Open Access

A novel point-of-care system for high-speed real-time polymerase chain reaction testing for epidermal growth factor receptor mutations in bronchial lavage fluids after transbronchial biopsy in patients with non-small cell lung cancer

  • Authors:
    • Tomohiro Sakamoto
    • Masahiro Kodani
    • Miyako Takata
    • Hiroki Chikumi
    • Masaki Nakamoto
    • Shizuka Nishii-Ito
    • Yasuto Ueda
    • Hiroki Izumi
    • Haruhiko Makino
    • Hirokazu Touge
    • Kenichi Takeda
    • Akira Yamasaki
    • Masaaki Yanai
    • Natsumi Tanaka
    • Tadashi Igishi
    • Eiji Shimizu
  • View Affiliations

  • Published online on: February 4, 2015     https://doi.org/10.3892/ijo.2015.2875
  • Pages: 1473-1480
  • Copyright: © Sakamoto et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epidermal growth factor receptor (EGFR) gene mutation testing is essential for choosing appropriate treatment options in patients with advanced non-small cell lung cancer (NSCLC). However, a time delay occurs between histological diagnosis and molecular diagnosis in clinical situations. To minimize this delay, we developed a novel point-of-care test for EGFR mutations, based on a high-speed real-time polymerase chain reaction (PCR) system designated here as ultrarapid PCR combined with highly accurate bronchoscopic sampling. We investigated whether our system for detecting EGFR mutations was valid by comparing test results with those obtained using a commercialized EGFR mutation test. We obtained small amounts of bronchial lavage fluids after transbronchial biopsies (TBBs) were performed on enrolled patients (n=168) who underwent endobronchial ultrasonography using a guide sheath (EBUS-GS). EGFR mutation analysis was performed by ultrarapid PCR immediately after EBUS-GS-TBBs were obtained (on the same day). After pathological diagnoses of NSCLC, EGFR mutation status in formalin-fixed, paraffin- embedded samples was confirmed by the PCR-invader method, and the concordance rates between the PCR methods were compared. The total diagnostic yield of EBUS-GS-TBB was 91.0%. The positive concordance rates for detecting 19del and L858R with the ultrarapid PCR and PCR-invader methods were both 100%. Negative concordance rates were 97.2 and 98.1%, respectively. We also demonstrated a dramatic effect of early erlotinib administration, based on ultrarapid PCR results, for a 52-year-old woman suffering from respiratory failure due to severe intrapulmonary metastases with poor performance status. In conclusion, ultrarapid PCR combined with EBUS-GS-TBB enabled rapid and reliable point-of-care testing for EGFR mutations.

Introduction

Over the last decade, the discovery of epidermal growth factor receptor (EGFR) gene mutations and the development of tyrosine kinase inhibitors (TKIs) have dramatically changed the treatment strategies for patients with advanced non-small cell lung cancer (NSCLC) (15). Therefore, EGFR mutation testing is essential for optimal treatment selection for advanced NSCLC patients. Several methods for detecting EGFR mutations mainly in formalin-fixed, paraffin-embedded (FFPE) samples have already been validated and applied in practice (611). However, these methods adopt relatively complex polymerase chain reaction (PCR) technologies with pre-designed fluorogenic probes, are packaged by manufacturers, and are often available through outside reference laboratories at relatively high rates. In Japan, the use of EGFR-TKIs for chemo-naïve patients has been limited to those with EGFR mutations since 2011. Despite this regulation, the majority of community and university hospitals still depend on outside laboratories for EGFR mutation testing. Accordingly, there is a time delay between histological diagnosis and molecular diagnosis in clinical situations. In general, obtaining PCR-based EGFR test results from outside laboratories requires 7–14 days after tumor sampling. In cases where immediate treatment is critical, failure to provide appropriate molecular targeted therapy due to delayed molecular diagnostic test results may cause fatal outcomes. Therefore, a quicker, simpler, and less expensive point-of-care EGFR mutation testing system is needed.

In the field of infectious diseases, a more rapid real-time PCR system for detecting pathogens has been developed (12). Similarly, we have developed a new, simple, high-speed real-time PCR system (referred to as ultrarapid PCR) for the detection of the 2 most common EGFR mutations. This assay involves a pair of mutation-specific primers used in combination with a newly developed PCR machine that is equipped with a novel thermo-control mechanism that makes ultrarapid PCR cycling possible.

In-frame deletion in exon 19 (E746-A750del) and the point mutation replacing leucine with arginine at codon 858 of exon 21 (L858R) represent >90% of oncogenic EGFR mutations. Large clinical trials have been conducted to establish the efficacy of EGFR-TKIs in targeting the resulting mutated EGFR proteins (15). Therefore, we designed a deletion-specific primer targeting the exon 19 E746-A750del mutation and a point mutation-specific primer for the exon 21 L858R mutation. PCR conditions were optimized for amplifying templates harboring each mutation.

Endobronchial ultrasonography using a guide sheath (EBUS-GS) combined with a virtual bronchoscopic navigation system (VBN) is very useful approach for collecting samples from peripheral pulmonary lesions (1320). However, a major disadvantage of EBUS-GS is the low sample volume that can be obtained, leading to reduced sensitivity in molecular testing. Therefore, we performed this validation study to determine whether ultrarapid PCR can detect EGFR mutations with liquid bronchial lavage fluid (BLF) samples after EBUS-GS-transbronchial biopsies (EBUS-GS-TBBs) were taken.

Materials and methods

Patients and samples

A total of consecutive 168 patients who underwent EBUS-GS-TBB at the Tottori University Hospital (Yonago, Japan) from November 2012 to December 2013 were enrolled prospectively (Fig. 1). Eligible patients had undiagnosed pulmonary lesions suspected to be lung cancer on chest computed tomography (CT) findings. Samples were prepared by mixing BLFs obtained during EBUS-GS-TBB procedures with saline solutions mixed with EBUS-GS-brush biopsy samples after they were stamped on glass slides. DNA was extracted from patient fluid samples using the QIAamp Blood Mini kit (Qiagen, Tokyo, Japan) (Fig. 2).

Ethical approval was obtained from the Tottori University Hospital and informed consent was obtained from all patients involved prior to performing bronchoscopies.

VBN and EBUS-GS-TBB procedures

VBNs were performed following approval from physicians and expert bronchoscopists, based on CT findings. CT scan data from multi-detector chest CTs (64- or 128-row; slice width, 0.5 mm) were acquired from all patients before EBUS-GS-TBB. Individual CT data sets from VBN/EBUS-GS group were transferred to a workstation on which VBN software (Bf-NAVI; Cybernet Systems, Co., Ltd., Tokyo, Japan) automatically created VBN images within 15 min. VBN images could be moved multi-directionally on a monitor beside the video-bronchoscopic monitor. All patients were anaesthetized with midazolam and examined using a P260F video bronchoscope (4.0 mm outer diameter; Olympus Corp., Tokyo, Japan). The bronchoscope was introduced into the targeted bronchus with VBN support or the guidance of 2 expert bronchoscopists based on CT axial images. Peripheral target lesions were visualized using a 20 MHz radial-type EBUS probe (external diameter, 1.4 mm; UM-S20-17S; Olympus) with a GS (K-201; Olympus) through a working channel. Ultrasound images were processed in an ultrasound scanner (EU-ME-1 or EU-ME2; Olympus). Pathological samples were collected using forceps and brushes through the GS. Biopsy samples were immediately fixed in formalin. After biopsies were obtained, the target area was washed with 20 ml of saline.

Mutation-specific PCR using an ultrarapid PCR machine

EGFR exon 19 E746-A750 deletion type 1 (2235-2249del; 5′-GGAATTAAGAGAAGC-3′) and exon 21 L858R (2573T>G) were detected using a novel high-speed real-time PCR machine, namely a Hyper-PCR UR104MK IV (Trust Medical Co., Ltd., Kasai, Japan), with allele-specific primers and SpeedSTAR HS DNA Polymerase (Takara Bio, Inc., Shiga, Japan). The UR104MK IV PCR machine utilized a novel temperature control technology. In this system, the PCR mixture is enclosed in a small vessel on a thin, flexible plastic disk and sealed with adhesive film, and the disk is rotated rapidly onto 3 separated heat elements. Rapid PCR can be accomplished by controlling the speed of rotation and the temperature of the 3 heat elements. The UR104MK also has the capacity for real-time monitoring of PCR reactions with a fluorescent probe and post-PCR melt curve analysis. The typical time for amplification and detection when using this machine was <10 min.

Optimized reaction mixtures contained 1.6 μl of 10X Fast buffer I (Takara), 1.3 μl of 2.5 mM dNTPs, 0.4 μl of each allele-specific primer (10 μM), 0.2 μl of SpeedSTAR HS DNA Polymerase (5 U/μl; Takara), 1 μl of template DNA, 1.6 μl of 1:2,000 SYBR-Green, and 9.5 μl of ddH2O in a volume of 16 μl. Furthermore, dimethylsulfoxide was added to obtain a final concentration of 5%. PCR thermal cycling conditions were as follows. To amplify E746-A750del type 1, we used 1 cycle of 94°C for 1 min, followed by 35 cycles of 98°C for 1.3 sec, 55°C for 5 sec, and 72°C for 3 sec. To amplify DNA sequences harboring the L858R point mutation, we used 1 cycle of 94°C for 1 min, followed by 30 cycles of 98°C for 1.3 sec, 68°C for 8 sec and 68°C for 8 sec.

Sensitivity assay

To validate the sensitivity of the PCR system, sensitivity assays were performed using DNA mixtures extracted from the following cell lines: PC9 (2235-2249del), H1975 (2573T>G) and N417 (wild-type). The PC9 cell line was obtained from the RIKEN Cell Bank (Tsukuba, Japan). The H1975 cell line was obtained from the American Type Culture Collection (Rockville, MD, USA). The N417 cell line was provided by Dr A.F. Gazdar and Dr H. Oie (National Cancer Institute-Navy Medical Oncology Branch, Bethesda, MD, USA). These cell lines were mixed in different ratios. Specifically, the PC9 and N417 cell lines were mixed in ratios of 1:0, 0.5:0.5, 0.1:0.9, 0.01:0.99 and 0:1, respectively, while the H1975 and N417 were mixed in ratios of 1:0, 0.5:0.5, 0.1:0.9, 0.01:0.99 and 0:1, respectively. Analysis of EGFR mutations was performed as described above.

Comparison of ultrarapid PCR with the PCR-invader method

EGFR mutation analysis was performed with BLF samples from all 168 patients, regardless of pathological diagnosis, by ultrarapid PCR immediately after EBUS-GS-TBB. After pathological diagnosis of NSCLC, the associated EGFR mutation statuses in FFPE samples were evaluated by the PCR-invader method (BML, Inc., Tokyo, Japan), which is used in clinical practice at our hospital. To assess the performance of ultrarapid PCR, we evaluated the concordance rates and calculated kappa coefficients for both the ultrarapid PCR and PCR-invader methods.

Statistical analysis

Average target lesion diameters and diagnostic yields were calculated for the VBN/EBUS-GS and EBUS-GS groups, respectively, and analyzed using the Mann-Whitney U test and the Chi-squared test between these 2 groups. All P-values were 2-sided. A P-value of <0.05 indicated statistical significance. Concordance rates and Cohen’s kappa coefficients were determined between the ultrarapid PCR and PCR-invader methods. Cohen’s kappa coefficient was calculated as kappa = (Po-Pe)/(1-Pe), where Po is the observed concordance rate and Pe is the expected probability of chance agreement (21). A kappa of zero means that there is no agreement beyond chance, and a kappa of 1.00 means that there is perfect agreement. Values ranging from 0.81 to 1.00 indicate near perfect agreements (22). All data were statistically analyzed using IBM SPSS Statistics, ver. 22.

Results

Sensitivity

The E746-A750del mutation was detected in mixed cell populations containing decreasing percentages (100-1%) of the E746-A750del-positive cell line (PC9) and increasing percentages of the N417 cell line containing 2 copies of the wild-type EGFR gene. Similarly, the L858R mutation was detected in cell line mixtures containing 100-1% of an L858R mutation-positive cell line (H1975) and N417 cells (Fig. 3).

Characteristics of patients and patient samples

VBN was combined with EBUS-GS for 83 out of the 168 patients enrolled in the present study. The median and average diameters of the target lesions were 25 and 30.6 mm, respectively (range, 8–150 mm). In the VBN/EBUS-GS group, the median and average diameters of target lesions were 19 and 20.5 mm, respectively (range, 8–54 mm). In the EBUS-GS group, the median and average diameters of target lesions were 34.5 and 38.6 mm, respectively (range, 8–150 mm; Table I). As shown in Fig. 1, lung cancer was diagnosed histologically in 121 patients, but not in 47 patients, including 5 patients with benign diseases and 6 patients with metastatic tumors. Thirteen out of the 41 patients who were not diagnosed with NSCLC using EBUS-GS-TBB specimens were later diagnosed with NSCLC by re-examination or using another sampling method. Twenty-three patients were provided follow-up with imaging examinations at fixed intervals, and did not show enlargement of peripheral small lesions after EBUS-GS-TBB. After these 23 patients were excluded from the analysis, the total diagnostic yield obtained with EBUS-GS-TBB samples was 91.0% (132/145 cases). In the EBUS-GS-TBB group, the diagnostic yield was 94.6% (70/74 cases), while the diagnostic yield of the VBN/EBUS-GS-TBB was 87.3% (62/71 cases; Table I). Although target lesion diameters were significantly different (P<0.001; Mann-Whitney U test), diagnostic yields were similar in the 2 groups (P=0.18; Chi-squared test).

Table I

Comparison of target lesions diameters and diagnostic yields between VBN/EBUS-GS-TBB and EBUS-GS-TBB.

Table I

Comparison of target lesions diameters and diagnostic yields between VBN/EBUS-GS-TBB and EBUS-GS-TBB.

VBN/EBUS-GS-TBB (N=83)EBUS-GS-TBB (N=85)P-value
Diameter (mm)
 Median19.034.5
 Average20.538.6<0.001b
 Range8–548–150
Diagnostic yielda87.3% (62/71 cases)94.6% (70/74 cases)0.18c

a The diagnostic yield of EBUS-GS-TBB was 91.0% (132/145). The diagnostic yield was calculated for all patients, except for 23 patients that were provided follow-up with imaging examinations at fixed intervals and for whom enlargement of peripheral small lesions after EBUS-GS-TBB was not observed.

b Mann-Whitney U test;

c Chi-squared test.

The median age of the 121 lung cancer patients was 70 years (range, 37–97), and all of the patients were Japanese. NSCLC specimens were classified histologically as adeno-carcinoma in 89 patients (73.6%), squamous cell carcinoma in 22 patients (18.2%), large-cell neuroendocrine carcinoma (LCNEC) in 4 patients (3.3%), adenosquamous carcinoma in 2 patients (1.7%), large cell carcinoma in 2 patients (1.7%), small cell carcinoma in 1 patient (0.8%), and pleomorphic carcinoma in 1 patient (0.8%). The distribution of clinical stages at the time of diagnosis was as follows: 60 patients (49.6%) had stage I carcinoma, 13 patients (10.7%) had stage II, 15 patients (12.4%) had stage III, and 32 patients (26.4%) had stage IV. In 1 patient, the clinical stage was not classified (Table II).

Table II

Patient characteristics.

Table II

Patient characteristics.

CharacteristicsDiagnosed with lung cancer by EBUS-GS-TBBa (N=121)Not diagnosed with lung cancer by EBUS-GS-TBB (N=47)
Age (years)
 Median7071
 Range37–9765–87
Male gender, n (%)75 (64.1)29 (56.9)
Smoking status, n (%)
 Current smoker34 (28.1)7 (14.9)
 Former smoker48 (39.7)22 (46.8)
 Never smoker39 (32.2)18 (38.3)
Histologic type, n (%)
 Adenocarcinoma89 (73.6)
 Squamous cell carcinoma22 (18.2)
 Large cell carcinoma2 (1.7)
 Small cell carcinoma1 (0.8)
 Adenosquamous carcinoma2 (1.7)
 LCNEC4 (3.3)
 Pleomorphic1 (0.8)
Stage, n (%)
 I60 (49.6)
 II13 (10.7)
 III15 (12.4)
 IV32 (26.4)
 Not evaluated1 (0.8)

a A total of 121 patients were diagnosed bronchoscopically with lung cancer. Out of 121 cancers, 89 (73.6%) were adenocarcinoma and 32 (26.4%) were stage IV.

EGFR mutation detection by ultrarapid PCR

EGFR mutations in BLF samples were detected by ultrarapid PCR in 26 adenocarcinoma patients among the 120 NSCLC patients tested (21.7%), but were not detected in any of the 48 patients who were not diagnosed bronchoscopically with NSCLC. Eleven patients (42.3%) had an EGFR 19del mutation, and 15 patients (57.7%) had an L858R EGFR point mutation (Table III).

Table III

Comparison of ultrarapid PCR and PCR-invader test results found when detecting the 2 most common EGFR mutations in samples from 120 NSCLC patients.

Table III

Comparison of ultrarapid PCR and PCR-invader test results found when detecting the 2 most common EGFR mutations in samples from 120 NSCLC patients.

PCR-invader

Ultrarapid PCRMutation (+)Mutation (−)Total
19del
 Mutation (+)11011
 Mutation (−)3106109
Total14106120
L858R
 Mutation (+)15015
 Mutation (−)2103105
Total17103120
Comparison of the ultrarapid PCR and PCR-invader detection methods

EGFR mutations in FFPE tissues were detected in 36 adenocarcinoma patients among 120 NSCLC patients (30.0%) by the PCR-invader method (Table III). Two of these patients (5.6%) had an exon 18 G719A point mutation, 1 patient (2.8%) had a G719C point mutation and an exon 20 S768I point mutation, 1 patient (2.8%) had a G719S and a S768I mutation, 1 patient (2.8%) had a G719C mutation and an exon 21 L858R mutation, 8 patients (22.2%) had an E746-A750del type 1 mutation, 1 patient (2.8%) had an E746-A750del type 2 mutation, 6 patients (16.7%) had low-frequency mutations in exon 19, and 16 patients (44.4%) had an L858R mutation.

As shown in Table IV, positive concordance rates of 19del and L858R between ultrarapid PCR and PCR-invader were both 100%, while negative concordance rates were 97.2 and 98.1%, respectively. The kappa coefficients for detecting the 19del and L858R mutations between ultrarapid PCR and PCR-invader were 0.87 and 0.93, respectively. The average turnaround time for ultrarapid PCR was only 90 min, whereas that for the PCR-invader method by an outside laboratory was 9 days.

Table IV

Concordance rates and Cohen’s kappa coefficients between the ultrarapid PCR and PCR-invader methods.

Table IV

Concordance rates and Cohen’s kappa coefficients between the ultrarapid PCR and PCR-invader methods.

Concordance rate19del (%)L858 (%)
Positive100100
Negative97.298.1
Kappa coefficienta0.870.93

a A range from 0.81 to 1.00 corresponds to near perfect agreement.

Case report

A 52-year-old non-smoking female, without previous illness, was admitted to our hospital because of a dry cough and dyspnea at rest. Her performance status (PS) was 3 on admission. Her chest CT scan showed numerous bilateral diffuse granular lung shadows and a 20 mm-diameter nodular shadow on the lower right lobe (Fig. 4A). Whole body bone scintigraphy was performed later, revealing an abnormal accumulation in the fifth lumbar vertebra. Suspecting that she had advanced lung cancer, we immediately performed an EBUS-GS-TBB against the primary lesion of the lower right lobe. By 60 min after performing the EBUS-GS-TBB procedure, we obtained a positive result for the E746-A750del mutation by ultrarapid PCR. Because she had respiratory failure and a poor PS on admission, she was not eligible for cytotoxic chemotherapy. Therefore, it was deemed appropriate to initiate EGFR-TKI therapy as soon as possible. The following day, we started EGFR-TKI therapy (erlotinib 150 mg orally every 24 h), after obtaining a definitive pathological diagnosis of adenocarcinoma by an immunohistochemical method. Two weeks later, the diffuse and numerous granular shadows of bilateral lung field had mostly disappeared (Fig. 4B). Moreover, her respiratory failure and poor PS score were dramatically improved before PCR-invader results were provided.

Discussion

Bronchoscopy has been used to diagnose abnormal lung lesions for ~60 years. In recent years, the development of new diagnostic tools, such as EBUS, GS and VBN, has substantially improved diagnostic accuracy. Eberhardt et al (15) reported that the combination of EBUS and VBN improved the diagnostic yield in peripheral lung lesions, and VBN/EBUS is recommended for the diagnosis of lung peripheral lesions in guidelines of the European Society for Medical Oncology (23). Ishida et al (24) reported that the diagnostic yield of VBN combined EBUS-GS with small peripheral lesions (diameter <30 mm) was 80%. Similarly, we found high diagnostic yields in the present study despite the fact that most target lesions were small, especially in the VBN/EBUS-GS group. The appropriate decisions made regarding whether VBN should be used reinforced the diagnostic accuracy of EBUS-GS-TBBs for small peripheral lesions. Moreover, we usually collect at least 6 or more tissue samples. An advantage of EBUS-GS-TBB is that it is easy to obtain multiple biopsies through the fixed GS safely.

In this study, we validated ultrarapid PCR as a method for detecting the 2 most common EGFR mutations in liquid samples obtained by the EBUS-GS-TBB method. In many cases, even though these samples contain a very small amount of tumor cells, our method can detect the major EGFR mutations. Previous studies have shown similar results by molecular analysis of liquid samples collected by bronchoscopy. Yamaguchi et al (25) concluded that the analysis of EGFR, KRAS and TP53 mutations using curette lavage fluids obtained by bronchoscopy was possible. Furthermore, some reports have described the molecular analysis of lymph node samples obtained by EBUS guided trans-bronchial needle aspiration (2628) or trans-esophageal ultrasound scanning with fine needle aspiration (29,30). Likewise, Buttitta et al (31) reported that EGFR mutation analysis of bronchoalveolar lavage by next-generation sequencing was possible even in cases where conventional methods failed. Importantly, the accuracy of our method was remarkably high, although the BLF samples contained a small amount of tumor cells.

The greatest advantage of the ultrarapid PCR method is its speed. To the best of our knowledge, ultrarapid PCR is the fastest PCR system for detecting EGFR mutations at present. Ultrarapid PCR is completed within 10 min, while other methods take a few hours to detect mutations. This advantage can potentially have positive effects on treatment outcomes in cases requiring urgent treatment by early EGFR-TKI administration. Generally, the administration of cytotoxic agents for patients with poor PS is not recommended (32). However, some reports indicate that the use of EGFR-TKIs in patients with poor PS is effective and feasible because of their relatively mild toxicities (33). It is necessary to be careful in selecting therapeutic measures because TKIs are associated with an increased risk for developing interstitial pneumonitis in patients with poor PS scores (34). In addition, it will also be important to explore therapeutic opportunities for improving prognoses.

Most EGFR mutations are located in exon 18, 19, 20 and 21, with ~90% of these mutations occurring in exons 19 and 21 (35). In previous phase III trials with EGFR-TKIs, patients with hotspot mutations (exon 19 deletions or exon 21 L858R) were mostly recruited. The response rate of patients with these hotspot mutations was ~80% (2,5). In contrast, the response rate of patients with minor mutations, such as exon 18 point mutation G719X and exon 21 point mutation L861Q, was only 20% (36). Moreover, EGFR-TKIs had no proven survival benefit in patients with minor mutations (36). Therefore, we limited our search to these hotspot mutations in this study.

As demonstrated in our case report, ultrarapid PCR can deliver quick results in practical clinical situations. Patients with hotspot mutations in need of immediate care should receive EGFR-TKI treatment as soon as possible. Failures in providing appropriate molecular therapy due to molecular diagnosis delays should be avoided.

Despite the promising results obtained using ultrarapid PCR for detecting major EGFR mutations, a limitation of this method is that it can only detect known mutations. Detecting minor EGFR mutations in exon 18 and the T790M point mutation associated with drug resistance (exon 20) will require the development of additional probes. This current limitation reduces patients’ opportunities for rapid qualification for the third-generation EGFR-TKIs therapy, such as AZD9291 (37) by ultrarapid PCR alone. However, this problem may be solved by the development of additional primer sets for minor mutations in the near future.

In conclusion, it was demonstrated that ultrarapid PCR is an extremely quick and precise method for examining clinical liquid samples with a background of normal cells. The combination of ultrarapid PCR and EBUS-GS-TBB methods may enable point-of-care testing for NSCLC patient samples harboring EGFR mutations.

References

1 

Fukuoka M, Wu YL, Thongprasert S, et al: Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J Clin Oncol. 29:2866–2874. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Maemondo M, Inoue A, Kobayashi K, et al: Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 362:2380–2388. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Mitsudomi T, Morita S, Yatabe Y, et al: Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 11:121–128. 2010. View Article : Google Scholar

4 

Paez JG, Janne PA, Lee JC, et al: EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 304:1497–1500. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Zhou C, Wu YL, Chen G, 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. 12:735–742. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Asano H, Toyooka S, Tokumo M, et al: Detection of EGFR gene mutation in lung cancer by mutant-enriched polymerase chain reaction assay. Clin Cancer Res. 12:43–48. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Hoshi K, Takakura H, Mitani Y, et al: Rapid detection of epidermal growth factor receptor mutations in lung cancer by the SMart-Amplification Process. Clin Cancer Res. 13:4974–4983. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Hall JG, Eis PS, Law SM, et al: Sensitive detection of DNA polymorphisms by the serial invasive signal amplification reaction. Proc Natl Acad Sci USA. 97:8272–8277. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Nagai Y, Miyazawa H, Huqun, et al: Genetic heterogeneity of the epidermal growth factor receptor in non-small cell lung cancer cell lines revealed by a rapid and sensitive detection system, the peptide nucleic acid-locked nucleic acid PCR clamp. Cancer Res. 65:7276–7282. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Naoki K, Soejima K, Okamoto H, et al: The PCR-invader method (structure-specific 5′ nuclease-based method), a sensitive method for detecting EGFR gene mutations in lung cancer specimens; comparison with direct sequencing. Int J Clin Oncol. 16:335–344. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Sasaki H, Endo K, Konishi A, et al: EGFR mutation status in Japanese lung cancer patients: genotyping analysis using LightCycler. Clin Cancer Res. 11:2924–2929. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Fujimoto T, Konagaya M, Enomoto M, et al: Novel high-speed real-time PCR method (Hyper-PCR): results from its application to adenovirus diagnosis. Jpn J infect Dis. 63:31–35. 2010.PubMed/NCBI

13 

Asahina H, Yamazaki K, Onodera Y, Kikuchi E, Shinagawa N, Asano F and Nishimura M: Transbronchial biopsy using endobronchial ultrasonography with a guide sheath and virtual bronchoscopic navigation. Chest. 128:1761–1765. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Asano F, Matsuno Y, Shinagawa N, Yamazaki K, Suzuki T, Ishida T and Moriya H: A virtual bronchoscopic navigation system for pulmonary peripheral lesions. Chest. 130:559–566. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Eberhardt R, Anantham D, Ernst A, Feller-Kopman D and Herth F: Multimodality bronchoscopic diagnosis of peripheral lung lesions: a randomized controlled trial. Am J Respir Crit Care Med. 176:36–41. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Eberhardt R, Kahn N, Gompelmann D, Schumann M, Heussel CP and Herth FJ: LungPoint - a new approach to peripheral lesions. J Thorac Oncol. 5:1559–1563. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Fielding DI, Chia C, Nguyen P, Bashirzadeh F, Hundloe J, Brown IG and Steinke K: Prospective randomised trial of endobronchial ultrasound-guide sheath versus computed tomography-guided percutaneous core biopsies for peripheral lung lesions. Intern Med J. 42:894–900. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Gildea TR, Mazzone PJ, Karnak D, Meziane M and Mehta AC: Electromagnetic navigation diagnostic bronchoscopy: a prospective study. Am J Respir Crit Care Med. 174:982–989. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Seijo LM, de Torres JP, Lozano MD, Bastarrika G, Alcaide AB, Lacunza MM and Zulueta JJ: Diagnostic yield of electromagnetic navigation bronchoscopy is highly dependent on the presence of a Bronchus sign on CT imaging: results from a prospective study. Chest. 138:1316–1321. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Kurimoto N, Miyazawa T, Okimasa S, Maeda A, Oiwa H, Miyazu Y and Murayama M: Endobronchial ultrasonography using a guide sheath increases the ability to diagnose peripheral pulmonary lesions endoscopically. Chest. 126:959–965. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Kundel HL and Polansky M: Measurement of observer agreement. Radiology. 228:303–308. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Landis JR and Koch GG: The measurement of observer agreement for categorical data. Biometrics. 33:159–174. 1977. View Article : Google Scholar : PubMed/NCBI

23 

Vansteenkiste J, De Ruysscher D, Eberhardt WE, Lim E, Senan S, Felip E and Peters S; ESMO Guidelines Working Group. Early and locally advanced non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 24(Suppl 6): vi89–vi98. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Ishida T, Asano F, Yamazaki K, et al: Virtual bronchoscopic navigation combined with endobronchial ultrasound to diagnose small peripheral pulmonary lesions: a randamized trial. Thorax. 66:1072–1077. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Yamaguchi F, Kugawa S, Tateno H, Kokubu F and Fukuchi K: Analysis of EGFR, KRAS and P53 mutations in lung cancer using cells in the curette lavage fluid obtained by bronchoscopy. Lung Cancer. 78:201–206. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Jurado J, Saqi A, Maxfield R, et al: The efficacy of EBUS-guided transbronchial needle aspiration for molecular testing in lung adenocarcinoma. Ann Thorac Surg. 96:1196–1202. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Santis G, Angell R, Nickless G, Quinn A, Herbert A, Cane P, Spicer J, Breen R, McLean E and Tobal K: Screening for EGFR and KRAS mutations in endobronchial ultrasound derived trans-bronchial needle aspirates in non-small cell lung cancer using COLD-PCR. PLoS One. 6:e251912011. View Article : Google Scholar

28 

Tsai TH, Yang CY, Ho CC, et al: Multi-gene analyses from waste brushing specimens for patients with peripheral lung cancer receiving EBUS-assisted bronchoscopy. Lung Cancer. 82:420–425. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Lewandowska MA, Jozwicki W, Jochymski C and Kowalewski J: Application of PCR methods to evaluate EGFR, KRAS and BRAF mutations in a small number of tumor cells in cytological material from lung cancer patients. Oncol Rep. 30:1045–1052. 2013.PubMed/NCBI

30 

van Eijk R, Licht J, Schrumpf M, et al: Rapid KRAS, EGFR, BRAF and PIK3CA mutation analysis of fine needle aspirates from non-small-cell lung cancer using allele-specific qPCR. PLoS One. 6:e177912011. View Article : Google Scholar : PubMed/NCBI

31 

Buttitta F, Felicioni L, Del Grammastro M, et al: Effective assessment of egfr mutation status in bronchoalveolar lavage and pleural fluids by next-generation sequencing. Clin Cancer Res. 19:691–698. 2013. View Article : Google Scholar

32 

Pfister DG, Johnson DH, Azzoli CG, et al: American Society of Clinical Oncology treatment of unresectable non-small-cell lung cancer guideline: update 2003. J Clin Oncol. 22:330–353. 2004. View Article : Google Scholar

33 

Inoue A, Kobayashi K, Usui K, et al: First-line gefitinib for patients with advanced non-small-cell lung cancer harboring epidermal growth factor receptor mutations without indication for chemotherapy. J Clin Oncol. 27:1394–1400. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Kudoh S, Kato H, Nishiwaki Y, et al: Interstitial lung disease in Japanese patients with lung cancer: a cohort and nested case-control study. Am J Respir Crit Care Med. 177:1348–1357. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Mitsudomi T and Yatabe Y: Mutations of the epidermal growth factor receptor gene and related genes as determinants of epidermal growth factor receptor tyrosine kinase inhibitors sensitivity in lung cancer. Cancer Sci. 98:1817–1824. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Watanabe S, Minegishi Y, Yoshizawa H, et al: Effectiveness of gefitinib against non-small-cell lung cancer with uncommon EGFR mutations G719X and L861Q. J Thorac Oncol. 9:189–194. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Cross DA, Ashton SE, Ghiorghiu S, et al: AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Dicov. 4:1046–1061. 2014. View Article : Google Scholar

Related Articles

Journal Cover

April-2015
Volume 46 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sakamoto T, Kodani M, Takata M, Chikumi H, Nakamoto M, Nishii-Ito S, Ueda Y, Izumi H, Makino H, Touge H, Touge H, et al: A novel point-of-care system for high-speed real-time polymerase chain reaction testing for epidermal growth factor receptor mutations in bronchial lavage fluids after transbronchial biopsy in patients with non-small cell lung cancer. Int J Oncol 46: 1473-1480, 2015
APA
Sakamoto, T., Kodani, M., Takata, M., Chikumi, H., Nakamoto, M., Nishii-Ito, S. ... Shimizu, E. (2015). A novel point-of-care system for high-speed real-time polymerase chain reaction testing for epidermal growth factor receptor mutations in bronchial lavage fluids after transbronchial biopsy in patients with non-small cell lung cancer. International Journal of Oncology, 46, 1473-1480. https://doi.org/10.3892/ijo.2015.2875
MLA
Sakamoto, T., Kodani, M., Takata, M., Chikumi, H., Nakamoto, M., Nishii-Ito, S., Ueda, Y., Izumi, H., Makino, H., Touge, H., Takeda, K., Yamasaki, A., Yanai, M., Tanaka, N., Igishi, T., Shimizu, E."A novel point-of-care system for high-speed real-time polymerase chain reaction testing for epidermal growth factor receptor mutations in bronchial lavage fluids after transbronchial biopsy in patients with non-small cell lung cancer". International Journal of Oncology 46.4 (2015): 1473-1480.
Chicago
Sakamoto, T., Kodani, M., Takata, M., Chikumi, H., Nakamoto, M., Nishii-Ito, S., Ueda, Y., Izumi, H., Makino, H., Touge, H., Takeda, K., Yamasaki, A., Yanai, M., Tanaka, N., Igishi, T., Shimizu, E."A novel point-of-care system for high-speed real-time polymerase chain reaction testing for epidermal growth factor receptor mutations in bronchial lavage fluids after transbronchial biopsy in patients with non-small cell lung cancer". International Journal of Oncology 46, no. 4 (2015): 1473-1480. https://doi.org/10.3892/ijo.2015.2875