Multiplexed detection of predictive fusions and splicing variants in RNA from lung cancer tissue samples using a hybridization-based platform: narrative review
Review Article

Multiplexed detection of predictive fusions and splicing variants in RNA from lung cancer tissue samples using a hybridization-based platform: narrative review

Ana Giménez-Capitán1, Cristina Aguado1, Clara Mayo-de las-Casas1, Rafael Rosell1,2,3, Miguel Ángel Molina-Vila1

1Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain; 2Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain; 3Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Badalona, Barcelona, Spain

Contributions: (I) Conception and design: A Giménez-Capitán, MÁ Molina-Vila, C Mayo-de las-Casas; (II) Administrative support: A Giménez-Capitán, MÁ Molina-Vila, C Aguado; (III) Provision of study materials or patients: A Giménez-Capitán, MÁ Molina-Vila, C Aguado; (IV) Collection and assembly of data: A Giménez-Capitán, MÁ Molina-Vila, C Mayo-de las-Casas; (V) Data analysis and interpretation: A Giménez-Capitán, MÁ Molina-Vila, C Mayo-de las-Casas; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Ana Giménez-Capitán, BSc; Miguel Ángel Molina-Vila, PhD. Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Sabino Arana 5-19, 08028, Barcelona, Spain. Email: agimenez@panoncology.com; mamolina@panoncology.com.

Background and Objective: Cancer is one of the leading causes of disease-related casualties worldwide. More than 2 million new cases of lung cancer were detected in 2021 and this malignancy was the most common cause of cancer related death with 1.80 million casualties. Personalized medicine has revolutionized the therapeutic landscape of some hematological malignancies and solid tumors, particularly non-small cell lung cancer (NSCLC). Rearrangements of anaplastic lymphoma receptor tyrosine kinase, ROS protooncogene 1, receptor tyrosine kinase, RET proto-oncogene and neurotrophic receptor tyrosine kinase genes and MET proto-oncogene receptor tyrosine kinase, exon 14 splicing are present in 1–9% of NSCLC patients and their correct identification is key to select targeted therapies. NSCLC patients presenting these types of alterations can receive tyrosine kinase inhibitors (TKIs), which have demonstrated improved clinical benefit compared with standard chemotherapy. The nCounter system, a hybridization-based platform from NanoString Technology, has been tested in recent years for the detection of fusions and splicing variants in NSCLC. In this literature review, we summarize the published studies in this area.

Methods: We performed a search narrative of the scientific literature in PubMed database and selected all the articles in English from origin until October 5th, 2020 where nCounter was used for fusion and splicing variant detection.

Key Content and Findings: nCounter has been demonstrated to be a useful tool for fusion and splicing variant testing in NSCLC in the clinical setting. The technique has several advantages such as a fewer processing steps, short turnaround time and less hands-on time compared with gold standard methods [fluorescence in situ hybridization (FISH), immunohistochemistry (IHC)] or next-generation sequencing (NGS). In addition, it can be easily employed in formalin-fixed paraffin-embedded (FFPE) tumor samples and requires low quantities of tissue. Finally, nCounter has shown high sensitivity and specificity compared with gold standard methods for detection of clinically relevant fusions and splicing variants.

Conclusions: nCounter can be employed in the clinical setting for the detection of splicing variants and fusion transcripts in NSCLC.

Keywords: Lung cancer; gene fusions; splicing; hybridization-based platform


Received: 26 January 2022; Accepted: 14 June 2022; Published: 30 June 2022.

doi: 10.21037/jxym-22-6


Introduction

Cancer of the lung is one of the most common malignancies and the first cause of cancer-related deaths, representing almost 25% (1). Around 84% of lung tumors are adenocarcinomas, squamous cell carcinomas and large cell carcinomas, which are grouped as non-small cell lung cancers (NSCLCs). Several types of genetic alterations have been demonstrated to be oncogenic and are referred to as drivers, including point mutations, deletions, insertions and gene fusions. The 45% of driver alterations in NSCLC are somatic mutations in the KRAS proto-oncogene (KRAS), epidermal growth factor receptor (EGFR) and B-Raf proto-oncogene (BRAF) genes, while oncogenic gene fusions and splicing variants are present in 5–10% of patients.

Fusion gene and splicing variant occur when two different genes are juxtaposed or when particular exons of a mRNA are processed in different combinations, respectively. The most common are anaplastic lymphoma receptor tyrosine kinase (ALK), ROS protooncogene 1, receptor tyrosine kinase (ROS1), RET proto-oncogene (RET) and neurotrophic receptor tyrosine kinase (NRTK1/2/3) fusions and the MET proto-oncogene, receptor tyrosine kinase splicing (METΔex14) variant being mutually exclusive with other drivers (2). The development of the first tyrosine kinase inhibitors (TKIs) targeting ALK fusions represented a breakthrough advance in the NSCLC treatment landscape in the last decade. Several pre-clinical and clinical studies have demonstrated the clinical benefit of targeted therapies with TKIs in patients with ALK, ROS1, NTRK1/2/3, RET fusions rearrangements or METΔex14 splicing variant. These benefits include increased objective response rates (ORR), progression-free survival (PFS) and overall survival (OS) compared with chemotherapy and TKIs are currently the standard of care in first line treatment of the NSCLC patients harboring the alterations mentioned above. However, due to the emergence of drug resistance, patients ultimately relapse to TKIs and new generation inhibitors have been developed, targeting some mechanisms of resistance (3-6) (Table 1).

Table 1

Summary of inhibitors approved for fusion-positive NSCLC patients

Target Alteration Frequency Drug Reference
ALK Fusion 5–7% Crizotinib (3)
Ceritinib (7)
Alectinib (8)
Brigatinib (9)
Lorlatinib (10)
ROS1 Fusion 1–2% Crizotinib (3)
Ceritinib (7)
Entrectinib (5,11)
Lorlatinib (10)
RET Fusion 1–2% Selpercatinib (4)
Pralsetinib (4)
NTRK Fusion 1% Larotrectinib (6)
Entrectinib (5,11)
METΔex14 Splicing variant 3–4% Crizotinib (3)
Capmatinib (12)
Tepotinib (12)

NSCLC, non-small cell lung cancer; ALK, anaplastic lymphoma receptor tyrosine kinase; ROS1, ROS protooncogene 1, receptor tyrosine kinase; RET, RET proto-oncogene; NTRK, neurotrophic receptor tyrosine kinase genes; MET, MET proto-oncogene receptor tyrosine kinase.

The first ALK inhibitor (ALKi) approved by the Food and Drug Administration (FDA) for metastatic NSCLC was crizotinib in 2011, which targets ALK, ROS1 and c-MET (3). Two second-generation ALKis, ceritinib and alectinib, obtained FDA approval in 2014 and 2015 for patients progressing to crizotinib or intolerant to it (7). Based on the results of the randomized phase III ALEX trial, alectinib was also approved in November of 2017 for treatment-naïve ALK-positive patients (8). Thereupon, the FDA authorized brigatinib for those patients who had failed prior ALKi treatment (9,13). In this fast-growing therapeutic landscape, highly potent third generation ALKis, such as lorlatinib, have been recently developed to treat acquired resistance, improve the control of the disease, and target central nervous system (CNS) disease (10).

Regarding the rest of oncogenic fusions, ROS1 patients are currently treated with two inhibitors, crizotinib and entrectinib, that bind to ROS1 fusion protein (3,5,11). In the case of RET, the first multi-kinase inhibitors tested were cabozantinib, vandetanib and lenvatinib, with contrasting results. More recently, two selective RET inhibitors, selpercatinib and pralsetinib, demonstrated better clinical efficacy and good tolerability, being approved in 2020 (4,14,15). Finally, the kinase inhibitors larotrectinib and entrectinib were approved by the FDA in 2018 and 2019, respectively, for the treatment of patients with NTRK1-3 fusion-positive solid tumors (6,11,16,17).

In the case of MET exon 14 skipping mutation, several MET TKIs have been developed and are currently being tested in clinical trials (18-25). Two type Ib MET TKIs, tepotinib and capmatinib, have recently been approved by the FDA for the treatment of NSCLC patients harboring METΔex14 (12).

Although there are several publications of fusion detection using the nCounter methodology, the perception is that this platform has not managed to establish itself as a benchmark. In most clinical trials, the use of technologies such as next-generation sequencing (NGS), immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH) is preferred or often required for fusion detection. However, our laboratory has been using nCounter for several years and we have observed that this technology outperforms NGS (26) and should be universally accepted for testing fusions and splicing variants in tumor samples. Consequently, we performed a narrative review of the scientific literature about fusion and splicing variant detection using nCounter to support this point and we present the following article in accordance with Narrative Review reporting checklist (available at https://jxym.amegroups.com/article/view/10.21037/jxym-22-6/rc).


Methods

We performed a search narrative of the scientific literature in the PubMed database using the keywords “nCounter” and “fusion” and “non-small cell lung cancer” or “nCounter” and “splicing variant” and “non-small cell lung cancer”. The articles listed after both searches were individually examined, and those actually describing the use of nCounter for fusion and splicing variant detection were selected (Table 2 and Table S1).

Table 2

The search strategy summary

Items Specification
Date of search 2012/08/24–2020/11/27
Databases and other sources searched PubMed
Search terms used See Table S1
Timeframe 2012–2021
Inclusion and exclusion criteria Inclusion criteria: research articles and reviews about nCounter technology for fusion and splicing detection in NSCLC in FFPE tissue
Exclusion criteria: articles that have no performed the technique in FFPE tissue
Selection process It was conducted independently by Ana Giménez-Capitán and Miguel Ángel Molina-Vila, all authors attended a meeting to discuss the literature selection and obtained the consensus

FFPE, formalin-fixed paraffin-embedded; NSCLC, non-small cell lung cancer.


The nCounter technology

The nCounter is a hybridization-based platform (NanoString Technologies, Seattle, WA, USA) based in a fluorescent barcode that enables direct detection of hundreds (≤800) of different target molecules in a single assay. The technology can be used for gene expression profiling, detection of fusion and alternative splicing transcripts or protein analysis, can be easily incorporated into the diagnostic routine and is cost-effective compared to alternative techniques. Regarding gene expression and detection of altered transcripts, the panels can be commercial or custom-made.

The technology can be adapted for simultaneous analysis of multiple fusion transcripts, using a dual strategy aimed to detect possible imbalances in the 3'/5' expression of the wild type (WT) sequences and specific fusion junction targets (27). The nCounter protocol has 3 basic steps: (I) the RNA is hybridized with the specific probe pairs (reporter probe and capture probe); (II) the tripartite structure coated with streptavidin is bound to the surface of the sample cartridge and reporters are aligned by an electric current and immobilized for data collection; (III) fluorescent barcodes are counted by a digital analyzer, RNAs are identified and counts tabulated (27-29) (Figure 1).

Figure 1 nCounter system workflow (BioRender illustration software).

The technique has several advantages compared with gold standard methods such as FISH and IHC or other techniques such as NGS, such as a short turnaround time and needs less hands-on time (Table 3). In addition, it requires low amounts of RNA, which can be easily purified from a single tissue or cytology slide with a minimum area of 1.1 mm2 (27). This aspect is particularly relevant in the case of NSCLC patients, since biopsies are often scarce or the only sample available is a cytological specimen. Often the mRNA from formalin-fixed paraffin-embedded (FFPE) is degraded and with this system the sample can be direct measure without amplification step avoiding any bias. All of these considerations made an attractive platform for the clinical setting implementation (30). The main disadvantage of nCounter is that many laboratories only dispose of NGS and do not have the technology and the required equipment available. At the technical level, an advantage of NGS over nCounter is that NGS can determine the specific sequence of the fusion point and detect any deviation from the standard sequence, while nCounter cannot.

Table 3

Comparison of NanoString nCounter with Illumina MiSeq RNA-Seq, ThermoFisher Ion AmpliSeq RNA Fusion, Agenda Bioscience MassArray, IHC and FISH assay properties

Characteristics NanoString nCounter Illumina MiSeq RNA-Seq ThermoFisher Ion AmpliSeq RNA Fusion Agena Bioscience MassArray IHC FISH
Panel Elements Custom Panel or Vantage 3DTM Lung Fusion Panel TruSight RNA fusion panel RNA fusion lung cancer research panel V2 Lung FUSIONTM Panel Not apply Not apply
Processing steps RNA extraction, hybridization, purification and scan, data analysis* RNA extraction, reverse transcribe sample, fragmentation, cDNA library preparation, sequencing, data analysis RNA extraction, reverse transcribe sample, fragmentation, cDNA library preparation, sequencing, data analysis RNA extraction, reverse transcribe sample, PCR amplification, PCR primer extension, SpectroCHIP Array and Clean Resin, data analysis Cut FFPE tissue, automatic hybridization, slide evaluation Cut FFPE tissue, deparaffinization, tissue pretreatment, hybridization, washing, slide evaluation
Input requirements 6–50 ng total RNA* 10 ng total RNA 10 ng total RNA 10–40 ng of cDNA A slide of FFPE tissue A slide of FFPE tissue
Sensitivity <1 copy/cell <1 copy cell <1 copy cell <1 copy cell 50–100 cells 50–100 cells
Specificity Design of Capture and Reporter probes Rely on data analysis Primer design Primer design Rely on antibody to be used Rely on probes to be used
Assay time 24 hours* 2.5 days 2 days 8hours 24 hours 48 hours
Hands-on time 15 min* 11 hours 45 min (using Ion Chef) 1 hour 1 hour 3 hours
Up to sample per assay 12 8 samples per run 16 samples per Ion 318 Chip 96 1 1
Genes in the panel Custom personalized up 800 transcripts Vantage 3DTM Lung Fusion Panel: ALK, RET, ROS1, NTRK1 Targeting 507 genes ALK, RET, ROS1, NTRK1 ALK, RET, ROS1 NA NA
Number of genes or transcripts detected Custom Panel up to 800 genes or Commercial Vantage 3DTM Lung Fusion Panel has 63 probes: 35 for specific fusion detection and 24 for positional gene expression imbalance detection Gene fusion panel targeting 507 cancer-associated fusion genes and 7,690 exons Over 70 transcripts 31 transcripts 1 1
Analysis software Manual or nSolverTM analysis software RNA fusion analysis module Ion ReporterTM Software MassArray analysis software Manual analysis, pathology specialist Manual analysis, pathology specialist

*, advantages. NA, not applicable; RNA-Seq, RNA sequencing; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; FFPE, formalin-fixed paraffin-embedded; ALK, anaplastic lymphoma receptor tyrosine kinase; ROS1, ROS protooncogene 1, receptor tyrosine kinase; RET, RET proto-oncogene; MET, MET proto-oncogene receptor tyrosine kinase.

In this review, we will summarize the studies published using nCounter for the detection of fusion genes in NSCLC, which are summarized in Table 4. The same table also presents the sensitivity and specificity of the nCounter results versus orthogonal techniques such as FISH or IHC.

Table 4

Summary of nCounter sensibility and specificity vs. gold standard techniques

Study (author, year, country) Alteration Type of nCounter panel nCounter sensibility vs. FISH/IHC/PCR/NGS nCounter specificity vs. FISH/IHC/RT-PCR/NGS
Lira et al., 2013, Korea (29) ALK Custom panel, Elements assay FISH: 100% and IHC: 97.8% FISH and IHC: 98.8%
ROS1 FISH: 100% FISH: 100%
RET FISH: 100% FISH: 100%
Reguart et al., 2017, Spain (27) ALK Custom panel, Elements assay FISH: 87.5% and IHC: 98.5% FISH: 84.9% IHC: 97.2 %
RET Not reported not reported
ROS1 FISH: 85.9% and IHC: 87.2% FISH: 96.1% and IHC: 88.3%
Lindquist et al., 2017, Sweden (31) ALK Custom panel, Elements assay FISH: 100% FISH: 100%
RET FISH: 100% FISH: 100%
ROS1 FISH: 100% FISH: 100%
Rogers et al., 2017, Australia (32) ALK Custom panel, Elements assay FISH: 94% FISH: 97%
ROS1 FISH: 100% FISH: 100%
RET Not reported FISH: 100%
Evangelista et al., 2017, Brazil (33) ALK Custom panel, Elements assay FISH and/or IHC: 100% FISH and/or IHC: 100%
Aguado C et al., 2021, Spain (26) METΔex14 Custom panel, Elements assay RT-PCR: 54.2% RT-PCR: 100%
NGS: 100% NGS: 98.4%
Elfving et al., 2021, Sweden (34) NTRK TruSight Tumor 170 RNA assay No concordance with IHC No concordance with IHC

ALK, anaplastic lymphoma receptor tyrosine kinase; ROS1, ROS protooncogene 1, receptor tyrosine kinase; RET, RET proto-oncogene; MET, MET proto-oncogene receptor tyrosine kinase; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; FFPE, formalin-fixed paraffin-embedded; NGS, next-generation sequencing; IHC, immunohistochemistry.


Detection of ALK, ROS1 and RET gene fusions by nCounter

In 2012, Suehara and colleagues were the first group to report the detection of ALK, ROS1 and RET fusion using nCounter technology (35). The study included 75 lung adenocarcinoma RNA samples; 6 extracted from frozen tissue and 69 from FFPE blocks. Each sample was analyzed using 100 to 200 ng of total RNA using 5'/3' imbalance probes targeting two selected regions of 100 base pairs (pb) for each gene under study. Using serial dilutions of RNA from cell lines, they first determined that the positive tumor cell content should be >25% for the fusion to be detectable. In the case of the 75 samples, the nCounter assay correctly identified 24/24 positive cases. Furthermore, they identified aberrant 5' to 3' ratios in ROS1 and RET of novel Golgi associated PDZ and coiled-coil motif containing (GOPC) GOPC-ROS1 and kinesin family member 5B (KIF5B) KIF5B-RET fusions (35).

Next, Lira et al. [2013] developed an nCounter assay able to identify specific ALK fusions, which included 8 pairs of imbalance probes and 7 pairs of probes for ALK known fusion variants. The assay was validated in RNA (500 ng) isolated from 10 µm sections of FFPE blocks from 67 NSCLC samples, 34 positive and 33 negative (29), and was found to be highly concordant with FISH and IHC.

In 2014, the same group modified the technology for simultaneous screening of ALK, ROS1 and RET fusions. The new assay included 24 probe pairs targeting wild-type 3' and 5' regions of ALK, ROS1, and RET and 27 fusion-specific probe pairs. The assay was validated in 295 NSCLC specimens, ALK results were 100% and 97.8% concordant with FISH and IHC, respectively. Regarding ROS1 and RET, they observed 100% concordance with FISH (36).

In 2017, our group validated nCounter for routine detection of fusion transcripts (27). Our codeset included 24 imbalance probe pairs targeting ALK, ROS1 and RET; and 23 fusion-specific probe pairs. Using FFPE blocks derived from cell lines, we determined 25 ng of total RNA with >10% tumor cell content was sufficient for the detection of fusion transcripts. The assay was retrospectively validated in 108 FFPE samples from advanced NSCLC patients of them, 98 were successfully analyzed by nCounter (91%), which identified 55 fusion positive cases (32 ALK, 21 ROS1, and two RET). nCounter results were highly concordant with IHC (98.5%, CI 91.8–99.7) and FISH [87.5%, confidence interval (CI): 79.0–92.9] for ALK. Regarding ROS1, nCounter showed a similar agreement with IHC and FISH (87.2% and 85.9%).

Three additional groups published in 2017 their experiences in detection of ALK, ROS1 and RET fusions by nCounter. Lindquist et al. analyzed a Swedish cohort comprising 169 FFPE lung cancer blocks. The RNA was 100 to 250 ng and 80% of samples yielded valid results. Five ALK, two ROS1 and three RET positive cases were detected, agreement with FISH was 100% (31). Rogers et al. compared three platforms with FISH; nCounter, a Lung Fusion array (Agena Bioscience, San Diego, CA, USA) and a NGS fusion panel (Thermo Fisher Scientific, Waltham, MA, USA) (29,36). Valid results by nCounter were obtained for 48/51 surgically resected NSCLC samples; 17 tested were positive for ALK, two for ROS1 and one for RET. Overall agreement with FISH was 96% for nCounter, compared to 94% for the array and 86% for the NGS panel (32). Finally, Evangelista et al. tested the nCounter ALK-fusion panel developed by Lira et al. in 43 FFPE lung cancer biopsies from a Brazilian cohort (29,36). A total of 100 ng RNA was used for the analysis. The assay detected 13 ALK-positive samples with 100% agreement with FISH and/or IHC (33).


Detection of MET and NTRK alterations by nCounter

Li et al. [2016] pioneered the detection METΔex14 transcripts by nCounter, incorporating to the Lira assay probes for MET exons 13 and 14. When used to analyze an Asian population cohort (n=271), the assay detected 20 gene ALK fusions (7.4%), six ROS1 (2.2%) and RET (2.2%) fusions and seven METex14 skipping (2.5%) (37).

In 2020, our group performed an extensive retrospective validation of nCounter for the detection of MET alterations, not only METΔex14 but also MET overexpression. Of the 474 advanced NSCLC samples analyzed, 422 (89%) yielded valid results by nCounter, which identified 13 patients (3%) with METΔex14 and 15 (3.2%) overexpressing MET. The two subgroups displayed distinct phenotypes and rarely coexisted with other drivers. NGS failed to detect 3/8 (37.5%) METΔex14 samples positive by nCounter (26). Regarding patients with overexpressing MET mRNA, 92% had MET amplification by FISH and/or NGS. However, three FISH-negative patients showed high MET RNA expression by nCounter, one of them received MET TKI treatment deriving clinical benefit.

Next, our group performed a prospective study to demonstrate the feasibility and usefulness of embedding the RNA tissue-based nCounter panel described by Aguado et al. (26) in the clinical routine. In a cohort of 224 advanced NSCLC patients, nCounter testing yielded an informative result in 207 patients (92%). Driver alterations for ALK (n=7, 4%) and METex14 (n=9, 5%) were detected and patients treated with ALK or MET TKIs based on the nCounter results (38).

Novaes et al. (39) published in 2021 a new study in a Brazilian cohort lung of 142 FFPE lung adenocarcinoma samples, incorporating specific probes for NRTK1 fusion detection. Of them, 134 (94.4%) yielded valid results. ALK rearrangements were detected in 6.5% samples (21/325), while the frequency observed for RET and ROS1 rearrangements was 0.6% (2/325) and 0.3% (1/325), respectively. NTRK1 fusion results were not reported (39).

A more extensive study for NTRK rearrangements was published in 2021 by Elfving et al. comparing detection by IHC assay with nCounter and NGS (TruSight Tumor 170 RNA assay, Illumina, San Diego, CA, USA). A total of 688 NSCLC samples were first stained with the pan-TRK antibody clone EPR17341. Positive cases were further analyzed by the other techniques. However, nCounter or NGS could not confirm an NTRK fusion in any of the IHC positive cases (34).

In summary, all the studies conclude that nCounter platform is particularly useful for fusions and splicing variants detection. However, some of the published articles offer limited evidence at this respect and only a few of them report an extensive validation of the technique, not only using FFPE blocks obtained from cell lines but also comparing the nCounter results with gold standard techniques (NGS, FISH, IHC) in FFPE tumor samples [i.e., (27,29,36); see Table 4]. Also, the minimum amount of tissue sample, the limit of detection, the sensitivity and the specificity of nCounter for fusion and splicing variant detection are all described, being these data particularly useful for the reproducible implementation of the technique in the clinical setting.


Summary

The nCounter technique has demonstrated high sensitivity and specificity for detection of clinically relevant fusions and splicing variants compared with gold standard (FISH, IHC) and can be easily implemented in the clinical setting for multiplex detection of these alterations. nCounter can be used in FFPE tumor samples, requires low quantities of RNA, has a short turnaround time and needs less hands-on time than other techniques.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editors (Umberto Malapelle and Giancarlo Troncone) for the series “Predictive Molecular Pathology in Lung Cancer” published in Journal of Xiangya Medicine. The article has undergone external peer review.

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://jxym.amegroups.com/article/view/10.21037/jxym-22-6/rc

Peer Review File: Available at https://jxym.amegroups.com/article/view/10.21037/jxym-22-6/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (Available at https://jxym.amegroups.com/article/view/10.21037/jxym-22-6/coif). The series “Predictive Molecular Pathology in Lung Cancer” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Dela Cruz CS, Tanoue LT, Matthay RA. Lung cancer: epidemiology, etiology, and prevention. Clin Chest Med 2011;32:605-44. [Crossref] [PubMed]
  2. Farago AF, Azzoli CG. Beyond ALK and ROS1: RET, NTRK, EGFR and BRAF gene rearrangements in non-small cell lung cancer. Transl Lung Cancer Res 2017;6:550-9. [Crossref] [PubMed]
  3. Kazandjian D, Blumenthal GM, Chen HY, et al. FDA approval summary: crizotinib for the treatment of metastatic non-small cell lung cancer with anaplastic lymphoma kinase rearrangements. Oncologist 2014;19:e5-11. [Crossref] [PubMed]
  4. Approves Selpercatinib FDA. Pralsetinib May Soon Follow. Cancer Discov 2020;10:OF1. [Crossref] [PubMed]
  5. Drilon A, Siena S, Dziadziuszko R, et al. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020;21:261-70. [Crossref] [PubMed]
  6. Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020;21:531-40. [Crossref] [PubMed]
  7. Wu J, Savooji J, Liu D. Second- and third-generation ALK inhibitors for non-small cell lung cancer. J Hematol Oncol 2016;9:19. [Crossref] [PubMed]
  8. Peters S, Camidge DR, Shaw AT, et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:829-38. [Crossref] [PubMed]
  9. Camidge DR, Kim HR, Ahn MJ, et al. Brigatinib Versus Crizotinib in Advanced ALK Inhibitor-Naive ALK-Positive Non-Small Cell Lung Cancer: Second Interim Analysis of the Phase III ALTA-1L Trial. J Clin Oncol 2020;38:3592-603. [Crossref] [PubMed]
  10. Shaw AT, Felip E, Bauer TM, et al. Lorlatinib in non-small-cell lung cancer with ALK or ROS1 rearrangement: an international, multicentre, open-label, single-arm first-in-man phase 1 trial. Lancet Oncol 2017;18:1590-9. [Crossref] [PubMed]
  11. Sartore-Bianchi A, Pizzutilo EG, Marrapese G, et al. Entrectinib for the treatment of metastatic NSCLC: safety and efficacy. Expert Rev Anticancer Ther 2020;20:333-41. [Crossref] [PubMed]
  12. Mathieu LN, Larkins E, Akinboro O, et al. FDA Approval Summary: Capmatinib and Tepotinib for the Treatment of Metastatic NSCLC Harboring MET Exon 14 Skipping Mutations or Alterations. Clin Cancer Res 2022;28:249-54. [Crossref] [PubMed]
  13. Carcereny E, Fernández-Nistal A, López A, et al. Head to head evaluation of second generation ALK inhibitors brigatinib and alectinib as first-line treatment for ALK+ NSCLC using an in silico systems biology-based approach. Oncotarget 2021;12:316-32. [Crossref] [PubMed]
  14. Subbiah V, Shen T, Terzyan SS, et al. Structural basis of acquired resistance to selpercatinib and pralsetinib mediated by non-gatekeeper RET mutations. Ann Oncol 2021;32:261-8. [Crossref] [PubMed]
  15. Liu X, Shen T, Mooers BHM, et al. Drug resistance profiles of mutations in the RET kinase domain. Br J Pharmacol 2018;175:3504-15. [Crossref] [PubMed]
  16. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020;21:271-82. [Crossref] [PubMed]
  17. Haratake N, Seto T. NTRK Fusion-positive Non-small-cell Lung Cancer: The Diagnosis and Targeted Therapy. Clin Lung Cancer 2021;22:1-5. [Crossref] [PubMed]
  18. Angevin E, Spitaleri G, Rodon J, et al. A first-in-human phase I study of SAR125844, a selective MET tyrosine kinase inhibitor, in patients with advanced solid tumours with MET amplification. Eur J Cancer 2017;87:131-9. [Crossref] [PubMed]
  19. Bang YJ, Su WC, Schuler M, et al. Phase 1 study of capmatinib in MET-positive solid tumor patients: Dose escalation and expansion of selected cohorts. Cancer Sci 2020;111:536-47. [Crossref] [PubMed]
  20. Camidge DR, Otterson GA, Clark JW et al. Crizotinib in patients (pts) with MET-amplified non-small cell lung cancer (NSCLC): Updated safety and efficacy findings from a phase 1 trial. J Clin Oncol 2018;36:abstr 9062.
  21. Drilon A, Cappuzzo F, Ou SI, et al. Targeting MET in Lung Cancer: Will Expectations Finally Be MET? J Thorac Oncol 2017;12:15-26. [Crossref] [PubMed]
  22. Guo R, Luo J, Chang J, et al. MET-dependent solid tumours - molecular diagnosis and targeted therapy. Nat Rev Clin Oncol 2020;17:569-87. [Crossref] [PubMed]
  23. Paik PK, Felip E, Veillon R, et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N Engl J Med 2020;383:931-43. [Crossref] [PubMed]
  24. Wolf J, Seto T, Han JY, et al. Capmatinib in MET Exon 14-Mutated or MET-Amplified Non-Small-Cell Lung Cancer. N Engl J Med 2020;383:944-57. [Crossref] [PubMed]
  25. Wu YL, Cheng Y, Zhou J, et al. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): an open-label, phase 1b/2, multicentre, randomised trial. Lancet Respir Med 2020;8:1132-43. [Crossref] [PubMed]
  26. Aguado C, Teixido C, Román R, et al. Multiplex RNA-based detection of clinically relevant MET alterations in advanced non-small cell lung cancer. Mol Oncol 2021;15:350-63. [Crossref] [PubMed]
  27. Reguart N, Teixidó C, Giménez-Capitán A, et al. Identification of ALK, ROS1, and RET Fusions by a Multiplexed mRNA-Based Assay in Formalin-Fixed, Paraffin-Embedded Samples from Advanced Non-Small-Cell Lung Cancer Patients. Clin Chem 2017;63:751-60. [Crossref] [PubMed]
  28. Goytain A, Ng T. NanoString nCounter Technology: High-Throughput RNA Validation. Methods Mol Biol 2020;2079:125-39. [Crossref] [PubMed]
  29. Lira ME, Kim TM, Huang D, et al. Multiplexed gene expression and fusion transcript analysis to detect ALK fusions in lung cancer. J Mol Diagn 2013;15:51-61. [Crossref] [PubMed]
  30. Narrandes S, Xu W. Gene Expression Detection Assay for Cancer Clinical Use. J Cancer 2018;9:2249-65. [Crossref] [PubMed]
  31. Lindquist KE, Karlsson A, Levéen P, et al. Clinical framework for next generation sequencing based analysis of treatment predictive mutations and multiplexed gene fusion detection in non-small cell lung cancer. Oncotarget 2017;8:34796-810. [Crossref] [PubMed]
  32. Rogers TM, Arnau GM, Ryland GL, et al. Multiplexed transcriptome analysis to detect ALK, ROS1 and RET rearrangements in lung cancer. Sci Rep 2017;7:42259. [Crossref] [PubMed]
  33. Evangelista AF, Zanon MF, Carloni AC, et al. Detection of ALK fusion transcripts in FFPE lung cancer samples by NanoString technology. BMC Pulm Med 2017;17:86. [Crossref] [PubMed]
  34. Elfving H, Broström E, Moens LNJ, et al. Evaluation of NTRK immunohistochemistry as a screening method for NTRK gene fusion detection in non-small cell lung cancer. Lung Cancer 2021;151:53-9. [Crossref] [PubMed]
  35. Suehara Y, Arcila M, Wang L, et al. Identification of KIF5B-RET and GOPC-ROS1 fusions in lung adenocarcinomas through a comprehensive mRNA-based screen for tyrosine kinase fusions. Clin Cancer Res 2012;18:6599-608. [Crossref] [PubMed]
  36. Lira ME, Choi YL, Lim SM, et al. A single-tube multiplexed assay for detecting ALK, ROS1, and RET fusions in lung cancer. J Mol Diagn 2014;16:229-43. [Crossref] [PubMed]
  37. Li S, Choi YL, Gong Z, et al. Comprehensive Characterization of Oncogenic Drivers in Asian Lung Adenocarcinoma. J Thorac Oncol 2016;11:2129-40. [Crossref] [PubMed]
  38. Marin E, Reyes R, Arcocha A, et al. Prospective Evaluation of Single Nucleotide Variants by Two Different Technologies in Paraffin Samples of Advanced Non-Small Cell Lung Cancer Patients. Diagnostics (Basel) 2020;10:902. [Crossref] [PubMed]
  39. Novaes LAC, Sussuchi da Silva L, De Marchi P, et al. Simultaneous analysis of ALK, RET, and ROS1 gene fusions by NanoString in Brazilian lung adenocarcinoma patients. Transl Lung Cancer Res 2021;10:292-303. [Crossref] [PubMed]
doi: 10.21037/jxym-22-6
Cite this article as: Giménez-Capitán A, Aguado C, Mayo-de las-Casas C, Rosell R, Molina-Vila MÁ. Multiplexed detection of predictive fusions and splicing variants in RNA from lung cancer tissue samples using a hybridization-based platform: narrative review. J Xiangya Med 2022;7:17.

Download Citation