We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Diagnostic and economic value of biomarker testing for targetable mutations in non-small-cell lung cancer: a literature review

    Ying Zheng

    US Health Economics and Outcomes Research, EMD Serono, Inc., An affiliate of Merck KGaA, Rockland, MA 02370, USA

    ,
    Helene Vioix

    Global Evidence & Value Development, Merck Healthcare KGaA, Darmstadt, Germany

    ,
    Frank X Liu

    *Author for correspondence:

    E-mail Address: frank.liu@emdserono.com

    US Health Economics and Outcomes Research, EMD Serono, Inc., An affiliate of Merck KGaA, Rockland, MA 02370, USA

    ,
    Barinder Singh

    HEOR, Parexel, Access Consulting, Mohali, Punjab, India

    ,
    Sakshi Sharma

    HEOR, Parexel, Access Consulting, Mohali, Punjab, India

    &
    Deepti Sharda

    HEOR, Parexel, Access Consulting, Mohali, Punjab, India

    Published Online:https://doi.org/10.2217/fon-2021-1040

    Abstract

    We aimed to assess the diagnostic and economic value of next-generation sequencing (NGS) versus single-gene testing, and of liquid biopsy (LBx) versus tissue biopsy (TBx) in non-small-cell lung cancer biomarker testing through literature review. Embase and MEDLINE were searched to identify relevant studies (n = 43) from 2015 to 2020 in adults with advanced non-small-cell lung cancer. For NGS versus single-gene testing, concordance was 70–99% and sensitivity was 86–100%. For LBx versus TBx, specificity was 43–100% and sensitivity was ≥60%. Turnaround times were longer for NGS versus single-gene testing (but not vs sequential testing) and faster for LBx versus TBx. NGS was cost-effective, and LBx reduced US per-patient costs. NGS versus single-gene testing and LBx versus TBx were concordant. NGS and LBx may be cost-effective for initial screening.

    Plain language summary

    Patients with lung cancer with specific genetic mutations can benefit from medications that are specific to those mutations, known as targetable mutations. There are many methods to test for specific genetic mutations in patients with lung cancer. To detect genetic mutations, doctors can test the blood or urine, or they can test biopsy tissue; a small piece of the tumor removed from the lung. These tests can either look for mutations in one specific gene at a time, or they can use technology that reads the entire DNA sequence to observe multiple genes at once. In this review, we examined scientific reports to answer important questions about using genetic testing to find targetable mutations in patients with lung cancer. How accurate are different genetic tests? How fast can doctors get results from different genetic tests? How much do different genetic tests cost? We found that reading the entire DNA sequence was as accurate as testing one specific gene. Reading the entire DNA sequence takes more time than testing one specific gene, but it might reduce overall costs. Testing blood or urine was not as accurate as testing tissue, but it took less time for doctors to receive genetic test results and reduced costs.

    Lung cancer is the most common cancer worldwide and is the leading cause of cancer death for men and the second leading cause for women [1]. Non-small-cell lung cancer (NSCLC) affects approximately 85% of all patients with lung cancer [2]. These patients typically have a poor prognosis, with 5-year survival rates of 24% for all NSCLC patients and of only 6% among those with metastatic disease [3].

    Targeted therapy is an important treatment option for patients with NSCLC who have driver mutation-positive tumors. Current treatment guidelines recommend tyrosine kinase inhibitor therapy for patients with actionable driver mutations, including EGFR, BRAF and RAS mutations, ALK, ROS1, RET or NTRK fusions and MET tyrosine kinase abnormalities (i.e., high-level MET amplification and MET exon 14 skipping mutation) [4,5]. Genetic testing is thus required, either to guide appropriate selection of available therapies or to assess patient suitability for a clinical trial of a new therapy [6]. However, with each biomarker testing procedure performed using a biopsy from an individual patient, the amount of tissue available for further biomarker testing is reduced. As the number of potential genetic targets increases, prioritization of limited tissue is essential [7].

    Strategies to ensure maximum testing yield from available tissue and to limit invasive procedures for the patient include multigene sequencing with next-generation sequencing (NGS) and liquid biopsy (LBx) techniques, respectively. NGS is tissue-sparing compared with conventional sequencing because it allows identification of a panel of genes using a single sample, but it has not replaced conventional sequencing despite progressive cost reduction [8]. LBx techniques can be used to test for circulating tumor DNA (ctDNA), circulating tumor cells, exosomes, platelets and microRNAs [9]. The role of these biomarker techniques in NSCLC, including their diagnostic and economic value, has not been clearly defined.

    The overall objectives of this literature review were to assess the diagnostic evidence and economic impact of first, NGS versus single-gene testing and second, LBx compared with standard tissue biopsy (TBx) in adults with unresectable, advanced or metastatic NSCLC.

    Methods

    A literature review was conducted to identify publications related to NGS and LBx in adult patients (aged ≥18 years) with advanced, recurrent and/or metastatic NSCLC. The review was guided by the population, intervention, comparison, outcome, study type (PICOS) framework [10].

    Data sources

    Embase® and MEDLINE® were searched for records from 2015 to 2020 using targeted keyword searches (Supplementary Table 1). To supplement the database searches, conference abstracts (2017–2020) from American Society of Clinical Oncology (ASCO), European Society for Medical Oncology (ESMO), International Society for Pharmacoeconomics and Outcomes Research (ISPOR), American Association for Cancer Research (AACR), World Conferences on Lung Cancer (WCLC), International Association for the Study of Lung Cancer (IASLC) North American Congress on Lung Cancer (NACLC), IASLC Targeted Therapies and International Lung Cancer Congress were searched manually. Bibliographies of review articles were also searched manually, and the Tufts Cost–Effectiveness Analysis (CEA) Registry was searched for economic evidence [11,12].

    Study selection

    Eligible studies were controlled clinical trials (including both randomized and non-randomized), single-arm studies, observational studies (excluding case reports/series), systematic reviews, surveys and economic evaluations published in English. Studies had to include ≥100 adult patients of any race or sex with unresectable, advanced and/or metastatic NSCLC, regardless of histology, for any biomarker or mutation. Any study meeting the above criteria and reporting biomarker types, molecular testing methods and application, and their associated challenges and advantages in NSCLC, were considered.

    Review procedure

    All articles were downloaded into a systematic review database. First screening (titles and abstracts) and second screening (full-text papers) were conducted by a single reviewer, followed by a quality check from a second independent reviewer (Supplementary Figure 1). Data were extracted by a single reviewer and verified by an independent reviewer. Where more than one publication was identified describing a single trial, the data were compiled into a single entry in the data extraction table to avoid double counting of patients and studies.

    Results

    Literature review

    A total of 2602 records were identified, from which 375 full-text studies were screened (Figure 1). Of these 375 publications, the review included 43 relevant publications describing 42 studies. Fourteen studies examined NGS versus standard molecular testing techniques in NSCLC. Twenty-eight studies reported evidence on LBx versus TBx (29 publications describing 28 studies).

    Figure 1. Flow of studies included in the literature review.

    LBx: Liquid biopsy; NGS: Next-generation sequencing; NSCLC: Non-small-cell lung cancer.

    NGS versus standard molecular testing

    Six studies, two of them USA-based, reported diagnostic outcomes [13–18], while nine (five USA-based) reported economic evidence [17,19–27]. Of these, one study conducted in Singapore by Tan et al. reported both diagnostic and economic outcomes [17].

    Diagnostic evidence

    All studies reporting diagnostic outcomes were observational; two were retrospective and four were prospective. Among studies reporting demographic data (n = 4 studies with a range of 174–533 patients), the median age of patients ranged from 67 to 70 years, and 38–62% of patients were male [13,15–17]. Two studies from Europe [15,16] and one from Asia [17] reported concordance rates ranging from 70% to 99% for NGS versus standard molecular testing across clinically actionable mutations, including EGFR and ALK fusion (Table 1). Sensitivity for NGS versus standard molecular testing ranged from 86% to 100% for clinically actionable mutations and was reported as 55.6% in one study based on acquired resistant mutations [13–15,17]. Of the two USA-based studies included, one (Dagogo-Jack et al.) reported sensitivity and specificity of 98% and 100%, respectively, for a rapid EGFR PCR assay using NGS as the reference assay [13]. Other studies included did not report specificity data. The second USA-based study (Yu et al.) reported higher rates of test initiation and completion using less tissue compared with single-gene testing for four or more biomarkers [18]. Based on the two USA-based studies, median turnaround times were longer using NGS than with single-gene testing (14–17 vs 7–11 days), but this was not the case if multiple sequential single-gene tests were required (e.g., three single-gene tests ordered in sequence would require ∼21–∼33 days total turnaround time) [13,18].

    Table 1. Diagnostic accuracy of next-generation sequencing versus single-gene testing for clinically actionable mutations in patients with non-small-cell lung cancer.
    Study (year)CountryStudy designTest and comparatorBiomarker testsSample sizeSensitivity %§,Concordance %Ref.
    Dagogo Jack (2018)USAProspective observationalEGFR-specific PCR assay vs NGSEGFRL858R and exon 19 deletionNR98NR[13]
    Garcia (2018)FranceProspective observationalNGS vs OncoBEAMEGFRT790M19655.6#NR[14]
    Fernandes (2019)PortugalProspective observationalNGS vs Sanger sequencingEGFR11710099.1[15]
    Lindquist (2017)SwedenProspective observationalNGS vs single-gene testingEGFR, KRAS, NRAS and BRAF81NR96[16]
    Tan (2020)SingaporeRetrospective observationalNGS vs standard molecular testingEGFR exon 19 deletionNR93.970[17]
    ALK fusionNR85.794

    †One USA-based study (Yu, 2017) identified in the literature review did not report sensitivity or concordance for NGS vs conventional molecular testing [18].

    ‡ctDNA was used as a reference standard.

    §Specificity for NGS vs conventional molecular testing was reported in only one study (Dagogo-Jack, 2018), which reported a specificity of 100%.

    EGFR+ cases were assumed to be true positives. One patient was classified as EGFR+ by NGS but was unclassified by Sanger sequencing.

    #The detection rates for EGFRT790M were 10.2% and 18.3% by NGS and OncoBEAM, respectively. Sensitivity of NGS assumed all T790M+ cases were true positives, and thus describes the detection of an acquired resistance mutation.

    NGS: Next-generation sequencing; NR: Not reported.

    Economic impact

    Among the ten economic studies, six assessed cost–effectiveness [17,19,23–26], one reported costs [27], two assessed budget impacts [20,21] and one reported a cost–consequence analysis (Table 2) [22].

    Table 2. Details of studies reporting the economic impact of next-generation sequencing (base-case analysis) in advanced non-small-cell lung cancer.
    Study (year)CountryStudy designTest vs comparatorBiomarker testsCost yearStudy resultsRef.
    Steuten (2019)USACost–effectiveness analysisMGPS vs SMGTEGFR, ALK, BRAF, RET, ROS1, HER2, MET2017MGPS vs SMGT
    ICER:
    $148,478 per LYG
    LYs:
    1.2 vs 1.14
    Incremental cost:
    $67,110 vs $58,297
    [19]
    Pennell (2019)USABudget impact analysisNGS vs sequential testing, exclusionary testing and hotspot panel testingPD-1/PD-L1, EGFR, ALK, ROS1, KRAS, MET, RET, NTRK1, BRAF2017CMS (2066 patients)
    Cost savings for NGS vs:
    • Exclusionary testing: US$1,393,678
    • Sequential testing: US$1,530,869
    • Hotspot panel testing: US$2,140,795
    Commercially insured (156 patients)
    Cost savings for NGS vs:
    • Exclusionary testing: US$3809
    • Sequential testing: US$127,402
    • Hotspot panel testing: US$250,842
    [20]
    Yu (2018)USABudget impact analysisNGS vs single-gene testingEGFR, ALK, ROS1, BRAF, MET, HER2, RET2016Budget increase for NGS vs single-gene testing in 1-million-member plan model:
    • Over 5 years: $432,554
    • PMPM: US$0.0072
    [21]
    Dalal (2017)USACost–consequence analysisSequential testing
    Exclusionary mutation (i.e., KRAS) testing followed by sequential BRAF testing
    Upfront NGS, including BRAF testing
    BRAFV600ENRCMS reimbursement:
    • NGS cost: $623
    • Cost saving: $980 vs sequential and mutations panel
    • Cost saving: $1238 vs exclusionary strategy
    Sensitivity analysis (based on amounts reimbursed by third-party payers [commercial claims data]):
    • NGS ($2860) remained the least expensive option by $894–1044
    [22]
    Lopes (2020)USACost–effectiveness analysisctDNA NGS vs SoC (no additional genomic testing)NSCLC with incomplete tissue genotypingNRConservative drug costs:
    • ctDNA NGS increased the number of patients who received guideline-adherent treatment decisions
    • Clinical outcomes improved and were accompanied by meaningful cost savings
    • Per-patient cost savings were $1943
    Patients receiving NGS had, on average, an increased:
    • RR of 7.57%
    • PFS of 0.75 month
    • OS of 1.24 months
    [23]
    Ho (2019)BrazilCost–effectiveness analysesNGS panel of ctDNA vs ctDNA EGFR testingEGFR, ALK, ROS1, BRAFNRAnnual cost savings for NGS:
    1. EGFR and ALK: -4138.67 BRL$
    2. EGFR, ALK and ROS:
    -6245.10 BRL$
    3. EGFR, ALK, ROS-1 and BRAF:
    -5720.48 BRL$
    ICERs§:
    1: -15,595.77 (BRL$/PFS)
    2: -21,399.29 (BRL$/PFS)
    3: -18,006.42 (BRL$/PFS)
    [24]
    Ferreira (2018)BrazilCost–effectiveness analysesNGS vs sequential testing (RT-PCR and ddPCR)EGFRT790MNRICER (per positive T790M detected):
    Plasma PCR then tissue NGS if plasma negative vs plasma NGS + tissue NGS: US $21,193.66
    [25]
    Schluckebier (2017)BrazilCost–effectiveness analysesNGS vs diagnostic tests (RT-PCR and FISH)EGFR, ALK, ROS12016ICER (per correct case detected):
    NGS vs sequential: US $3381.82
    [26]
    Simarro (2019)SpainCost analysesNGS with Oncomine solid tumor (ThermoFisher) vs conventional methodsEGFR, ALK, ROS1NRCost for NGS:
    Total: €3369.84
    Per sample: €421.23
    Cost for conventional:
    Total: €2941.27
    Per sample: €367.66
    [27]
    Tan (2020)SingaporeCost–effectiveness analysesTargeted NGS panels (sequential singleplex, sequential multiplex and NGS-only testing) vs traditional assay (SoC)EGFR, ALK, ROS1, MET, RET2018Cost per patient:
    SoC only: SGD 2224.6
    Sequential (singleplex): SGD$ 1469.2
    NGS only: SGD 1579.4
    Sequential (multiplex): SGD$ 2622.7
    ICER (per percent increase in patients on targeted therapy):
    Sequential (singleplex):
    Dominant NGS only: SGD$ 110
    Sequential (multiplex): SGD$ 261
    [17]

    †Budget impact was calculated for a hypothetical 1,000,000-member health plan, with an expected 2066 Medicare-insured and 156 commercially insured testing-eligible patients with metastatic NSCLC.

    ‡Budget impact was calculated for a hypothetical 1,000,000-member health plan, with an expected 316 testing-eligible patients with advanced NSCLC.

    §ICERs reported in congress abstract. Units of PFS were not explicitly reported in the abstract. The horizon time for the model was 1 year.

    CMS: Centers for Medicare & Medicaid Services; ctDNA: Circulating tumor DNA; ddPCR: Droplet digital polymerase chain reaction; FISH: Fluorescence in situ hybridization; ICER: Incremental cost–effectiveness ratio; LY: Life-year; LYG: Life-year gained; MGPS: Multigene panel sequencing; NGS: Next-generation sequencing; NR: Not reported; NSCLC: Non-small-cell lung cancer; OS: Overall survival; PFS: Progression-free survival; PMPM: Per member per month; RR: Response rate; SMGT: Single-marker genetic testing; SoC: Standard of care.

    Among the five USA-based studies that reported economic evidence, two found tumor tissue NGS versus sequential exclusionary testing or hotspot panel testing (excluding treatment costs) to be cost saving [20,22], with one (Dalal et al.) reporting the shortest wait time, earlier initiation of effective targeted therapy and lower costs in patients receiving upfront NGS [22]. Two studies found tumor tissue NGS versus single-gene testing (including treatment costs) to be associated with increased budget, although that was balanced by evidence of cost–effectiveness for NGS testing [19,21]. In the study conducted by Yu et al. the budget increase over single-gene testing was minimal, at US$0.0072 per member per month, but NGS was expected to identify more patients with activating mutations, enabling better selection for targeted therapy [21]. Another study, by Steuten et al., found ctDNA NGS versus standard of care molecular testing (no additional genomic testing) to be cost-effective, with 8% more patients identified with targetable mutations and expected survival increasing by 0.06 years versus single-gene testing [19].

    Studies conducted in other regions (e.g., Europe, Asia) were aligned with USA-based studies regarding economic and diagnostic outcomes. In Spain, Simarro et al. reported that NGS implementation was feasible and could be done at reasonable cost [27]. In Singapore, Tan et al. found that routine upfront NGS was cost-effective compared with sequential sequencing [17].

    LBx versus TBx

    Of the 25 studies included that compared LBx with TBx (both NGS and single-gene assays) [28–52], four were from the USA [32–35], five were from Europe [36–40], 12 were from Asia [41–52] and four were global [28–31]. The majority (88%) of diagnostic studies were observational. Among studies reporting demographic data (n = 16 studies with a range of 102–1026 patients), the median age of patients ranged from 57 to 70 years (n = 11), and 32–70% of patients were male (n = 16) [32–38,40,45–51].

    Three studies comparing economic outcomes between LBx and TBx were a cost study by Arnaud et al. from the USA [53], a cost–consequence analysis from Italy by Gancitano et al. [54] and a Canadian cost–effectiveness and budget impact analysis by Ontario Health [55].

    Diagnostic evidence

    Measures of diagnostic accuracy, including sensitivity, concordance, positive predictive values (PPVs) and negative predictive values (NPVs), for LBx versus TBx in detecting clinically actionable genes were examined across studies (Table 3).

    Table 3. Diagnostic accuracy of liquid biopsy versus tissue biopsy for clinically actionable mutations in patients with non-small-cell lung cancer.
    Study nameCountryStudy designIntervention/assaySample sizeSpecificity %Sensitivity %Concordance %PPV %NPV %Ref.
    Goldman (2018)Global (USA, Poland, France, Australia)Non-randomized trialTumor (EGFR exon 19 deletions) vs plasma47597.2§84.688.8NRNR[28]
    Tumor (EGFRL858R) vs plasma97.7§8192.7NRNR
    Tumor (EGFRT790M) vs plasma42.5§80.977.0NRNR
    Tu (2019)Global (USA, Australia)Prospective observationalConcordance between plasma and tissue NGS
    (ROS1, RET, BRAF, MET and HER2)
    399NRNR94.7NRNR[29]
    Schwartzberg (2018)Global#Prospective observationalEGFR alterations140 (LBx),
    117 (concurrent TBx)
    100§76.794NRNR[30]
    ALK alterationsNRNR95.7NRNR
    Zhou (2017)Global (Australia, China, Korea)Single-arm trialEGFRT790M (ddPCR)2497356NRNRNR[31]
    EGFRL858R (ddPCR)9962NRNRNR
    EGFR exon 19 deletions (ddPCR)9866NRNRNR
    EGFRT790M (Cobas plasma)2408342NRNRNR
    EGFRL858R (Cobas plasma)10065NRNRNR
    EGFR exon 19 deletions (Cobas plasma)9786NRNRNR
    EGFRT790M (SuperARMS)2497849NRNRNR
    Sacher (2016)USAProspective observationalEGFR exon 19 deletion5010082NR100NR[32]
    EGFRL858R3210074NR100NR
    Li (2019)USAProspective observationalConfirmed mutation status of actionable alternations
    (EGFR, KRAS, ALK, MET, ERBB2, BRAF, ROS1 and RET)
    9110075NRNRNR[33]
    Thompson (2016)USAProspective observationalEGFR variants;
    ≤2 weeks between LBx and TBx
    50NRNR100NRNR[34]
    EGFR variants;
    ≤2 months between LBx and TBx
    NRNR92NRNR
    EGFR variants;
    ≤6 weeks between LBx and TBx
    NRNR94NRNR
    EGFR variants;
    >6 weeks between LBx and TBx
    NRNR60NRNR
    Leighl (2019)USAProspective observationalcfDNA vs tissue;
    EGFR exon 19 deletion
    28210081.898.210098[35]
    Remon (2019)FranceProspective observationalCore gene variants (36-gene panel, including EGFR, KRAS, ALK, MET, ERBB2, BRAF and ROS1)35 (tissue and liquid), 8 (tissue), 10 (liquid)96.781.495.277.897.6[36]
    Papadopoulou (2019)GreeceProspective observationalClinically significant genes (23-gene panel, including EGFR, KRAS, ALK, MET, ERBB2, BRAF and MET)12188.2484.2186.1188.89NR[37]
    Minari (2020)ItalyRetrospective observationalEGFRT790M52NRNR46.1NRNR[38]
    Spasic (2019)SerbiaProspective observationalEGFR mutation testing104NRNR93NRNR[39]
    Mayo de Las Casas (2017)SpainProspective observationalSerum cfDNA vs tissue (EGFR)2681005790NRNR[40]
    Plasma cfDNA vs tissue (EGFR)1007090NRNR
    Mok (2015)ChinaRCTOverall EGFR mutation positive969675889485[41]
    Yang (2018)ChinaProspective observationalctDNA samples (EGFR)114775970NRNR[42]
    Ma (2016)ChinaProspective observationalEGFR mutation1299760NR9378[34]
    Zhang (2017)ChinaProspective observationalOverall (EGFR)10610094.8799.06NRNR[43]
    Zhang (2018)ChinaProspective observationalEGFRL588R (biopsy vs plasma DNA)62NRNR82NRNR[45]
    EGFR exon 19 deletions (biopsy vs plasma DNA)68NRNR75NRNR
    EGFRT790M (biopsy vs plasma DNA)3NRNR100NRNR
    All EGFR mutations (biopsy vs plasma DNA)NRNRNRNR10051.7
    He (2017)ChinaProspective observationalEGFRL858R58NRNR83NRNR[46]
    EGFR exon 19 deletion48NRNR72NRNR
    EGFR+106NRNR78NRNR
    EGFRT790M26NRNR100NRNR
    Veldore (2018)IndiaProspective observationalOverall EGFR mutated4510091.1196.9710095.6[47]
    Wulandari (2020)IndonesiaProspective observationalEGFR ctDNA12490.948.370.9782.3566.67[48]
    Takahama (2016)JapanProspective observationalTKI-sensitizing mutations (EGFR)4187.575.878NRNR[49]
    Itotani (2019)JapanProspective observationalcfDNA gene mutation (EGFR, KRAS, BRAF, ERBB2, MET, ALK, RET and ROS1)121NR75§NRNRNR[50]
    Hur (2019)South KoreaProspective observationalBALF EV-based EGFR genotyping, overall13786.775.9NR78.884.7[51]

    †Studies with available specificity, sensitivity, concordance, PPV and/or NPV data are shown.

    ‡Summary outcomes and/or outcomes associated with key clinically actionable mutations are shown. One study from China (Cao, 2020) identified in the literature review did not report quantitative sensitivity or specificity data [44].

    §Reported as negative percent agreement.

    ¶Reported as positive percent agreement.

    #The geographic location of the patient population was not clear; the study was categorized as global based on the authors having affiliations with institutions in multiple countries.

    ARMS: Amplification-refractory mutation system; BALF: Bronchoalveolar lavage fluid; cfDNA: Cell-free DNA; ctDNA: Circulating tumor DNA; EV: Extravesicular; FFPE: Formalin-fixed paraffin-embedded; LBx: Liquid biopsy; NGS: Next-generation sequencing; NPV: Negative predictive value; NR: Not reported; NSCLC: Non-small-cell lung cancer; PPV: Positive predictive value; TBx: Tissue biopsy; TKI: Tyrosine kinase inhibitor.

    The range of specificity values reported across mutations and across studies was 42.5–100%, with 11 of 17 studies reporting a specificity ≥90% for all tests [28,30–33,35–37,40–43,47–49,51,52]. Specificity values <90% for EGFRT790M were observed in two of two studies reporting specificity specifically for this variant [28,31]. Sensitivity values varied widely across mutations and across studies (range: 0–100%), and 14 of 18 studies reported sensitivity of ≥60% for all tests [28,30–33,35–37,40–43,47–52].

    Concordance rates using LBx to detect targetable mutations were reported in 18 studies [28–30,34–43,45–49]. Concordance rates for all mutations tested were ≥70% in 16 studies and >90% in seven [28–30,35–37,39–43,45–49]. In ten studies reporting PPV for LBx versus TBx, the range of PPVs for clinically actionable mutations was 77.8–100% [32,34–37,41,45,47,48,51].

    In two USA-based studies, 100% PPV was reported for all clinically actionable mutations except EGFRT790M (79%) [32,35]. In seven studies that reported NPV of LBx versus TBx, the range of NPVs for clinically actionable mutations was 52–98% [34,35,41,45,47,48,51]. Faster turnaround times were reported for LBx versus TBx (range across three studies: 2–10 vs 5–25 days) [29,32,35].

    Economic impact

    Three economic studies found that incorporating LBx into the treatment pathway was associated with lower testing cost per patient (Table 4) [53–55].

    Table 4. Details of studies reporting the economic evidence for liquid biopsy in advanced non-small-cell lung cancer.
    Study (year)CountryStudy designTest and comparatorMutationsCost yearStudy resultsRef.
    Arnaud (2016)USACost analyses• Tissue-based biopsy: CT-guided fine-needle aspiration
    • Tissue-based biopsy: electromagnetic navigational bronchoscopy)
    • Blood-based GeneStrat test
    EGFR sensitizing and resistance, ALK, KRAS and BRAFNRCT-guided fine-needle aspiration:
    • Procedure Medicare costs: US$3253.52
    • Pathology cost: US$876.57
    • Total cost of biopsy and biomarker testing: US$4130.09
    Electromagnetic navigational bronchoscopy:
    • Procedure Medicare costs: US$7407.05
    • Pathology cost: US$876.57
    • Total cost of biopsy and biomarker testing: US$8283.62
    GeneStrat:
    • Procedure Medicare costs: US$3.00
    • Pathology cost: US$833.45
    • Total cost of biopsy and biomarker testing: US$836.45
    [53]
    Ontario Health (2020)CanadaCost/cost–effectiveness/ budget impact analyses• LBx as a triage test
    • LBx alone
    • TBx
    EGFRT790M2018Estimated costs:
    • LBx: CAN$700
    • TBx: CAN$2500
    Long-term cost of treatment and care:
    • LBx as a triage test was the most effective and most costly strategy, followed by LBx alone
    • ICERs of LBx as a triage test compared with LBx alone and of LBx alone compared with TBx alone were >CAN$100,000 per QALY
    Budget impact of LBx as a triage test:
    • Approximately CAN$60,000 in Year 1 to CAN$3 million in Year 5 in treatment costs
    [55]
    Gancitano (2018)ItalyCost–consequence analyses• Tissue pathway
    • Combined pathway
    • Potential pathway
    EGFRNRCosts per correctly identified case:
    • The potential pathway had the greatest number of correctly identified cases and the lowest average cost per correctly identified case
    • Tissue pathway: €1004
    • Combined pathway: €732
    • Potential pathway: €685
    [54]

    †Combined pathway, first line: TBx, if outcome is unknown proceed with LBx; second line: LBx, if outcome negative proceed with TBx.

    ‡Potential pathway, first line: TBx and LBx for tissue-ineligible patients, if outcome of TBx is unknown proceed with LBx; second line: LBx, if outcome is negative proceed with TBx.

    CT: Computed tomography; ICER: Incremental cost–effectiveness ratio; LBx: Liquid biopsy; NR: Not reported; QALY: Quality adjusted life year; TBx: Tissue biopsy.

    A USA-based study conducted from a Medicare reimbursement perspective compared the clinical costs and complications of LBx with TBx (computed tomography [CT]-guided biopsy and navigational bronchoscopy) in a hypothetical biomarker case that tested for EGFR, KRAS, ALK and BRAF mutations [53]. The authors found that LBx was significantly less expensive, having a lower total cost of biopsy and biomarker testing (US$836) compared with CT-guided biopsy (US$4130) and navigational bronchoscopy (US$8284). Overall, LBx was associated with cost reductions of ≥US$3200 per patient. LBx also reduced time from screening to treatment and was associated with fewer complications for patients compared with CT-guided biopsy or navigational bronchoscopy, which was associated with pneumothorax, lung collapse, hemorrhage and respiratory distress in some patients.

    A cost–consequence analysis was performed in a hypothetical cohort with EGFR mutations in Italy [54]. This model compared three different diagnostic pathways: TBx (for first- and second-line treatment), combined (TBx for first line and LBx for second line if the outcome was unknown) and potential (TBx or LBx for tissue-ineligible patients as first line and LBx as second line). The potential pathway provided the greatest number of correctly identified cases. The average cost per correctly identified case in the potential pathway (€685) was lower than for the combined pathway (€732) or the TBx pathway (€1004). Overall, the addition of LBx was associated with a cost reduction of approximately €300 per patient in this setting. These authors concluded that a correct diagnostic pathway is essential to optimize cancer therapies. This analysis also highlights the value of upfront LBx in the diagnostic pathway for both first- and second-line treatment.

    A recent Canadian Health Technology Assessment reported the cost–effectiveness of TBx alone, LBx alone or LBx as a triage test in a cohort with EGFRT790M mutations [55]. LBx alone or as a triage test was less costly and more effective (i.e., resulted in fewer tissue biopsies and more correct decisions) than TBx in terms of test-related costs and effects only. With regard to lifetime costs, LBx as a triage test was the most effective and produced the most life-years and quality-adjusted life-years, but it was the costliest of the three options. However, this was largely driven by higher long-term treatment cost as more patients were correctly identified to receive targeted therapy [55].

    Discussion

    This targeted literature search conducted from January 2015 to March 2020 identified studies reporting diagnostic and economic aspects of NGS versus single-gene testing and LBx versus TBx. The studies were global, with most conducted in North America (including the USA and Canada), followed by Europe and Asia. Overall, the studies suggest that NGS is highly concordant with conventional molecular testing in patients with NSCLC. Two studies reporting <90% concordance (55.6% [14] and 82% [37]) between NGS and single-gene tests used specialized single-gene tests for EGFR testing, and the discordances (missed by NGS) were mainly in the low mutant allelic fractions. While less sensitive than conventional methods, NGS resulted in broader genomic coverage, which may reveal diverse mechanisms of resistance among patients with advanced NSCLC.

    NGS can also measure tumor mutational burden (TMB), an emerging biomarker to select patients for immunotherapy, and will likely need to be used in conjunction with PD-L1 immunohistochemistry [5]. The sequencing of targeted therapies and immunotherapies as recommended in treatment guidelines will continue to evolve as the treatment landscape changes, but clinicians may make use of the wider range of genetic information available with NGS to facilitate the selection of the right therapy for an individual patient. The potential role of additional genetic screening – via NGS or single-gene testing – in a patient whose disease develops resistance to initial therapy needs to be clarified in future studies.

    In terms of cost and cost–effectiveness, NGS leads to a greater proportion of patients assigned to targeted therapy and increased life-years gained while being cost neutral or cost saving. NGS was generally found to be cost-effective at typical thresholds. With current treatment guidelines recommending targeted therapies for eight specific genetic biomarkers plus additional recommendations based on PD-L1 and TMB status [5], the additive costs of multiple single-gene tests should be considered. This review provides indirect evidence on the question of how the costs of NGS compare with those of multiple single-gene tests, and future studies on the costs of biomarker testing in NSCLC may provide clearer evidence.

    In all studies, concordance between LBx and TBx for all mutations tested was generally high (≥70%), with six studies reporting >90% concordance. LBx exhibits high specificity to detect targetable mutations in patients with NSCLC, but it may have lower sensitivity than TBx. Overall, the LBx studies reported shorter turnaround times from blood sample collection to report delivery compared with TBx. The faster turnaround time and high PPVs of LBx enable faster treatment decisions in patients with NSCLC who have targetable mutations. LBx may also provide additional genetic material for subsequent testing at the time when a patient develops resistance to initial therapy, limiting invasive procedures and potentially improving patient experience and outcomes. Upfront cost savings may be achieved using LBx as an initial screening method in complement to TBx, although identification of a greater number of cases may lead to increased treatment costs.

    Several limitations to this review should be noted. This review includes only English-language papers published as journal articles in the last 5 years. While unpublished or non-English-language studies may contain valid results that may conflict with the conclusions of this review, the broad search strategy used here and the large number of citations screened make this unlikely. The review excludes diagnostic studies with small sample size (≤100 patients); however, these are likely to be exploratory studies that could introduce bias. Additional parameters that inform the quality of biomarker testing (e.g., test failure rate) were not included in the data extraction. Only three economic evaluations were found comparing LBx and TBx, suggesting that data may be limited in this area.

    Future perspective

    The findings of our review may have implications regarding recommendations for the timing of LBx and NGS in future NSCLC treatment guidelines. Among studies included in this review, the results of NGS and conventional single-gene testing were highly concordant. Comparisons of TBx and LBx indicated that these techniques also generally have high concordance. NGS and LBx separately showed benefits in terms of correctly identifying more patients for targeted therapy, enabling faster turnaround and quicker treatment decisions. In terms of cost and cost–effectiveness, these methods were associated with reductions in short-term treatment-related costs. In the long term, increased use of NGS could result in a minimal increase of budget largely driven by more patients receiving targeted treatment.

    Executive summary

    Concordance & turnaround time for next-generation sequencing versus standard molecular testing

    • High concordance was found between next-generation sequencing (NGS) and single-gene testing methods.

    • Turnaround times were longer for NGS versus single-gene testing, but not longer versus sequential testing.

    Economic impact of NGS versus standard molecular testing

    • NGS was found to be cost-effective in the USA in identifying patients with non-small-cell lung cancer with targetable mutations.

    Concordance & turnaround time for liquid biopsy versus tissue biopsy

    • Liquid biopsy has high specificity but lower sensitivity for targetable mutations than tissue biopsy.

    • Turnaround times were faster with liquid biopsy versus tissue biopsy.

    Economic impact of liquid biopsy versus tissue biopsy

    • Liquid biopsy was found to reduce per-patient costs in the USA and may be cost-effective as an initial screening method.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fon-2021-1040

    Author contributions

    Y Zheng, H Vioix and F Liu contributed to conceptualization, methodology, formal analysis and writing of this manuscript. B Singh, S Sharma and D Sharda contributed to the methodology, investigation, data curation, formal analysis and writing of this manuscript.

    Financial & competing interests disclosure

    This literature review and manuscript were sponsored by EMD Serono Inc., an affiliate Merck KGaA. Y Zheng is a former employee of EMD Serono Inc., Rockland, MA, USA, an affiliate of Merck KGaA. F Liu is an employee of EMD Serono Inc., MA, USA, an affiliate of Merck KGaA. H Vioix is an employee of Merck Healthcare KGaA, Darmstadt, Germany. B Singh, S Sharma and D Sharda are employees of Parexel. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Editorial and writing support was provided by B Ricca of Parexel International and was funded by EMD Serono, Inc., an affiliate of Merck KGaA.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Bade BC, Dela Cruz CS. Lung cancer 2020: epidemiology, etiology, and prevention. Clin. Chest Med. 41(1), 1–24 (2020).
    • 2. Duma N, Santana-Davila R, Molina JR. Non-small cell lung cancer: epidemiology, screening, diagnosis, and treatment. Mayo Clin. Proc. 94(8), 1623–1640 (2019).
    • 3. Cancer.Net. Lung cancer – non-small cell: statistics (2020). www.cancer.net/cancer-types/lung-cancer-non-small-cell/statistics
    • 4. Hanna NH, Schneider BJ, Temin S et al. Therapy for stage IV non-small-cell lung cancer without driver alterations: ASCO and OH (CCO) joint guideline update. J. Clin. Oncol. 38(14), 1608–1632 (2020).
    • 5. Network NCC. NCCN Clinical Practice Guidelines in Oncology: non-small cell lung cancer, Version 5.2021 (2021). www.nccn.org/professionals/physician_gls/pdf/nscl.pdf
    • 6. Lindeman N, Cagle P, Aisner D 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 Association for Molecular Pathology. J. Thorac. Oncol. 13(3), 323–358 (2018).
    • 7. Ruiz-Cordero R, Devine WP. Targeted therapy and checkpoint immunotherapy in lung cancer. Surg. Pathol. Clin. 13(1), 17–33 (2020).
    • 8. Colomer R, Mondejar R, Romero-Laorden N, Alfranca A, Sanchez-Madrid F, Quintela-Fandino M. When should we order a next generation sequencing test in a patient with cancer? EClinicalMedicine 25, 100487 (2020).
    • 9. Saarenheimo J, Eigeliene N, Andersen H, Tiirola M, Jekunen A. The value of liquid biopsies for guiding therapy decisions in non-small cell lung cancer. Front. Oncol. 9, 129 (2019).
    • 10. Higgins J, Thomas J, Chandler J et al. Cochrane Handbook for Systematic Reviews of Interventions. John Wiley & Sons Chichester, UK (2019).
    • 11. Center for Evaluation of Value and Risk in Health. Tufts Medical Center Cost Effectiveness Analysis Registry (2018). https://cevr.tuftsmedicalcenter.org/databases/cea-registry
    • 12. Thorat T, Cangelosi M, Neumann PJ. Skills of the trade: the Tufts cost–effectiveness analysis registry. J. Benefit Cost Anal. 3(1), 1–9 (2012).
    • 13. Dagogo-Jack I, Azzolli CG, Fintelmann F et al. Clinical utility of rapid EGFR genotyping in advanced lung cancer. JCO Precis. Oncol. 2, 1–13 (2018). •• Turnaround times for next-generation sequencing (NGS) versus a rapid single-gene assay for EGFR mutations were compared in this prospective study of 243 US patients with non-small-cell lung cancer (NSCLC).
    • 14. Garcia J, Delherme A, Geigeur F et al. Comparison of OncoBEAM and NGS methods to detect plasma EGFR T790M mutations at progression of NSCLC. Ann. Oncol. 29(Suppl. 8), VIII31–VIII21 (2018).
    • 15. Fernandes MGO, Jacob M, Martins N et al. Targeted gene next-generation sequencing panel in patients with advanced lung adenocarcinoma: paving the way for clinical implementation. Cancers 11(9), 1229 (2019).
    • 16. Lindquist K, Karlsson A, Levéen P et al. Clinical framework for next generation sequencing based analysis of treatment predictive mutations and multiplexed gene fusion detection in non-small cell lung cancer. Oncotarget 8(21), 34796 (2017).
    • 17. Tan A, Lai G, San Tan G et al. Utility of incorporating next-generation sequencing (NGS) in an Asian non-small cell lung cancer (NSCLC) population: incremental yield of actionable alterations and cost–effectiveness analysis. Lung Cancer 139, 207–215 (2020).
    • 18. Yu T, Tradonsky A, Layton A. Practical implications of single-gene versus NGS testing in advanced NSCLC. J. Clin. Oncol. 35(Suppl. 15), e23106 (2017). • This retrospective analysis of data from a large US commercial laboratory examined turnaround times between NGS and single-gene tests.
    • 19. Steuten L, Goulart B, Meropol NJ, Pritchard D, Ramsey SD. Cost effectiveness of multigene panel sequencing for patients with advanced non-small-cell lung cancer. JCO Clin. Cancer Inform. 3, 1–10 (2019).
    • 20. Pennell N, Mutebi A, Zhou Z-Y 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. 3, 1–9 (2019). •• This budget impact analysis found cost savings for NGS over sequential testing, exclusionary testing and hotspot panel testing in US patients with NSCLC.
    • 21. Yu T, Morrison C, Gold E, Tradonsky A, Arnold R. Budget impact of next-generation sequencing for molecular assessment of advanced non-small cell lung cancer. Value Health 21(11), 1278–1285 (2018).
    • 22. Dalal A, Zhou Z, Tang W et al. Cost consequence analysis of different gene mutation testing modalities for BRAF V600E mutation in advanced non-small cell lung cancer using a decision analytic model. Value Health 20(9), A581–A582 (2017). • This cost–consequence analysis showed cost savings for upfront NGS compared with sequential testing to identify BRAFV600E mutation (including sequential testing prior to BRAF testing) in US patients with NSCLC.
    • 23. Lopes G, Liu E, Raymond V, Scott J, Ho V, Gandara D. PPM2 Comprehensive genomic testing in advanced non-small cell lung cancer (NSCLC): a cost–effectiveness analysis of plasma-based circulating tumor DNA (ctDNA) next-generation sequencing (NGS) to inform first-line treatment decisions. Value Health 23, S326 (2020).
    • 24. Ho R, Sebastião M, D'Innocenzo M, Nussbaum M. PPM4 Cost–effectiveness analysis of next generation sequencing panel of circulating tumor DNA in the diagnosis of patient with metastatic non-squamous non-small cell lung cancer in Brazil. Value Health 22, S333 (2019).
    • 25. Ferreira C, Custodio M, Montenegro G, Caetano R, Bonan L. P2. 01-32 Economic evaluation of diagnostic platforms for T790M detection in post EGFR-TKI NSCLC in Brazil. J. Thorac. Oncol. 13(10), S676–S677 (2018).
    • 26. Schluckebier L, Caetano R, Aran V, Ferreira C. Cost–effectiveness analysis comparing companion diagnostic tests for EGFR, ALK and ROS-1 versus next-generation sequence (NGS) in advanced adenocarcinoma lung cancer patients. J. Clin. Oncol. 35(Suppl. 15), 9068 (2017).
    • 27. Simarro J, Murria R, Pérez-Simó G et al. Development, implementation and assessment of molecular diagnostics by next generation sequencing in personalized treatment of cancer: experience of a public reference healthcare hospital. Cancers 11(8), 1196 (2019).
    • 28. Goldman J, Karlovich C, Sequist L et al. EGFR genotyping of matched urine, plasma, and tumor tissue in patients with non–small-cell lung cancer treated with rociletinib, an EGFR tyrosine kinase inhibitor. JCO Precis. Oncol. 2, 1–13 (2018).
    • 29. Tu H, Xu C, Tong-Li C et al. P1. 01-122 A Clinical utility study of plasma DNA next generation sequencing guided treatment of uncommon drivers in advanced non-small-cell lung cancers. J. Thorac. Oncol. 14(10), S410 (2019).
    • 30. Schwartzberg L, Gonzalez E, Isla D et al. Clinical concordance study of a 17-gene liquid biopsy NGS panel for non-small cell lung cancer (NSCLC). J. Clin. Oncol. 36(Suppl. 15), e21065 (2018).
    • 31. Zhou C, Wang M, Cheng Y et al. 1331P Detection of EGFR T790M in Asia-Pacific patients (pts) with EGFR mutation-positive advanced non-small cell lung cancer (NSCLC): circulating tumour (ct) DNA analysis across 3 platforms. Ann. Oncol. 28(Suppl. 5), V474–V475 (2017).
    • 32. Sacher A, Paweletz C, Dahlberg S et al. Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer. JAMA Oncol. 2(8), 1014–1022 (2016).
    • 33. Li B, Janku F, Jung B et al. Ultra-deep next-generation sequencing of plasma cell-free DNA in patients with advanced lung cancers: results from the Actionable Genome Consortium. Annals Oncol. 30(4), 597–603 (2019).
    • 34. Thompson J, Yee S, Troxel A 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. 22(23), 5772–5782 (2016).
    • 35. Leighl N, Page R, Raymond V 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. 25(15), 4691–4700 (2019). •• This prospective observational study in 282 US patients with NSCLC assessed the diagnostic accuracy and turnaround time for assays to detect EGFR exon 19 deletions using cfDNA compared with tissue biopsy (TBx).
    • 36. Remon J, Lacroix L, Jovelet C et al. Real-world utility of an amplicon-based next-generation sequencing liquid biopsy for broad molecular profiling in patients with advanced non–small-cell lung cancer. JCO Precis. Oncol. 3, 1–14 (2019).
    • 37. Papadopoulou E, Tsoulos N, Tsantikidi K et al. Clinical feasibility of NGS liquid biopsy analysis in NSCLC patients. PLoS One 14(12), e0226853 (2019).
    • 38. Minari R, Mazzaschi G, Bordi P et al. Detection of EGFR-activating and T790M mutations by liquid biopsy in patients with EGFR-mutated non-small-cell lung cancer who have progressed to first-and second-generation tyrosine kinase inhibitors: a multicenter real-life retrospective study. Clin. Lung Cancer 21(5), e464–e473 (2020).
    • 39. Spasic J, Krivokuca A, Boljevic I et al. P1. 03-07 Advantages and setbacks of EGFR mutation testing from liquid biopsy of advanced lung adenocarcinoma patients in Serbia. J. Thorac. Oncol. 14(10), S419–S420 (2019).
    • 40. Mayo-de-Las-Casas C, Jordana-Ariza N, Garzón-Ibañez M et al. Large scale, prospective screening of EGFR mutations in the blood of advanced NSCLC patients to guide treatment decisions. Ann. Oncol 28(9), 2248–2255 (2017).
    • 41. Mok T, Wu Y-L, Lee J et al. Detection and dynamic changes of EGFR mutations from circulating tumor DNA as a predictor of survival outcomes in NSCLC patients treated with first-line intercalated erlotinib and chemotherapy. Clin. Cancer Res. 21(14), 3196–3203 (2015).
    • 42. Yang Y, Song X, Guo W. P2. 01-130 Detection of actionable mutation status in advanced non-small cell lung cancer by next-generation sequencing of circulating tumor DNA. J. Thorac. Oncol. 13(10), S715 (2018).
    • 43. Zhang M, Gong Z, Li C et al. Assessment of EGFR mutations in plasma from advanced NSCLC patients by modified cell-free DNA extraction method and blocker PCR assays. J. Clin. Oncol. 35(Suppl. 15), e23115 (2017).
    • 44. Cao Z, Wu W, Zhang W et al. ALK and ROS1 rearrangement tested by ARMS-PCR in non-small-cell lung cancer patients via cytology specimens: the experience of Shanghai Pulmonary Hospital. Diagn. Cytopathol. 48(6), 524–530 (2020).
    • 45. Zhang H, He B, Cui J, Zhao M, Zhang Z. Comparison of circulating DNA from plasma and urine for EGFR mutations in NSCLC patients. Cancer Biomark. 23(3), 427–436 (2018).
    • 46. He J, Tan W, Ma J. Circulating tumor cells and DNA for real-time EGFR detection and monitoring of non-small-cell lung cancer. Future Oncol. 13(9), 787–797 (2017).
    • 47. Veldore V, Choughule A, Routhu T et al. Validation of liquid biopsy: plasma cell-free DNA testing in clinical management of advanced non-small cell lung cancer. Lung Cancer (Auckl.) 9,1–11 (2018).
    • 48. Wulandari L, Soegiarto G, Febriani A, Fatmawati F, Sahrun. Comparison of detection of epidermal growth factor receptor (EGFR) gene mutation in peripheral blood plasma (liquid biopsy) with cytological specimens in lung adenocarcinoma patients. Indian J. Surg. Oncol. 12(Suppl. 1), 65–71 (2020).
    • 49. Takahama T, Sakai K, Takeda M et al. Detection of the T790M mutation of EGFR in plasma of advanced non–small cell lung cancer patients with acquired resistance to tyrosine kinase inhibitors (West Japan Oncology Group 8014LTR study). Oncotarget 7(36), 58492 (2016).
    • 50. Itotani R, Matsumoto S, Udagawa H et al. 1557P A large scale prospective concordance study of oncogene driver detection between plasma-and tissue-based NGS analysis in advanced non-small cell lung cancer (NSCLC). Ann. Oncol. 30(Suppl. 5), v641–v642 (2019).
    • 51. Hur JY, Lee JS, Kim IA, Kim HJ, Kim WS, Lee KY. Extracellular vesicle-based EGFR genotyping in bronchoalveolar lavage fluid from treatment-naive non-small cell lung cancer patients. Transl. Lung Cancer Res. 8(6), 1051 (2019).
    • 52. Ma M, Shi C, Qian J, Teng J, Zhong H, Han B-H. Comparison of plasma and tissue samples in epidermal growth factor receptor mutation by ARMS in advanced non-small cell lung cancer. Gene 591(1), 58–64 (2016).
    • 53. Arnaud A. Costs and outcomes comparison of tissue and blood based biopsies for the purpose of biomarker testing. Value Health 19(3), A143–A144 (2016).
    • 54. Gancitano G, Gerolami GA, Ravasio R, Cortinovis L, Dionisi M. PCN134 Use of liquid biopsy in metastatic non-small cell lung cancer with EGFR mutation: a cost consequence analysis Value Health 21, S37 (2018). • This cost analysis demonstrated reduced costs associated with blood-based biomarker testing compared with tissue-based biopsy techniques.
    • 55. Ontario Health (Quality). Cell-free circulating tumour DNA blood testing to detect EGFR T790M mutation in people with advanced non-small cell lung cancer: a health technology assessment. Ont. Health Technol. Assess. Ser. 20(5), 1–176 (2020).