then click Next. Select the number of samples that will be extracted.
    4. Then select each sample, click Edit, enter Sample ID, and then click Save.
  2. Preparate FFPE DNA and RNA samples using GPI
    1. Prepare FFPE tissue samples (4 cutting sections of 10 µm with a microtome) or perform macrodissection directly on FFPE blocks. Place each FFPE tissue sample in separated and identified FFPE sample processing tubes (Table of Materials).
    2. Centrifuge at 2000 x g for 1 min to collect the tissue at the bottom of the tubes.
    3. Add to each FFPE sample processing tube a protease digest master mix prepared from a nucleic acid purification kit and incubate at 60 °C for at least 60 min (Table of Materials). Incubate the samples at 90 °C for 60 min.
    4. After incubation, let the samples cool down for 5 min at room temperature (RT).
    5. For each FFPE sample processing tube, lift the inner tube, lock it by turning left, and then centrifugate the samples at 2000 x g for 10 min.
    6. Unlock the inner tubes by turning right, separate them from the outer tubes, and discard them. Keep the samples in the outer tubes on ice until loading on the API.
    7. Prepare a DNase digestion master mix and load it in prefilled FFPE DNA and RNA purification plate 2. Then, load the prepared samples in FFPE DNA and RNA purification plate 1 (Table of Materials).
  3. Start the purification run on API.
    1. On the API screen, click Run, select the Run Plan, then click Next.
    2. Follow the on-screen indications to load the API with the required consumables and reagents needed for the purification run (Table of Materials).
    3. When all the reagents are loaded, click Next. Close the API door and click Start.
    4. At the end of the run, click « unload » on the touchscreen and immediately remove the 48-well nucleic acid archive plate containing the purified sample DNA and RNA.
    5. Export the quantitation results. Seal the plate and store purified samples at-80 °C. Transfer the 96 well plate to the sequencer to be analyzed.
  4. Create a sample and create a run.
    1. Open and sign in to the software, then choose in the menu bar Samples and click Create Sample.
    2. Enter the sample's name, sex, and percentage of tumor cells, and complete the required fields and optional fields if needed.
    3. Save the details (the sex and the percentage of tumor cells are important for analyzing copy number variants [CNVs]).
    4. Choose Runs and Nucleic Acid to Result in the menu bar.
    5. Enter the run name in the Setup step. Click Next.
    6. Select assay in the Assays step. Click Next.
    7. Select the samples in the Samples step to run with the assay.
    8. Then, in the Selected Assays pane, click Assign. Click Next.
    9. Review the sample positions on the sample input plate. Click Next.
    10. Review the run plan summary, and then Save & Print or Save.

2. Automated NGS on the sequencer (Processing time: 30 min)

  1. Load the sample plate.
    NOTE: The Optimal nucleic acid concentration is 0.5 ng/µL. The validated nucleic acid concentration range is 0.33-1 ng/µL. For DNA and RNA, 1 ng/µL concentration was validated in the lab.
    1. The sequencer requires 20 µL total volume. Ensure there is sufficient volume for sample loading.
    2. Add positive controls (DNA and RNA) and NTC (DNA and RNA) to the 1st stage of the purification instrument without being extracted. Use DNA and RNA controls for each run to validate the run and validate the batches used. Add DNA and RNA controls to the plate at the end of the API run.
  2. Load the sequencer and start a run.
    1. Remove all the reagents from their boxes in the refrigerator or freezer and thaw at RT for a minimum period of 30 min (Table of Materials) and a maximum of 12 h.
    2. In the laboratory, keep the sequencer always turned ON following the advice given by the supplier during the on-site training. Sign in to the system.
    3. Load the plate from the purification instrument, which contains samples, after sealing the plate with a sheet of adhesive PCR plate foil (Table of Materials).
    4. Install all consumables in the deck. The sequencer provides step-by-step instructions to load each required consumable in a highlighted position (Table of Materials).
    5. Check that there are no precipitations on tube 3 of the Strip 2-HD. Flick the tube or gently vortex the strip to dissolve the precipitate if needed.
    6. Strip 1 and Strip 3 contain magnetic beads. This step is very critical: ensure that the beads are resuspended. There should be no beads left in the upper part of the tube.
    7. Turn the strip upside down and back 3-4 times to remove the beads. Hold the strip at one end with the strip seal oriented upward. Afterward, swiftly swing the strip downward using a quick, centrifugal arm movement, and finish by giving a sharp flick of the wrist.
    8. Centrifuge at 300 x g for 15 s. Repeat if needed. Centrifuge the strips with adapters and strip holders to minimize failures observed randomly in connection with residues of balls in the upper part of the strips.
    9. Close the system and click Next. Install the sequencing reagents bay doors (Table of Materials). Close the door. The sequencer automatically starts the run.
  3. Cleaning
    1. For cleaning after the run, when the run is complete, click Next.
    2. The door opens successively. Follow the on-screen instructions to empty the waste and remove all consumables. Click Next.
    3. Close the deck when finished, click Next. A 2 min UV cleaning starts. The sequencer is ready to start a new run.

3. Analysis of the results using the integrated software (Analysis time by patient [samples ADN and ARN]: 15 min)

NOTE: The technique is accredited by the French Accreditation Committee (COFRAC) ISO 15189 (https://www.cofrac.fr/)

  1. Data and results
    1. Select a run result or a sample result in the Results menu. In the Results/Run Results screen, all the runs are listed.
    2. Search the list of results by filtering in a column header.
    3. Click Results and click Sample Results. Then, click the name of the sample of interest in the sample name column to view sequencing results for a unique sample.
    4. Click Columns in the top left corner of the screen or deselect the columns from the table.
  2. View the QC results and view the amplicon coverage.
    1. Click the QC tab in the Results screen.
    2. For DNA, ensure that the median absolute pairwise differences (MAPDs) are between 0 and 0.5 to validate the copy number variants (CNV) analysis. The mapped reads must be greater than 1.5 million.
    3. For RNA, check if the mapped reads are between 150,000 and 400,000. Below this, there is a risk of false negatives, and beyond that, there is a risk of false positives.
    4. Click a sample name to open the Key Findings tab.
    5. Scroll to the amplicon Coverage Graphs.
    6. Review the coverage graphs. Define the minimal threshold for the amplicon coverage, as the supplier does not provide it. To define a range and a threshold, mix a control and a wild type (WT) sample. Then, observe the smallest value of the amplicon coverage for one or more selected mutations.
  3. View variant results.
    1. To export the data in tabular format, click Export in the upper right corner of the screen.
    2. To view the single nucleotid variant/insertion-deletion variant (SNV/INDEL) result, click the Variants tab, then click SNVs/Indels. Click Edit Filters and select No Filter.
      NOTE: The sensitivity threshold has been defined at 5% according to the recommendations of the INCA (French National Cancer Institute) group (https://www.e-cancer.fr/Professionnels-de-sante/Les-therapies-ciblees/Les-plateformes-de-genetique-moleculaire-des-cancers/Le-programme-d-assurance-qualite-des-plateformes).
    3. View Fusion results and view RNA Exon Variants: Click the Variants tab, then click Fusions.
    4. In the top right, click Visualization and RNA Exon Variant, then review the RNA Exon Variants plot.
    5. With this kit, exon skipping of the EGFR, MET, and AR genes can be detected. Click Visualization; it is easier to analyze.
    6. View RNA exon tile fusion imbalance: Click the Variants tab, then click Fusions.
    7. In the top right, click Visualization and RNA Exon Tile Fusion Imbalance, then review the RNA Exon Tile Fusion Imbalance plots. An imbalance fusion is a fusion between a known partner and a partner that is not covered by the panel.
    8. View CNV results: Click CNVs in the Variants tab to display the data.
      NOTE: It is necessary to be vigilant with the results of CNV by informing during the preparation of the samples of the gender and the percentage of tumor cells of the sample. The AR gene is only carried by the X chromosome. If the sex is not specified, the male sex could encounter losses, leading to false positive results in male patients.
  4. Review plugin results.
    1. Review Coverage Analysis plugin results: In the top right, click and select download files.
    2. The Coverage Analysis plugin generates a Coverage Analysis Report.
    3. Review molecular Coverage Analysis plugin results: In the top right, click and select download files. This analysis verifies if all the amplicons are covered and confirms the copy number variants (CNVs) results provided by the software.
  5. View assay metrics and the run report
    1. Click the Run Report tab in the Run Summary.
    2. To view the Run Report, select the Select Sample option in the dropdown menu.
    3. Assay metrics and the run report: Find the sequencing metrics at the top of the screen, followed by sample-specific metrics in the Run Samples table.
      NOTE: Information in the customer support archive (CSA) file can help troubleshoot results, but they can't be directly analyzed. The CSA files summarize the data of the entire run data and highlight the causes of an issue in case of a failed run.
    4. Download a run report: Click the Reports tab.
    5. Click Download Report to download a run report summary in PDF format.
  6. Generate variant report.
    1. Enable Generate Report in the Setup step to automatically generate a variant report for each sample during data analysis of a run.
    2. After generating the variant report for a sample, the system makes the report available in two places: First, a link is made available in the Results/Sample Results screen. Click the link to download the PDF.
    3. Second, go to the Variant Report pane in the Reports tab and click on the Download Report button. Sign the variant reports electronically or manually.
      NOTE: Electronically signed reports have (Sign off) after the sample name in the Sample Results screen.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

Using the procedure presented here, described in detail in our recent publications21, we developed an optimal workflow for the assessment of molecular alteration as a reflex testing in routinely performed clinical practice for diagnosis in patients with NS-NSCLC using an ultra-fast amplicon-based next-generation sequencing approach. The molecular workflow of the method is shown in Figure 1. The list of genes included in the panel is shown in Supplementary Figure 1.

Molecular analysis was performed in 259 patients with NS-NSCLC. It included 153 bronchial biopsies, 47 transthoracic biopsies, 39 surgical specimens, and 25 cellblocks from 13 pleural effusion and 7 EBUS (Table 1). The following driver mutant genes were observed in 242 valid or available NGS DNA/RNA analyses: KRAS (25.4%), EGFR (18.2%), BRAF (6.3%), ERBB2 (1.7%) and MET (1.7%). The following gene fusions were reported: ALK (4.3%), ROS1 (2.3%), RET (1.9%) and NTRK (0.8%). Following alterations were detected with an incidence below 1% (e.g., IDH1, CDKN2A, FGFR3, KIT, MTOR, FGFR4. NTRK3, NRAS, PIK3CA, IDH2, ERBB3, ERBB4, AR, CHECK2, SMO, PTEN). For DNA, 10 cases (4%) failed, whereas 5 cases (2%) failed for RNA. Failure was due to poor quality of DNA/RNA and/or low amount of DNA/RNA. Failed analysis occurred only with samples having less than 10% tumor cellularity. Thus, we defined the tumor cellularity cut-off at 10%. For DNA NGS results, 10 cases (4%) did not pass due to either the low quality of nucleic acids (6 cases) or insufficient nucleic acid amounts (4 cases with <13 ng of DNA). Similarly, five cases (2%) failed for RNA sequencing results due to both inadequate RNA quality and a low quantity of RNA (<13 ng of RNA). The average proportion of tumor cells in the unsuccessful cases was limited (15%, ranging from 10% to 20%), in contrast to the successful cases where tumor cell percentages ranged from 30% to 90%. Among the 15 failed samples, 11 originated from small tissue biopsies (with an average area of 2 mm2), while 4 were cytological specimens (such as endobronchial ultrasound [EBUS]).

The sensitivity of the method for the detection of SNVs and INDELs was 93.44%, for CNVs was 100%, and for fusions was 91.67% with a minimum tumor cell content of 10% (Table 1).

The mean TAT from endoscopy sampling from the molecular report was 72 h (range: 48-96 h), with a mean TAT from extraction to report of 24 h.

The analytical validation of the above-described NGS workflow was performed on 30 NS-NSCLC cases. The validation demonstrated 100% concordance with the gold standard methods previously used at our center and listed in detail in the original publication21: RT-PCR EGFR Mutation Test; RT-PCR KRAS Mutation Test; ALK IHC, ALK FISH; ROS1 IHC; ROS1 FISH; BRAFV600E; and DNA/RNA NGS methods.

The multi-step procedure from sampling to the molecular report of an EGFR/TP53-mutant lung adenocarcinoma is pictured in Figure 2. The representation of the mutation found in the biopsy sample using integrative genomic viewer (IGV) software is shown in Supplementary Figure 2. An example of a generated report from the reporting software is shown in Supplementary Figure 3.

Figure 1
Figure 1: Diagram of the panel workflow used in this study. This figure has been modified with permission from Ilié et al.21. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Multiple steps from sampling to molecular biology report. (A) Three bronchial biopsy samples received in the laboratory from the endoscopy room. (B) Hematoxylin/Eosin/Safran-stained slide of the bronchial biopsy sample showing a lung adenocarcinoma with about 40%-50% of tumor cells (100x magnification). (C) Report from the reporting software showing an EGFR mutation associated with a TP53 mutation in the tumor sample. Please click here to view a larger version of this figure.

Total number of patients 259
Bronchial biopsies 153
Transthoracic biopsies 47
Surgical specimens 39
Pleural effusion  13
EBUS 7
Driver mutant genes
KRAS  25.40%
EGFR  18.20%
BRAF 6.30%
ERBB2 1.70%
MET 1.70%
Gene fusions 
ALK  4.30%
ROS1 2.30%
RET  1.95%
NTRK 0.80%
Failure - DNA cases 10 cases (4%)
Failure - RNA cases 5 cases (2%)
Tumor cellularity cut-off  10%
Percentage range of tumor cells in the failed cases  10%–20%
Percentage range of tumor cells in successful cases  30%–90%
Sensitivity of the method 
Detection of SNVs and INDELs  93.44%
Detection of CNVs  100%
Detection of Fusions 91.67%

Table 1: Outcomes of molecular analysis. Molecular analysis was performed in 259 patients with NS-NSCLC, including 153 bronchial biopsies, 47 transthoracic biopsies, 39 surgical specimens, and 25 cellblocks from 13 pleural effusion and 7 EBUS.

Supplementary Figure 1: The genes included in the NGS oncomine precision assay GX panel. Please click here to download this File.

Supplementary Figure 2: Representation of generated reads and mutations found in the biopsy sample using IGV software. (A) EGFR gene and (B) TP53 gene. Please click here to download this File.

Supplementary Figure 3: Molecular report generated by the reporting software. Please click here to download this File.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

The development of an ultra-fast amplicon-based NGS approach as reflex testing for molecular alteration assessment at diagnosis of any stage NS-NSLC is an optimal option for the detection of all guideline-recommended and emerging biomarkers in NS-NSCLC5,22,23. While sequential methods (IHC, PCR, FISH) focus only on specific genes and can result in tissue material exhaustion, this NGS workflow allows a specific and sensitive evaluation of the status of 50 genes, including mutations, fusions, and amplifications, even with a low amount of nucleic acid (minimum 13 ng for each DNA and RNA sequencing according to this study).

In addition, this method provides quick and relevant results for the management of patients with NS-NSCLC. Compared to a bespoke approach, the absence of an initial selection of cases to be analyzed, depending on the tumor stage, and without waiting for the physician's request enables them to save precious time. In this line, it has been shown that NGS testing of advanced NS-NSCLC directly affects patient overall survival24,25. In addition, these molecular results are frequently obtained simultaneously from the IHC results, especially for the evaluation of the percentage of PD-L1 positive tumor cells26. The physician can get the "molecular profile" of the tumor with all the biomarkers required for the best therapeutic choice of first-line treatment27. In specific situations, the physician can enroll the patients into a clinical trial according to the genomic results28,29,30,31.

Both DNA and RNA analysis with the panel can be performed on 2-14 patients per run, excluding controls (1 positive and 1 negative control). It is possible to make three runs per week, which means that 42 patients can benefit from an ultra-fast NGS profiling of their tumor. The purificator system, which performs the fully automated extraction and quantification of DNA and RNA before library preparation, enables rapid evaluation of the feasibility of the sequencing analyses.

The ability to quantify the extracted DNA and RNA prior to library preparation, and the option to work with small amounts of nucleic acid (starting from a minimum of 10 ng of DNA or RNA, as per the specifications) allows a rapid assessment of sequencing analysis viability.

An NGS approach can help optimize the management of the sample workflow in thoracic oncology by being more comprehensive in the detection of actionable biomarkers. It has been shown that the PCR-based method has intrinsic limitations in detecting some rare mutations, especially EGFR exon 20 insertion mutations32. Thus, the NGS approach enables a higher proportion of patients to receive biomarker-focused therapies. In this context, it has been demonstrated that NGS reflex testing for all advanced NS-NSCLC is cost-effective33. Moreover, the fully automated nucleic acid extraction, library preparation, and sequencing integrated with this amplicon-based NGS System is a significant time saver for laboratory staff.

However, implementing ultra-fast next-generation sequencing (NGS) as a reflex testing for diagnosing NS-NSCLC could encounter certain constraints in daily clinical practice. The gene panel's scope (encompassing 50 genes) is fewer compared to the extensive panels that comprise several hundred genes34. The panel includes all the recommended genes that are associated with a targeted therapy in thoracic oncology. Furthermore, some genes of interest with a potential prognostic value are not included. For instance, KEAP1, STK11, SMARCA4, and RB1 are not present on the panel used in the present work. These genes can be important in the future for the selection of patients to benefit from the FDA-approved KRASG12C inhibitors sotorasib9. The use of ultra-fast NGS with amplicon sequencing technology may also introduce the potential for missing rare fusion gene partners35. In addition, the imbalance analyses revealed some false positive ALK rearrangement initially36. The threshold for detecting ALK imbalance fusions was increased in the new modified assay. However, it is recommended to validate an imbalance result with an orthogonal technique (IHC and/or FISH).

Above all, this workflow raises questions related to the economic sustainability and the reimbursement of the NGS test, which can vary greatly depending on the country's health system, especially in Europe37. For the cost-effectiveness approach, it is imperative to run enough samples at the same time to save on consumables, which is sometimes not possible, as it depends on the sample recruitment and the laboratory35. Moreover, the possibility of generating an unnecessary additional workload for the laboratory staff should be discussed and requires good communication between physicians, surgeons, pathologists, and technical staff.

The simultaneous use of both DNA and RNA NGS is ideal but remains debatable. Knowing that most of the genomic alterations for targeted therapy are mutually exclusive38, it could be possible to first perform DNA and then RNA sequencing analysis. However, this strategy would consequently lead to a longer TAT to generate a report.

The adequacy of both the quantity and quality of extracted nucleic acids could represent a limitation in ensuring the robustness of NGS analyses, especially in cases involving hybrid capture sequencing technology and extensive panels that require a higher amount of material with higher TAT6. In this regard, this amplicon-based NGS System offers the advantage of requiring only 13 ng of DNA and RNA for the panel and has a very low overall failure rate. This is highly valuable in thoracic oncology since the size of samples is getting smaller and smaller20,39,40.

The indications of molecular testing are continuously changing in thoracic oncology and, for some gene alterations, are now recommended in adjuvant settings15. In our opinion, it is highly probable that several additional targeted therapies will emerge for the treatment of early-stage NS-NSCLC in routine clinical practice, reinforcing the need to promptly search for biomarkers at diagnosis. Taken together, the current progress in precision medicine in lung cancer will ineluctably favor an ultra-fast NGS approach that allows a more comprehensive, rapid, and material-saving molecular profiling of tumors.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

Christophe Bontoux participates in paid speaker activities and receives benefits from Thermo Fisher Scientific. Paul Hofman participates in paid speaker activities and receives benefits and funding from Thermo Fisher Scientific.

Acknowledgments

We thank Thermo Fisher Scientific for giving us the possibility to use their device and materials.

Materials

Name Company Catalog Number Comments
96 well hard shell plate clear Thermo Fisher Scientific (Waltham, Massachusetts, USA) 4483354
Adhesive PCR Plate Foil Thermo Fisher Scientific (Waltham, Massachusetts, USA) AB0626
AutoLys M tube  Thermo Fisher Scientific (Waltham, Massachusetts, USA) A38738 FFPE sample processing tubes
Genexus Barcodes 1-32 HD Thermo Fisher Scientific (Waltham, Massachusetts, USA) A40261
Genexus GX5 Chip and Genexus Coupler Thermo Fisher Scientific (Waltham, Massachusetts, USA) A40269
Genexus Pipette Tips Thermo Fisher Scientific (Waltham, Massachusetts, USA) A40266
Genexus Purification Instrument Thermo Fisher Scientific (Waltham, Massachusetts, USA) A48148 Automated purification instrument (API)
Genexus Sequencing Kit Thermo Fisher Scientific (Waltham, Massachusetts, USA) A40271
Genexus Templating Strips 3-GX5 and 4 Thermo Fisher Scientific (Waltham, Massachusetts, USA) A40263
Genexus Integrated Sequencer Thermo Fisher Scientific (Waltham, Massachusetts, USA) A45727
Ion Torrent  Genexus FFPE DNA/RNA Purification Combo Kit Thermo Fisher Scientific (Waltham, Massachusetts, USA) A45539
Oncomine  Precision Assay GX (OPA) Panel (included Strips 1 and 2-HD) Thermo Fisher Scientific (Waltham, Massachusetts, USA) A46291

DOWNLOAD MATERIALS LIST

References

  1. Howlader, N., et al. The effect of advances in lung-cancer treatment on population mortality. New England Journal of Medicine. 383 (7), 640-649 (2020).
  2. Melosky, B., et al. The rapidly evolving landscape of novel targeted therapies in advanced non-small cell lung cancer. Lung Cancer. 160, 136-151 (2021).
  3. Hanna, N. H., et al. Therapy for stage IV non-small-cell lung cancer with driver alterations: ASCO and OH (CCO) joint guideline update. Journal of Clinical Oncology. 39 (9), 1040-1091 (2021).
  4. Kerr, K. M., et al. The evolving landscape of biomarker testing for non-small cell lung cancer in Europe. Lung Cancer. 154, 161-175 (2021).
  5. Mosele, F., et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO precision medicine working group. Annals of Oncology. 31 (11), 1491-1505 (2020).
  6. Kazdal, D., Hofman, V., Christopoulos, P., Ilié, M., Stenzinger, A., Hofman, P. Fusion-positive non-small cell lung carcinoma: Biological principles, clinical practice, and diagnostic implications. Genes Chromosomes and Cancer. 61 (5), 244-260 (2022).
  7. Li, B. T., et al. Trastuzumab deruxtecan in HER2 -mutant non-small-cell lung cancer. New England Journal of Medicine. 386 (3), 241-251 (2022).
  8. Bontoux, C., Hofman, V., Brest, P., Ilié, M., Mograbi, B., Hofman, P. Daily practice assessment of KRAS status in NSCLC patients: A new challenge for the thoracic pathologist is right around the corner. Cancers. 14 (7), 1628 (2022).
  9. Skoulidis, F., et al. Sotorasib for lung cancers with KRAS p.G12C mutation. New England Journal of Medicine. 384 (25), 2371-2381 (2021).
  10. Hellyer, J. A., et al. Impact of tumor suppressor gene co-mutations on differential response to EGFR TKI therapy in EGFR L858R and Exon 19 deletion lung cancer. Clinical Lung Cancer. 23 (3), 264-272 (2022).
  11. Mograbi, B., Heeke, S., Hofman, P. The importance of stk11/lkb1 assessment in non-small cell lung carcinomas. Diagnostics. 11 (2), 196 (2021).
  12. Nadal, E., et al. Two patients with advanced-stage lung adenocarcinoma with radiologic complete response to nivolumab treatment harboring an STK11/LKB1 mutation. JCO Precision Oncology. 4, 1239-1245 (2022).
  13. Smeltzer, M. P., et al. The international association for the study of lung cancer global survey on molecular testing in lung cancer. Journal of Thoracic Oncology. 15 (9), 1434-1448 (2020).
  14. Ahern, E., Solomon, B. J., Hui, R., Pavlakis, N., O'Byrne, K., Hughes, B. G. M. Neoadjuvant immunotherapy for non-small cell lung cancer: Right drugs, right patient, right time. Journal for ImmunoTherapy of Cancer. 9 (6), e002248 (2021).
  15. Wu, Y. -L., et al. Osimertinib in resected EGFR-mutated non-small-cell lung cancer. New England Journal of Medicine. 383 (18), 1711-1723 (2020).
  16. Hanna, N. H., et al. Therapy for stage IV non-small-cell lung cancer without driver alterations: ASCO and OH (CCO) joint guideline update. J Clin Oncol. 38 (14), 1608-1632 (2020).
  17. de Maglio, G., et al. The storm of NGS in NSCLC diagnostic-therapeutic pathway: How to sun the real clinical practice. Critical Reviews in Oncology/Hematology. 169, 103561 (2022).
  18. DiStasio, M., Chen, Y., Rangachari, D., Costa, D. B., Heher, Y. K., VanderLaan, P. A. Molecular testing turnaround time for non-small cell lung cancer in routine clinical practice confirms feasibility of CAP/IASLC/AMP guideline recommendations: A single-center analysis. Clinical Lung Cancer. 18 (5), e349-e356 (2017).
  19. Heeke, S., et al. Use of the ion PGM and the genereader NGS systems in daily routine practice for advanced lung adenocarcinoma patients: A practical point of view reporting a comparative study and assessment of 90 patients. Cancers. 10 (4), 88 (2018).
  20. Hofman, P. The challenges of evaluating predictive biomarkers using small biopsy tissue samples and liquid biopsies from non-small cell lung cancer patients. Journal of Thoracic Disease. 11, S57-S64 (2019).
  21. Ilié, M., et al. Setting up an ultra-fast next-generation sequencing approach as reflex testing at diagnosis of non-squamous non-small cell lung cancer; experience of a single center (LPCE, Nice, France). Cancers. 14 (9), 2258 (2022).
  22. Zacharias, M., et al. Reflex testing in non-small cell lung carcinoma using DNA-and RNA-based next-generation sequencing-a single-center experience. Translational Lung Cancer Research. 10 (11), 4221-4234 (2021).
  23. Miller, T. E., et al. Clinical utility of reflex testing using focused nextgeneration sequencing for management of patients with advanced lung adenocarcinoma. Journal of Clinical Pathology. 71 (12), 1108-1115 (2018).
  24. Al-Ahmadi, A., et al. Next generation sequencing of advanced non-small cell lung cancer: utilization based on race and impact on survival. Clinical Lung Cancer. 22 (1), 16.e1-22.e1 (2021).
  25. Kim, J. H., Yoon, S., Lee, D. H., Jang, S. J., Chun, S. M., Kim, S. W. Real-world utility of next-generation sequencing for targeted gene analysis and its application to treatment in lung adenocarcinoma. Cancer Medicine. 10 (10), 3197-3204 (2021).
  26. Sheffield, B. S., et al. Point of care molecular testing: Community-based rapid next-generation sequencing to support cancer care. Current Oncology. 29 (3), 1326-1334 (2022).
  27. Camidge, D. R., Doebele, R. C., Kerr, K. M. Comparing and contrasting predictive biomarkers for immunotherapy and targeted therapy of NSCLC. Nature Reviews Clinical Oncology. 16 (6), 341-355 (2019).
  28. Rosas, D., Raez, L. E., Russo, A., Rolfo, C. Neuregulin 1 gene (Nrg1). A potentially new targetable alteration for the treatment of lung cancer. Cancers. 13 (20), 5038 (2021).
  29. Shapiro, G. I., et al. A Phase 1 study of RO6870810, a novel bromodomain and extra-terminal protein inhibitor, in patients with NUT carcinoma, other solid tumours, or diffuse large B-cell lymphoma. British Journal of Cancer. 124 (4), 744-753 (2021).
  30. Schoenfeld, A. J., et al. The genomic landscape of SMARCA4 alterations and associations with outcomes in patients with lung cancer. Clinical Cancer Research. 26 (21), 5701-5708 (2021).
  31. Zhang, K., et al. Identification of deleterious NOTCH mutation as novel predictor to efficacious immunotherapy in NSCLC. Clinical Cancer Research. 26 (14), 3649-3661 (2020).
  32. Shen, C. I., et al. Real-world evidence of the intrinsic limitations of PCR-based EGFR mutation assay in non-small cell lung cancer. Scientific Reports. 12 (1), 13566 (2022).
  33. Zou, D., et al. Diagnostic value and cost-effectiveness of next-generation sequencing-based testing for treatment of patients with advanced/metastatic non-squamous non-small-cell lung cancer in the United States. Journal of Molecular Diagnostics. 24 (8), 901-914 (2022).
  34. Zhong, Y., Xu, F., Wu, J., Schubert, J., Li, M. M. Application of next generation sequencing in laboratory medicine. Annals of Laboratory Medicine. 41 (1), 25-43 (2020).
  35. Bruno, R., Fontanini, G. Next generation sequencing for gene fusion analysis in lung cancer: A literature review. Diagnostics. 10 (8), 521 (2020).
  36. Hofman, V., et al. Ultra-fast gene fusion assessment for non-squamous non-small cell lung cancer. JTO Clinical and Research Reports. 4 (2), 100457 (2022).
  37. Horgan, D., et al. Personalized medicine perspective identifying the steps required to effectively implement next-generation sequencing in oncology at a national level in Europe. Journal of Personalized Medicine. 12 (1), 72 (2022).
  38. Cohen, D., et al. Optimizing mutation and fusion detection in NSCLC by sequential DNA and RNA sequencing. Journal of Thoracic Oncology. 15 (6), 1000-1014 (2020).
  39. Goswami, R. S., et al. Identification of factors affecting the success of next-generation sequencing testing in solid tumors. American Journal of Clinical Pathology. 145 (2), 222-237 (2016).
  40. Ilie, M., Hofman, P. Pitfalls in Lung Cancer Molecular Pathology: How to Limit them in Routine Practice. Current Medicinal Chemistry. 19 (16), 2638-2651 (2012).

Tags

Ultra-Fast Amplicon-Based Next-Generation Sequencing Non-squamous Non-small Cell Lung Cancer Molecular Alterations Targeted Therapy Detection Of Molecular Abnormalities Advanced Or Metastatic NS-NSCLC Targeted Therapies Overall Survival Mechanisms Of Resistance Novel Therapies Treatment Response Molecular Characterization Short Turnaround Time (TAT) International Guidelines Tissue Biopsies Genomic Analysis Less Invasive Methods And Protocols Pathologists Efficient And Rapid Diagnosis Strategy
Ultra-Fast Amplicon-Based Next-Generation Sequencing in Non-Squamous Non-Small Cell Lung Cancer
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Bontoux, C., Lespinet-Fabre, V.,More

Bontoux, C., Lespinet-Fabre, V., Bordone, O., Tanga, V., Allegra, M., Salah, M., Lalvée, S., Goffinet, S., Benzaquen, J., Marquette, C. H., Ilié, M., Hofman, V., Hofman, P. Ultra-Fast Amplicon-Based Next-Generation Sequencing in Non-Squamous Non-Small Cell Lung Cancer. J. Vis. Exp. (199), e65190, doi:10.3791/65190 (2023).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter