Purpose: Epidermal growth factor receptor (EGFR) mutations, especially deletional mutations in exon 19 (DEL) and L858R, predict gefitinib sensitivity in patients with non–small cell lung cancer (NSCLC). In this study, we validated EGFR mutation detection using high-resolution melting analysis (HRMA) and evaluated the associations between EGFR mutations and clinical outcomes in advanced NSCLC patients treated with gefitinib on a larger scale.

Experimental Design: The presence of DEL or L858R was evaluated using HRMA and paraffin-embedded tissues and/or cytologic slides from 212 patients. In 66 patients, the results were compared with direct sequencing data.

Results: HRMA using formalin-fixed tissues had a 92% sensitivity and a 100% specificity. The analysis was successfully completed in 207 patients, and DEL or L858R mutations were detected in 85 (41%) patients. The response rate (78% versus 8%), time-to-progression (median, 9.2 versus 1.6 months), and overall survival (median, 21.7 versus 8.7 months) were significantly better in patients with EGFR mutations (P < 0.001). Even among the 34 patients with stable diseases, the time-to-progression was significantly longer in patients with EGFR mutations. Patients with DEL (n = 49) tended to have better outcomes than those with L858R (n = 36); the response rates were 86% and 67%, respectively (P = 0.037), and the median time-to-progression was 10.5 and 7.4 months, respectively (P = 0.11).

Conclusions: HRMA is a precise method for detecting DEL and L858R mutations and is useful for predicting clinical outcomes in patients with advanced NSCLC treated with gefitinib.

Gefitinib (Iressa; AstraZeneca) is an orally active, selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor. Phase II studies have shown gefitinib antitumor activity in patients with advanced non–small cell lung cancer (NSCLC; refs. 1, 2). Several studies have shown that the response rate to gefitinib is higher in women, patients with adenocarcinoma, never smokers, and Japanese or East Asians (13); subsequently, somatic mutations in the kinase domain of EGFR were suggested to be a determinant of gefitinib sensitivity (4, 5). Since then, many retrospective studies have consistently revealed that EGFR mutations, mainly in-frame deletions including amino acids at codons 747 to 749 in exon 19 (DEL) and a missense mutation at codon 858 (L858R) in exon 21, are associated with tumor response, time-to-progression, and overall survival in NSCLC patients treated with gefitinib (68).

In our previous study, which clearly showed a correlation between EGFR mutations and gefitinib sensitivity in patients with recurrent NSCLC after surgical resection of the primary tumor (6), we used methanol-fixed, paraffin-embedded surgical specimens and did laser capture microdissection and direct sequencing, which we considered to be the most precise methods available for identifying mutations at that time. However, these methods are not useful in clinical practice for the treatment of advanced NSCLC for two reasons. First, the diagnostic samples of advanced NSCLC tumors, unlike surgical specimens, contain a small amount of tumor cells and are highly contaminated with normal cells. Second, laser capture microdissection and direct sequencing require special instruments and cost time and money. Recently, high-resolution melting analysis (HRMA) using the dye LCGreen I (Idaho Technology) was introduced as an easy, quick, and precise method for mutation screening (9), and we established a method for detecting DEL and L858R mutations using HRMA. Our cell line study revealed that DEL and L858R mutations could be detected using HRMA in the presence of 10% and 0.1% mutant cells, respectively (10). We also showed that the two major mutations could be identified by HRMA using DNA extracted from archived Papanicolaou-stained cytologic slides with 88% sensitivity and 100% specificity (10).

In this study, we validated EGFR mutation detection by HRMA using DNA extracted from archived paraffin-embedded tissues. We also did the HRMA in advanced NSCLC patients treated with gefitinib on a larger scale using archived tissues and/or cytologic slides.

Patients. Among 364 consecutive patients with NSCLC who began receiving gefitinib monotherapy (250 mg/d) at the National Cancer Center Hospital between July 2002 and December 2004, 212 patients were retrospectively analyzed using HRMA. One hundred fifty-two patients were excluded from the analysis because tumor samples were not available (n = 126) or their informed consent to the genetic analysis was not obtained (n = 26).

High-resolution melting analysis. On a protocol approved by the Institutional Review Board of the National Cancer Center Hospital, we did the following genetic analyses. Formalin-fixed, paraffin-embedded tissues and/or Papanicolaou-stained cytologic slides containing sufficient tumor cells (at least 1% of nucleated cells) were selected after microscopic examination by a pathologist (K.T.). The detailed analysis method has been described previously (10). Briefly, DNA was extracted from the tissues and/or cytologic slides using a QIAamp DNA Micro kit (Qiagen). PCR was done using dye LCGreen I and primers designed to amplify a region containing E746-I759 of EGFR [DEL-specific primer, AAAATTCCCGTCGCTATC (forward) and AAGCAGAAACTCACATCG (reverse)] or L858 of EGFR [L858R-specific primer, AGATCACAGATTTTGGGC (forward) and ATTCTTTCTCTTCCGCAC (reverse)] on a LightCycler (Roche Diagnostics). The PCR products were denatured at 95°C for 5 min and cooled to 40°C to form heteroduplexes. The LightCycler capillary was then transferred to an HR-1 (Idaho Technology), a HRMA instrument, and heated at a transition rate of 0.3°C per second. Data were acquired and analyzed using the accompanying software (Idaho Technology). After normalization and temperature adjustment steps, melting curve shapes from 78.5°C to 85.5°C were compared between samples and control samples. Human Genomic DNA (Roche Diagnostics) was used as a control sample with wild-type (WT) EGFR. Samples revealing skewed or left-shifted curves from those of control samples were judged to have mutations. All analyses were done in a blinded fashion.

Clinical validation of HRMA. Direct sequencing with and without laser capture microdissection had been done in 66 patients with recurrent NSCLC after surgery in the previous study (6). In these patients, HRMA was done using both formalin-fixed and methanol-fixed surgical specimens without laser capture microdissection, and the results were compared with the results of direct sequencing with laser capture microdissection, which we considered to be the gold standard method.

Radiologic evaluation. One board-certified radiologist (U.T.) who was unaware of the patients' mutational statuses reviewed the baseline, the first follow-up, and confirmatory imaging studies and classified the tumor responses into complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) using standard bidimensional measurements (11). In patients without measurable lesions, significant clinical benefit and disease progression were defined as clinical PR and clinical PD, respectively. Patients who died before the follow-up imaging studies were classified as PD. SD was subdivided into minor response (MR), long SD, and short SD. MR was defined as a ≥25% decrease in the sum of the products of the perpendicular diameters of all measurable lesions, and long SD meant that SD lasted for >6 months. Responders were defined as patients with CR, PR, or clinical PR.

Statistical analysis. The associations among EGFR mutations, patient characteristics, and tumor responses to gefitinib were assessed using a χ2 test. The differences in time-to-progression and overall survival according to the patient subgroups were compared using Kaplan-Meier curves and log-rank tests. The starting point of the time-to-progression and overall survival was the first administration of gefitinib. Multivariate analyses using logistic regression models and Cox proportional hazard models were done to assess the association between the clinical outcomes and the following factors: age (<70 versus ≥70 years), sex, smoking history (never smokers versus others), histology (adenocarcinoma versus others), performance status (0/1 versus 2/3), stage (recurrence after surgery versus III/IV), prior chemotherapy (yes versus no), and the mutational status of EGFR (mutant versus WT). All analyses were done using the SPSS statistical package (SPSS version 11.0 for Windows; SPSS, Inc.).

Patient characteristics. The patient characteristics are listed in Table 1. All the patients were East Asians: 210 Japanese, 1 Korean, and 1 Chinese. The median follow-up time for the survivors was 29.7 months (range, 10.7-49.8 months).

Table 1.

Patient characteristics (N = 212)

n (%)
Age (y)  
    Median (range) 62 (29-84) 
Sex  
    Women 92 (43) 
    Men 120 (57) 
Smoking history*  
    Never smokers 96 (45) 
    Former smokers 38 (18) 
    Current smokers 78 (37) 
Histology  
    Adenocarcinoma 193 (91) 
    Others 19 (9) 
Performance status  
    0 59 (28) 
    1 123 (58) 
    2 22 (10) 
    3 8 (4) 
Stage  
    III 42 (20) 
    IV 75 (35) 
    Recurrence after surgery 95 (45) 
Gefitinib therapy  
    First line 89 (42) 
    Second line 66 (31) 
    Third or more line 57 (27) 
n (%)
Age (y)  
    Median (range) 62 (29-84) 
Sex  
    Women 92 (43) 
    Men 120 (57) 
Smoking history*  
    Never smokers 96 (45) 
    Former smokers 38 (18) 
    Current smokers 78 (37) 
Histology  
    Adenocarcinoma 193 (91) 
    Others 19 (9) 
Performance status  
    0 59 (28) 
    1 123 (58) 
    2 22 (10) 
    3 8 (4) 
Stage  
    III 42 (20) 
    IV 75 (35) 
    Recurrence after surgery 95 (45) 
Gefitinib therapy  
    First line 89 (42) 
    Second line 66 (31) 
    Third or more line 57 (27) 
*

Never smokers were defined as patients who have never had a smoking habit and former smokers were defined as patients who had stopped smoking at least 1 y before diagnosis.

At the beginning of gefitinib therapy.

Clinical validation of HRMA. The clinical validation of the HRMA results using various samples is shown in Table 2. The sensitivity of HRMA using DNA extracted from formalin-fixed tissues was 92%, significantly higher than that of direct sequencing without laser capture microdissection but lower than that of HRMA using methanol-fixed tissues. The specificity and positive predictive values were 100% in all the analyses.

Table 2.

Clinical validation of HRMA and direct sequencing without laser capture microdissection

HRMA without LCM
Direct sequencing without LCM (6)
Formalin-fixed tissuesMethanol-fixed tissuesCytologic slides (10)
n 66 66 29 66 
Successfully analyzed, n (%) 63 (95) 66 (100) 28 (97) 66 (100) 
True positive 34 36 14 28 
True negative 26 29 12 29 
False positive 
False negative 
Sensitivity (%) 92 97 88 76 
Specificity (%) 100 100 100 100 
Positive predictive value (%) 100 100 100 100 
Negative predictive value (%) 90 97 86 76 
HRMA without LCM
Direct sequencing without LCM (6)
Formalin-fixed tissuesMethanol-fixed tissuesCytologic slides (10)
n 66 66 29 66 
Successfully analyzed, n (%) 63 (95) 66 (100) 28 (97) 66 (100) 
True positive 34 36 14 28 
True negative 26 29 12 29 
False positive 
False negative 
Sensitivity (%) 92 97 88 76 
Specificity (%) 100 100 100 100 
Positive predictive value (%) 100 100 100 100 
Negative predictive value (%) 90 97 86 76 

NOTE: The results of these analyses were compared with those of direct sequencing with LCM (used as the “gold standard” method). True positive is defined as the correct detection of deletional mutations in exon 19 or L858R.

Abbreviation: LCM, laser capture microdissection.

Mutational analysis. HRMA was completed in 207 patients. Five patients could not be successfully analyzed because of incomplete PCR. Of the 207 patients, 130 were analyzed using tissue samples (96 samples were obtained by thoracotomy, 17 by mediastinoscopic lymph node biopsy, 9 by thoracoscopic lung or pleural biopsy, 5 by resection or biopsy of distant metastases, and 3 by transbronchial lung biopsy), and 117 were analyzed using cytology samples (43 samples were obtained by bronchial brushing or washing, 40 from pleural effusion, 9 by transbronchial needle aspiration, 8 from pericardial effusion, 7 by needle aspiration of superficial lymph nodes, 6 by percutaneous needle aspiration of lung tumors, and 4 from sputum). In 40 patients who were analyzed using both tissue and cytology samples, 4 had inconsistent results; mutations were detected only in tissue samples and not in cytology samples (3 patients) or vice versa (1 patient). These four patients were judged to have mutations because false-negative results were more common than false-positive results in the validation of HRMA. Consequently, DEL and L858R mutations were detected in 49 (24%) and 36 (17%) patients, respectively, and these mutations were mutually exclusive. The other 122 (59%) patients were classified as having WT EGFR in this study, although some of them may have had minor mutations. As shown in Table 3, EGFR mutations were detected more frequently in women, never smokers, and patients with adenocarcinoma. Patient characteristics were not significantly different between patients with DEL mutations and those with an L858R mutation.

Table 3.

EGFR mutations among patient subgroups

nEGFR mutations
P
DELL858RTotal%
Total 207 49 36 85 41  
Sex       
    Women 89 31 17 48 54 0.001 
    Men 118 18 19 37 31  
Smoking history       
    Never smokers 93 30 19 49 53 0.002* 
    Former smokers 38 12 10 22 58  
    Current smokers 76 14 18  
Histology       
    Adenocarcinoma 189 48 35 83 44 0.007 
    Others 18 1 1 11  
nEGFR mutations
P
DELL858RTotal%
Total 207 49 36 85 41  
Sex       
    Women 89 31 17 48 54 0.001 
    Men 118 18 19 37 31  
Smoking history       
    Never smokers 93 30 19 49 53 0.002* 
    Former smokers 38 12 10 22 58  
    Current smokers 76 14 18  
Histology       
    Adenocarcinoma 189 48 35 83 44 0.007 
    Others 18 1 1 11  
*

Comparison between never smokers and others.

Pleomorphic carcinoma.

Adenosquamous carcinoma.

EGFR mutations and clinical outcomes. The association of the mutational status of EGFR and the response to gefitinib is shown in Table 4. The response rate was significantly higher in patients with EGFR mutations than in those with WT EGFR (78% versus 8%; P < 10−23). Among patients with EGFR mutations, those with DEL mutations had a higher response rate than those with an L858R mutation (86% versus 67%; P = 0.037). Tumor responses were classified as SD in 11 patients with EGFR mutations and in 23 patients with WT EGFR. Among the patients with SD, a MR and/or a long SD (>6 months) were observed more frequently (91% versus 26%; P = 0.0004) and the time-to-progression was significantly longer (median, 6.9 versus 4.4 months; P = 0.019) in the patients with EGFR mutations than in the patients with WT EGFR.

Table 4.

EGFR mutations and response to gefitinib

Responders
SD
PDResponse rate (%)P
CRPRMRLong SDShort SD
WT 10 17 89 10/122 (8)  
Mutant 64* 8 66/85 (78) <10−23 
    DEL 42 42/49 (86)  
    L858R 22 24/36 (67) 0.037 
Total 74 18 97 76/207 (37)  
Responders
SD
PDResponse rate (%)P
CRPRMRLong SDShort SD
WT 10 17 89 10/122 (8)  
Mutant 64* 8 66/85 (78) <10−23 
    DEL 42 42/49 (86)  
    L858R 22 24/36 (67) 0.037 
Total 74 18 97 76/207 (37)  
*

Including four clinical responders without measurable lesions.

Including a patient who had no measurable lesions at baseline.

As shown in Fig. 1, the time-to-progression (median, 9.2 versus 1.6 months; P < 0.0001) and overall survival (median, 21.7 versus 8.7 months; P = 0.0001) were significantly longer in patients with EGFR mutations than in those with WT EGFR. Patients with DEL mutations tended to have a longer time-to-progression (median, 10.5 versus 7.4 months; P = 0.11) and overall survival (median, 24.0 versus 20.4 months; P = 0.22) than those with an L858R mutation, although the difference did not reach statistical significance.

Fig. 1.

Kaplan-Meier plot of time-to-progression (A) and overall survival (B) for patients with or without EGFR mutations. Kaplan-Meier plot of time-to-progression (C) and overall survival (D) for patients with DEL or L858R mutations. TTP, time-to-progression; MST, median survival time.

Fig. 1.

Kaplan-Meier plot of time-to-progression (A) and overall survival (B) for patients with or without EGFR mutations. Kaplan-Meier plot of time-to-progression (C) and overall survival (D) for patients with DEL or L858R mutations. TTP, time-to-progression; MST, median survival time.

Close modal

Clinical outcomes among subgroups of patients are shown in Table 5. In the univariate analysis, sex, smoking history, and histology were significant predictive factors for gefitinib sensitivity.

Table 5.

Clinical outcomes among subgroups of patients

nResponse rate (%)PMedian TTP (mo)PMST (mo)P
Total 207 37  3.7  14.5  
Sex        
    Women 89 51 <0.001 5.6 0.17 18.3 0.15 
    Men 118 26  2.3  9.6  
Smoking history        
    Never smokers 93 51 <0.001* 6.2 0.073* 16.9 0.22* 
    Former smokers 38 47  5.2  14.5  
    Current smokers 76 14  2.2  9.1  
Histology        
    Adenocarcinoma 189 40 0.004 4.3 0.060 15.1 0.10 
    Others 18  1.6  4.9  
EGFR mutations        
    DEL/L858R 85 78 <0.001 9.2 <0.001 21.7 <0.001 
    WT 122  1.6  8.7  
nResponse rate (%)PMedian TTP (mo)PMST (mo)P
Total 207 37  3.7  14.5  
Sex        
    Women 89 51 <0.001 5.6 0.17 18.3 0.15 
    Men 118 26  2.3  9.6  
Smoking history        
    Never smokers 93 51 <0.001* 6.2 0.073* 16.9 0.22* 
    Former smokers 38 47  5.2  14.5  
    Current smokers 76 14  2.2  9.1  
Histology        
    Adenocarcinoma 189 40 0.004 4.3 0.060 15.1 0.10 
    Others 18  1.6  4.9  
EGFR mutations        
    DEL/L858R 85 78 <0.001 9.2 <0.001 21.7 <0.001 
    WT 122  1.6  8.7  

Abbreviations: TTP, time-to-progression; MST, median survival time.

*

Comparison between never smokers and others.

In the multivariate analyses, the mutational status of EGFR was an independent predictive factor of response [odds ratio, 38.9; 95% confidence interval (95% CI), 15.7-96.5; P < 0.001], time-to-progression (hazard ratio, 0.33; 95% CI, 0.24-0.45; P < 0.001), and overall survival (hazard ratio, 0.48; 95% CI, 0.34-0.67; P < 0.001). A poor performance status (2/3) was an independent predictor of a shorter time-to-progression (hazard ratio, 1.80; 95% CI, 1.19-2.72; P = 0.006) and overall survival (hazard ratio, 3.97; 95% CI, 2.56-6.16; P < 0.001), and a history of prior chemotherapy was another independent predictor of a shorter overall survival (hazard ratio, 1.59; 95% CI, 1.14-2.23; P = 0.006). However, other clinical characteristics, including sex, smoking history, and histology, were not independent predictive factors for any clinical outcomes.

In the current study, we showed the practicality of our new HRMA method for detecting two major EGFR mutations, DEL and L858R. The sensitivity and specificity of the analysis were 92% and 100%, respectively, when archived formalin-fixed, paraffin-embedded tissues were used without laser capture microdissection. Given the similar results that were obtained when Papanicolaou-stained cytologic slides were used (10), DEL and L858R mutations can likely be detected from such archived samples with about a 90% sensitivity and 100% specificity. Because the mutations were detected by HRMA even when only a small proportion (0.1% or 10%) of mutant cells existed (10), laser capture microdissection or other enrichment procedures are not needed in most cases. This is a major advantage of HRMA over direct sequencing because direct sequencing requires laser capture microdissection for accurate evaluation (6). However, there remained some risk of indeterminate or false-negative results because the DNA might have degenerated during sampling or the preservation of the archived samples. In fact, an analysis using methanol-fixed tissues, which are known to preserve DNA better than formalin-fixed tissues (12), was stable with no indeterminate and fewer false-negative results. Thus, an even higher sensitivity can be expected when fresh tumor samples are used. In any event, HRMA was successfully used to identify EGFR mutations and, more importantly, predict the clinical outcomes of gefitinib-treated patients with a high sensitivity and specificity.

Although the detection of EGFR mutations can provide patients with NSCLC and their physicians with critical information for optimal decision making, such tests are not common in clinical settings mainly because of the difficulty and impracticality of direct sequencing. Recently, highly sensitive nonsequencing methods to detect EGFR mutations in small tumor samples contaminated with normal cells have been reported (10, 1321). Among them, HRMA has the advantages of being able to identify the mutations with less labor, time, and expense; PCR and the melting analysis can be done in the same capillary tube within a few hours, and the running cost is only about 1 U.S. dollar per sample. HRMA is expected to be one of the most practical methods for detecting EGFR mutations in clinical settings.

We analyzed consecutive gefitinib-treated patients in a single institution on a larger scale than any other previous report. The mutational analysis by HRMA was successful in 207 patients and confirmed strong and independent associations between the two major EGFR mutations and clinical outcomes. Clinical predictors, such as sex, smoking history, and histology, added little predictive information to that provided by the mutational analysis. We believe that the mutational status of EGFR is the most important predictor of clinical outcomes in gefitinib-treated patients.

Among the patients without the two major mutations, 8% were responders. This result may be due to false-negative HRMA results, other EGFR mutations, or other determinants of gefitinib sensitivity. As for other EGFR mutations, the direct sequencing of exons 18 to 24 was done in four responders without DEL or L858R mutations, and one of them had G719C and S768I mutations. Although missense mutations at codon 719 of EGFR (G719C, G719S, or G719A) may be associated with gefitinib sensitivity, the predictive significance of these mutations is unclear because the number of reported patients is small (6). At present, we consider the accurate detection of the two major EGFR mutations to be sufficient for optimal decision making.

Recently, the EGFR copy number was reported to be another predictor of gefitinib sensitivity (6, 22, 23), and Cappuzzo et al. (22) suggested that this factor was a stronger predictor of overall survival than EGFR mutations. Our previous study also showed that the EGFR copy number evaluated by quantitative PCR was associated with response; however, an increased EGFR copy number was concentrated in patients with EGFR mutations and was not an independent predictor of response and overall survival (6). In the current study, we showed that EGFR mutations were associated with better outcomes even among patients with SD. The interpretation of this result is difficult because a long SD might be caused by intrinsic characteristics independent of treatment; however, this result suggested that EGFR mutations predicted not only “super responders” but also “non–super responders” who gained a clinical benefit. Contrary to these findings, Cappuzzo et al. (22) showed that EGFR mutations predicted only responders and were not associated with overall survival, whereas EGFR copy number was associated with both response and SD and was an independent predictor of overall survival. Although the reason of these discrepancies is unclear, we consider that if EGFR mutations are accurately identified, EGFR copy number adds little information for patient selection, at least in Japanese patients.

About the outcomes of patients with DEL or L858R mutations, our larger scale study produced results similar to those of some previous studies, which indicated that DEL mutations were associated with better outcomes after EGFR tyrosine kinase inhibitor treatment than an L858R mutation (2427). Further investigations are needed to clarify the difference in the biological characteristics of the two mutations. However, in the current study, the difference was small and even patients with an L858R mutation had favorable outcomes: the response rate was 67%, the median time-to-progression was 7.4 months, and the median survival time was 20.4 months. We now think that both DEL and L858R mutations should be treated equally in clinical decision-making.

In conclusion, the detection of DEL and L858R mutations using HRMA is accurate and practical. Using HRMA, we confirmed a strong association between the two major EGFR mutations and clinical outcomes in patients with advanced NSCLC treated with gefitinib.

Grant support: A program for the Promotion of Fundamental Studies in Health Sciences of the Pharmaceuticals and Medical Devices Agency; a Health and Labour Science Research grant from the Ministry of Health, Labor and Welfare, Japan; and a Grant-in-Aid for Young Scientists from the Ministry of Education, Culture, Sports, Science and Technology, Japan.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: This study was presented at the 42nd Annual Meeting of the American Society of Clinical Oncology, Atlanta, Georgia, June 2-6, 2006.

We thank Kiyoaki Nomoto, Karin Yokozawa, Chizu Kina, and Sachiko Miura for their technical support.

1
Fukuoka M, Yano S, Giaccone G, et al. A multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small cell lung cancer (The IDEAL 1 Trial).
J Clin Oncol
2003
;
21
:
2237
–46.
2
Kris MG, Natale RB, Herbst RS, et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial.
JAMA
2003
;
290
:
2149
–58.
3
Takano T, Ohe Y, Kusumoto M, et al. Risk factors for interstitial lung disease and predictive factors for tumor response in patients with advanced non-small cell lung cancer treated with gefitinib.
Lung Cancer
2004
;
45
:
93
–104.
4
Lynch TJ, Bell DW, Sordella R, 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
–39.
5
Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy.
Science
2004
;
304
:
1497
–500.
6
Takano T, Ohe Y, Sakamoto H, et al. Epidermal growth factor receptor gene mutations and increased copy numbers predict gefitinib sensitivity in patients with recurrent non-small-cell lung cancer.
J Clin Oncol
2005
;
23
:
6829
–37.
7
Mitsudomi T, Kosaka T, Endoh H, et al. Mutations of the epidermal growth factor receptor gene predict prolonged survival after gefitinib treatment in patients with non-small cell lung cancer with postoperative recurrence.
J Clin Oncol
2005
;
23
:
2513
–20.
8
Han SW, Kim TY, Hwang PG, et al. Predictive and prognostic impact of epidermal growth factor receptor mutation in non-small-cell lung cancer patients treated with gefitinib.
J Clin Oncol
2005
;
23
:
2493
–501.
9
Wittwer CT, Reed GH, Gundry CN, Vandersteen JG, Pryor RJ. High-resolution genotyping by amplicon melting analysis using LCGreen.
Clin Chem
2003
;
49
:
853
–60.
10
Nomoto K, Tsuta K, Takano T, et al. Detection of EGFR mutations in archived cytologic specimens of non-small cell lung cancer using high-resolution melting analysis.
Am J Clin Pathol
2006
;
126
:
1
–8.
11
Green S, Weiss GR. Southwest Oncology Group standard response criteria, endpoint definitions, and toxicity criteria.
Invest New Drugs
1992
;
10
:
239
–53.
12
Noguchi M, Furuya S, Takeuchi T, et al. Modification formalin and methanol fixation methods for molecular biological and morphological analyses.
Pathol Int
1997
;
47
:
685
–91.
13
Marchetti A, Martella C, Felicioni L, et al. EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment.
J Clin Oncol
2005
;
23
:
857
–65.
14
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
2005
;
65
:
7276
–82.
15
Pan Q, Pao W, Ladanyi M. Rapid polymerase chain reaction-based detection of epidermal growth factor receptor gene mutations in lung adenocarcinomas.
J Mol Diagn
2005
;
7
:
396
–403.
16
Yatabe Y, Hida T, Horio Y, et al. A rapid, sensitive assay to detect EGFR mutation in small biopsy specimens from lung cancer.
J Mol Diagn
2006
;
8
:
335
–41.
17
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
2006
;
12
:
43
–8.
18
Jänne PA, Borras AM, Kuang Y, et al. A rapid and sensitive enzymatic method for epidermal growth factor receptor mutation screening.
Clin Cancer Res
2006
;
12
:
751
–8.
19
Sasaki H, Endo K, Konishi A, et al. EGFR Mutation status in Japanese lung cancer patients: genotyping analysis using LightCycler.
Clin Cancer Res
2005
;
11
:
2924
–9.
20
Kimura H, Kasahara K, Kawaishi M, et al. Detection of epidermal growth factor receptor mutations in serum as a predictor of the response to gefitinib in patients with non-small-cell lung cancer.
Clin Cancer Res
2006
;
12
:
3915
–21.
21
Endo K, Konishi A, Sasaki H, et al. Epidermal growth factor receptor gene mutation in non-small cell lung cancer using highly sensitive and fast Taqman PCR assay.
Lung Cancer
2005
;
50
:
375
–84.
22
Cappuzzo F, Hirsch FR, Rossi E, et al. Epidermal growth factor receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer.
J Natl Cancer Inst
2005
;
97
:
643
–55.
23
Hirsch FR, Varella-Garcia M, McCoy J, et al. Increased epidermal growth factor receptor gene copy number detected by fluorescence in situ hybridization associates with increased sensitivity to gefitinib in patients with bronchioloalveolar carcinoma subtypes: a Southwest Oncology Group study.
J Clin Oncol
2005
;
23
:
6838
–45.
24
Riely GJ, Pao W, Pham DK, et al. Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib.
Clin Cancer Res
2006
;
12
:
839
–44.
25
Jackman DM, Yeap BY, Sequist LV, et al. Exon 19 deletion mutations of epidermal growth factor receptor are associated with prolonged survival in non-small cell lung cancer patients treated with gefitinib or erlotinib.
Clin Cancer Res
2006
;
12
:
3908
–14.
26
Paz-Ares L, Sanchez JM, García-Velasco A, et al. A prospective phase II trial of erlotinib in advanced non-small cell lung cancer (NSCLC) patients (p) with mutations in the tyrosine kinase (TK) domain of the epidermal growth factor receptor (EGFR) [abstract 7020].
Proc Am Soc Clin Oncol
2006
;
24
:
369s
.
27
Hirsch FR, Franklin WA, McCoy J, et al. Predicting clinical benefit from EGFR TKIs: not all EGFR mutations are equal [abstract 7072].
Proc Am Soc Clin Oncol
2006
;
24
:
382s
.