Next Article in Journal
Patient-Specific Mathematical Model of the Clear Cell Renal Cell Carcinoma Microenvironment
Previous Article in Journal
IL-26 in the Lung and Its Role in COPD Inflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Profiling in Non-Squamous Non-Small Cell Lung Carcinoma: Towards a Switch to Next-Generation Sequencing Reflex Testing

1
Centre Antoine-Lacassagne, Department of Radiation Oncology, Côte d’Azur University, 06000 Nice, France
2
Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
3
Laboratory of Clinical and Experimental Pathology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, Biobank BB-0033-00025, 06000 Nice, France
4
CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
5
Department of Pulmonary Medicine and Thoracic Oncology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, 06000 Nice, France
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(10), 1684; https://doi.org/10.3390/jpm12101684
Submission received: 31 August 2022 / Revised: 7 October 2022 / Accepted: 8 October 2022 / Published: 9 October 2022
(This article belongs to the Section Methodology, Drug and Device Discovery)

Abstract

:
Molecular diagnosis of lung cancer is a constantly evolving field thanks to major advances in precision oncology. The wide range of actionable molecular alterations in non-squamous non-small cell lung carcinoma (NS-NSCLC) and the multiplicity of mechanisms of resistance to treatment resulted in the need for repeated testing to establish an accurate molecular diagnosis, as well as to track disease evolution over time. While assessing the increasing complexity of the molecular composition of tumors at baseline, as well as over time, has become increasingly challenging, the emergence and implementation of next-generation sequencing (NGS) testing has extensively facilitated molecular profiling in NS-NSCLC. In this review, we discuss recent developments in the molecular profiling of NS-NSCLC and how NGS addresses current needs, as well as how it can be implemented to address future challenges in the management of NS-NSCLC.

1. Introduction

Lung cancer is the leading cause of cancer-related death, as well as the third most frequent cancer in Europe with an increasing incidence [1]. Non-small cell lung cancer (NSCLC) is the most frequent lung cancer, with two predominant histologies, non-squamous (NS) and squamous cell carcinoma (SCC). Smoking remains the leading risk factor for non-small cell lung cancer, though cases related to low or no smoking burden are increasingly clinically relevant [2,3]. Importantly, while targetable alterations are more commonly found in patients with lower smoking burden, targetable molecular alterations must be assessed independently of smoking status for any patient with NS-NSCLC, as these alterations are found in ~50% of cases in non-smokers and up to 20% of cases occurring in former or current-smoking patients [4].
Personalized medicine revolutionized NS-NSCLC in 2004, with the discovery that specific mutations in the epidermal growth factor receptor (EGFR) sensitized tumors to a targeted treatment with the tyrosine kinase inhibitors (TKI) erlotinib and gefitinib [5,6,7]. Following approvals in a later treatment setting, the first-line administration of EGFR TKIs became standard in 2009 with the approval of gefitinib [8], in patients with NS-NSCLC harboring an EGFR deletion 19 or L858R substitution, followed by the approval of erlotinib [9] and afatinib [5].
Despite durable responses with the administration of EGFR-TKIs, resistance systematically occurs, and the occurrence of the EGFR T790M resistance mutation has been described as a major driver of resistance [10]. This led to the development and approval of third-generation EGFR TKIs like osimertinib, which are currently recommended for first-line use in patients with EGFR mutant NS-NSCLC with exon 19 deletion or L858R mutation [11]. However, resistance remains a major challenge in this setting, with increasingly complex and diverse resistance mechanisms [12]. As an example, C797S mutations, which confer resistance to osimertinib treatment, are currently being targeted with the development of fourth-generation TKIs [13,14].
Independently of EGFR, multiple additional drivers have been defined in NS-NSCLC, and many drugs have been approved in the last few years that directly target those alterations. Anaplastic lymphoma kinase (ALK) and ROS-1 proto-oncogene rearrangement, BRAF proto-oncogene mutation, V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations, neurotrophic tyrosine receptor kinase (NTRK) fusion, MET proto-oncogene mutation or amplification, RET proto-oncogene mutation or fusion, Neuregulin 1 (NRG1) fusion, and Human Epidermal Growth Factor Receptor-2 (HER-2) mutation are all described in NS-NSCLC, with corresponding drugs investigated or approved.
Those developments reshape the classification of lung cancer based on molecular classification and enabled an increasingly complex personalized approach in thoracic oncology, which improved the overall survival but also the quality of life of these patients [15].
Consequently, tumor genetic testing has become standard of care for metastatic and locally advanced NS-NSCLC [4]. Indeed, the Food and Drug Administration (FDA) and the European Medical Agency (EMA) promote the principles of precision medicine and claim that a specific and approved companion biomarker is mandatory for the safe administration of a targeted drug [16]. EGFR, ALK, KRAS, and ROS1 testing are mandatory at initial diagnosis, while BRAF mutations, MET exon 14 mutations, RET fusion, and NTRK fusion testing are optional at initial diagnosis and mandatory before second-line therapy, and other MET mutations, HER2 mutations, and NRG1 fusions should be tested before second-line treatment [17]. For example, second-line trastuzumab–deruxtecan demonstrated clinical benefit in metastatic HER2-positive patients, with a recent FDA approval for this indication highlighting the constantly evolving need for biomarker testing [18]. In addition, EGFR-mutation testing for pII and pIII stage NS-NSCLC patients is anticipated to become mandatory based on the results of the ADAURA study positioning the use of osimertinib as adjuvant therapy in this population [19].
After an initial response, drug resistance and disease progression occur in most cases, and therefore companion biomarkers must not only identify a therapeutic target at diagnosis, but also the techniques must be repeated, as therapeutic resistance mechanisms should be investigated during disease progression. Several methods are used to identify these molecular alterations, by analyzing protein expression levels using immunohistochemistry (IHC), by assessing mutations in tumor DNA, and by assessing the fusion of certain genes of interest (i.e., ALK, ROS1, NTRK, RET) using IHC and notably fluorescence in situ hybridization (FISH) [15]. A reference method for the study of mutations in tumor DNA was historically Sanger sequencing, described in 1977, which has been superseded by the routine use of real-time PCR (qPCR) [20]. A still commonly used strategy is based on sequential exclusion testing looking for mutations that are mutually exclusive with others or using hotspot screening to search for exploitable molecular alterations. In contrast, next-generation sequencing (NGS) allows for broad molecular profiling and allows for the simultaneous detection of both common targetable and rare mutations at the tumor DNA level and are able to identify translocations at the RNA level [20].
Practically, EGFR including the T790M mutation, BRAF and KRAS mutations, are commonly analyzed by qPCR; ALK rearrangement can be analyzed either by IHC or fluorescence in situ hybridization (FISH); ROS1 rearrangements relies on IHC and has to be confirmed by FISH analysis, and more recently DNA and RNA NGS are used for a larger panel of molecular alterations including MET exon 14; RET; NTRK1,2,3; and NRG1 [17]. In addition to MET exon 14 skipping assessment by NGS, the evaluation of the MET gene copy number (amplification) by FISH (gold-standard) or NGS should be an integral part of the diagnostic work-up, due to its clinical relevance [21]. Indeed, MET amplifications may occur de novo or as a mechanism of resistance to EGFR TKIs, but clinical trials have demonstrated that this could be targeted using capmatinib [22] or tepotinib [23] as second-line therapy [24].

2. Approved Companion Tests (Figure 1)

2.1. EGFR

The EGFR gene codes for a membrane receptor with tyrosine kinase activity belonging to the HER family [25]. The activation of EGFR is dependent on ligand binding to the extracellular domain resulting in a conformational change of the receptor allowing its hetero or homo-dimerization [26], leading to the phosphorylation of the tyrosine kinase domain. This activation allows various downstream signaling pathways of cell proliferation. The two main types of EGFR mutations are represented by in-frame exon 19 deletions, which are the most frequent, and L858R exon 21 substitution [27], and are targetable by first (erlotinib, gefitinib)-, second (afatinib, dacomitinib)-, and third (osimertinib)-generation TKIs now approved for first-line treatment in the European Union [28]. Afatinib is additionally approved for treating NSCLC with G719X, S768I, and L861Q mutations.
Figure 1. Approved diagnostic tools in lung cancer. Strengths and weaknesses of diagnostic tools appear respectively in blue and red font.
Figure 1. Approved diagnostic tools in lung cancer. Strengths and weaknesses of diagnostic tools appear respectively in blue and red font.
Jpm 12 01684 g001
qPCR is the most routinely used tool to detect deletion 19, exon 20 mutations, exon 21 L858R substitution, and T790M mutation and allows for a particularly rapid result [29]. However, some allele-specific PCR kits are likely to miss some rare EGFR mutations like exon 18 alterations (E709X, E709A, E709G, E709K, E709V, delE709-T710insD, G719X, G719S, G719A, G719C, and G719D), exon 19 in-frame insertions, exon 20 alterations (A763_Y764insFQEA insertion, S768I), and exon 21 mutation L861Q [30,31,32].
In order to use EGFR TKI, diagnosis by qPCR based on the Cobas® or Therascreen® assay is approved for the detection of EGFR mutations and used routinely, but only Cobas is approved for the use of osimertinib in patients with EGFR-mutated non-small cell lung cancer (NSCLC) with T790M mutations, and only Therascreen® assay is approved for the use of dacomitinib. Of note, Therascreen® assay is approved for using afatinib in L861Q, G719X and S768I mutations.
Next-generation sequencing (NGS), either using tissue or circulating tumor DNA (ctDNA) [33] based on the FoundationOne CDX assay (Foundation Medicine, Cambridge, MA, USA)® and FoundationOne Liquid CDX® assay for ctDNA EGFR-mutations assessment and Guardant 360 (Guardant Health, Redwood City, CA, USA)®, are approved for the use of TKIs targeting exon 19 deletions and L858R exon 21 substitution (gefitinib, erlotinib, afatinib, osimertinib). Other approved tests are the ONCO/Reveal Dx Lung & Colon Cancer Assay (O/RDx-LCCA) (Pillar Biosciences, Inc., Natick, MA, USA)® for gefitinib, erlotinib, afatinib, and dacomitinib and the Oncomine Dx Target Test (ThermoFisher Scientific, Waltham, MA, USA)® for gefitinib, but also the promising new drugs amivantanab and mobocertinib, initially developed to target EGFR exon 20 insertion at second-line treatment and now investigated in other mutations as well [34]. Amivantamab is studied in the CHRYSALIS Phase I Study [35], with or without association with lazertinib in the CHRYSALIS-2 Phase Ib/II Study [36], and in association with different regiments of chemotherapy in the PAPILLON Phase III Study [37]; while mobocertinib has been studied [38] for targeting Exon 20 EGFR insertions, amivantamab recently obtained approval by the FDA and EMA, while mobocertinib was approved by the FDA.

2.2. ALK

The ALK gene is located on chromosome 2 and codes for a receptor tyrosine kinase and was first identified in anaplastic large cell lymphoma [39]. Although many variants have been described with varying frequencies, the rearrangement of the ALK gene with EML4 (Echinoderm microtubule associated protein like 4) is the most common mechanism and occurs due to a rearrangement on the same chromosome [40,41]. This rearrangement leads to the expression of a chimeric protein, with the constitutive activation of ALK leading to cell proliferation [40], and can be targeted by the ALK TKIs crizotinib and ceritinib or more recently alectinib, brigatinib, and lorlatinib [42].
The first studies evaluating ALK rearrangement were based on FISH; however, IHC is increasingly used. Nevertheless, confirmation is recommended using FISH (if available) to confirm the gene fusion event in case of a positive IHC [43].
ALK antibodies include ALK1, 5A4, D5F3, and SP8 with better diagnostic performance in terms of sensitivity and specificity for 5A4 (Novocastra®) and D5F3 (Ventana®) [44,45], resulting in their approval as companion diagnostics for the Ventana ALK (D5F3) CDx® IHC Assay. FoundationOne CDx and FoundationOne Liquid (Foundation Medicine, Cambridge, MA, USA)® are approved [46] for administrating ALK TKIs. NGS from RNA as an alternative to DNA-based sequencing has also been investigated and implemented [47] and may be critical for the comprehensive detection of ALK gene-specific fusion partners.

2.3. ROS1

ROS1 is a tyrosine kinase receptor of the insulin receptor family that activates the MAPK signaling pathway through the phosphorylation of RAS. Its hyperactivation leads to cell growth and proliferation [48], and this receptor can be successfully targeted by crizotinib and entrectinib [49].
There are four approaches described to detect ROS1 rearrangements: IHC, FISH, qPCR, and next-generation sequencing (NGS) [50,51]. IHC can be used as screening tool and needs confirmation by another method, either FISH, qPCR, or NGS [50,51,52,53]. Nevertheless, due to the low frequency of this alteration, ROS1 rearrangements are not always routinely tested at baseline when NGS is not used and thus are assessed only after eliminating an alternative molecular alteration. Consequently, samples may have been exhausted by previous tests, exposing the risk of insufficient material, particularly for IHC or FISH techniques [54].
Consequently, two NGS companion tests are approved, either Oncomine Dx Target Test (ThermoFisher Scientific, Waltham, MA, USA)® [55] or FoundationOne CDx (Foundation Medicine, Cambridge, MA, USA)® [56].

2.4. BRAF

The BRAF gene encodes an intra-cytoplasmic serine/threonine kinase. BRAF mutations, mostly non-V600E, account for 1–4% of NS-NSCLC [57] and lead to the activation of the MAPK pathway downstream of EGFR and RAS, and this kinase can be targeted by the combination of the BRAF inhibitor dabrafenib and the MEK inhibitor trametimib [58,59].
The use of IHC is a promising approach; however, the antibody available for BRAF (VE1) [60] limits the detection to only exon 15 V600E mutations, while other alterations can be found [61,62,63].
Oncomine Dx Target Test (ThermoFisher Scientific, Waltham, MA, USA)® and FoundationOne CDx (Foundation Medicine, Cambridge, MA, USA)® are approved NGS companion tests for using dabrafenib–trametinib.

2.5. KRAS

The RAS protein is located downstream of the EGFR signaling cascade and can also interact with other EGFR downstream pathways (MAPK, PI3K-AKT-mTOR), thus bypassing its activation. This protein has an important role in cell growth, differentiation, and apoptosis control and is active when bound to GTP. KRAS is the most commonly mutated oncogene in NSCLC, and this mutation is associated with a poor prognosis [64], with the G12C being the most frequent (40–50% of KRAS mutations), followed by G12V (19% of KRAS mutations) [65,66]. These mutations are particularly observed in the lung adenocarcinoma (25 to 40%) of patients with high smoking exposure. While long considered to be undruggable, recently the approval of sotorasib revolutionized the targeting of KRAS mutant NSCLC, with adagrasib as another G12C targeting TKI expected to receive FDA approval following promising results from the phase II Krystal-1 trial as well [67]. Furthermore, there are many additional drugs targeting other non-G12C mutants currently under development [68].
The two companion tests approved are the Therascreen KRAS RGQ PCR Kit (Qiagen, Manchester, Ltd., UK)® qPCR test and the ctDNA Guardant360 CDx (Guardant Health, Inc., Redwood City, CA, USA)® NGS test for using sotorasib [69].

2.6. MET

The MET gene is located on chromosome 7 and codes for a receptor with tyrosine kinase activity whose ligand is hepatocyte growth factor. This gene is involved in the carcinogenesis of NSCLC, with several mechanisms leading to its activation by gene amplification in 4% of cases, activating the mutation and deletions of exon 14 and the overexpression of hepatocyte growth factor [70]. The occurrence of MET amplification increases to ~20% in 3rd TKI resistant EGFR-mutated NSCLC, constituting an important mechanism of resistance [70]. Presently, MET exon 14 skipping can successfully be targeted using tepotinib [71] and capmatinib [22].
FoundationOne CDx (Foundation Medicine, Cambridge, MA, USA)® and FoundationOne Liquid CDx® are two approved NGS companion tests for this alteration for using capmatinib, but there is currently no companion test specifically approved for using tepotinib.
However, an exclusive MET determination based on NGS cannot completely replace an assessment of MET gene amplification status based on FISH [21], which allows the precise assessment of amplification level normalized to the level of concomitant polysomy, although neither MET amplification test is approved.

2.7. RET

The RET gene translates into a proto-oncogene transmembrane receptor tyrosine kinase, leading to the activation of downstream signaling pathways such as RAS, MAPK, and PI3K-AKT-mTOR. The juxtaposition of the C-terminal region of the RET protein with the N-terminal portion of another protein by fusion, of which KIF5B is the most common fusion partner [72], leads to its self-sustained constitutive activation [73] and can be successfully targeted using pralsetinib [74] or selpercatinib [75].
The only approved companion test is the Oncomine Dx Target Test (ThermoFisher Scientific, Waltham, MA, USA)® NGS test for using pralsetinib, whereas selpercatinib does not yet ahve a companion diagnostic test to date.

2.8. NTRK

The NTRK 1, 2, and 3 genes allow the production of three proteins, TRKA, TRKB and TRKC, which affect cell differentiation, survival and proliferation, and migration. The NTRK fusion protein results from a DNA fragment insertion into another part of the genome. This fusion protein has emerged as a target, with approved treatments by the EMA and the FDA for larotrectinib [76] and entrectinib [77], by accelerated approval for all adult solid tumors harboring an NTRK fusion.
The only approved companion test is FoundationOne CDx (Foundation Medicine, Cambridge, MA, USA)® NGS companion test, for using larotrectinib or entrectinib [78].

3. From Multiple Companion Biomarkers to NGS?

Many companion tests exist for NS-NSCLC therapeutic targets, though not all are being approved. However, the initiation of a targeted therapy is feasible even if the test used is not the native companion test, as shown in [79] where the switching of PCR assays for common EGFR-activating mutations did not affect the outcome.
Considering that most oncogenic driver mutations that can be targeted are mutually exclusive, sequential testing, in which each gene is tested in a stepwise approach, has long been the standard procedure in the molecular pathology of NSCLC [52].
However, this approach is increasingly challenged by the development of novel therapies, dramatically broadening the number of genes to be tested [80]. In NSCLC, where tissue is often limited, all available specimens will be exhausted before tissue testing is complete. Furthermore, this approach often leads to incomplete biomarker testing, limiting the possibility of initiating adequate treatment [81,82].
Additionally, many tests do not cover all relevant regions in the respective genes and thus often miss out on the detection of certain mutations. For example, there are several studies currently investigating the treatment of Exon 20 mutations in both EGFR and HER2, with multiple drugs currently being investigated in this setting [83]. Most qPCR tests only have limited coverage in those regions, thus limiting the detection of mutations and, consequently, the initiation of targeted treatments in this setting [84,85].
A recent study investigating the structural implications of a broad range of EGFR mutations highlighted that many EGFR mutations result in comparable structural changes and comparable sensitivity to certain TKI drug classes [31]. Since many of these mutations cannot be investigated with qPCR, the implementation of NGS is critical in the clinical setting.
Indeed, the use of NGS as a primary test has dramatically increased during past years [86], though additional efforts are critical to implementing NGS widely as a principal testing method at baseline in NSCLC. This approach is not only suitable for primary tumor tissue but is routinely used in the transbronchial needle aspiration of lymph node [87], plasma [88], and pleural effusions [89] and could also be used from bronchoalveolar lavage fluid [90] and is also suitable for cerebrospinal fluid [91]. Besides, there are promising data for the screening of residual disease of EGFR mutant NSCLC in saliva [92] and urine [93].
The rationale to extend the use of NGS for NSCLC molecular diagnostic is also highlighted by ESCAT (ESMO Scale for Clinical Actionability of Molecular Targets). This collaborative tool aims to classify the molecular alterations that can be found in tumor tissue [94] or in ctDNA [95] into six tiers, depending on the clinical significance of the molecular alterations, in order to harmonize practices. Briefly, tier I corresponds to alterations that are known to be targetable; tier II corresponds to alterations that are non-targetable according to current recommendations but are promising in clinical trials; tier III corresponds to alterations that are targetable in another tumor type or for similar targets; tier IV corresponds to alterations for which preclinical models are promising; tier V corresponds to alterations that could be co-targeted, and tier X corresponds to alterations with no evidence of clinical significance. Importantly, studies have provided evidence that the ESCAT classification of genomic alterations is implementable in clinical practice [96,97].
In the past, especially increased costs and longer turn-around times have disfavored NGS over sequential testing utilizing IHC/FISH or qPCR. Comparisons on the cost efficacy of NGS are challenging due to a rapidly changing field and due to the fact that the assessment of genetic tests has traditionally been regulated mostly at the national level in the EU [98], leading to a certain heterogeneity of the pathology workflow between countries and institutions. In France, for example, regional molecular genetics centers have been created to allow performing selected molecular tests free of charge for all cancer patients in their region, with financial support assisting in the implementation of NGS [15]. Issues surrounding the systematic implementation of NGS at the European level are discussed in [99], highlighting difficulties linked to the governance of collaboration between private and public institutions, to the adaptation of the necessary and often complex infrastructure, to the standardization of sequencing panels which might include in-house developed assays, to the often diverse training of personnel, and to the financial cost of the tests. Importantly, cost effectiveness was investigated in multiple studies, and NGS has been considered cost-effective in NSCLC when multiple genes require parallel assessment [100,101,102]. However, there are still ongoing debates on the most cost-effective use of NGS. For example, while performing NGS at initial diagnosis as well as at disease progression under treatment is technically feasible and routinely performed, reimbursement for NGS in France is limited to one single test. Multiple testing is thus often covered by research funding or results in additional out-of-pocket costs for the patients. Similar regulations may exist in other countries as well, and consequently reimbursement regulations need adaptions in order to allow a scientifically driven and effective biomarker-driven treatment in NSCLC.
Two approaches can be currently considered for NGS [103]. The “bespoke testing” approach, initiated “on-demand” by the clinician, is essentially aimed at advanced-stage patients in order to initiate a targeted therapy. However, this approach might delay treatment duration or might restrict the testing to certain population, due to delay in the request for testing and the tissue exhaustion due to multiple requests at different times. By contrast, the “reflex testing” approach is triggered by a pathologist based on a histological diagnosis, with the advantage of managing currently available tissue more efficiently, with an early result, which, however, leads to increased testing requirements and requires additional resources and infrastructure.
In addition to tissue-based testing approaches, liquid biopsies using circulating tumor DNA (ctDNA) are also critical for clinical recommendations and are thus recommended by the IASLC [104]. However, the importance of liquid biopsies compared to tissue have been reviewed extensively, and the following paragraph should only give a brief overview [105,106,107,108]. Importantly, liquid biopsies can help to adjust treatment decisions under active treatment through the detection of resistance mechanisms but can also replace tissue-based testing when specimen have been exhausted or are too limited [109], a situation in which the validation of NGS assays designated for low DNA input is crucial. It can further help as a prognostic tool through the determination of minimal residual disease (MRD) [107]. Consequently, it is critical to further ensure that liquid biopsies are implemented in NSCLC and become standard when tissue biopsies are absent or not possible.
Turn-around times is another challenge, since the quick reporting of results is critical to ensuring the administration of an appropriate treatment. Delayed reporting often leads to the initiation of untargeted approaches with dismal outcomes [110] and should thus be avoided. Nevertheless, turn-around time for NGS has been reduced and is comparable to sequential approaches [86]. Importantly, novel technologies dramatically reduced turn-around times to ~24 h [111]. While novel technologies constantly reduce turn-around times in NGS, qPCR, especially highly automated assays, still offer significantly faster results, and it was demonstrated that a combination of ultra-fast qPCR for EGFR combined with subsequent NGS can be implemented, allowing both quick reports on common oncogenes and broad testing in a timely and cost-effective manner [112]. The type of NGS test is also of particular concern, with assays commonly relying on either amplicon-based [113] or hybrid-capture-based tests [114]. While both principles allow reproducible tests, the optimal solution is also dependent on the size of the panel used, with NGS panels commonly spanning a few (~10) to hundreds or thousands of genes, which can also be achieved with in-house solutions [111,115]. However, for larger panels with eg > 500 genes, validation is critical, and an outsourced approach might be more relevant, especially when demand is low. Access to clinical trials and needed research might be critical to determining the optimal size, as only few gene targets are actually associated with an approved therapy and consequently, larger panels might be more suitable in academic centers but unnecessary for routine clinical care [116].
Limitations to NGS still exist, and, in particular, the detection of gene fusion events using NGS lacks sensitivity compared to IHC/FISH methods, which often warrants the additional use of those methods in patients with high prevalence of those alterations [114]. Moreover, the detection of amplifications still poses challenges in NGS compared to FISH analysis [21]. Consequently, further development to refine sensitivities in these setting is critical but should not prohibit the use of NGS.

4. Conclusions

In the era of precision medicine, NGS appears fundamental to offer the most appropriate personalized therapy for NS-NSCLC patients and is poised to substitute for sequential testing using individual or low multiplexing gene tests. Indeed, research leading to new targeted therapies is increasing very quickly and the new international guidelines recommends the use of NGS in order to test all of the actionable alterations simultaneously, including established but also emerging targets.

Author Contributions

N.P.: original draft writing and graphical design; S.H.: supervision, administration, resources, reviewing and editing; C.B.: reviewing and editing; J.B. (Jacques Boutros): reviewing and editing; M.I.: reviewing and editing; V.H.: reviewing and editing; C.-H.M.: reviewing and editing; P.H.: reviewing and editing; J.B. (Jonathan Benzaquen): supervision, administration, resources, reviewing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

S.H. received speaker/consulting fees from Qiagen and Boehringer Ingelheim and owns various intellectual property on the treatment of lung cancer. V.H. conducted an advisory board for BMS. P.H. conducted advisory boards for Janssen, BMS, AstraZeneca, Abbvie, Roche, Pfizer, Lilly, Biocartis, Guardant Health; received honorarias from Janssen, BMS, AstraZeneca, Abbvie, Guardant Health, Pierre Fabre, Illumina, Thermo Fisher Scientific, Lilly, Biocartis, Roche, Pfizer, Bayer, MSD, Novartis; received financial research support from BMS, Thermo Fisher Scientific, and Biocartis. J.B. (Jonathan Benzaquen) received speaker fees/board honorarias from AstraZeneca and Sanofi.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. O’Keeffe, L.M.; Taylor, G.; Huxley, R.R.; Mitchell, P.; Woodward, M.; Peters, S.A.E. Smoking as a risk factor for lung cancer in women and men: A systematic review and meta-analysis. BMJ Open 2018, 8, e021611. [Google Scholar] [CrossRef] [Green Version]
  3. Pelosof, L.; Ahn, C.; Gao, A.; Horn, L.; Madrigales, A.; Cox, J.; McGavic, D.; Minna, J.D.; Gazdar, A.F.; Schiller, J. Proportion of Never-Smoker Non-Small Cell Lung Cancer Patients at Three Diverse Institutions. J. Natl. Cancer Inst. 2017, 109, djw295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sholl, L.M.; Aisner, D.L.; Varella-Garcia, M.; Berry, L.D.; Dias-Santagata, D.; Wistuba, I.I.; Chen, H.; Fujimoto, J.; Kugler, K.; Franklin, W.A.; et al. Multi-institutional oncogenic driver mutation analysis in lung adenocarcinoma: The lung cancer mutation consortium experience. J. Thorac. Oncol. 2015, 10, 768–777. [Google Scholar] [CrossRef] [Green Version]
  5. Smolle, E.; Pichler, M. Non-smoking-associated lung cancer: A distinct entity in terms of tumor biology, patient characteristics and impact of hereditary cancer predisposition. Cancers 2019, 11, 204. [Google Scholar] [CrossRef] [Green Version]
  6. Travis, W.D.; Rekhtman, N. Pathological diagnosis and classification of lung cancer in small biopsies and cytology: Strategic management of tissue for molecular testing. Semin. Respir. Crit. Care Med. 2011, 32, 22–31. [Google Scholar] [CrossRef]
  7. Walsh, S.L.F.; Lederer, D.J.; Ryerson, C.J.; Kolb, M.; Maher, T.M.; Nusser, R.; Poletti, V.; Richeldi, L.; Vancheri, C.; Wilsher, M.L.; et al. Diagnostic likelihood thresholds that define a working diagnosis of idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2019, 200, 1146–1153. [Google Scholar] [CrossRef]
  8. Mok, T.S.; Wu, Y.-L.; Thongprasert, S.; Yang, C.-H.; Chu, D.-T.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; et al. Gefitinib or Carboplatin–Paclitaxel in Pulmonary Adenocarcinoma. N. Engl. J. Med. 2009, 361, 947–957. [Google Scholar] [CrossRef] [PubMed]
  9. Zhou, C.; Wu, Y.L.; Chen, G.; Feng, J.; Liu, X.Q.; Wang, C.; Zhang, S.; Wang, J.; Zhou, S.; Ren, S.; et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): A multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011, 12, 735–742. [Google Scholar] [CrossRef]
  10. Pao, W.; Miller, V.A.; Politi, K.A.; Riely, G.J.; Somwar, R.; Zakowski, M.F.; Kris, M.G.; Varmus, H. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005, 2, 0225–0235. [Google Scholar] [CrossRef]
  11. Papadimitrakopoulou, V.A.; Mok, T.S.; Han, J.Y.; Ahn, M.J.; Delmonte, A.; Ramalingam, S.S.; Kim, S.W.; Shepherd, F.A.; Laskin, J.; He, Y.; et al. Osimertinib versus platinum–pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis. Ann. Oncol. 2020, 31, 1536–1544. [Google Scholar] [CrossRef] [PubMed]
  12. Leonetti, A.; Sharma, S.; Minari, R.; Perego, P.; Giovannetti, E.; Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 2019, 121, 725–737. [Google Scholar] [CrossRef]
  13. Schalm, S.S.; Dineen, T.; Lim, S.M.; Park, C.-W.; Hsieh, J.; Woessner, R.; Zhang, Z.; Wilson, K.; Eno, M.; Wilson, D.; et al. 1296P BLU-945, a highly potent and selective 4th generation EGFR TKI for the treatment of EGFR T790M/C797S resistant NSCLC. Ann. Oncol. 2020, 31, S839. [Google Scholar] [CrossRef]
  14. 14. Finlay, M.R.V.; Barton, P.; Bickerton, S.; Bista, M.; Colclough, N.; Cross, D.A.E.; Evans, L.; Floc’h, N.; Gregson, C.; Guérot, C.M.; et al. Potent and Selective Inhibitors of the Epidermal Growth Factor Receptor to Overcome C797S-Mediated Resistance. J. Med. Chem. 2021, 64, 13704–13718. [Google Scholar] [CrossRef] [PubMed]
  15. Hofman, P.; Rouleau, E.; Sabourin, J.C.; Denis, M.; Deleuze, J.F.; Barlesi, F.; Laurent-Puig, P. Predictive molecular pathology in non–small cell lung cancer in France: The past, the present and the perspectives. Cancer Cytopathol. 2020, 128, 601–610. [Google Scholar] [CrossRef] [PubMed]
  16. Brown, N.A.; Aisner, D.L.; Oxnard, G.R. Precision Medicine in Non–Small Cell Lung Cancer: Current Standards in Pathology and Biomarker Interpretation. Am. Soc. Clin. Oncol. Educ. B 2018, 38, 708–715. [Google Scholar] [CrossRef]
  17. Aisner, D.L.; Riely, G.J. Non-small cell lung cancer: Recommendations for biomarker testing and treatment. J. Natl. Compr. Cancer Netw. 2021, 19, 610–613. [Google Scholar] [CrossRef]
  18. Li, B.T.; Smit, E.F.; Goto, Y.; Nakagawa, K.; Udagawa, H.; Mazières, J.; Nagasaka, M.; Bazhenova, L.; Saltos, A.N.; Felip, E.; et al. Trastuzumab Deruxtecan in HER2 -Mutant Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2022, 386, 241–251. [Google Scholar] [CrossRef]
  19. Wu, Y.-L.; Tsuboi, M.; He, J.; John, T.; Grohe, C.; Majem, M.; Goldman, J.W.; Laktionov, K.; Kim, S.-W.; Kato, T.; et al. Osimertinib in Resected EGFR -Mutated Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 1711–1723. [Google Scholar] [CrossRef] [PubMed]
  20. Aggarwal, C.; Bubendorf, L.; Cooper, W.A.; Illei, P.; Borralho Nunes, P.; Ong, B.H.; Tsao, M.S.; Yatabe, Y.; Kerr, K.M. Molecular testing in stage I–III non-small cell lung cancer: Approaches and challenges. Lung Cancer 2021, 162, 42–53. [Google Scholar] [CrossRef] [PubMed]
  21. Schubart, C.; Stöhr, R.; Tögel, L.; Fuchs, F.; Sirbu, H.; Seitz, G.; Seggewiss-bernhardt, R.; Leistner, R.; Sterlacci, W.; Vieth, M.; et al. Met amplification in non-small cell lung cancer (Nsclc)—A consecutive evaluation using next-generation sequencing (ngs) in a real-world setting. Cancers 2021, 13, 5023. [Google Scholar] [CrossRef] [PubMed]
  22. Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Hida, T.; de Jonge, M.; Orlov, S.V.; et al. Capmatinib in MET Exon 14–Mutated or MET -Amplified Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef] [PubMed]
  23. Le, X.; Paz-Ares, L.G.; Van Meerbeeck, J.; Viteri, S.; Cabrera Galvez, C.; Vicente Baz, D.; Kim, Y.-C.; Kang, J.-H.; Schumacher, K.-M.; Karachaliou, N.; et al. Tepotinib in patients (pts) with advanced non-small cell lung cancer (NSCLC) with MET amplification (MET amp). J. Clin. Oncol. 2021, 39, 9021. [Google Scholar] [CrossRef]
  24. Jørgensen, J.T.; Mollerup, J. Companion Diagnostics and Predictive Biomarkers for MET-Targeted Therapy in NSCLC. Cancers 2022, 14, 2150. [Google Scholar] [CrossRef]
  25. Mendelsohn, J.; Baselga, J. Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J. Clin. Oncol. 2003, 21, 2787–2799. [Google Scholar] [CrossRef]
  26. Schlessinger, J. Ligand-induced, receptor-mediated dimerization and activation of EGF receptor. Cell 2002, 110, 669–672. [Google Scholar] [CrossRef] [Green Version]
  27. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating Mutations in the Epidermal Growth Factor Receptor Underlying Responsiveness of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef] [PubMed]
  28. Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.; Chewaskulyong, B.; et al. Overall Survival with Osimertinib in Untreated, EGFR -Mutated Advanced NSCLC. N. Engl. J. Med. 2020, 382, 41–50. [Google Scholar] [CrossRef] [PubMed]
  29. Dagogo-Jack, I.; Azzolli, C.G.; Fintelmann, F.; Mino-Kenudson, M.; Farago, A.F.; Gainor, J.F.; Jiang, G.; Piotrowska, Z.; Heist, R.S.; Lennes, I.T.; et al. Clinical Utility of Rapid EGFR Genotyping in Advanced Lung Cancer. JCO Precis. Oncol. 2018, 2, 1–13. [Google Scholar] [CrossRef] [PubMed]
  30. Gristina, V.; Malapelle, U.; Galvano, A.; Pisapia, P.; Pepe, F.; Rolfo, C.; Tortorici, S.; Bazan, V.; Troncone, G.; Russo, A. The significance of epidermal growth factor receptor uncommon mutations in non-small cell lung cancer: A systematic review and critical appraisal. Cancer Treat. Rev. 2020, 85, 101994. [Google Scholar] [CrossRef] [PubMed]
  31. Robichaux, J.P.; Le, X.; Vijayan, R.S.K.; Hicks, J.K.; Heeke, S.; Elamin, Y.Y.; Lin, H.Y.; Udagawa, H.; Skoulidis, F.; Tran, H.; et al. Structure-based classification predicts drug response in EGFR-mutant NSCLC. Nature 2021, 597, 732–737. [Google Scholar] [CrossRef] [PubMed]
  32. Harrison, P.T.; Vyse, S.; Huang, P.H. Rare epidermal growth factor receptor (EGFR) mutations in non-small cell lung cancer. Semin. Cancer Biol. 2020, 61, 167–179. [Google Scholar] [CrossRef] [PubMed]
  33. Aggarwal, C.; Rolfo, C.D.; Oxnard, G.R.; Gray, J.E.; Sholl, L.M.; Gandara, D.R. Strategies for the successful implementation of plasma-based NSCLC genotyping in clinical practice. Nat. Rev. Clin. Oncol. 2021, 18, 56–62. [Google Scholar] [CrossRef] [PubMed]
  34. Olivier, T.; Prasad, V. Amivantamab and Mobocertinib in Exon 20 insertions EGFR Mutant Lung Cancer, Challenge to the Current Guidelines. Transl. Oncol. 2022, 23, 101475. [Google Scholar] [CrossRef]
  35. Park, K.; Haura, E.B.; Leighl, N.B.; Mitchell, P.; Shu, C.A.; Girard, N.; Viteri, S.; Han, J.Y.; Kim, S.W.; Lee, C.K.; et al. Amivantamab in egfr exon 20 insertion- mutated non-small-cell lung cancer progressing on platinum chemotherapy: Initial results from the chrysalis phase i study. J. Clin. Oncol. 2021, 39, 3391–3402. [Google Scholar] [CrossRef]
  36. Shu, C.A.; Goto, K.; Cho, B.C.; Griesinger, F.; Yang, J.C.-H.; Felip, E.; Xie, J.; Chen, J.; Mahoney, J.; Thayu, M.; et al. CHRYSALIS-2: A phase 1/1b study of lazertinib as monotherapy and in combination with amivantamab in patients with EGFR-mutant NSCLC. J. Clin. Oncol. 2021, 39, TPS9132. [Google Scholar] [CrossRef]
  37. Agrawal, T.; Artis, E.; Xie, J.; Bhattacharya, A.; Haddish-Berhane, N.; Gopen, T.; Curtin, J.C.; Karkera, J.; Roshak, A.; Knoblauch, R.E.; et al. P76.74 PAPILLON: Randomized Phase 3 Study of Amivantamab Plus Chemotherapy vs Chemotherapy Alone in EGFR Exon20ins NSCLC. J. Thorac. Oncol. 2021, 16, S621. [Google Scholar] [CrossRef]
  38. Zhou, C.; Ramalingam, S.S.; Kim, T.M.; Kim, S.W.; Yang, J.C.H.; Riely, G.J.; Mekhail, T.; Nguyen, D.; Garcia Campelo, M.R.; Felip, E.; et al. Treatment Outcomes and Safety of Mobocertinib in Platinum-Pretreated Patients with EGFR Exon 20 Insertion-Positive Metastatic Non-Small Cell Lung Cancer: A Phase 1/2 Open-label Nonrandomized Clinical Trial. JAMA Oncol. 2021, 7, 1772–1781. [Google Scholar] [CrossRef]
  39. Morris, S.W.; Kirstein, M.N.; Valentine, M.B.; Dittmer, K.G.; Shapiro, D.N.; Saltman, D.L.; Look, A.T. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma. Science 1994, 263, 1281–1284. [Google Scholar] [CrossRef]
  40. Soda, M.; Choi, Y.L.; Enomoto, M.; Takada, S.; Yamashita, Y.; Ishikawa, S.; Fujiwara, S.I.; Watanabe, H.; Kurashina, K.; Hatanaka, H.; et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007, 448, 561–566. [Google Scholar] [CrossRef]
  41. Chia, P.L.; Dobrovic, A.; Dobrovic, A.; John, T. Prevalence and natural history of ALK positive non-small-cell lung cancer and the clinical impact of targeted therapy with ALK inhibitors. Clin. Epidemiol. 2014, 6, 423–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Pan, Y.; Deng, C.; Qiu, Z.; Cao, C.; Wu, F. The Resistance Mechanisms and Treatment Strategies for ALK-Rearranged Non-Small Cell Lung Cancer. Front. Oncol. 2021, 11, 3945. [Google Scholar] [CrossRef]
  43. Kozuma, Y.; Toyokawa, G.; Seto, T. ALK testing methods: Is there a winner or loser? Expert Rev. Anticancer Ther. 2019, 19, 237–244. [Google Scholar] [CrossRef]
  44. Mino-Kenudson, M.; Chirieac, L.R.; Law, K.; Hornick, J.L.; Lindeman, N.; Mark, E.J.; Cohen, D.W.; Johnson, B.E.; Jänne, P.A.; Iafrate, A.J.; et al. A novel, highly sensitive antibody allows for the routine detection of ALK-rearranged lung adenocarcinomas by standard immunohistochemistry. Clin. Cancer Res. 2010, 16, 1561–1571. [Google Scholar] [CrossRef] [Green Version]
  45. Wynes, M.W.; Sholl, L.M.; Dietel, M.; Schuuring, E.; Tsao, M.S.; Yatabe, Y.; Tubbs, R.R.; Hirsch, F.R. An international interpretation study using the ALK IHC antibody D5F3 and a sensitive detection kit demonstrates high concordance between ALK IHC and ALK FISH and between evaluators. J. Thorac. Oncol. 2014, 9, 631–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. 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 Chromosom. Cancer 2022, 61, 244–260. [Google Scholar] [CrossRef]
  47. Xia, P.; Zhang, L.; Li, P.; Liu, E.; Li, W.; Zhang, J.; Li, H.; Su, X.; Jiang, G. Molecular characteristics and clinical outcomes of complex ALK rearrangements identified by next-generation sequencing in non-small cell lung cancers. J. Transl. Med. 2021, 19, 308. [Google Scholar] [CrossRef] [PubMed]
  48. Chin, L.P.; Soo, R.A.; Soong, R.; Ou, S.H.I. Targeting ROS1 with anaplastic lymphoma kinase inhibitors: A promising therapeutic strategy for a newly defined molecular subset of non-small-cell lung cancer. J. Thorac. Oncol. 2012, 7, 1625–1630. [Google Scholar] [CrossRef] [Green Version]
  49. Moliner, L.; Arriola, E. ROS1 non-small cell lung cancer patients treatment approach. Precis. Cancer Med. 2021, 4, 25. [Google Scholar]
  50. Garrido, P.; Conde, E.; de Castro, J.; Gómez-Román, J.J.; Felip, E.; Pijuan, L.; Isla, D.; Sanz, J.; Paz-Ares, L.; López-Ríos, F. Updated guidelines for predictive biomarker testing in advanced non-small-cell lung cancer: A National Consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology. Clin. Transl. Oncol. 2020, 22, 989–1003. [Google Scholar] [CrossRef] [Green Version]
  51. Conde, E.; Hernandez, S.; Benito, A.; Caminoa, A.; Garrido, P.; Lopez-Rios, F. Screening for ROS1 fusions in patients with advanced non-small cell lung carcinomas using the VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody. Expert Rev. Mol. Diagn. 2021, 21, 437–444. [Google Scholar] [CrossRef]
  52. Lindeman, N.I.; Cagle, P.T.; Aisner, D.L.; Arcila, M.E.; Beasley, M.B.; Bernicker, E.H.; Colasacco, C.; Dacic, S.; Hirsch, F.R.; Kerr, K.; et al. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors guideline from the college of American pathologists, the international association for the study of lung cancer, and the a. Arch. Pathol. Lab. Med. 2018, 142, 321–346. [Google Scholar] [CrossRef] [Green Version]
  53. Hofman, V.; Rouquette, I.; Long-Mira, E.; Piton, N.; Chamorey, E.; Heeke, S.; Vignaud, J.M.; Yguel, C.; Mazières, J.; Lepage, A.-L.; et al. Multicenter Evaluation of a Novel ROS1 Immunohistochemistry Assay (SP384) for Detection of ROS1 Rearrangements in a Large Cohort of Lung Adenocarcinoma Patients. J. Thorac. Oncol. 2019, 14, 1204–1212. [Google Scholar] [CrossRef] [PubMed]
  54. Bubendorf, L.; Büttner, R.; Al-Dayel, F.; Dietel, M.; Elmberger, G.; Kerr, K.; López-Ríos, F.; Marchetti, A.; Öz, B.; Pauwels, P.; et al. Testing for ROS1 in non-small cell lung cancer: A review with recommendations. Virchows Arch. 2016, 469, 489–503. [Google Scholar] [CrossRef]
  55. Ito, K.; Sakata, S.; Otsubo, K.; Nakamura, A.; Teraoka, S.; Matsumoto, N.; Shiraishi, Y.; Haratani, K.; Tamiya, M.; Ikeda, S.; et al. MO4-5 A multicenter retrospective study of Oncomine Dx Target Test multi-CDx system for genetic profiling of NSCLC: WJOG13019L. Ann. Oncol. 2021, 32, S297. [Google Scholar] [CrossRef]
  56. Schwartzberg, L.S.; Li, G.; Tolba, K.; Bourla, A.B.; Schulze, K.; Gadgil, R.; Fine, A.; Lofgren, K.T.; Graf, R.P.; Oxnard, G.R.; et al. Complementary roles for tissue and blood based comprehensive genomic profiling for detection of actionable driver alterations in advanced NSCLC. JTO Clin. Res. Rep. 2022, 3, 100386. [Google Scholar] [CrossRef] [PubMed]
  57. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Planchard, D.; Besse, B.; Groen, H.J.M.; Hashemi, S.M.S.; Mazieres, J.; Kim, T.M.; Quoix, E.; Souquet, P.J.; Barlesi, F.; Baik, C.; et al. Phase 2 Study of Dabrafenib Plus Trametinib in Patients With BRAF V600E-Mutant Metastatic NSCLC: Updated 5-Year Survival Rates and Genomic Analysis. J. Thorac. Oncol. 2022, 17, 103–115. [Google Scholar] [CrossRef]
  59. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M., Jr.; Zhang, P.; Mookerjee, B.; et al. Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non-small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef]
  60. Gow, C.H.; Hsieh, M.S.; Lin, Y.T.; Liu, Y.N.; Shih, J.Y. Validation of immunohistochemistry for the detection of BRAF V600E-mutated lung adenocarcinomas. Cancers 2019, 11, 866. [Google Scholar] [CrossRef] [Green Version]
  61. Sasaki, H.; Shimizu, S.; Tani, Y.; Shitara, M.; Okuda, K.; Hikosaka, Y.; Moriyama, S.; Yano, M.; Fujii, Y. Usefulness of immunohistochemistry for the detection of the BRAF V600E mutation in Japanese lung adenocarcinoma. Lung Cancer 2013, 82, 51–54. [Google Scholar] [CrossRef] [PubMed]
  62. Hofman, V.; Benzaquen, J.; Heeke, S.; Lassalle, S.; Poudenx, M.; Long, E.; Lantéri, E.; Bordone, O.; Lespinet, V.; Tanga, V.; et al. Real-world assessment of the BRAF status in non-squamous cell lung carcinoma using VE1 immunohistochemistry: A single laboratory experience (LPCE, Nice, France). Lung Cancer 2020, 145, 58–62. [Google Scholar] [CrossRef] [PubMed]
  63. Ilie, M.; Long, E.; Hofman, V.; Dadone, B.; Marquette, C.H.; Mouroux, J.; Vignaud, J.M.; Begueret, H.; Merlio, J.P.; Capper, D.; et al. Diagnostic value of immunohistochemistry for the detection of the BRAFV600E mutation in primary lung adenocarcinoma caucasian patients. Ann. Oncol. 2013, 24, 742–748. [Google Scholar] [CrossRef]
  64. Keohavong, P.; DeMichele, M.A.A.; Melacrinos, A.C.; Landreneau, R.J.; Weyant, R.J.; Siegfried, J.M. Detection of K-ras mutations in lung carcinomas: Relationship to prognosis. Clin. Cancer Res. 1996, 2, 411–418. [Google Scholar]
  65. Judd, J.; Karim, N.A.; Khan, H.; Naqash, A.R.; Baca, Y.; Xiu, J.; VanderWalde, A.M.; Mamdani, H.; Raez, L.E.; Nagasaka, M.; et al. Characterization of KRAS mutation subtypes in non-small cell lung cancer. Mol. Cancer Ther. 2021, 20, 2577–2584. [Google Scholar] [CrossRef]
  66. 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 2022, 14, 1628. [Google Scholar] [CrossRef]
  67. Jänne, P.A.; Riely, G.J.; Gadgeel, S.M.; Heist, R.S.; Ou, S.-H.I.; Pacheco, J.M.; Johnson, M.L.; Sabari, J.K.; Leventakos, K.; Yau, E.; et al. Adagrasib in Non–Small-Cell Lung Cancer Harboring a KRAS G12C Mutation. N. Engl. J. Med. 2022, 387, 120–131. [Google Scholar] [CrossRef] [PubMed]
  68. Xie, M.; Xu, X.; Fan, Y. KRAS-Mutant Non-Small Cell Lung Cancer: An Emerging Promisingly Treatable Subgroup. Front. Oncol. 2021, 11, 672612. [Google Scholar] [CrossRef]
  69. Skoulidis, F.; Li, B.T.; Dy, G.K.; Price, T.J.; Falchook, G.S.; Wolf, J.; Italiano, A.; Schuler, M.; Borghaei, H.; Barlesi, F.; et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N. Engl. J. Med. 2021, 384, 2371–2381. [Google Scholar] [CrossRef]
  70. Sattler, M.; Reddy, M.M.; Hasina, R.; Gangadhar, T.; Salgia, R. The role of the c-Met pathway in lung cancer and the potential for targeted therapy. Ther. Adv. Med. Oncol. 2011, 3, 171–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non–Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef] [PubMed]
  72. Ju, Y.S.; Lee, W.C.; Shin, J.Y.; Lee, S.; Bleazard, T.; Won, J.K.; Kim, Y.T.; Kim, J.-I.; Kang, J.H.; Seo, J.S. A transforming KIF5B and RET gene fusion in lung adenocarcinoma revealed from whole-genome and transcriptome sequencing. Genome Res. 2012, 22, 436–445. [Google Scholar] [CrossRef] [PubMed]
  73. Tsuta, K.; Kohno, T.; Yoshida, A.; Shimada, Y.; Asamura, H.; Furuta, K.; Kushima, R. RET-rearranged non-small-cell lung carcinoma: A clinicopathological and molecular analysis. Br. J. Cancer 2014, 110, 1571–1578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Gainor, J.F.; Curigliano, G.; Kim, D.W.; Lee, D.H.; Besse, B.; Baik, C.S.; Doebele, R.C.; Cassier, P.A.; Lopes, G.; Tan, D.S.W.; et al. Pralsetinib for RET fusion-positive non-small-cell lung cancer (ARROW): A multi-cohort, open-label, phase 1/2 study. Lancet Oncol. 2021, 22, 959–969. [Google Scholar] [CrossRef]
  75. Drilon, A.; Oxnard, G.R.; Tan, D.S.W.; Loong, H.H.F.; Johnson, M.; Gainor, J.; McCoach, C.E.; Gautschi, O.; Besse, B.; Cho, B.C.; et al. Efficacy of Selpercatinib in RET Fusion–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 813–824. [Google Scholar] [CrossRef] [PubMed]
  76. Drilon, A.; Laetsch, T.W.; Kummar, S.; DuBois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion–Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef]
  77. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; 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, 271–282. [Google Scholar] [CrossRef]
  78. Marchiò, C.; Scaltriti, M.; Ladanyi, M.; Iafrate, A.J.; Bibeau, F.; Dietel, M.; Hechtman, J.F.; Troiani, T.; López-Rios, F.; Douillard, J.Y.; et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann. Oncol. 2019, 30, 1417–1427. [Google Scholar] [CrossRef] [Green Version]
  79. Uchibori, K.; Takano, N.; Manabe, R.; Tsugitomi, R.; Ogusu, S.; Tozuka, T.; Sakamoto, H.; Yoshida, H.; Amino, Y.; Ariyasu, R.; et al. Clinical influence of switching companion diagnostic tests for EGFR-TKs from Therascreen to Cobas v2. Thorac. Cancer 2021, 12, 906–913. [Google Scholar] [CrossRef]
  80. Gregg, J.P.; Li, T.; Yoneda, K.Y. Molecular testing strategies in non-small cell lung cancer: Optimizing the diagnostic journey. Transl. Lung Cancer Res. 2019, 8, 286–301. [Google Scholar] [CrossRef] [Green Version]
  81. Griesinger, F.; Eberhardt, W.; Nusch, A.; Reiser, M.; Zahn, M.O.; Maintz, C.; Bernhardt, C.; Losem, C.; Stenzinger, A.; Heukamp, L.C.; et al. Biomarker testing in non-small cell lung cancer in routine care: Analysis of the first 3,717 patients in the German prospective, observational, nation-wide CRISP Registry (AIO-TRK-0315). Lung Cancer 2021, 152, 174–184. [Google Scholar] [CrossRef] [PubMed]
  82. Adizie, J.B.; Tweedie, J.; Khakwani, A.; Peach, E.; Hubbard, R.; Wood, N.; Gosney, J.R.; Harden, S.V.; Beckett, P.; Popat, S.; et al. Biomarker Testing for People With Advanced Lung Cancer in England. JTO Clin. Res. Rep. 2021, 2, 100176. [Google Scholar] [CrossRef] [PubMed]
  83. Friedlaender, A.; Subbiah, V.; Russo, A.; Banna, G.L.; Malapelle, U.; Rolfo, C.; Addeo, A. EGFR and HER2 exon 20 insertions in solid tumours: From biology to treatment. Nat. Rev. Clin. Oncol. 2022, 19, 51–69. [Google Scholar] [CrossRef] [PubMed]
  84. Lin, H.M.; Yin, Y.; Crossland, V.; Wu, Y.; Ou, S.H.I. EGFR Testing Patterns and Detection of EGFR Exon 20 Insertions in the United States. JTO Clin. Res. Rep. 2022, 3, 100285. [Google Scholar] [CrossRef]
  85. Bauml, J.M.; Viteri, S.; Minchom, A.; Bazhenova, L.; Ou, S.; Schaffer, M.; Le Croy, N.; Riley, R.; Mahadevia, P.; Girard, N. FP07.12 Underdiagnosis of EGFR Exon 20 Insertion Mutation Variants: Estimates from NGS-based Real-World Datasets. J. Thorac. Oncol. 2021, 16, S208–S209. [Google Scholar] [CrossRef]
  86. Hess, L.M.; Krein, P.M.; Haldane, D.; Han, Y.; Sireci, A.N. Biomarker Testing for Patients With Advanced/Metastatic Nonsquamous NSCLC in the United States of America, 2015 to 2021. JTO Clin. Res. Rep. 2022, 3, 100336. [Google Scholar] [CrossRef]
  87. Xie, F.; Zheng, X.; Mao, X.; Zhao, R.; Ye, J.; Zhang, Y.; Sun, J. Next-Generation Sequencing for Genotyping of Endobronchial Ultrasound-Guided Transbronchial Needle Aspiration Samples in Lung Cancer. Ann. Thorac. Surg. 2019, 108, 219–226. [Google Scholar] [CrossRef]
  88. Heitzer, E.; van den Broek, D.; Denis, M.G.; Hofman, P.; Hubank, M.; Mouliere, F.; Paz-Ares, L.; Schuuring, E.; Sültmann, H.; Vainer, G.; et al. Recommendations for a practical implementation of circulating tumor DNA mutation testing in metastatic non-small-cell lung cancer. ESMO Open 2022, 7, 100399. [Google Scholar] [CrossRef]
  89. Patel, A.; Hissong, E.; Rosado, L.; Burkhardt, R.; Cong, L.; Alperstein, S.A.; Siddiqui, M.T.; Park, H.J.; Song, W.; Velu, P.D.; et al. Next-Generation Sequencing of Cell-Free DNA Extracted From Pleural Effusion Supernatant: Applications and Challenges. Front. Med. 2021, 8, 662312. [Google Scholar] [CrossRef]
  90. Otake, S.; Goto, T.; Higuchi, R.; Nakagomi, T.; Hirotsu, Y.; Amemiya, K.; Oyama, T.; Mochizuki, H.; Omata, M. The Diagnostic Utility of Cell-Free DNA from Ex Vivo Bronchoalveolar Lavage Fluid in Lung Cancer. Cancers 2022, 14, 1764. [Google Scholar] [CrossRef]
  91. Shen, F.; Liang, N.; Fan, Z.; Zhao, M.; Kang, J.; Wang, X.; Hu, Q.; Mu, Y.; Wang, K.; Yuan, M.; et al. Genomic Alterations Identification and Resistance Mechanisms Exploration of NSCLC With Central Nervous System Metastases Using Liquid Biopsy of Cerebrospinal Fluid: A Real-World Study. Front. Oncol. 2022, 12, 889591. [Google Scholar] [CrossRef] [PubMed]
  92. Kim, C.; Xi, L.; Cultraro, C.M.; Wei, F.; Jones, G.; Cheng, J.; Shafiei, A.; Pham, T.H.T.; Roper, N.; Akoth, E.; et al. Longitudinal circulating tumor dna analysis in blood and saliva for prediction of response to osimertinib and disease progression in egfr-mutant lung adenocarcinoma. Cancers 2021, 13, 3342. [Google Scholar] [CrossRef] [PubMed]
  93. 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. 2016, 11, 1690–1700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Mateo, J.; Chakravarty, D.; Dienstmann, R.; Jezdic, S.; Gonzalez-Perez, A.; Lopez-Bigas, N.; Ng, C.K.Y.; Bedard, P.L.; Tortora, G.; Douillard, J.Y.; et al. A framework to rank genomic alterations as targets for cancer precision medicine: The ESMO Scale for Clinical Actionability of molecular Targets (ESCAT). Ann. Oncol. 2018, 29, 1895–1902. [Google Scholar] [CrossRef]
  95. Pascual, J.; Attard, G.; Bidard, F.-C.; Curigliano, G.; De Mattos-Arruda, L.; Diehn, M.; Italiano, A.; Lindberg, J.; Merker, J.D.; Montagut, C.; et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: A report from the ESMO Precision Medicine Working Group. Ann. Oncol. 2022, 33, 750–768. [Google Scholar] [CrossRef]
  96. Martin Romano, P.; Mezquita, L.; Lacroix, L.; Rouleau, E.; Varga, A.; Baldini, C.; Postel-Vinay, S.; Friboulet, L.; Geraud, A.; Aldea, M.; et al. 1930O Genomic alterations in solid tumours according to ESMO scale for clinical actionability of molecular targets (ESCAT). Ann. Oncol. 2020, 31, S1092–S1093. [Google Scholar] [CrossRef]
  97. Verdaguer, H.; Saurí, T.; Acosta, D.A.; Guardiola, M.; Sierra, A.; Hernando, J.; Nuciforo, P.; Miquel, J.M.; Molero, C.; Peiró, S.P.; et al. ESMO Scale for Clinical Actionability of Molecular Targets Driving Targeted Treatment in Patients with Cholangiocarcinoma. Clin. Cancer Res. 2022, 28, 1662–1671. [Google Scholar] [CrossRef]
  98. Kalokairinou, L.; Howard, H.C.; Slokenberga, S.; Fisher, E.; Flatscher-Thöni, M.; Hartlev, M.; van Hellemondt, R.; Juškevičius, J.; Kapelenska-Pregowska, J.; Kováč, P.; et al. Legislation of direct-to-consumer genetic testing in Europe: A fragmented regulatory landscape. J. Community Genet. 2018, 9, 117–132. [Google Scholar] [CrossRef] [Green Version]
  99. Horgan, D.; Curigliano, G.; Rieß, O.; Hofman, P.; Büttner, R.; Conte, P.; Cufer, T.; Gallagher, W.M.; Georges, N.; Kerr, K.; et al. Identifying the Steps Required to Effectively Implement Next-Generation Sequencing in Oncology at a National Level in Europe. J. Pers. Med. 2022, 12, 72. [Google Scholar] [CrossRef]
  100. Steuten, L.; Goulart, B.; Meropol, N.J.; Pritchard, D.; Ramsey, S.D. Cost Effectiveness of Multigene Panel Sequencing for Patients With Advanced Non–Small-Cell Lung Cancer. JCO Clin. Cancer Inform. 2019, 3, 1–10. [Google Scholar] [CrossRef]
  101. Schluckebier, L.; Caetano, R.; Garay, O.U.; Montenegro, G.T.; Custodio, M.; Aran, V.; Gil Ferreira, C. Cost-effectiveness analysis comparing companion diagnostic tests for EGFR, ALK, and ROS1 versus next-generation sequencing (NGS) in advanced adenocarcinoma lung cancer patients. BMC Cancer 2020, 20, 875. [Google Scholar] [CrossRef]
  102. Pennell, N.A.; Mutebi, A.; Zhou, Z.-Y.; Ricculli, M.L.; Tang, W.; Wang, H.; Guerin, A.; Arnhart, T.; Dalal, A.; Sasane, M.; et al. Economic Impact of Next-Generation Sequencing Versus Single-Gene Testing to Detect Genomic Alterations in Metastatic Non–Small-Cell Lung Cancer Using a Decision Analytic Model. JCO Precis. Oncol. 2019, 3, 1–9. [Google Scholar] [CrossRef] [PubMed]
  103. Thunnissen, E.; Weynand, B.; Udovicic-Gagula, D.; Brcic, L.; Szolkowska, M.; Hofman, P.; Smojver-Ježek, S.; Anttila, S.; Calabrese, F.; Kern, I.; et al. Lung cancer biomarker testing: Perspective from Europe. Transl. Lung Cancer Res. 2020, 9, 887–897. [Google Scholar] [CrossRef] [PubMed]
  104. Rolfo, C.; Mack, P.; Scagliotti, G.V.; Aggarwal, C.; Arcila, M.E.; Barlesi, F.; Bivona, T.; Diehn, M.; Dive, C.; Dziadziuszko, R.; et al. Liquid Biopsy for Advanced NSCLC: A Consensus Statement From the International Association for the Study of Lung Cancer. J. Thorac. Oncol. 2021, 16, 1647–1662. [Google Scholar] [CrossRef] [PubMed]
  105. Rijavec, E.; Coco, S.; Genova, C.; Rossi, G.; Longo, L.; Grossi, F. Liquid biopsy in non-small cell lung cancer: Highlights and challenges. Cancers 2020, 12, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Li, W.; Liu, J.-B.; Hou, L.K.; Yu, F.; Zhang, J.; Wu, W.; Tang, X.M.; Sun, F.; Lu, H.M.; Deng, J.; et al. Liquid biopsy in lung cancer: Significance in diagnostics, prediction, and treatment monitoring. Mol. Cancer 2022, 21, 25. [Google Scholar] [CrossRef]
  107. Bonanno, L.; Dal Maso, A.; Pavan, A.; Zulato, E.; Calvetti, L.; Pasello, G.; Guarneri, V.; Conte, P.F.; Indraccolo, S. Liquid biopsy and non-small cell lung cancer: Are we looking at the tip of the iceberg? Br. J. Cancer 2022, 127, 383–393. [Google Scholar] [CrossRef]
  108. Guibert, N.; Pradines, A.; Mazieres, J.; Favre, G. Current and future applications of liquid biopsy in nonsmall cell lung cancer from early to advanced stages. Eur. Respir. Rev. 2020, 29, 190052. [Google Scholar] [CrossRef] [Green Version]
  109. Choudhury, Y.; Tan, M.H.; Shi, J.L.; Tee, A.; Ngeow, K.C.; Poh, J.; Goh, R.R.; Mong, J. Complementing Tissue Testing With Plasma Mutation Profiling Improves Therapeutic Decision-Making for Patients With Lung Cancer. Front. Med. 2022, 9, 758464. [Google Scholar] [CrossRef]
  110. Mileham, K.F.; Schenkel, C.; Bruinooge, S.S.; Freeman-Daily, J.; Basu Roy, U.; Moore, A.; Smith, R.A.; Garrett-Mayer, E.; Rosenthal, L.; Garon, E.B.; et al. Defining comprehensive biomarker-related testing and treatment practices for advanced non-small-cell lung cancer: Results of a survey of U.S. oncologists. Cancer Med. 2022, 11, 530–538. [Google Scholar] [CrossRef]
  111. Ilié, M.; Hofman, V.; Bontoux, C.; Heeke, S.; Lespinet-Fabre, V.; Bordone, O.; Lassalle, S.; Lalvée, S.; Tanga, V.; Allegra, 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 2022, 14, 2258. [Google Scholar] [CrossRef] [PubMed]
  112. Momeni-Boroujeni, A.; Salazar, P.; Zheng, T.; Mensah, N.; Rijo, I.; Dogan, S.; Yao, J.Y.; Moung, C.; Vanderbilt, C.; Benhamida, J.; et al. Rapid EGFR Mutation Detection Using the Idylla Platform: Single-Institution Experience of 1200 Cases Analyzed by an In-House Developed Pipeline and Comparison with Concurrent Next-Generation Sequencing Results. J. Mol. Diagn. 2021, 23, 310–322. [Google Scholar] [CrossRef] [PubMed]
  113. Thompson, J.C.; Yee, S.S.; Troxel, A.B.; Savitch, S.L.; Fan, R.; Balli, D.; Lieberman, D.B.; Morrissette, J.D.; Evans, T.L.; Bauml, J.; et al. Detection of therapeutically targetable driver and resistance mutations in lung cancer patients by next-generation sequencing of cell-free circulating tumor DNA. Clin. Cancer Res. 2016, 22, 5772–5782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Cohen, D.; Hondelink, L.M.; Solleveld-Westerink, N.; Uljee, S.M.; Ruano, D.; Cleton-Jansen, A.M.; von der Thüsen, J.H.; Ramai, S.R.S.; Postmus, P.E.; Graadt van Roggen, J.F.; et al. Optimizing Mutation and Fusion Detection in NSCLC by Sequential DNA and RNA Sequencing. J. Thorac. Oncol. 2020, 15, 1000–1014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Nong, L.; Zhang, Z.; Xiong, Y.; Zheng, Y.; Li, X.; Li, D.; He, Q.; Li, T. Comparison of next-generation sequencing and immunohistochemistry analysis for targeted therapy-related genomic status in lung cancer patients. J. Thorac. Dis. 2019, 11, 4992–5003. [Google Scholar] [CrossRef]
  116. Durães, C.; Gomes, C.P.; Costa, J.L. Demystifying the Discussion of Sequencing Panel Size in Oncology Genetic Testing. Eur. Med. J. 2022, 7, 68–77. [Google Scholar]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pujol, N.; Heeke, S.; Bontoux, C.; Boutros, J.; Ilié, M.; Hofman, V.; Marquette, C.-H.; Hofman, P.; Benzaquen, J. Molecular Profiling in Non-Squamous Non-Small Cell Lung Carcinoma: Towards a Switch to Next-Generation Sequencing Reflex Testing. J. Pers. Med. 2022, 12, 1684. https://doi.org/10.3390/jpm12101684

AMA Style

Pujol N, Heeke S, Bontoux C, Boutros J, Ilié M, Hofman V, Marquette C-H, Hofman P, Benzaquen J. Molecular Profiling in Non-Squamous Non-Small Cell Lung Carcinoma: Towards a Switch to Next-Generation Sequencing Reflex Testing. Journal of Personalized Medicine. 2022; 12(10):1684. https://doi.org/10.3390/jpm12101684

Chicago/Turabian Style

Pujol, Nina, Simon Heeke, Christophe Bontoux, Jacques Boutros, Marius Ilié, Véronique Hofman, Charles-Hugo Marquette, Paul Hofman, and Jonathan Benzaquen. 2022. "Molecular Profiling in Non-Squamous Non-Small Cell Lung Carcinoma: Towards a Switch to Next-Generation Sequencing Reflex Testing" Journal of Personalized Medicine 12, no. 10: 1684. https://doi.org/10.3390/jpm12101684

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