Next Article in Journal
Engineering Multigenerational Host-Modulated Microbiota against Soilborne Pathogens in Response to Global Climate Change
Previous Article in Journal
Contractile and Structural Properties of Detrusor from Children with Neurogenic Lower Urinary Tract Dysfunction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Next Generation Sequencing Technology in Lung Cancer Diagnosis

by
Calin Cainap
1,2,
Ovidiu Balacescu
3,
Simona Sorana Cainap
4,5,* and
Laura-Ancuta Pop
6
1
Department of Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
2
Ion Chiricuta Institute of Oncology, 400015 Cluj-Napoca, Romania
3
Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute Ion Chiricuta, 400015 Cluj-Napoca, Romania
4
Pediatric Clinic no 2, Department of Pediatric Cardiology, Emergency County Hospital for Children, 400177 Cluj-Napoca, Romania
5
Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400013 Cluj-Napoca, Romania
6
Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
Biology 2021, 10(9), 864; https://doi.org/10.3390/biology10090864
Submission received: 27 July 2021 / Revised: 26 August 2021 / Accepted: 31 August 2021 / Published: 3 September 2021
(This article belongs to the Section Cancer Biology)

Abstract

:

Simple Summary

Lung cancer is still one of the most commonly diagnosed and deadliest cancers in the world. Its diagnosis at an early stage is highly necessary and will improve the standard of care of this disease. The aim of this article is to review the importance and applications of next generation sequencing in lung cancer diagnosis. As observed in many studies, next generation sequencing has been proven as a very helpful tool in the early detection of different types of cancers, including lung cancer, and has been used in the clinic, mainly due to its many advantages, such as low cost, speed, efficacy, low quantity usage of biological samples, and diversity.

Abstract

Lung cancer is still one of the most commonly diagnosed cancers, and one of the deadliest. The high death rate is mainly due to the late stage of diagnosis and low response rate to therapy. Previous and ongoing research studies have tried to discover new reliable and useful cbiomarkers for the diagnosis and prognosis of lung cancer. Next generation sequencing has become an essential tool in cancer diagnosis, prognosis, and evaluation of the treatment response. This article aims to review the leading research and clinical applications in lung cancer diagnosis using next generation sequencing. In this scope, we identified the most relevant articles that present the successful use of next generation sequencing in identifying biomarkers for early diagnosis correlated to lung cancer diagnosis and treatment. This technique can be used to evaluate a high number of biomarkers in a short period of time and from small biological samples, which makes NGS the preferred technique to develop clinical tests for personalized medicine using liquid biopsy, the new trend in oncology.

1. Lung Cancer

Lung cancer remains one of the most common cancers diagnosed in 2020, and one of the deadliest cancer types. In Europe, the lung cancer incidence rate is 97.6 (men) and 38.3 (women), with a mortality rate of 81.7 (men) and 29 (women), respectively [1]. In Romania, the incidence is lower in women (28.5) and higher in men (105.3) than the European incidence rate. The same trend is also observed in mortality, 24.8 (women) and 95.6 in men [1]. An increased number of deaths due to lung cancer is mainly due to late-stage diagnosis, mostly because this cancer shows no symptoms in its early stages.
There are several risk factors associated with lung cancer, such as smoking, air pollution, radon exposure, occupational exposure to different chemicals, heredity susceptibility, radiation and diet [2]. Considering these risk factors, it has been observed that specific subtypes are correlated to exposure to specific risk factors. These subtypes are small cell lung cancer and non-small cell lung cancer. Small cell lung cancer (SCLC) represents about 15% of lung cancers and is correlated mainly to smoking. Non-small cell lung cancer (NSCLC) has three main subtypes and accounts for 85% of lung cancer diagnoses. These subtypes are adenocarcinoma, squamous cell carcinoma and large cell carcinoma [3].
Lung cancer is diagnosed at the late stage mainly because in the early stage it presents no symptoms, and patients approach a doctor only when they experience chest pain, persistent cough, and weight loss. Due to this fact, it is very important to identify reliable methods for screening patients with a high risk for lung cancer. In 2018, the National Comprehensive Cancer Network (NCCN) defined low-dose computer tomography (LDCT) as an early screening method for high-risk lung cancer patients [4]. In a German trial, the LDCT screening helped reduce mortality in women with lung cancer [5]. Two main disadvantages of this technique are that it is recommended for a specific range of patients, mainly people that are smoking or are between 50 and 80 years old, and that there are difficulties in evaluating the correct size and number of lung nodules on CT scans, and sometimes these nodules are benign. To overcome these limitations, new methods for lung screening and diagnosis have been developed [6,7]. It is possible to improve lung cancer screening and diagnosis by using LDCT in combination with different biomarkers, either from serum or blood [8]. On the other hand, due to a lower treatment success rate for late-stage lung cancer, its mortality is relatively high. Moreover, lung cancer patients who smoke present a higher number of somatic mutations, which can give rise to a higher number of cancer-driven mutations [9]. Computer tomography is still the main method used for lung cancer screening and diagnosis [10], and the application of genetic testing is mainly used for treatment selection and guidance. Studies have shown that genetic testing using NGS has helped identify at least one actionable target that could be used for targeted therapy [11,12,13]. Additionally, it was observed that patients treated with targeted therapies show better survival and response rates [14]. For lung cancer patients, mainly NSCLC, treatment is still based on chemotherapy for initial stages, but in local, advanced or metastatic disease, biomarker testing for different genes (EGFR, ALK, KRAS, ROS1, BRAF, NTRK1/2/3, MET, RET and PD-L1) helps patients benefit from specifically targeted therapies (anti-EGFR, anti-ALK or anti-ROS) and immune checkpoint inhibitor therapy [15].
Therefore, by using and corroborating the data provided by next generation sequencing (NGS) assessment, the early diagnosis and guidance of treatment for lung cancer have become more precise.

2. Next Generation Sequencing

Next generation sequencing (NGS) is a comprehensive technology used for sequence (DNA) and gene expression (RNA species) analysis [16,17,18]. The NGS technique was developed to overcome the Sanger sequencing limitation, but it evolved into being used in all areas of genomic research, starting with DNA, RNA, miRNA, ChIP and methylation sequencing [19,20,21]. As with any technique, NGS has multiple advantages that have made it an essential tool in all areas of research and in the clinic [22]. However, even after over 15 years of development, this technique has some disadvantages, such as the need for powerful bioinformatics tools and specialized personnel for both experimental and data analysis [20]. Some advantages and disadvantages of NGS are presented in Table 1.
The data provided by NGS have proven valuable and reliable for both research and in the clinic to improve the diagnosis, prognosis, and treatment of several diseases [23,24,25,26], and are widely used in the oncology field [27,28]. In lung cancer, this technique has been used for early diagnosis biomarker identification, targeted treatment decisions, and identification of causative mutations [29,30,31,32,33,34].

3. NGS in Lung Cancer Diagnosis

Lung cancer diagnosis is challenging in the early stages because patients do not present any symptoms, or symptoms are shared with other pulmonary diseases. In addition, classic techniques for lung cancer diagnosis have many false-negative results due to different reasons, such as quality and quantity of the samples or sensitivity of the test [35]. Here, NGS can be beneficial due to its high sensitivity and specificity, using low amounts of sample. Additionally, NGS can determine an increased number of alterations simultaneously from the same quantity of sample. Therefore, NGS has been applied with success in the identification of lung cancer-specific mutations in paraffin-embedded tissue samples, with a higher rate than standard PCR testing [36,37]. Recently, studies have shown that NGS can effectively be used to identify specific lung cancer mutations in circulating tumor DNA, in a liquid biopsy sample [38,39,40,41]. The main applications of NGS in the clinic are related to genomics, transcriptomics and epigenomics. When using whole genome, whole exome or targeted DNA analysis, specific information on point mutations, copy number alterations, small indels or structural variance alterations can be identified. RNA seq analysis can provide information related to gene fusions, alternative splicing, differential expression or RNA editing, while Bisuphite seq or ChIP seq are used for the identification of the methylation profile, histone modification or transcription factor binding alterations.
These important advantages demonstrated by NGS in the evaluation of the alterations related to lung cancer diagnosis have created a new opportunity for the development of commercial kits and assays specific to lung cancer. One such kit is the NextDaySeq-Lung panel, developed by Beijing ACCB Biotech (Beijing, China), with primers for the amplification of EGFR exon 18, 19, 20, 21, KRAS exon 2, 3, PIK3CA exon 9, 20, and BRAF exon 11, 15. The mutations in the KRAS gene can predict the efficiency of EGFR-tyrosine kinase inhibitors [42]. Recently, it was observed that most patients that developed resistance to TKIs have different EGFR mutations [43,44]. Mutations in BRAF can be correlated to response to BRAF/MEK inhibitors in NSCLC patients [45,46,47], while PIK3CA mutations could render SCLC patients sensible to triciribine treatment [48]. In addition, there is a study that uses alpelisib, a PIK3CA inhibitor, for breast cancer PIK3CA mutated patients, who have shown better survival than that of other treatment [49], which could be implemented in lung cancer as well. The NextDaySeq-Lung panel has been used in several studies and has demonstrated better results than Sanger sequencing or qRT-PCR [50,51]. Other gene panels specific for lung cancer focus on fusion alterations, based on RNA sequencing, and evaluate translocations, chromosomal inversions or interstitial deletions. One such panel is the Ion Ampliseq RNA fusion lung cancer panel offered by ThermoFisher Scientific, Waltham, USA, which targets 70 known fusion transcripts of ALK, RET, ROS1, and NTRK. This panel has shown high sensitivity and good concordance with the typical methods used for fusion testing [52]. For fusion testing, RNA seq has proven to be more sensitive and is used in parallel with DNA seq for mutation evaluation [53,54]. In addition, NGS has successfully been used to identify lung cancer patients that had MET exon14 skipping alterations [55]. Some other NGS lung cancer panels are presented in Table 2.
It is well known that cancer is considered a genetic disorder in which somatic mutations accumulate and give cancer cells the ability to over proliferate and avoid apoptosis [56,57]. Lung cancer is one of the cancers that exhibit a high degree of mutation burden and a high number of driven mutations [9]. Consequently, NGS is extremely useful, due to its many advantages, and the development of different NGS panels is implemented in the clinical setting. In lung cancer diagnosis, NGS is employed mainly in evaluating the gene alteration in key genes involved in the development of lung cancer. These genes are EGFR, BRAF, KRAS, HER2, ROS, ALK, PIK3CA, NTRK, RET and MET [58]. One example is using CGH NGS-based assay for assessing 51 FFPE samples of adenocarcinoma to evaluate its efficiency compared to standard mutation testing. The authors observed that 58% of wild-type patients presented alterations in one of these genes when using the NGS approach, making them suitable for targeted therapy [35]. In other studies, NGS was implemented for NSCLC diagnosis due to the small quantity of tissue samples, which is not suitable for traditional testing methods. Hagemman et al. successfully sequenced 209 samples of NSCLC using a 28 gene NGS panel and identified actionable mutations in 46% of the tested samples [36]. In the same line, Moskalev et al. used the 454 NGS system to evaluate EGFR and KRAS mutation in NSCLC samples with a low number of tumor cells. They were able to identify mutations with an allele frequency of 0.2–1.5%. When reevaluating 16 cases with low tumor cells that were wild type by Sanger, seven of them presented mutations in the EGFR gene at a frequency of 0.9–10% [37]. Another study compared an NGS panel, Sanger sequencing, and qRT-PCR in evaluating mutation in 138 NSCLC FFPE samples. The authors observed that NGS and qRT-PCR have a higher sensitivity than Sanger sequencing. NGS is better than qRT-PCR because it also provides information about the mutation sequence and allele frequency, and identifies mutations that are not in the hotspot area [50]. Liang et al. used a DNA methylation profile to develop a blood-based test for the early diagnosis of lung cancer. Their method presented a sensitivity of 75% for stage 1A and 85.7 for stage 1B lung cancer [59]. NGS has proven to be more sensitive and specific than FISH or IHC when analyzing fusion alterations in lung cancer, which are the main methods used for fusion detection. Lin et al. observed a positive rate of 92.7% for ALK rearrangement when using NGS, 82.4% for FISH and 94.5% for IHC, and a concordance of 87.3% of NGS results with IHC results. They also concluded that IHC fusion testing is better for screening, while NGS fusion testing is more accurate for predicting the clinical benefits of crizotinib treatment [60]. Another benefit of NGS is the fact that it also provides information on the exact fusion alteration, which is very important in evaluating the treatment and outcome of patients [61]. To overcome the problem of harvesting tissue samples from early-stage lung cancer sample patients, new challenges related to identifying novel non-invasive biomarkers are under investigation. One such example is the use of miRNA for the diagnosis of lung cancer. miRNA sequencing was used to identify specific miRNAs for adenocarcinoma and SCLC. Jin et al. were able to identify miR-181-5p, miR-30a-3p, miR-30e-3p and miR-361-5p as being specific for adenocarcinoma, and miR-10b-5p, miR-15b-5p and miR-320b for SCLC (small cell lung cancer) [62]. In addition, taking advantage of the many benefits of NGS, oncology researchers have developed liquid biopsy testing for lung cancer diagnosis [63]. Leighl et al. observed a very high concordance for NGS results from cfDNA and tissue DNA in untreated metastatic NSCLC [64]. The same was observed by Mack et al. when analyzing 8388 cases of NSCLC [65]. NGS testing was successfully recommended in lung cancer diagnosis by different expert panels [66] and oncology organizations [67]. Gray et al. performed a thorough survey of the relevant literature regarding liquid biopsy and observed that the advantages of NGS have helped to develop different assays using liquid biopsy samples for the early diagnosis, treatment selection, minimal disease detection, monitoring treatment efficacy and evaluation of tumor burden in lung cancer [63]. Sueoka-Aragane et al. observed that the analysis of ctDNA by NGS could be a promising tool for the evaluation of the efficacy of osimertinib in NSCLC with EGFR T790M mutation [68]. Table 3 presents studies correlated to the performance of NGS technology in the diagnosis and screening of lung cancer [69,70].
The research area using NGS for lung cancer diagnosis has been extensively developed and has made this technique valuable for different clinical trials on lung cancer. Data from clinicaltrial.gov include 98 trials on lung cancer that use NGS (https://clinicaltrials.gov/ct2/results?cond=Lung+Cancer&term=next+generation+sequencing&cntry&state=&city=&dist=, accessed on 25 July 2021). Some of these trials are already complete; others are recruiting or enrolling. There are clinical trials that evaluate the possibility of NGS to identify mutations in very small samples (NGS NCT02420405), or improve the personalized treatment (NCT02281214). Table 4 presents data on the clinical trials using NGS for lung cancer diagnosis.
In addition, recent studies have shown that by implementing NGS-based testing, clinics can reduce the cost required for evaluating biomarkers specifically for targeted treatments or agnostic therapy implementation. NGS-based testing can reduce total testing cost by EUR 30–1249 depending on how comprehensive the analysis is, when compared to RT-PCR technology [80].

4. Conclusions

NGS has successfully been used both in research and in the clinic, and has become one of the main tools in lung cancer diagnosis, showing better results that standard techniques used for lung cancer diagnosis, and being able to identify lung cancer-specific alteration in a variety of biological samples such as blood, plasma, fresh frozen or FFPE tissue, urine or other bodily fluids, even where the nucleic acid content is limited and where classic methods fail. In addition, the cost of NGS is lower than that of standard testing methods, which makes this technique appealing for the implementation of different agnostic therapies, targeted therapies and immune checkpoint inhibitor therapies. Its success was demonstrated in different clinical studies that were developed to obtain better methods for lung cancer diagnosis. The NGS technique has become the primary tool for investigating different types of samples and different subtypes of lung cancer, being implemented in mutation evaluation and fusion alteration identification, due to its great advantage over FISH and IHC, which are techniques that can have inconsistent results due to the expertise of the pathologist, and cannot be used on other types of samples, except tissue. As with any technique, NGS still has its limitations, mostly related to the amount of data obtained, and the need for a big data storage capacity and a good bioinformatics team.
Nevertheless, the advantages of NGS make it ideal to be used for evaluating a high number of biomarkers in a short period of time, from small biological samples, and at a low price. Therefore, NGS should be the preferred technique to develop clinical tests for personalized medicine using liquid biopsy, the new trend in oncology.

Author Contributions

Conceptualization, C.C. and L.A.P.; data curation, O.B. and S.S.C.; writing—original draft preparation, C.C., O.B., S.S.C. and L.A.P.; writing—review and editing, O.B. and L.P.; visualization, S.S.C. and L.P.; supervision, L.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the FEDR through the Competitiveness Operational Programme 2014-2020, POC-P_40_318, contract no. 10/01.09.2016: Grant partnership for the transfer of knowledge in biogenomics applications in oncology and related fields, BIOGENONCO.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would also like to thank Sanogenetic (Voluntari, Romania) for their help with this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

cfDNA: circulating free DNA; CGH: comparative genomic hybridization; ChIP Seq: chromatin immune-precipitation and DNA sequencing; CT: computer tomography; ddPCR: digital droplet polymerase chain reaction; DNA: deoxiribonucleic acid; FFPE: formalin fixed paraffin embedded; FISH: fluorescence in situ hybridization; GWAS: genome wide association studies; IHC: imunohisto chemistry; IPH: idiopathic pulmonary fibrosis; LC: lung cancer; LDCT: low dose computer tomography; miRNA: micro RNA; NCCN: National Comprehensive Cancer Network; NGS: next generation sequencing; NSCLC: non-small cell lung cancer; PCR: polymerase chain reaction; qRT-PCR: quantitative real time polymerase chain reaction; RNA: ribonucleic acid; SCLC: small cell lung cancer.

References

  1. System, E.-E.C.I. Lung cancer incidance and mortality 2020. 2021. Available online: https://ecis.jrc.ec.europa.eu (accessed on 26 July 2021).
  2. Mao, Y.; Yang, D.; He, J.; Krasna, M.J. Epidemiology of Lung Cancer. Surg. Oncol. Clin. N. Am. 2016, 25, 439–445. [Google Scholar] [CrossRef] [PubMed]
  3. Travis, W.D.; Brambilla, E.; Burke, A.P.; Marx, A.; Nicholson, A.G. Introduction to The 2015 World Health Organization Classification of Tumors of the Lung, Pleura, Thymus, and Heart. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2015, 10, 1240–1242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Wood, D.E.; Kazerooni, E.A.; Baum, S.L.; Eapen, G.A.; Ettinger, D.S.; Hou, L.; Jackman, D.M.; Klippenstein, D.; Kumar, R.; Lackner, R.P.; et al. Lung Cancer Screening, Version 3.2018, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. JNCCN 2018, 16, 412–441. [Google Scholar] [CrossRef]
  5. Becker, N.; Motsch, E.; Trotter, A.; Heussel, C.P.; Dienemann, H.; Schnabel, P.A.; Kauczor, H.U.; Maldonado, S.G.; Miller, A.B.; Kaaks, R.; et al. Lung cancer mortality reduction by LDCT screening-Results from the randomized German LUSI trial. Int. J. Cancer 2020, 146, 1503–1513. [Google Scholar] [CrossRef] [PubMed]
  6. Han, D.; Heuvelmans, M.A.; Oudkerk, M. Volume versus diameter assessment of small pulmonary nodules in CT lung cancer screening. Transl. Lung Cancer Res. 2017, 6, 52–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Cainap, C.; Pop, L.A.; Balacescu, O.; Cainap, S.S. Early diagnosis and screening in lung cancer. Am. J. Cancer Res. 2020, 10, 1993–2009. [Google Scholar]
  8. Chu, G.C.W.; Lazare, K.; Sullivan, F. Serum and blood based biomarkers for lung cancer screening: A systematic review. BMC Cancer 2018, 18, 181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [Green Version]
  10. Oudkerk, M.; Devaraj, A.; Vliegenthart, R.; Henzler, T.; Prosch, H.; Heussel, C.P.; Bastarrika, G.; Sverzellati, N.; Mascalchi, M.; Delorme, S.; et al. European position statement on lung cancer screening. Lancet. Oncol. 2017, 18, e754–e766. [Google Scholar] [CrossRef]
  11. Johnson, D.B.; Dahlman, K.H.; Knol, J.; Gilbert, J.; Puzanov, I.; Means-Powell, J.; Balko, J.M.; Lovly, C.M.; Murphy, B.A.; Goff, L.W.; et al. Enabling a genetically informed approach to cancer medicine: A retrospective evaluation of the impact of comprehensive tumor profiling using a targeted next-generation sequencing panel. Oncologist 2014, 19, 616–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Tan, O.; Shrestha, R.; Cunich, M.; Schofield, D.J. Application of next-generation sequencing to improve cancer management: A review of the clinical effectiveness and cost-effectiveness. Clin. Genet. 2018, 93, 533–544. [Google Scholar] [CrossRef] [Green Version]
  13. Meric-Bernstam, F.; Brusco, L.; Shaw, K.; Horombe, C.; Kopetz, S.; Davies, M.A.; Routbort, M.; Piha-Paul, S.A.; Janku, F.; Ueno, N.; et al. Feasibility of Large-Scale Genomic Testing to Facilitate Enrollment Onto Genomically Matched Clinical Trials. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015, 33, 2753–2762. [Google Scholar] [CrossRef]
  14. Schwaederle, M.; Zhao, M.; Lee, J.J.; Lazar, V.; Leyland-Jones, B.; Schilsky, R.L.; Mendelsohn, J.; Kurzrock, R. Association of Biomarker-Based Treatment Strategies With Response Rates and Progression-Free Survival in Refractory Malignant Neoplasms: A Meta-analysis. JAMA Oncol. 2016, 2, 1452–1459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Consortium, N. NCCN Guidlines-Non Small Cell Lung Cancer; NCCN: Plymouth Meeting, PA, USA, 2021. [Google Scholar]
  16. Cirulli, E.T.; Goldstein, D.B. Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nat. Rev. Genet. 2010, 11, 415–425. [Google Scholar] [CrossRef]
  17. Sultan, M.; Schulz, M.H.; Richard, H.; Magen, A.; Klingenhoff, A.; Scherf, M.; Seifert, M.; Borodina, T.; Soldatov, A.; Parkhomchuk, D.; et al. A global view of gene activity and alternative splicing by deep sequencing of the human transcriptome. Science 2008, 321, 956–960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Ju, Y.S.; Kim, J.I.; Kim, S.; Hong, D.; Park, H.; Shin, J.Y.; Lee, S.; Lee, W.C.; Kim, S.; Yu, S.B.; et al. Extensive genomic and transcriptional diversity identified through massively parallel DNA and RNA sequencing of eighteen Korean individuals. Nat. Genet. 2011, 43, 745–752. [Google Scholar] [CrossRef] [PubMed]
  19. Behjati, S.; Tarpey, P.S. What is next generation sequencing? Arch. Dis. Child. Educ. Pract. Ed. 2013, 98, 236–238. [Google Scholar] [CrossRef]
  20. Levy, S.E.; Myers, R.M. Advancements in Next-Generation Sequencing. Annu. Rev. Genom. Hum. Genet. 2016, 17, 95–115. [Google Scholar] [CrossRef] [Green Version]
  21. Slatko, B.E.; Gardner, A.F.; Ausubel, F.M. Overview of Next-Generation Sequencing Technologies. Curr. Protoc. Mol. Biol. 2018, 122, e59. [Google Scholar] [CrossRef] [PubMed]
  22. Pereira, M.A.; Malta, F.S.V.; Freire, M.C.M.; Couto, P.G.P. Application of Next-Generation Sequencing in the Era of Precision Medicine. In Applications of RNA-Seq and Omics Strategies—From Microorganisms to Human Health; Marchi, F., Cirillo, P., Mateo, E.C., Eds.; IntechOpen: London, UK, 2017. [Google Scholar] [CrossRef] [Green Version]
  23. Spaulding, T.P.; Stockton, S.S.; Savona, M.R. The evolving role of next generation sequencing in myelodysplastic syndromes. Br. J. Haematol. 2020, 188, 224–239. [Google Scholar] [CrossRef]
  24. Besser, J.; Carleton, H.A.; Gerner-Smidt, P.; Lindsey, R.L.; Trees, E. Next-generation sequencing technologies and their application to the study and control of bacterial infections. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2018, 24, 335–341. [Google Scholar] [CrossRef] [Green Version]
  25. Pipis, M.; Rossor, A.M.; Laura, M.; Reilly, M.M. Next-generation sequencing in Charcot-Marie-Tooth disease: Opportunities and challenges. Nat. Rev. Neurol. 2019, 15, 644–656. [Google Scholar] [CrossRef]
  26. Mellis, R.; Chandler, N.; Chitty, L.S. Next-generation sequencing and the impact on prenatal diagnosis. Expert Rev. Mol. Diagn. 2018, 18, 689–699. [Google Scholar] [CrossRef] [PubMed]
  27. Cha, Y.J.; Koo, J.S. Next-generation sequencing in thyroid cancer. J. Transl. Med. 2016, 14, 322. [Google Scholar] [CrossRef] [Green Version]
  28. Sabour, L.; Sabour, M.; Ghorbian, S. Clinical Applications of Next-Generation Sequencing in Cancer Diagnosis. Pathol. Oncol. Res. POR 2017, 23, 225–234. [Google Scholar] [CrossRef]
  29. Karnes, H.E.; Duncavage, E.J.; Bernadt, C.T. Targeted next-generation sequencing using fine-needle aspirates from adenocarcinomas of the lung. Cancer Cytopathol. 2014, 122, 104–113. [Google Scholar] [CrossRef]
  30. Zhang, Y.; Li, J.; Hua, P.; Liu, N.; Li, Q.; Zhu, X.; Jiang, L.; Zheng, K.; Su, X. Targeted next-generation sequencing in cytology specimens for molecular profiling of lung adenocarcinoma. Int. J. Clin. Exp. Pathol. 2018, 11, 3647–3655. [Google Scholar] [PubMed]
  31. Chen, Y.; Shi, J.X.; Pan, X.F.; Feng, J.; Zhao, H. Identification of candidate genes for lung cancer somatic mutation test kits. Genet. Mol. Biol. 2013, 36, 455–464. [Google Scholar] [CrossRef] [PubMed]
  32. Wu, K.; Huang, R.S.; House, L.; Cho, W.C. Next-generation sequencing for lung cancer. Future Oncol. 2013, 9, 1323–1336. [Google Scholar] [CrossRef]
  33. Tuononen, K.; Mäki-Nevala, S.; Sarhadi, V.K.; Wirtanen, A.; Rönty, M.; Salmenkivi, K.; Andrews, J.M.; Telaranta-Keerie, A.I.; Hannula, S.; Lagström, S.; et al. Comparison of targeted next-generation sequencing (NGS) and real-time PCR in the detection of EGFR, KRAS, and BRAF mutations on formalin-fixed, paraffin-embedded tumor material of non-small cell lung carcinoma-superiority of NGS. Genes Chromosomes Cancer 2013, 52, 503–511. [Google Scholar] [CrossRef]
  34. Buttitta, F.; Felicioni, L.; Del Grammastro, M.; Filice, G.; Di Lorito, A.; Malatesta, S.; Viola, P.; Centi, I.; D’Antuono, T.; Zappacosta, R.; et al. Effective assessment of egfr mutation status in bronchoalveolar lavage and pleural fluids by next-generation sequencing. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 691–698. [Google Scholar] [CrossRef] [Green Version]
  35. Lim, S.M.; Kim, E.Y.; Kim, H.R.; Ali, S.M.; Greenbowe, J.R.; Shim, H.S.; Chang, H.; Lim, S.; Paik, S.; Cho, B.C. Genomic profiling of lung adenocarcinoma patients reveals therapeutic targets and confers clinical benefit when standard molecular testing is negative. Oncotarget 2016, 7, 24172–24178. [Google Scholar] [CrossRef] [Green Version]
  36. Hagemann, I.S.; Devarakonda, S.; Lockwood, C.M.; Spencer, D.H.; Guebert, K.; Bredemeyer, A.J.; Al-Kateb, H.; Nguyen, T.T.; Duncavage, E.J.; Cottrell, C.E.; et al. Clinical next-generation sequencing in patients with non-small cell lung cancer. Cancer 2015, 121, 631–639. [Google Scholar] [CrossRef]
  37. Moskalev, E.A.; Stöhr, R.; Rieker, R.; Hebele, S.; Fuchs, F.; Sirbu, H.; Mastitsky, S.E.; Boltze, C.; König, H.; Agaimy, A.; et al. Increased detection rates of EGFR and KRAS mutations in NSCLC specimens with low tumour cell content by 454 deep sequencing. Virchows Arch. Int. J. Pathol. 2013, 462, 409–419. [Google Scholar] [CrossRef] [Green Version]
  38. Taylor, C.; Chacko, S.; Davey, M.; Lacroix, J.; MacPherson, A.; Finn, N.; Wajnberg, G.; Ghosh, A.; Crapoulet, N.; Lewis, S.M.; et al. Peptide-Affinity Precipitation of Extracellular Vesicles and Cell-Free DNA Improves Sequencing Performance for the Detection of Pathogenic Mutations in Lung Cancer Patient Plasma. Int. J. Mol. Sci. 2020, 21, 9083. [Google Scholar] [CrossRef] [PubMed]
  39. Govind, K.B.; Koppaka, D.; Dasappa, L.; Jacob, L.A.; Babu, S.M.; Lokesh, N.K.; Haleshappa, R.A.; Rajeev, L.K.; Saldanha, S.C.; Abhishek, A.; et al. Detection of clinically relevant epidermal growth factor receptor pathway mutations in circulating cell-free tumor DNA using next generation sequencing in squamous cell carcinoma lung. South Asian J. Cancer 2019, 8, 247–249. [Google Scholar] [CrossRef]
  40. Yamamoto, G.; Kikuchi, M.; Kobayashi, S.; Arai, Y.; Fujiyoshi, K.; Wakatsuki, T.; Kakuta, M.; Yamane, Y.; Iijima, Y.; Mizutani, H.; et al. Routine genetic testing of lung cancer specimens derived from surgery, bronchoscopy and fluid aspiration by next generation sequencing. Int. J. Oncol. 2017, 50, 1579–1589. [Google Scholar] [CrossRef] [PubMed]
  41. Li, H.; Xie, Y.; Lin, Y.; Yu, T.; Yin, Z. Different Gene Mutation Spectrum of the Paired CSF and Plasma Samples in Lung Adenocarcinoma with Leptomeningeal Metastases: The Liquid Biopsy Based on Circulating Tumor DNA. Zhongguo Fei Ai Za Zhi = Chin. J. Lung Cancer 2020, 23, 646–654. [Google Scholar] [CrossRef]
  42. Eberhard, D.A.; Johnson, B.E.; Amler, L.C.; Goddard, A.D.; Heldens, S.L.; Herbst, R.S.; Ince, W.L.; Jänne, P.A.; Januario, T.; Johnson, D.H.; et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 5900–5909. [Google Scholar] [CrossRef] [PubMed]
  43. Lam, T.C.; Tsang, K.C.; Choi, H.C.; Lee, V.H.; Lam, K.O.; Chiang, C.L.; So, T.H.; Chan, W.W.; Nyaw, S.F.; Lim, F.; et al. Combination atezolizumab, bevacizumab, pemetrexed and carboplatin for metastatic EGFR mutated NSCLC after TKI failure. Lung Cancer 2021, 159, 18–26. [Google Scholar] [CrossRef]
  44. Riudavets, M.; Bosch-Barrera, J.; Cabezón-Gutiérrez, L.; Diz Taín, P.; Hernández, A.; Alonso, M.; Blanco, R.; Gálvez, E.; Insa, A.; Mielgo, X.; et al. Efficacy of nintedanib plus docetaxel in patients with refractory advanced epidermal growth factor receptor mutant lung adenocarcinoma. Clin. Transl. Oncol. Off. Publ. Fed. Span. Oncol. Soc. Natl. Cancer Inst. Mex. 2021. [Google Scholar] [CrossRef]
  45. Pratilas, C.A.; Hanrahan, A.J.; Halilovic, E.; Persaud, Y.; Soh, J.; Chitale, D.; Shigematsu, H.; Yamamoto, H.; Sawai, A.; Janakiraman, M.; et al. Genetic predictors of MEK dependence in non-small cell lung cancer. Cancer Res. 2008, 68, 9375–9383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Facchinetti, F.; Lacroix, L.; Mezquita, L.; Scoazec, J.Y.; Loriot, Y.; Tselikas, L.; Gazzah, A.; Rouleau, E.; Adam, J.; Michiels, S.; et al. Molecular mechanisms of resistance to BRAF and MEK inhibitors in BRAF(V600E) non-small cell lung cancer. Eur. J. Cancer 2020, 132, 211–223. [Google Scholar] [CrossRef]
  47. Sánchez-Torres, J.M.; Viteri, S.; Molina, M.A.; Rosell, R. BRAF mutant non-small cell lung cancer and treatment with BRAF inhibitors. Transl. Lung Cancer Res. 2013, 2, 244–250. [Google Scholar] [CrossRef]
  48. Shibata, T.; Kokubu, A.; Tsuta, K.; Hirohashi, S. Oncogenic mutation of PIK3CA in small cell lung carcinoma: A potential therapeutic target pathway for chemotherapy-resistant lung cancer. Cancer Lett. 2009, 283, 203–211. [Google Scholar] [CrossRef] [PubMed]
  49. André, F.; Ciruelos, E.M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I.A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y.S.; et al. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: Final overall survival results from SOLAR-1. Ann. Oncol. Off. J. Eur. Soc. Med Oncol. 2021, 32, 208–217. [Google Scholar] [CrossRef] [PubMed]
  50. Gao, J.; Wu, H.; Shi, X.; Huo, Z.; Zhang, J.; Liang, Z. Comparison of Next-Generation Sequencing, Quantitative PCR, and Sanger Sequencing for Mutation Profiling of EGFR, KRAS, PIK3CA and BRAF in Clinical Lung Tumors. Clin. Lab. 2016, 62, 689–696. [Google Scholar] [CrossRef]
  51. Xu, X.; Yang, Y.; Li, H.; Chen, Z.; Jiang, G.; Fei, K. Assessment of the clinical application of detecting EGFR, KRAS, PIK3CA and BRAF mutations in patients with non-small cell lung cancer using next-generation sequencing. Scand. J. Clin. Lab. Investig. 2016, 76, 386–392. [Google Scholar] [CrossRef]
  52. Vaughn, C.P.; Costa, J.L.; Feilotter, H.E.; Petraroli, R.; Bagai, V.; Rachiglio, A.M.; Marino, F.Z.; Tops, B.; Kurth, H.M.; Sakai, K.; et al. Simultaneous detection of lung fusions using a multiplex RT-PCR next generation sequencing-based approach: A multi-institutional research study. BMC Cancer 2018, 18, 828. [Google Scholar] [CrossRef]
  53. Benayed, R.; Offin, M.; Mullaney, K.; Sukhadia, P.; Rios, K.; Desmeules, P.; Ptashkin, R.; Won, H.; Chang, J.; Halpenny, D.; et al. High Yield of RNA Sequencing for Targetable Kinase Fusions in Lung Adenocarcinomas with No Mitogenic Driver Alteration Detected by DNA Sequencing and Low Tumor Mutation Burden. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 4712–4722. [Google Scholar] [CrossRef] [PubMed]
  54. Volckmar, A.L.; Leichsenring, J.; Kirchner, M.; Christopoulos, P.; Neumann, O.; Budczies, J.; Morais de Oliveira, C.M.; Rempel, E.; Buchhalter, I.; Brandt, R.; et al. Combined targeted DNA and RNA sequencing of advanced NSCLC in routine molecular diagnostics: Analysis of the first 3000 Heidelberg cases. Int. J. Cancer 2019, 145, 649–661. [Google Scholar] [CrossRef] [PubMed]
  55. Frampton, G.M.; Ali, S.M.; Rosenzweig, M.; Chmielecki, J.; Lu, X.; Bauer, T.M.; Akimov, M.; Bufill, J.A.; Lee, C.; Jentz, D.; et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015, 5, 850–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Peña-Diaz, J.; Bregenhorn, S.; Ghodgaonkar, M.; Follonier, C.; Artola-Borán, M.; Castor, D.; Lopes, M.; Sartori, A.A.; Jiricny, J. Noncanonical Mismatch Repair as a Source of Genomic Instability in Human Cells. Mol. Cell 2017, 67, 162. [Google Scholar] [CrossRef]
  57. Pfeifer, G.P. Environmental exposures and mutational patterns of cancer genomes. Genome Med. 2010, 2, 54. [Google Scholar] [CrossRef] [Green Version]
  58. Zhao, J.; Xiong, J. Advances on driver oncogenes of non-small cell lung cancer. Zhongguo Fei Ai Za Zhi = Chin. J. Lung Cancer 2015, 18, 42–47. [Google Scholar] [CrossRef]
  59. Liang, W.; Zhao, Y.; Huang, W.; Gao, Y.; Xu, W.; Tao, J.; Yang, M.; Li, L.; Ping, W.; Shen, H.; et al. Non-invasive diagnosis of early-stage lung cancer using high-throughput targeted DNA methylation sequencing of circulating tumor DNA (ctDNA). Theranostics 2019, 9, 2056–2070. [Google Scholar] [CrossRef] [PubMed]
  60. Lin, C.; Shi, X.; Yang, S.; Zhao, J.; He, Q.; Jin, Y.; Yu, X. Comparison of ALK detection by FISH, IHC and NGS to predict benefit from crizotinib in advanced non-small-cell lung cancer. Lung Cancer 2019, 131, 62–68. [Google Scholar] [CrossRef]
  61. Christopoulos, P.; Endris, V.; Bozorgmehr, F.; Elsayed, M.; Kirchner, M.; Ristau, J.; Buchhalter, I.; Penzel, R.; Herth, F.J.; Heussel, C.P.; et al. EML4-ALK fusion variant V3 is a high-risk feature conferring accelerated metastatic spread, early treatment failure and worse overall survival in ALK(+) non-small cell lung cancer. Int. J. Cancer 2018, 142, 2589–2598. [Google Scholar] [CrossRef] [Green Version]
  62. Jin, X.; Chen, Y.; Chen, H.; Fei, S.; Chen, D.; Cai, X.; Liu, L.; Lin, B.; Su, H.; Zhao, L.; et al. Evaluation of Tumor-Derived Exosomal miRNA as Potential Diagnostic Biomarkers for Early-Stage Non-Small Cell Lung Cancer Using Next-Generation Sequencing. Clin. cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 5311–5319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gray, J.; Thompson, J.C.; Carpenter, E.L.; Elkhouly, E.; Aggarwal, C. Plasma Cell-Free DNA Genotyping: From an Emerging Concept to a Standard-of-Care Tool in Metastatic Non-Small Cell Lung Cancer. Oncologist 2021. [Google Scholar] [CrossRef]
  64. Leighl, N.B.; Page, R.D.; Raymond, V.M.; Daniel, D.B.; Divers, S.G.; Reckamp, K.L.; Villalona-Calero, M.A.; Dix, D.; Odegaard, J.I.; Lanman, R.B.; et al. Clinical Utility of Comprehensive Cell-free DNA Analysis to Identify Genomic Biomarkers in Patients with Newly Diagnosed Metastatic Non-small Cell Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 4691–4700. [Google Scholar] [CrossRef] [Green Version]
  65. Mack, P.C.; Banks, K.C.; Espenschied, C.R.; Burich, R.A.; Zill, O.A.; Lee, C.E.; Riess, J.W.; Mortimer, S.A.; Talasaz, A.; Lanman, R.B.; et al. Spectrum of driver mutations and clinical impact of circulating tumor DNA analysis in non-small cell lung cancer: Analysis of over 8000 cases. Cancer 2020, 126, 3219–3228. [Google Scholar] [CrossRef]
  66. Cheema, P.K.; Gomes, M.; Banerji, S.; Joubert, P.; Leighl, N.B.; Melosky, B.; Sheffield, B.S.; Stockley, T.; Ionescu, D.N. Consensus Recommendations for Optimizing Biomarker Testing to Identify and Treat Advanced EGFR-Mutated Non-Small-Cell Lung Cancer. Curr. Oncol. 2020, 27, 321–329. [Google Scholar] [CrossRef] [PubMed]
  67. Mosele, F.; Remon, J.; Mateo, J.; Westphalen, C.B.; Barlesi, F.; Lolkema, M.P.; Normanno, N.; Scarpa, A.; Robson, M.; Meric-Bernstam, 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. Ann. Oncol. Off. J. Eur. Soc. Med Oncol. 2020, 31, 1491–1505. [Google Scholar] [CrossRef]
  68. Sueoka-Aragane, N.; Nakashima, C.; Yoshida, H.; Matsumoto, N.; Iwanaga, K.; Ebi, N.; Nishiyama, A.; Yatera, K.; Kuyama, S.; Fukuda, M.; et al. The role of comprehensive analysis with circulating tumor DNA in advanced non-small cell lung cancer patients considered for osimertinib treatment. Cancer Med. 2021, 10, 3873–3885. [Google Scholar] [CrossRef]
  69. Wang, B.; Chen, R.; Wang, C.; Guo, J.; Yuan, M.; Chen, H.; Xia, X.; Zhong, D. Identification of novel ALK fusions using DNA/RNA sequencing in immunohistochemistry/RT-PCR discordant NSCLC patients. Hum. Pathol. 2021, 114, 90–98. [Google Scholar] [CrossRef] [PubMed]
  70. Drilon, A.; Wang, L.; Arcila, M.E.; Balasubramanian, S.; Greenbowe, J.R.; Ross, J.S.; Stephens, P.; Lipson, D.; Miller, V.A.; Kris, M.G.; et al. Broad, Hybrid Capture-Based Next-Generation Sequencing Identifies Actionable Genomic Alterations in Lung Adenocarcinomas Otherwise Negative for Such Alterations by Other Genomic Testing Approaches. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 3631–3639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Beadling, C.; Wald, A.I.; Warrick, A.; Neff, T.L.; Zhong, S.; Nikiforov, Y.E.; Corless, C.L.; Nikiforova, M.N. A Multiplexed Amplicon Approach for Detecting Gene Fusions by Next-Generation Sequencing. J. Mol. Diagn. JMD 2016, 18, 165–175. [Google Scholar] [CrossRef] [Green Version]
  72. Dacic, S.; Villaruz, L.C.; Abberbock, S.; Mahaffey, A.; Incharoen, P.; Nikiforova, M.N. ALK FISH patterns and the detection of ALK fusions by next generation sequencing in lung adenocarcinoma. Oncotarget 2016, 7, 82943–82952. [Google Scholar] [CrossRef] [Green Version]
  73. Moskalev, E.A.; Frohnauer, J.; Merkelbach-Bruse, S.; Schildhaus, H.U.; Dimmler, A.; Schubert, T.; Boltze, C.; König, H.; Fuchs, F.; Sirbu, H.; et al. Sensitive and specific detection of EML4-ALK rearrangements in non-small cell lung cancer (NSCLC) specimens by multiplex amplicon RNA massive parallel sequencing. Lung Cancer 2014, 84, 215–221. [Google Scholar] [CrossRef] [PubMed]
  74. Pfarr, N.; Stenzinger, A.; Penzel, R.; Warth, A.; Dienemann, H.; Schirmacher, P.; Weichert, W.; Endris, V. High-throughput diagnostic profiling of clinically actionable gene fusions in lung cancer. Genes Chromosomes Cancer 2016, 55, 30–44. [Google Scholar] [CrossRef]
  75. Clavé, S.; Rodon, N.; Pijuan, L.; Díaz, O.; Lorenzo, M.; Rocha, P.; Taus, Á.; Blanco, R.; Bosch-Barrera, J.; Reguart, N.; et al. Next-generation Sequencing for ALK and ROS1 Rearrangement Detection in Patients With Non-small-cell Lung Cancer: Implications of FISH-positive Patterns. Clin. Lung Cancer 2019, 20, e421–e429. [Google Scholar] [CrossRef]
  76. Pekar-Zlotin, M.; Hirsch, F.R.; Soussan-Gutman, L.; Ilouze, M.; Dvir, A.; Boyle, T.; Wynes, M.; Miller, V.A.; Lipson, D.; Palmer, G.A.; et al. Fluorescence in situ hybridization, immunohistochemistry, and next-generation sequencing for detection of EML4-ALK rearrangement in lung cancer. Oncologist 2015, 20, 316–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Jiang, Z.; Li, C.; Lu, H. Next generation sequencing detection in archival surgically resected lung adenocarcinoma specimens harbouring the anaplastic lymphoma kinase fusion protein. JPMA. J. Pak. Med Assoc. 2021, 71, 531–536. [Google Scholar] [CrossRef]
  78. Reckamp, K.L.; Melnikova, V.O.; Karlovich, C.; Sequist, L.V.; Camidge, D.R.; Wakelee, H.; Perol, M.; Oxnard, G.R.; Kosco, K.; Croucher, P.; et al. A Highly Sensitive and Quantitative Test Platform for Detection of NSCLC EGFR Mutations in Urine and Plasma. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2016, 11, 1690–1700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Liu, C.; Liu, C.; Zou, X.; Shao, L.; Sun, Y.; Guo, Y. Next-generation sequencing facilitates differentiating between multiple primary lung cancer and intrapulmonary metastasis: A case series. Diagn. Pathol. 2021, 16, 21. [Google Scholar] [CrossRef] [PubMed]
  80. Pruneri, G.; De Braud, F.; Sapino, A.; Aglietta, M.; Vecchione, A.; Giusti, R.; Marchiò, C.; Scarpino, S.; Baggi, A.; Bonetti, G.; et al. Next-Generation Sequencing in Clinical Practice: Is It a Cost-Saving Alternative to a Single-Gene Testing Approach? PharmacoEconomics—Open 2021, 5, 285–298. [Google Scholar] [CrossRef]
Table 1. Advantages and disadvantages of NGS technology.
Table 1. Advantages and disadvantages of NGS technology.
AdvantagesDisadvantages
Low priceNeed for specialized software and computers for data analysis
Short time from library preparation to resultsNo standardization or availability of standardized material for clinical application
Variety of applicationsStill expensive in some developing countries
Useful both in research and clinic
High number of commercially available NGS platforms and specialized kits
Table 2. Some commercially available NGS panels for lung cancer testing.
Table 2. Some commercially available NGS panels for lung cancer testing.
NameCompanyType of SequencingGene TargetedTarget Approach for Gene Fusion AnalysisInput Nucleic Acid (ng)Type of Test
AccuFusionParagon Genomics, Hayward, USA RNA fusionALK, CIT, EML4, FGFR1, MBIP, MET, NRG1, NTRK1, NTRK3, PDGFRA, RET, ROS1, TACC3.Amplicon based10Diagnosis and treatment selection
OmniFusionParagon Genomics, Hayward, USARNA fusionALK, CIT, MBIP, MET, NRG1, NTRK1, NTRK3, PDGFRA, RET, ROS1, TACC3Amplicon based25Diagnosis and treatment selection
Ion AmpliSeq™ RNA Fusion Lung Cancer PanelThermoFisher Scientific, Waltham, USARNA fusionALK, RET, ROS1, and NTRKAmplicon based10Diagnosis and treatment selection
QuantideX® NGS RNA Lung Cancer KitAsuragen, Austin, USARNA expression and fusionALK, ROS1, RET, FGFR3NTRK1, NTRK3, NRG1, FGFR1, FGFR2, MBIP, PDGFRA, MET, ABCB1, BRCA1, CD274, CDKN2A, CTLA4, ERCC1, ESR1, IFNGR, ISG15, MSLN, PDCD1, PDCD1LG2, PTEN, RRM1, TDP1, TERT, TLET3, TOP1, TUBB3, TYMSAmplicon based10Treatment selection
TruSight RNA fusion panelIllumina, San Diego, USARNA seq507 fusion-associated genesHybrid capture based10 total RNA20–100 FFPE RNATreatment selection
Archer fusion plex Comprehensive Thyroid and LungArcherDX Inc, Illumina, San Diego, USARNA seqgene fusions, SNV, indels, splicing and gene expression in 36 genesAMP based10 ngDiagnosis
Archer fusion plex Lung kitArcherDX Inc, Illumina, San Diego, USADNA and RNA seqEGFR vIII and MET exon 14 skipping events along with prominent ALK, BRAF, FGFR, NRG1, NTRK, RET, and ROS1 fusions and select point mutations in 14 key gene targets associated with lung cancerAMP based10 ngDiagnosis
Lung Cancer-Targeted Gene Panel, TumorMAYO Clinic, Scottsdale, USADNAEGFR, BRAF, KRAS, HRAS, NRAS, ALK, ERBB2, and METAmplicon basedNADiagnosis and management of lung cancer
Ion AmpliSeq™ Colon and Lung Research Panel v2ThermoFisher Scientific, Waltham, USADNAKRAS, EGFR, BRAF, PIK3CA, AKT1, ERBB2, PTEN, NRAS, STK11, MAP2K1, ALK, DDR2, CTNNB1, MET, TP53, SMAD4, FBXW7, FGFR3, NOTCH1, ERBB4, FGFR1, FGFR2Amplicon based10Diagnosis and treatment selection
AmpliSeq for Illumina Colon and Lung Research PanelIllumina, San Diego, USADNAKRAS, EGFR, BRAF, PIK3CA, AKT1, ERBB2, PTEN, NRAS, STK11, MAP2K1, ALK, DDR2, CTNNB1, MET, TP53, SMAD4, FBXW7, FGFR3, NOTCH1, ERBB4, FGFR1, and FGFR2Amplicon based10Diagnosis and treatment selection
Table 3. Studies describing the implication of NGS in lung cancer diagnosis.
Table 3. Studies describing the implication of NGS in lung cancer diagnosis.
SamplesCorrelation with Other TechniquesNGS MethodType of Lung CancerSpecificity (%)Sensitivity (%)Ref.
31 tissues lung samples negative for mutations by FISH or PCR8/31 presented actionable mutationsBroad, hybrid capture-based NGSAdenocarcinomaNANA[70]
40 FFPE tissue with known fusion (test), 59 FFPE fusion-negative (validation)Good concordance with FISH, PCR or SangerRNA seq gene fusionNA93–10086–100[71]
28 fusion positive FISH sample16 were positive in NGSRNA fusion and DNA seqNSCLCNANA[72]
32 FFPEGood concordance with FISH and qRT-PCRRNA seqNSCLC100100[73]
50 FFPE (35 test positive for different fusion alterations, 15 negative)
109 FFPE (validation)
Good concordance with FISHRNA fusion and DNA seqNSCLC100100[74]
31 FFPE positive for rearrangement by FISH26 were positive in NGS and were confirmed by IHCRNA fusion and DNA seqNSCLCNANA[75]
51 tested with FISH, IHC and NGS8 samples positive by NGS and IHC, only 4 by FISHDNA seqAdenocarcinoma100100[76]
19 FFPE tested with IHC and NGSGood concordance between NGS and IHCDNA seqAdenocarcinomaNANA[77]
63 tissue, urine and plasmaNGS testing of urine and plasma presented more EGFR mutated positive samples that tissue samples tested by RT-PCRDNA seqNSCLC94 for urine
96–100 for plasma
80–93 for urine
87–100 for plasma
[78]
3 cases with multiple resected tumorsNGS revealed different molecular characteristics that the normal pathological diagnosisDNA seqAdenocarcinomaNANA[79]
Table 4. Clinical trials on lung cancer diagnosis using NGS technology.
Table 4. Clinical trials on lung cancer diagnosis using NGS technology.
Trial NoConditionScope of the TrialSample TypeNumber of PatientsResults
NCT03558165Lung adenocarcinoma stage IVDiagnostic test: oncomine comprehensive assayFFPE tissue100NA
NCT02420405Non-squamous NSCLC stage IIIA-IVRoutine gene testing by NGS for diagnosisTissue78NA
NCT02297087Incurable SCLCStandard of care based on target(s) identified via GWAS for diagnosis and treatmentBlood and tissue12NA
NCT02281214Bronchial adenocarcinoma with metastases, epidermoid cancer of the lungsNGS testing for treatment selection and prognosticBlood and tissue165NA
NCT03257735NSCLC with brain metastasisConsistency of gene mutation status between different types of samples using NGSCerebrospinal fluid, blood and tissue50NA
NCT04849481NSCLCLarge-scale NGS analysis for novel treatment strategies and deciphering the mechanisms of drug resistanceTissue500NA
NCT03244904SCLCNGS analysis for biomarkers for SCLCBlood and tissue80NA
NCT02416726Non-squamous NSCLCNGS for gene profile comparison between different types of samplesBlood and tissue35NA
NCT04260295Lung cancer and non-lung cancer patientsNGS for identification of microorganisms in lungsTissue300NA
NCT02705404Multifocal lung cancerNGS for differentiation of primary tumors from metastatic tumorsBlood, cytology and tissue100NA
NCT02705404NSCLCTargeted NGS for mutation profile concordance in different types of samplesBlood, fresh frozen and FFPE tissue45NA
NCT03833934NSCLCNGS testing for evaluation of ALK resistant mutationsPlasma300NA
NCT03220230NSCLCConcordance between NGS and IHC ALK statusTissue and blood4240Accuracy 95.9% for 1450 participants, sensitivity 54.2% for 83 participants, specificity 98.4% for 1367 participants
NCT03658460NSCLCGene testing using NGS with focus on immuno- oncology markersTissue100NA
NCT02273336Lung cancerNGS testing for treatment selectionTissue, blood and cytology40NA
NCT02941003Lung adenocarcinomaNGS for early stage diagnosisTissue540NA
NCT04238130NSCLCNGS assessment of mutation profile in personalized analysis of cancerPlasma200NA
NCT02169349Stage IIIb and IV NSCLCNGS evaluation cfDNA for diagnosis, treatment and disease progressionPlasma100NA
NCT02299622NSCLC, head and neck cancer, esophageal cancerNGS testing for evaluation of mutation profileTissue200NA
NCT02778854NSCLCGenetic detection of driver mutation using ddPCR and NGS for evaluation of the efficacy of liquid biopsy in diagnosis and prognosisTissue, plasma and other biological liquids200NA
NCT03486262Lung carcinoma patients with/withoutidiopathic pulmonary fibrosis (IPH)NGS testing for genetic alterations identification in lung cancer patients with IPH and without IPHTissue100NA
NCT02113852NSCLCNGS study for identification and characterization of genetic and transcriptomic alterationTissue and blood250NA
NCT03771404Operable (stages I-IIIA) NSCLC PatientsNGS evaluation of the genetic landscape of each patient in order to determine heterogeneity in early stage NSCLCBlood and tissue50NA
NCT04698681Stage IV non-squamous NSCLCNGS evaluation for tumor mutations identification in the KEAP1 or NRF2/NFE2L2 genes in order to determine potential eligibility for a biomarker selected clinical trialBlood200NA
NCT04266483NSCLCMolecular typing of lung cancer in ChinaBlood and tissue2500NA
NCT04624373Stage IV lung cancerMolecular analysis to investigate the sensitivity of cytology supernatant DNA for genotypingSupernatant, blood and tissue50NA
NCT02718651NSCLCNew diagnostic test to detect ALK rearrangements using NGSplasma70NA
NCT03576937Non-squamous NSCLCComparison of blood-based mutational profile with tissue mutational profile for diagnosisBlood and tissue207NA
NCT03248089Non-squamous NSCLCInvestigation of the efficacy of cfDNA genotyping for diagnosisBlood and tissue186NA
NCT03317080I-IV lung cancer eligible for surgery.Use of liquid biopsy for lung cancer detectionBlood1500NA
NCT04025515Asian patients with NSCLCComprehensive molecular profiling of “actionable” alterations in lung cancer specimens in order to determine the prevalence of each genetic subtype in the local population.Tissue500NA
NCT03706625Immune-suppressed patients suffering from HIV-related NSCLCIdentify novel biomarkers such as tumor mutational profiling and immunomutanome in immunosuppressed patientsTissue170NA
NCT03651986Patients with benign and malignant pulmonary nodulesDevelopment of a blood-based assay for early differentiation of benign and malignant pulmonary nodulesBlood10,560NA
NCT02906943Several cancer including lung cancerNGS evaluation of different types of cancer for biomarker identificationFFPE tissue10,000NA
NCT03609918NSCLCTo build NSCLC gene mutation profile in China and find related correlation between gene mutation panel and clinical outcomeFresh frozen tissues and FFPE tissues513NA
NCT03029065Lung cancer patients with brain metastasesTo determine whether cfDNA can be used for concomitant diagnosis to improve the treatment efficacy and prognosis of patients with brain (meningeal) metastasisTissue, plasma and cerebrospinal fluids50NA
NCT03971175Lung cancer and relapse NSCLCTo evaluate accuracy of molecular genetic characterisation of NSCLCTissue, cytology and liquid biopsy540NA
NCT04692935Lung adenocarcinoma from asian and Caucasian patientsEvaluation of the mutational profile by raceTissue450NA
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cainap, C.; Balacescu, O.; Cainap, S.S.; Pop, L.-A. Next Generation Sequencing Technology in Lung Cancer Diagnosis. Biology 2021, 10, 864. https://doi.org/10.3390/biology10090864

AMA Style

Cainap C, Balacescu O, Cainap SS, Pop L-A. Next Generation Sequencing Technology in Lung Cancer Diagnosis. Biology. 2021; 10(9):864. https://doi.org/10.3390/biology10090864

Chicago/Turabian Style

Cainap, Calin, Ovidiu Balacescu, Simona Sorana Cainap, and Laura-Ancuta Pop. 2021. "Next Generation Sequencing Technology in Lung Cancer Diagnosis" Biology 10, no. 9: 864. https://doi.org/10.3390/biology10090864

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop